1
|
Lee S. Effect of Tunicamycin on Viability, Motility, Reactive Oxygen Species, Nitric Oxide, and Lipid Peroxidation in Boar Sperm. Animals (Basel) 2025; 15:1422. [PMID: 40427299 PMCID: PMC12108351 DOI: 10.3390/ani15101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Tunicamycin induces endoplasmic reticulum stress in mammalian cells. Our study aimed to investigate the effect of tunicamycin on the motility and viability of sperm, reactive oxygen species, nitric oxide, and lipid peroxidation in boar sperm. We treated 1.0, 2.0, 5.0, and 10 μM of tunicamycin in boar semen, and experimental treatments were performed. The viability (55.44%, 53.20, and 40.00%, p < 0.05) and motility (73.28%, 71.48%, and 54.48%, p < 0.05) of sperm at 2.0, 5.0, and 10.0 μM were decreased by tunicamycin, and the levels of reactive oxygen species and lipid peroxidation in tunicamycin-treated boar semen were increased (p < 0.05). However, the nitric oxide level was not changed by tunicamycin. Based on the results, we indicated that tunicamycin induces cell death by increasing oxidative stress in boar sperm, which may be the cause of decreased sperm viability and motility. Thus, we suggest that tunicamycin may induce cell death due to oxidative stress by reactive oxygen species and lipid peroxidation.
Collapse
Affiliation(s)
- Seunghyung Lee
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
2
|
Li M, Duan M, Yang Y, Li X, Li D, Gao W, Ji X, Bai J. Combination of brefeldin A and tunicamycin induces apoptosis in HepG2 cells through the endoplasmic reticulum stress-activated PERK-eIF2α-ATF4-CHOP signaling pathway. LIVER RESEARCH (BEIJING, CHINA) 2025; 9:49-56. [PMID: 40206437 PMCID: PMC11977284 DOI: 10.1016/j.livres.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/09/2024] [Accepted: 01/15/2025] [Indexed: 04/11/2025]
Abstract
Background and aims Hepatocellular carcinoma (HCC) is a malignant tumor with a high mortality rate, but there are still no effective treatments. The aim of this study was to investigate the anticancer potential of the combined use of brefeldin A (BFA) and tunicamycin (TM) in HepG2 cells, as well as the underlying mechanisms. Methods HepG2 cells were treated with different concentrations of BFA (0.1-2.5 mg/L) and TM (1-5 mg/L) for 24 h. DMSO (0.1 %, v/v) was used as a vehicle control. Cell viability and cell migration were measured using MTT assay and scratch wound assay, respectively. Apoptosis was detected using flow cytometry and acridine orange (AO) staining. The protein and mRNA levels of various factors involved in apoptosis (poly (ADP-ribose) polymerase-1 (PARP-1), caspase-12, caspase-3, and stearoyl-CoA desaturase 1) and endoplasmic reticulum (ER) stress (binding immunoglobulin protein (BiP), protein kinase R-like endoplasmic reticulum kinase (PERK), p-PERK, phosphorylation of eukaryotic translation initiation factor 2alpha (p-eIF2α), activating transcription factor (ATF) 4, and C/EBP homologous protein (CHOP)) were measured using Western blotting and qRT-PCR, respectively. Results Both BFA and TM alone significantly reduced the viability of HepG2 cells in a dose-dependent way. The co-incubation with TM (1 mg/L) further significantly reduced the viability of HepG2 cells treated with BFA (0.25 mg/L) alone (P < 0.05). BFA significantly increased the protein and mRNA levels of caspase-3 and PARP-1 (P < 0.05) compared to control and DMSO-treated cells, indicating that BFA induced apoptosis in HepG2 cells by increasing the expression of caspase-3 and PARP-1. The induction of apoptosis by BFA could be further significantly enhanced by co-incubation with TM. In addition, BFA significantly increased the mRNA levels of BiP, PERK and ATF4 (P < 0.05) compared to control and DMSO-treated cells. After co-incubation of BFA and TM, the protein levels of BiP, p-PERK, p-eIF2α and CHOP were significantly increased, indicating that TM could enhance BFA-induced ER stress in HepG2 cells through the PERK-eIF2α-ATF4-CHOP pathway. Conclusions BFA could induce apoptosis and ER stress, and TM could enhance the ability of BFA to induce apoptosis and ER stress in HepG2 cells through the PERK-eIF2ɑ-ATF4-CHOP pathway. The findings highlight the therapeutic potential of the combined use of BFA and TM in treating HCC.
