1
|
Wang M, An Q, Li Z, Huang Z, Huang K, Li G, Ma Q, Zhao L. The alkylglycerone phosphate synthase sustains the resistance of gastric cancer cells to ferroptosis induced by Apatinib. Gastric Cancer 2025:10.1007/s10120-025-01610-0. [PMID: 40186794 DOI: 10.1007/s10120-025-01610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Apatinib is a targeted therapy used in the treatment of advanced gastric cancer. However, many gastric cancer patients develop resistance to Apatinib, and the mechanisms underlying this resistance remain unclear. Previous studies have shown that Apatinib can induce ferroptosis in gastric cancer cells. More recent research suggests that polyunsaturated ether phospholipids are closely associated with tumor cell sensitivity to ferroptosis, and may represent key molecules involved in the resistance of tumor cells to ferroptosis. METHODS We established Apatinib-resistant gastric cancer cell lines and assessed their tolerance to ferroptosis. We identified key enzymes responsible for the ferroptosis tolerance observed in drug-resistant cells using lipidomics and transcriptomics analysis. Molecular and biological experiments were conducted to elucidate the molecular mechanisms underlying Apatinib resistance mediated by ferroptosis tolerance in gastric cancer cells. RESULTS Apatinib resistance is closely linked to ferroptosis resistance, which is driven by a reduction in the levels of polyunsaturated ether phospholipids-phospholipids that are particularly susceptible to oxidation and induce ferroptosis. The downregulation of key enzymes involved in polyunsaturated ether phospholipid synthesis, such as AGPS, mediates tolerance to both ferroptosis and Apatinib in gastric cancer cells, both in vitro and in vivo. Mechanistically, the expression of AGPS in tumor cells is regulated by the transcription factor ELK1. Drug-resistant cells acquire Apatinib tolerance by downregulating both ELK1 and AGPS expression. CONCLUSIONS Apatinib-resistant gastric cancer cells exhibit reduced expression of the transcription factor ELK1, which regulates the expression of AGPS. This reduction contributes to the resistance and malignancy of gastric cancer cells.
Collapse
Affiliation(s)
- Minghao Wang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qiyuan An
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Zhiwei Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhicheng Huang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Kaihua Huang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Guoxin Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qiang Ma
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Liying Zhao
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Wang Y, Xu S, Liu J, Qi P. A Novel Peroxisome-Related Gene Signature Predicts Breast Cancer Prognosis and Correlates with T Cell Suppression. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:887-911. [PMID: 39678026 PMCID: PMC11639899 DOI: 10.2147/bctt.s490154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
Background Peroxisomes are increasingly linked to cancer development, yet the prognostic role of peroxisome-related genes (PRGs) in breast cancer remains unclear. Objective This study aimed to construct a prognostic model based on PRG expression in breast cancer to clarify their prognostic value and clinical implications. Methods Transcriptomic data from TCGA and GEO were used for training and validation cohorts. TME characteristics were analyzed with ESTIMATE, MCP-counter, and CIBERSORT algorithms. qPCR validated mRNA expression levels of risk genes, and data analysis was conducted in R. Results Univariate and multivariate Cox regression identified a 7-gene PRG risk signature (ACBD5, ACSL5, DAO, NOS2, PEX3, PEX10, and SLC27A2) predicting breast cancer prognosis in training (n=1069), internal validation (n=327), and external validation (merged from four GEO datasets, n=640) datasets. While basal and Her2 subtypes had higher risk scores than luminal subtypes, a significant prognostic impact of the PRG risk signature was seen only in luminal subtypes. The high-risk subgroup exhibited a higher frequency of focal synonymous copy number alterations (SCNAs), arm-level amplifications and deletions, and single nucleotide variations. These increased genomic aberrations were associated with greater immune suppression and reduced CD8+ T cell infiltration. Bulk RNA sequencing and single-cell analyses revealed distinct expression patterns of peroxisome-related genes (PRGs) in the breast cancer TME: PEX3 was primarily expressed in malignant and stromal cells, while ACSL5 showed high expression in T cells. Additionally, the PRG risk signature demonstrated efficacy comparable to that of well-known biomarkers for predicting immunotherapy responses. Drug sensitivity analysis revealed that the PRG high-risk subgroup was sensitive to inhibitors of BCL-2 family proteins (BCL-2, BCL-XL, and MCL1) and other kinases (PLK1, PLK1, BTK, CHDK1, and EGFR). Conclusion The PRG risk signature serves as a promising biomarker for evaluating peroxisomal activity, prognosis, and responsiveness to immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Yunxiang Wang