Collapse
Affiliation(s)
- Minghong Li
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Occupational Disease Control, Ansteel Group General Hospital, Anshan, Liaoning, China
| | - Mengyi Duan
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ying Yang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xingdao Li
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Dan Li
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenting Gao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaotong Ji
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianying Bai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
- MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Taiyuan, Shanxi, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
3
|
Bharti R, Dey G, Khan D, Myers A, Huffman OG, Saygin C, Braley C, Richards E, Sangwan N, Willard B, Lathia JD, Fox PL, Lin F, Jha BK, Brown JM, Yu JS, Dwidar M, Joehlin-Price A, Vargas R, Michener CM, Longworth MS, Reizes O. Cell surface CD55 traffics to the nucleus leading to cisplatin resistance and stemness by inducing PRC2 and H3K27 trimethylation on chromatin in ovarian cancer. Mol Cancer 2024; 23:121. [PMID: 38853277 PMCID: PMC11163727 DOI: 10.1186/s12943-024-02028-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Platinum resistance is the primary cause of poor survival in ovarian cancer (OC) patients. Targeted therapies and biomarkers of chemoresistance are critical for the treatment of OC patients. Our previous studies identified cell surface CD55, a member of the complement regulatory proteins, drives chemoresistance and maintenance of cancer stem cells (CSCs). CSCs are implicated in tumor recurrence and metastasis in multiple cancers. METHODS Protein localization assays including immunofluorescence and subcellular fractionation were used to identify CD55 at the cell surface and nucleus of cancer cells. Protein half-life determinations were used to compare cell surface and nuclear CD55 stability. CD55 deletion mutants were generated and introduced into cancer cells to identify the nuclear trafficking code, cisplatin sensitivity, and stem cell frequency that were assayed using in vitro and in vivo models. Detection of CD55 binding proteins was analyzed by immunoprecipitation followed by mass spectrometry. Target pathways activated by CD55 were identified by RNA sequencing. RESULTS CD55 localizes to the nucleus of a subset of OC specimens, ascites from chemoresistant patients, and enriched in chemoresistant OC cells. We determined that nuclear CD55 is glycosylated and derived from the cell surface pool of CD55. Nuclear localization is driven by a trafficking code containing the serine/threonine (S/T) domain of CD55. Nuclear CD55 is necessary for cisplatin resistance, stemness, and cell proliferation in OC cells. CD55 S/T domain is necessary for nuclear entry and inducing chemoresistance to cisplatin in both in vitro and in vivo models. Deletion of the CD55 S/T domain is sufficient to sensitize chemoresistant OC cells to cisplatin. In the nucleus, CD55 binds and attenuates the epigenetic regulator and tumor suppressor ZMYND8 with a parallel increase in H3K27 trimethylation and members of the Polycomb Repressive Complex 2. CONCLUSIONS For the first time, we show CD55 localizes to the nucleus in OC and promotes CSC and chemoresistance. Our studies identify a therapeutic mechanism for treating platinum resistant ovarian cancer by blocking CD55 nuclear entry.
Collapse
Affiliation(s)
- Rashmi Bharti
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Goutam Dey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Debjit Khan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Alex Myers
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Olivia G Huffman
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Caner Saygin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
- Present address: Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Chad Braley
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Elliott Richards
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
- Reproductive, Endocrinology, and Infertility, Obstetrics and Gynecology Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Naseer Sangwan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
- Microbiome Analytics and Composition Core Facility, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Paul L Fox
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Feng Lin
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Babal Kant Jha
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Center for Immunotherapy & Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - J Mark Brown
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA
| | - Jennifer S Yu
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA
| | - Mohammed Dwidar
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Microbial Culturing and Engineering Facility, Cleveland Clinic, Cleveland, OH, USA
| | - Amy Joehlin-Price
- Anatomic Pathology, Pathology and Lab Medicine Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Roberto Vargas
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Gynecologic Oncology, Obstetrics and Gynecologic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Chad M Michener
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Gynecologic Oncology, Obstetrics and Gynecologic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Michelle S Longworth
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue Cleveland Clinic, Cleveland, OH, 44195, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
4
|
Schuurmans F, Wagemans KE, Adema GJ, Cornelissen LAM. Tumor glucose metabolism and the T cell glycocalyx: implication for T cell function. Front Immunol 2024; 15:1409238. [PMID: 38881904 PMCID: PMC11176483 DOI: 10.3389/fimmu.2024.1409238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
The T cell is an immune cell subset highly effective in eliminating cancer cells. Cancer immunotherapy empowers T cells and occupies a solid position in cancer treatment. The response rate, however, remains relatively low (<30%). The efficacy of immunotherapy is highly dependent on T cell infiltration into the tumor microenvironment (TME) and the ability of these infiltrated T cells to sustain their function within the TME. A better understanding of the inhibitory impact of the TME on T cells is crucial to improve cancer immunotherapy. Tumor cells are well described for their switch into aerobic glycolysis (Warburg effect), resulting in high glucose consumption and a metabolically distinct TME. Conversely, glycosylation, a predominant posttranslational modification of proteins, also relies on glucose molecules. Proper glycosylation of T cell receptors influences the immunological synapse between T cells and tumor cells, thereby affecting T cell effector functions including their cytolytic and cytostatic activities. This review delves into the complex interplay between tumor glucose metabolism and the glycocalyx of T cells, shedding light on how the TME can induce alterations in the T cell glycocalyx, which can subsequently influence the T cell's ability to target and eliminate tumor cells.
Collapse
Affiliation(s)
| | | | | | - Lenneke A. M. Cornelissen
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
5
|
Cheng Q, Hu X, Zhang X, Yang D, Zhao G, Sun L, Jiang M, Yang L, Cai J, Wang B, Zhang M, Han F, Li Y, Nie H. N-glycosylation at N57/100/110 affects CD44s localization, function and stability in hepatocellular carcinoma. Eur J Cell Biol 2023; 102:151360. [PMID: 37703748 DOI: 10.1016/j.ejcb.2023.151360] [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: 04/21/2023] [Revised: 08/23/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
The glycosylation levels of proteins in cancer cells are closely related to cancer invasion and migration. CD44 is a transmembrane glycoprotein that is significantly overexpressed in a variety of tumor cells and has been proven to promote the migration and motility of cancer cells, but the effect of its N-glycosylation modification on CD44 protein function in tumors is less studied. Here, we investigated the effect of six N-glycan chains (N25/57/100/110/120/255) on CD44s localization, function and stability in hepatocarcinoma cells. When the six sites were mutated, we found that CD44s lost its membrane localization in Huh7 and MHCC-97H cells. On this basis, we identified three glycosylation sites on CD44s (N57, N100 and N110) that played key roles in intracellular localization. When N57, N100 and N110 were mutated together, CD44 localized to the cytoplasm, while another three-site mutant (N25/N120/N255) was still anchored to the membrane. In addition, the ability of CD44-N57Q/N100Q/N110Q to promote the metastasis and invasion of Huh7 and 97H cells was weakened compared with that of CD44-N25Q/N120Q/N255Q. Furthermore, CD44-N57Q/N100Q/N110Q accumulated abnormally in the ER, and a high level of the ER stress (ERS) marker BiP was detected at the same time compared with wild-type CD44. When the lysosome inhibitor CQ was added, the content of mutant protein that triggered ERS significantly increased, which indicated that the degradation mode of CD44-N57Q/N100Q/N110Q after ERS was mainly through the lysosomal pathway (ERLAD). The results revealed that the N-glycosylation sites N57, N100 and N110 mutated on CD44s affected its function and degraded it by lysosomes after triggering ERS. These findings provide data for new studies on ER-related degradation, further promote the study of the glycan chain function of CD44 and furnish new ideas for the treatment of liver cancer metastasis.
Collapse
Affiliation(s)
- Qixiang Cheng
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xibo Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaoqing Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China; Hebei Normal University of Science & Technology, Qinhuangdao, Hebei 066004, China
| | - Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Guiping Zhao
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Liping Sun
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Meiyi Jiang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Bing Wang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Mengmeng Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Fang Han
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Yu Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Huan Nie
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
6
|
Lin S, Cao Y, Zhu K, Yang C, Zhu X, Zhang H, Zhang R. Identification of a Novel Prognostic Signature Based on N-Linked Glycosylation and Its Correlation with Immunotherapy Response in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2023; 10:1749-1765. [PMID: 37841372 PMCID: PMC10575065 DOI: 10.2147/jhc.s417407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Background The complex tumor microenvironment of hepatocellular carcinoma (HCC) has led to a low response to immune checkpoints inhibitors (ICIs) and a poor prognosis. PD-L1, as one of the indications for ICIs, is rich in glycosylation modifications, which result in untimely ICIs. Our study constructed a prognostic model based on N-linked glycosylation related genes for predicting the prognosis and the response to ICIs. Methods The list of N-linked glycosylation related genes is from the AmiGO2 database. The patients in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) cohorts were enrolled. The Cox regression was performed to develop a prognostic model and patients were divided into a low- and high-risk subgroups. The role of signature in HCC was well investigated by prognostic analysis, gene set enrichment analysis, and immune infiltration analysis. 21 recurrent HCC patients who received postoperative adjuvant ICIs were recruited to evaluate the relationship between immunotherapy response and the signature. In vitro studies were conducted to investigate the oncogenic effects of DDOST, STT3A and TMEM165 in HCC. Results 59 N-linked glycosylation related differentially expressed genes were screened from HCC and normal tissues in the TCGA cohort. The prognostic model was developed with DDOST, STT3A and TMEM165. The risk score could be an independent prognostic factor. Patients in the high-risk subgroup showed a worse prognosis than patients in the low-risk one. ssGSEA showed that patients in the low-risk subgroup tended to be in the immune-activated state, with higher levels of B cell and macrophage cell infiltrations and lower levels of regulatory T cell (Treg) infiltrations in both TCGC and GEO cohorts. Immunohistochemistry studies showed that DDOST, STT3A and TMEM165 are highly expressed in tumor tissues and patients with a high-risk score correlated with poor progression free survival and worse immunotherapeutic response. Furthermore, the proliferation of HCC cells was reduced after the knockdown of DDOST, as well as upon the knockdown of STT3A and TMEM165. Conclusion In this study, we establish that the risk model based on N-linked glycosylation related genes could efficiently predict the prognosis and tumor microenvironment immune state of HCC patients, and the risk score could serve as a novel indicator of immunotherapy.
Collapse
Affiliation(s)
- Shusheng Lin
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yi Cao
- Emergency Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ke Zhu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong TCRCure Biopharma Technology Co., Ltd, Guangzhou, People’s Republic of China
| | - Caini Yang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xiangping Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Honghua Zhang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Rui Zhang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
7
|
Cheng H, Wang S, Gao D, Yu K, Chen H, Huang Y, Li M, Zhang J, Guo K. Nucleotide sugar transporter SLC35A2 is involved in promoting hepatocellular carcinoma metastasis by regulating cellular glycosylation. Cell Oncol (Dordr) 2023; 46:283-297. [PMID: 36454514 DOI: 10.1007/s13402-022-00749-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
PURPOSE Recently, aberrant glycosylation has been recognized to be relate to malignant behaviors of cancer and outcomes of patients with various cancers. SLC35A2 plays an indispensable role on glycosylation as a nucleotide sugar transporter. However, effects of SLC35A2 on malignant behaviors of cancer cells and alteration of cancer cells surface glycosylation profiles are still not fully understood, particularly in hepatocellular carcinoma (HCC). Hence, from a glycosylation perspective, we investigated the effects of SLC35A2 on metastatic behaviors of HCC cells. METHODS SLC35A2 expression in clinical samples and HCC cells was examined by immunohistochemical staining or Western blot/quantitative PCR and was regulated by RNA interference or vectors-mediated transfection. Effects of SLC35A2 expression alteration on metastatic behaviors and membrane glycan profile of HCC cells were observed by using respectively invasion, migration, cell adhesion assay, in vivo lung metastatic nude mouse model and lectins microarray. Co-location among proteins in HCC cells was observed by fluorescence microscope and detected by an in vitro co-immunoprecipitation assay. RESULTS SLC35A2 was upregulated in HCC tissues, and is associated with poor prognosis of HCC patients. SLC35A2 expression alteration significantly affected the invasion, adhesion, metastasis and membrane glycan profile and led to the dysregulated expressions or glycosylation of cell adhesion-related molecules in HCC cells. Mechanistically, the maintenance of SLC35A2 activity is critical for the recruitment of the key galactosyltransferase B4GalT1, which is responsible for complex glycoconjugate and lactose biosynthesis, to Golgi apparatus in HCC cells. CONCLUSION SLC35A2 plays important roles in promoting HCC metastasis by regulating cellular glycosylation modification and inducing the cell adhesive ability of HCC cells.