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Sheng Xu
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Junfeng Liu
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Pan Qi
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| |
Collapse
|
3
|
Papin M, Fontaine D, Goupille C, Figiel S, Domingo I, Pinault M, Guimaraes C, Guyon N, Cartron PF, Emond P, Lefevre A, Gueguinou M, Crottès D, Jaffrès PA, Ouldamer L, Maheo K, Fromont G, Potier-Cartereau M, Bougnoux P, Chantôme A, Vandier C. Endogenous ether lipids differentially promote tumor aggressiveness by regulating the SK3 channel. J Lipid Res 2024; 65:100544. [PMID: 38642894 PMCID: PMC11127165 DOI: 10.1016/j.jlr.2024.100544] [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: 11/22/2023] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/22/2024] Open
Abstract
SK3 channels are potassium channels found to promote tumor aggressiveness. We have previously demonstrated that SK3 is regulated by synthetic ether lipids, but the role of endogenous ether lipids is unknown. Here, we have studied the role of endogenous alkyl- and alkenyl-ether lipids on SK3 channels and on the biology of cancer cells. Experiments revealed that the suppression of alkylglycerone phosphate synthase or plasmanylethanolamine desaturase 1, which are key enzymes for alkyl- and alkenyl-ether-lipid synthesis, respectively, decreased SK3 expression by increasing micro RNA (miR)-499 and miR-208 expression, leading to a decrease in SK3-dependent calcium entry, cell migration, and matrix metalloproteinase 9-dependent cell adhesion and invasion. We identified several ether lipids that promoted SK3 expression and found a differential role of alkyl- and alkenyl-ether lipids on SK3 activity. The expressions of alkylglycerone phosphate synthase, SK3, and miR were associated in clinical samples emphasizing the clinical consistency of our observations. To our knowledge, this is the first report showing that ether lipids differentially control tumor aggressiveness by regulating an ion channel. This insight provides new possibilities for therapeutic interventions, offering clinicians an opportunity to manipulate ion channel dysfunction by adjusting the composition of ether lipids.
Collapse
Affiliation(s)
- Marion Papin
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Delphine Fontaine
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Caroline Goupille
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France; Department of Gynecology, CHRU Bretonneau, Tours, France
| | - Sandy Figiel
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Isabelle Domingo
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Michelle Pinault
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Cyrille Guimaraes
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Nina Guyon
- CRCINA-INSERM 1232, Equipe « Apoptose et Progression tumorale », Nantes, France
| | | | - Patrick Emond
- iBrain, UMR 1253, INSERM, Université de Tours, Tours, France; Nuclear medicine in vitro department, CHRU Bretonneau, Tours, France
| | - Antoine Lefevre
- iBrain, UMR 1253, INSERM, Université de Tours, Tours, France
| | - Maxime Gueguinou
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - David Crottès
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Paul-Alain Jaffrès
- Laboratoire Chimie Electrochimie Moléculaires et Chimie Analytique (CEMCA), UMR 6521, CNRS, University of Brest, Brest, France
| | - Lobna Ouldamer
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France; Department of Gynecology, CHRU Bretonneau, Tours, France
| | - Karine Maheo
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Gaëlle Fromont
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France; Department of Pathology, CHRU Bretonneau, Tours, France
| | - Marie Potier-Cartereau
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Philippe Bougnoux
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Aurélie Chantôme
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Christophe Vandier
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France.