Collapse
Affiliation(s)
- Hongxia Cheng
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Zhongshan Hospital, Building19, No. 180, Fenglin Road, 20032, Shanghai, People's Republic of China
| | - Sikai Wang
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Zhongshan Hospital, Building19, No. 180, Fenglin Road, 20032, Shanghai, People's Republic of China
| | - Dongmei Gao
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Zhongshan Hospital, Building19, No. 180, Fenglin Road, 20032, Shanghai, People's Republic of China
| | - Kangkang Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Huaping Chen
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yilan Huang
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Zhongshan Hospital, Building19, No. 180, Fenglin Road, 20032, Shanghai, People's Republic of China
| | - Miaomiao Li
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Zhongshan Hospital, Building19, No. 180, Fenglin Road, 20032, Shanghai, People's Republic of China
| | - Jubo Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Kun Guo
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Zhongshan Hospital, Building19, No. 180, Fenglin Road, 20032, Shanghai, People's Republic of China.
- Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
8
|
Mennerich D, Kubaichuk K, Raza GS, Fuhrmann DC, Herzig KH, Brüne B, Kietzmann T. ER-stress promotes VHL-independent degradation of hypoxia-inducible factors via FBXW1A/βTrCP. Redox Biol 2022; 50:102243. [PMID: 35074541 PMCID: PMC8792260 DOI: 10.1016/j.redox.2022.102243] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
Metabolic adaptation and signal integration in response to hypoxic conditions is mainly regulated by hypoxia-inducible factors (HIFs). At the same time, hypoxia induces ROS formation and activates the unfolded protein response (UPR), indicative of endoplasmic reticulum (ER) stress. However, whether ER stress would affect the hypoxia response remains ill-defined. Here we report that feeding mice a high fat diet causes ER stress and attenuates the response to hypoxia. Mechanistically, ER stress promotes HIF-1α and HIF-2α degradation independent of ROS, Ca2+, and the von Hippel-Lindau (VHL) pathway, involving GSK3β and the ubiquitin ligase FBXW1A/βTrCP. Thereby, we reveal a previously unknown function of the GSK3β/HIFα/βTrCP1 axis in ER homeostasis and demonstrate that inhibition of the HIF-1 and HIF-2 response and genetic deficiency of GSK3β affects proliferation, migration, and sensitizes cells for ER stress promoted apoptosis. Vice versa, we show that hypoxia affects the ER stress response mainly through the PERK-arm of the UPR. Overall, we discovered previously unrecognized links between the HIF pathway and the ER stress response and uncovered an essential survival pathway for cells under ER stress.
Collapse
Affiliation(s)
- Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland
| | - Kateryna Kubaichuk
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland
| | - Ghulam S Raza
- Research Unit of Biomedicine, and Biocenter Oulu, Oulu University Hospital and Medical Research Center, FI-90014, Oulu, Finland
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, D-60590, Frankfurt, Germany
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, and Biocenter Oulu, Oulu University Hospital and Medical Research Center, FI-90014, Oulu, Finland
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, D-60590, Frankfurt, Germany
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, FI-90014, Oulu, Finland.
| |
Collapse
|
9
|
Pibuel MA, Poodts D, Díaz M, Molinari YA, Franco PG, Hajos SE, Lompardía SL. Antitumor effect of 4MU on glioblastoma cells is mediated by senescence induction and CD44, RHAMM and p-ERK modulation. Cell Death Discov 2021; 7:280. [PMID: 34628469 PMCID: PMC8502173 DOI: 10.1038/s41420-021-00672-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/23/2021] [Indexed: 01/10/2023] Open
Abstract
The extracellular matrix plays a key role in cancer progression. Hyaluronan, the main glycosaminoglycan of the extracellular matrix, has been related to several tumor processes. Hyaluronan acts through the interaction with cell membrane receptors as CD44 and RHAMM and triggers signaling pathways as MEK/ERK. 4-methylumbelliferone (4MU), a well-known hyaluronan synthesis inhibitor, is a promising alternative for cancer therapy. 4MU is a coumarin derivative without adverse effects that has been studied in several tumors. However, little is known about its use in glioblastoma (GBM), the most malignant primary brain tumor in adults. Glioblastoma is characterized by fast growth, migration and tissue invasiveness, and a poor median survival of the patients after treatment. Several reports linked glioblastoma progression with HA levels and even with CD44 and RHAMM expression, as well as MEK/ERK activation. Previously, we showed on a murine GBM cell line that HA enhances GBM migration, while 4MU markedly inhibits it. In this work we showed for the first time, that 4MU decreases cell migration and induces senescence in U251 and LN229 human GBM cell lines. Furthermore, we observed that HA promotes GBM cell migration on both cell lines and that such effects depend on CD44 and RHAMM, as well as MEK/ERK signaling pathway. Interestingly, we observed that the exogenous HA failed to counteract the effects of 4MU, indicating that 4MU effects are independent of HA synthesis inhibition. We found that 4MU decreases total CD44 and RHAMM membrane expression, which could explain the effect of 4MU on cell migration. Furthermore, we observed that 4MU increases the levels of RHAMM inside the cell while decreases the nucleus/cytoplasm relation of p-ERK, associated with 4MU effects on cell proliferation and senescence induction. Overall, 4MU should be considered as a promising therapeutic alternative to improve the outcome of patients with GBM.
Collapse
Grants
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°0289 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- PIP N°053 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- UBACYT 20020170100454BA Universidad de Buenos Aires (University of Buenos Aires)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
- PICT-2017- 2971 Ministry of Science, Technology and Productive Innovation, Argentina | Agencia Nacional de Promoción Científica y Tecnológica (National Agency for Science and Technology, Argentina)
Collapse
Affiliation(s)
- Matías Arturo Pibuel
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina.
| | - Daniela Poodts
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Yamila Azul Molinari
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET; Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Paula Gabriela Franco
- Instituto de Química y Fisicoquímica Biológicas (IQUIFIB)-CONICET; Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvia Elvira Hajos
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvina Laura Lompardía
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)- CONICET; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Capital Federal, Argentina
| |
Collapse
|
10
|
Huang T, Tian W, Zhou Q, Li J, Jiang Z, Chen J, Ge C, Tian H. Upregulation of Rpn10 promotes tumor progression via activation of the NF-κB pathway in clear cell renal cell carcinoma. Acta Biochim Biophys Sin (Shanghai) 2021; 53:988-996. [PMID: 34133712 DOI: 10.1093/abbs/gmab078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Indexed: 01/08/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a central role in regulating protein homeostasis in tumor progression. The proteasome subunit Rpn10 is associated with the progression of several tumor types. However, little is known regarding the role of Rpn10 in clear cell renal cell carcinoma (ccRCC). In this study, we found that overexpression of Rpn10 increased ccRCC cell proliferation, migration, and invasion. Silencing Rpn10 expression resulted in decreased cell proli-feration, migration, and invasion in ccRCC cells. Knockdown of Rpn10 inhibits tumor growth and cell proliferation in vivo. Furthermore, we demonstrated that Rpn10 increased cell proliferation, migration, and invasion via regulation of the nuclear factor kappa B (NF-κB) pathway. Rpn10 directly promoted inhibitor of nuclear factor-kappa B alpha (IκBα) degradation through the UPS. Moreover, we observed that upregulation of Rpn10 or downregulation of IκBα in ccRCC was associated with poor prognosis. We found that the combination of these two parameters was a more powerful predictor of poor prognosis than either parameter alone. Collectively, these findings provide evidence that Rpn10 promotes the progression of ccRCC by regulation of the NF-κB pathways and is a prognostic indicator for patients with ccRCC.
Collapse
Affiliation(s)
- Tingting Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Wei Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Qingqing Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jiajun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Zhiyuan Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jinsi Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
11
|
Li B, Zhou M, Wang J, Xu H, Yang M. Suppressing ERK Pathway Impairs Glycochenodeoxycholate-Mediated Survival and Drug-Resistance in Hepatocellular Carcinoma Cells. Front Oncol 2021; 11:663944. [PMID: 34327135 PMCID: PMC8313996 DOI: 10.3389/fonc.2021.663944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/22/2021] [Indexed: 12/02/2022] Open
Abstract
Glycochenodeoxycholate (GCDA), a toxic component in bile salts, is involved in carcinogenesis of gastrointestinal tumors. The objective of this research was to study the function of ERK1/2 in the GCDA-mediated survival and drug-resistance in hepatocellular carcinoma cells (HCCs). Firstly, extracellular signal-regulated kinase 1/2 (ERK1/2) was detected extensively expressed in liver cancer cells, and silencing ERK1/2 by RNA interference could suppress GCDA-stimulated survival and promote apoptosis. Furthermore, phosphorylation of endogenous ERK1/2 could be potently stimulated by GCDA in combination with enhanced chemoresistance in QGY-7703 hepatocellular carcinoma cells. The GCDA-mediated proliferation and chemoresistance could be impaired by PD98059, which acted as an inhibitor to block the phosphorylation of ERK1/2. Mechanistically, PD98059 was able to potently suppress GCDA-stimulated nuclear aggregation of ERK1/2 and p-ERK1/2, upregulate pro-survival protein Mcl-1 and downregulate pro-apoptotic protein Bim. The results of this study indicated that disruption of ERK1/2 by blocking phosphorylation or nuclear translocation may put forward new methods for solving the problem of GCDA-related proliferation and drug-resistance in liver cancer treatment.
Collapse
Affiliation(s)
- Bingxin Li
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| | - Maojun Zhou
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, National Center for Geriatrics Clinical Research, State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Jue Wang
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongjuan Xu
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| | - Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, National Health Commission (NHC) Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Choi J, Jo M, Lee E, Lee DY, Choi D. Dienogest regulates apoptosis, proliferation, and invasiveness of endometriotic cyst stromal cells via endoplasmic reticulum stress induction. Mol Hum Reprod 2021; 26:30-39. [PMID: 31814016 DOI: 10.1093/molehr/gaz064] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/25/2019] [Indexed: 02/06/2023] Open
Abstract
Dienogest, a specific progesterone receptor agonist, is used in the treatment of endometriosis. However, it is still unclear as to the mechanisms of therapeutic effects on endometriosis. Our recent study showed that endometriosis may be the result of aberrant endoplasmic reticulum (ER) stress induction due to progesterone resistance. This finding suggests that the regulation of ER stress induction may play a key role in treatment of endometriosis. Therefore, the anti-endometriotic effects of dienogest may be mediated by regulation of ER stress. To test this hypothesis, we elucidate whether dienogest affects endometriotic stromal cell apoptosis, proliferation and invasiveness by modulating ER stress-induced CCAAT/enhancer-binding protein homologous protein (CHOP) expression. Specifically, PRKR-like ER kinase (PERK)/eukaryotic initiation factor 2α (eIF2α)/activating transcription factor 4 (ATF4), inositol-requiring kinase 1 (IRE1)/TNF receptor-associated factor 2 (TRAF2)/apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) signaling, and downstream CHOP were evaluated to determine the involved ER stress-mediated regulation mechanism of CHOP expression. Our results show that progesterone treatment did not have any significant effects on ER stress, apoptosis, proliferation, and invasion in estrogen-treated endometriotic cyst stromal cells (ECSCs). However, dienogest treatment upregulated the induction of ER stress. It also led to increased apoptosis, and decreased proliferation and invasiveness. These dienogest-induced changes in apoptosis, proliferation and invasiveness were reversed by the ER stress inhibitor salubrinal. Furthermore, dienogest-induced ER stress increased CHOP expression through activation of both PERK/elf2α/ATF4 and IRE1/TRAF2/ASK1/JNK signaling. This upregulation was blocked by transfection with PERK and IRE1 siRNA, which decreased apoptosis and increased the proliferation and invasiveness of dienogest-treated ECSCs. Taken together, our findings indicate that dienogest enhances ER stress induction in endometriotic stromal cells, which affects apoptosis, proliferation and invasiveness via CHOP upregulation.
Collapse
Affiliation(s)
- JongYeob Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul 06351, Korea
| | - MinWha Jo
- Center for Clinical Research, Samsung Biomedical Research Institute, 50 Irwon-dong, Gangnam-gu, Seoul 06351, Korea
| | - EunYoung Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul 06351, Korea
| | - Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul 06351, Korea
| | - DooSeok Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul 06351, Korea
| |
Collapse
|
13
|
Sun D, Zhu Y, Zhu J, Tao J, Wei X, Wo Y, Hou H. Primary resistance to first-generation EGFR-TKIs induced by MDM2 amplification in NSCLC. Mol Med 2020; 26:66. [PMID: 32611363 PMCID: PMC7329552 DOI: 10.1186/s10020-020-00193-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction Targeted therapy for NSCLC is rapidly evolving. EGFR-TKIs benefit NSCLC patients with sensitive EGFR mutations and significantly prolong survival. However, 20–30% of patients demonstrate primary resistance to EGFR-TKIs, which leads to the failure of EGFR-TKI treatment. The mechanisms of primary resistance to EGFR-TKIs require further study. Methods Targeted sequencing was used for the detection of genomic alterations among patients in our center. Regular cell culture and transfection with plasmids were used to establish NSCLC cell lines over-expressing MDM2 and vector control. We used the MTT assays to calculate the inhibition rate after exposure to erlotinib. Available datasets were used to determine the role of MDM2 in the prognosis of NSCLC. Results Four patients harboring concurrent sensitive EGFR mutations and MDM2 amplifications demonstrated insensitivity to EGFR-TKIs in our center. In vitro experiments suggested that MDM2 amplification induces primary resistance to erlotinib. Over-expressed MDM2 elevated the IC50 value of erlotinib in HCC2279 line and reduced the inhibition rate. In addition, MDM2 amplification predicted a poor prognosis in NSCLC patients and was associated with a short PFS in those treated with EGFR-TKIs. The ERBB2 pathway was identified as a potential pathway activated by MDM2 amplification could be the focus of further research. Conclusion MDM2 amplification induces the primary resistance to EGFR-TKIs and predicts poor prognosis in NSCLC patients. MDM2 may serve as a novel biomarker and treatment target for NSCLC. Further studies are needed to confirm the mechanism by which amplified MDM2 leads to primary resistance to EGFR-TKIs.
Collapse
Affiliation(s)
- Dantong Sun
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Yan Zhu
- Department of Medical Oncology, The Municipal Hospital of Qingdao, Qingdao, 266000, Shandong, China
| | - Jingjuan Zhu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Junyan Tao
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Xiaojuan Wei
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Yang Wo
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Helei Hou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong, China.
| |
Collapse
|
14
|
Wang Y, Zhang L, He Z, Deng J, Zhang Z, Liu L, Ye W, Liu S. Tunicamycin induces ER stress and inhibits tumorigenesis of head and neck cancer cells by inhibiting N-glycosylation. Am J Transl Res 2020; 12:541-550. [PMID: 32194902 PMCID: PMC7061826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/06/2020] [Indexed: 06/10/2023]
Abstract
Glycosylation plays an important role in the genesis of various cancers. The inhibition of glycosylation disturbs the protein folding machinery, causing the accumulation of unfolded proteins in the cell endoplasmic reticulum (ER) and inducing ER stress. Tunicamycin (TM) is an inhibitor of glycosylation that has shown marked antitumor activity. In this study, we investigated the effect of TM on the tumorigenesis of head and neck cancer cells. The effects of TM on cell proliferation, colony formation and tumorsphere formation in vitro and tumorigenicity in vivo were investigated in head and neck cancer cells. ER stress was determined by the evaluation of PERK, PDI, IRE1-α, BIP, Ero1-Lα and calnexin expression using western blotting and immunofluorescence. We found that TM inhibited colony formation and tumorsphere formation of head and neck cancer cells in vitro and suppressed tumor growth in vivo. After incubation with TM, the expression of the cancer stem cell markers CD44 and Bmi-1 was reduced, and the expression of the ER stress markers BIP, Ero1-Lα and calnexin was elevated. Moreover, the EGFR signaling pathway was inhibited, and nonglycosylated EGFR degradation was accelerated with TM treatment. Our results suggest that inhibition of glycosylation by TM may be a novel treatment strategy for use with HNSCC patients.
Collapse
Affiliation(s)
- Yang Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| | - Ling Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| | - Zhiyan He
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| | - Jiong Deng
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| | - Liu Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| | - Weimin Ye
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| | - Shuli Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai, China
| |
Collapse
|
15
|
Yamahana H, Takino T, Endo Y, Yamada H, Suzuki T, Uto Y. A novel celecoxib analog UTX-121 inhibits HT1080 cell invasion by modulating membrane-type 1 matrix metalloproteinase. Biochem Biophys Res Commun 2020; 521:137-144. [PMID: 31629465 DOI: 10.1016/j.bbrc.2019.10.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/10/2019] [Indexed: 11/12/2022]
|
16
|
Zhou Q, Huang T, Jiang Z, Ge C, Chen X, Zhang L, Zhao F, Zhu M, Chen T, Cui Y, Li H, Yao M, Li J, Tian H. Upregulation of SNX5 predicts poor prognosis and promotes hepatocellular carcinoma progression by modulating the EGFR-ERK1/2 signaling pathway. Oncogene 2019; 39:2140-2155. [DOI: 10.1038/s41388-019-1131-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
|
17
|
Song Y, Ye M, Zhou J, Wang Z, Zhu X. Targeting E-cadherin expression with small molecules for digestive cancer treatment. Am J Transl Res 2019; 11:3932-3944. [PMID: 31396310 PMCID: PMC6684918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/28/2019] [Indexed: 06/10/2023]
Abstract
Digestive system cancers, mainly including gastric cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal cancer, are major public health problems and lead to serious cancer-related deaths worldwide. Clinically, treatment strategies of these cancers include surgery, chemotherapy, and immunotherapy. Although successful resection and chemotherapeutic drugs have improved the treatment level, the survival rate of patients with advanced digestive system cancers remains still low primarily due to tumor metastasis. E-cadherin, the prototypical member of the type-1 classical cadherins, has been characrized as an important molecule in epithelial-mesenchymal transition (EMT) process. Loss of E-cadherin is able to induce EMT process, which is associated with cancer stem cells and drug resistance in human cancer. Therefore, restoring E-cadherin could be a useful strategy for reversal of EMT and overcoming drug resistance. In this review, we describe pharmacological small molecules targeting E-cadherin expression for the treatment of digestive system cancers, which have emerged in the recent 5 years. We hope these compounds could be potentially used for treating cancer in the near future.
Collapse
Affiliation(s)
- Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Junhan Zhou
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Zhiwei Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
- Center of Scientific Research, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| |
Collapse
|
18
|
Mori M, Mori T, Yamamoto A, Takagi S, Ueda M. Proliferation of poorly differentiated endometrial cancer cells through autocrine activation of FGF receptor and HES1 expression. Hum Cell 2019; 32:367-378. [PMID: 30963412 DOI: 10.1007/s13577-019-00249-1] [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: 06/04/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Patients with poorly differentiated endometrial cancer show poor prognosis, and effective molecular target-based therapies are needed. Endometrial cancer cells proliferate depending on the activation of HES1 (hairy and enhancer of split-1), which is induced by several pathways, such as the Notch and fibroblast growth factor receptor (FGFR) signaling pathways. In addition, aberrant, ligand-free activation of the FGFR signaling pathway resulting from mutations in FGFR2 was also reported in endometrial cancer. However, a clinical trial showed that there was no difference in the effectiveness of FGFR inhibitors between patients with and without the FGFR2 mutation, suggesting a presence of another signaling pathway for the FGFR activation. Here, we investigated the signaling pathway regulating the expression of HES1 and proliferation of poorly and well-differentiated endometrial cancer cell lines Ishikawa and HEC-50B, respectively. Whereas Ishikawa cells proliferated and expressed HES1 in a Notch signaling-dependent manner, Notch signaling was not involved in HES1 and proliferation of HEC-50B cells. The FGFR inhibitor, NVP-BGJ398, decreased HES1 expression and proliferation of HEC-50B cells; however, HEC50B cells had no mutations in the FGFR2 gene. Instead, HEC-50B cells highly expressed ligands for FGFR2, suggesting that FGFR2 is activated by an autocrine manner, not by ligand-free activation. This autocrine pathway activated Akt downstream of FGFR for cell proliferation. Our findings suggest the usefulness of HES1 as a marker for the proliferation signaling and that FGFR inhibitor may be effective for poorly differentiated endometrial cancers that harbor wild-type FGFR.
Collapse
Affiliation(s)
- Michihiro Mori
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan. .,Kake Institute of Cytopathology, Okayama, Japan.
| | - Toshinori Mori
- Department of Clinical Laboratory, Mihara Medical Associations Hospital, Hiroshima, Japan.,Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Aina Yamamoto
- Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Shoji Takagi
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan.,Kake Institute of Cytopathology, Okayama, Japan
| | - Masatsugu Ueda
- Faculty of Health Sciences, Kio University, Nara, Japan.,Graduate School of Health Sciences, Kio University, Nara, Japan
| |
Collapse
|
19
|
Rpn10 promotes tumor progression by regulating hypoxia-inducible factor 1 alpha through the PTEN/Akt signaling pathway in hepatocellular carcinoma. Cancer Lett 2019; 447:1-11. [PMID: 30673593 DOI: 10.1016/j.canlet.2019.01.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/08/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
The ubiquitin-proteasome pathway plays a pivotal role in tumor progression. Rpn10 is the major ubiquitin (Ub) receptor of the 26S proteasome. Mounting evidence shows that Rpn10 is associated with the progression of several tumor types. However, little is known regarding the mechanistic role of Rpn10 in hepatocellular carcinoma (HCC). In this study, we found that the upregulation of Rpn10 in HCC was associated with poor prognosis. The ectopic overexpression of Rpn10 increased HCC cell proliferation, whereas silencing Rpn10 expression resulted in decreased cell proliferation. Furthermore, we demonstrated that knockdown of Rpn10 induced cell cycle arrest at G1 phase in HCC cells. In addition, we found that Rpn10 increased cell proliferation via regulation of the PTEN/Akt pathways. Knockdown of Rpn10 induced suppression of cell proliferation could be reversed by overexpressing active Akt in HCC cells. Rpn10 directly promoted PTEN degradation through the ubiquitin-proteasome system. The transcription factor HIF1α directly bound to the Rpn10 promoter and increased its expression in HCC tissue. Moreover, we observed a significant correlation between HIF1α expression and Rpn10 levels in HCC patients and found that the combination of these two parameters was a more powerful predictor of poor prognosis than either parameter alone. Collectively, these findings highlight the molecular mechanism of Rpn10 expression in HCC and provide valuable information for cancer prognosis and treatment.
Collapse
|
20
|
Liu Z, Yang J, Ge C, Zhao F, Li H, Yao M, Li J, Tian H. Inhibitor of binding/differentiation 2 (Id2) is regulated by CCAAT/enhancer-binding protein-α (C/EBPα) and promotes the proliferation of hepatocellular carcinoma. Am J Cancer Res 2018; 8:2254-2266. [PMID: 30555742 PMCID: PMC6291656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/02/2018] [Indexed: 06/09/2023] Open
Abstract
Inhibitor of DNA binding/differentiation (Id2) is an important regulator involved in the initiation and progression of cancer. However, the function and mechanism of the regulation of Id2 in hepatocellular carcinoma (HCC) was unclear. In the present study, we found that the overexpression of Id2 increased HCC cell proliferation in vitro and in vivo. Knockdown of Id2 inhibited HCC cell proliferation in vitro and in vivo. Furthermore, knockdown of Id2 enhanced sorafenib-induced apoptosis in HCC. Conversely, overexpression of Id2 weakened sorafenib-induced apoptosis in HCC. In addition, the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) bound to the Id2 promoter and decreased its expression in HCC cells. Therefore, all results suggest that Id2 promotes the proliferation of HCC cells by inhibiting cell apoptosis. Id2 may serve as a potential target in HCC therapy.
Collapse
Affiliation(s)
- Zheng Liu
- Shanghai Medical College, Fudan UniversityShanghai 200032, PR China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Jing Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Hong Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of MedicineShanghai 200032, PR China
| |
Collapse
|
21
|
Luo G, Feng R, Sun Y, Zheng L, Wang Y, Chen Y, Wen T. Dendritic cell factor 1 inhibits proliferation and migration and induces apoptosis of neuroblastoma cells by inhibiting the ERK signaling pathway. Oncol Rep 2018; 41:103-112. [PMID: 30365123 PMCID: PMC6278510 DOI: 10.3892/or.2018.6796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 10/09/2018] [Indexed: 12/11/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor that affects mainly children and has extremely high mortality and recurrence rates. A previous study revealed that dendritic cell factor 1 (DCF1), also called transmembrane protein 59, could activate apoptosis in glioma cells. In the present study, we applied immunofluorescence, western blot analysis, flow cytometry and cell tumorigenicity to investigate the DCF1 mechanisms involved in NB apoptosis. DCF1 was overexpressed in Neuro-2a and SK-N-SH cells through instantaneous transfection. The data revealed that overexpression of DCF1 could inhibit cell proliferation, migration, invasion and promote cell apoptosis in vitro, and suppress NB growth in vivo. The ERK1/2 signaling pathway, which promotes cell survival, was the target of DCF1 in neuroblastoma cells. All the results indicated that DCF1 could be a potential therapeutic target for the understanding and treatment of NB.
Collapse
Affiliation(s)
- Guanghong Luo
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Ruili Feng
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Yangyang Sun
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Lili Zheng
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Yajiang Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Yanlu Chen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| |
Collapse
|
22
|
Hou H, Ge C, Sun H, Li H, Li J, Tian H. Tunicamycin inhibits cell proliferation and migration in hepatocellular carcinoma through suppression of CD44s and the ERK1/2 pathway. Cancer Sci 2018; 109:1088-1100. [PMID: 29377347 PMCID: PMC5891198 DOI: 10.1111/cas.13518] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/02/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022] Open
Abstract
Tunicamycin (TM) is an N‐linked glycosylation (NLG) inhibitor with strong antitumor activity, the exact underlying molecular mechanism of which remains to be elucidated. In our previous studies, we found that TM reversed drug resistance and improved the efficacy of combination treatments for hepatocellular carcinomas (HCC). Here, we investigated the effects of TM on HCC cell proliferation and migration as well as the mechanism of those effects. Our results showed that TM inhibited cell proliferation and migration as well as induced apoptosis of hepatocellular carcinoma cells. TM inhibited proliferation of HCC cells by inducing cell apoptosis and cell cycle arrest at the G2/M phase. Meanwhile, TM inhibited migration of HCC cells by suppressing CD44s‐mediated epithelial‐mesenchymal transition (EMT). TM inhibited migration and invasion of HCC cells by decreasing CD44 expression and altering its glycosylation. In addition, CD44s is involved in promoting EMT and is associated with a poor prognosis in HCC patients. Overexpression of CD44s promoted tumor migration and activated phosphorylation of ERK1/2 in HCC cells, whereas TM inhibited CD44s overexpression‐associated cell migration. The ability of TM to inhibit cell migration and invasion was enhanced or reversed in CD44s knockdown cells and cells overexpressing CD44s, respectively. The MEK/ERK inhibitor U0126 and TM inhibited hyaluronic acid‐induced cell migration in HCC cells. Furthermore, TM inhibited exogenous transforming growth factor beta (TGF‐β)‐mediated EMT by an ERK1/2‐dependent mechanism and restored the TGF‐β‐mediated loss of E‐cadherin. In summary, our study provides evidence that TM inhibits proliferation and migration of HCC cells through inhibition of CD44s and the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Helei Hou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hefen Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|