| |
Collapse
|
4
|
Zhang Y, Huang Z, Li K, Xie G, Feng Y, Wang Z, Li N, Liu R, Ding Y, Wang J, Yang J, Jia Z. TrkA promotes MDM2-mediated AGPS ubiquitination and degradation to trigger prostate cancer progression. J Exp Clin Cancer Res 2024; 43:16. [PMID: 38200609 PMCID: PMC10782585 DOI: 10.1186/s13046-023-02920-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/30/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND As a novel necrosis manner, ferroptosis has been increasingly reported to play a role in tumor progression and treatment, however, the specific mechanisms underlying its development in prostate cancer remain unclear. Growing evidence showed that peroxisome plays a key role in ferroptosis. Herein, we identified a novel mechanism for the involvement of ferroptosis in prostate cancer progression, which may provide a new strategy for clinical treatment of prostate cancer. METHODS Label-Free Mass spectrometry was used to screen and identify candidate proteins after ferroptosis inducer-ML210 treatment. Immunohistochemistry was undertaken to explore the protein expression of AGPS in prostate cancer tissues compared with normal tissues. Co-immunoprecipitation and GST pull-down were used to identify the directly binding of AGPS to MDM2 in vivo and in vitro. CCK8 assay and colony formation assay were used to illustrate the key role of AGPS in the progression of prostate cancer in vitro. The xenograft model was established to verify the key role of AGPS in the progression of prostate cancer in vivo. RESULTS AGPS protein expression was downregulated in prostate cancer tissues compared with normal tissues from the first affiliated hospital of Zhengzhou University dataset. Lower expression was correlated with poorer overall survival of patients compared to those with high expression of AGPS. In addition, AGPS can promote ferroptosis by modulating the function of peroxisome-resulting in the lower survival of prostate cancer cells. Furthermore, it was shown that AGPS can be ubiquitinated and degraded by the E3 ligase-MDM2 through the proteasomal pathway. Meanwhile, kinase TrkA can promote the combination of AGPS and MDM2 by phosphorylating AGPS at Y451 site. It was verified that kinase TrkA inhibitor-Larotrectinib can increase the susceptibility of prostate cancer cells to ferroptosis, which leads to the inhibition of prostate cancer proliferation to a great extent in vitro and in vivo. CONCLUSION Based on these findings, we proposed the combination of ferroptosis inducer and TrkA inhibitor to synergistically exert anti-tumor effects, which may provide a new strategy for the clinical treatment of prostate cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenlin Huang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Keqiang Li
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, China
| | - Guoqing Xie
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, China
| | - Yuankang Feng
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zihao Wang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ningyang Li
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruoyang Liu
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yinghui Ding
- Department of Otorhinolaryngology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jun Wang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Jinjian Yang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhankui Jia
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Papin M, Bouchet AM, Chantôme A, Vandier C. Ether-lipids and cellular signaling: A differential role of alkyl- and alkenyl-ether-lipids? Biochimie 2023; 215:50-59. [PMID: 37678745 DOI: 10.1016/j.biochi.2023.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/17/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Ether-lipids (EL) are specific lipids bearing a characteristic sn-1 ether bond. Depending on the ether or vinyl-ether nature of this bond, they are present as alkyl- or alkenyl-EL, respectively. Among EL, alkenyl-EL, also referred as plasmalogens in the literature, attract most of the scientific interest as they are the predominant EL species in eukaryotic cells, thus less is known about alkyl-EL. EL have been implicated in various signaling pathways and alterations in their quantity are frequently observed in pathologies such as neurodegenerative and cardiovascular diseases or cancer. However, it remains unknown whether both alkyl- and alkenyl-EL play the same roles in these processes. This review summarizes the roles and mechanisms of action of EL in cellular signaling and tries to discriminate between alkyl- and alkenyl-EL. We also focus on the involvement of EL-mediated alterations of cellular signaling in diseases and discuss the potential interest for EL in therapy.
Collapse
Affiliation(s)
- Marion Papin
- Nutrition, Croissance, Cancer (N2C) UMR 1069, University of Tours, INSERM, 37000, Tours, France.
| | | | - Aurélie Chantôme
- Nutrition, Croissance, Cancer (N2C) UMR 1069, University of Tours, INSERM, 37000, Tours, France
| | - Christophe Vandier
- Nutrition, Croissance, Cancer (N2C) UMR 1069, University of Tours, INSERM, 37000, Tours, France; Lifesome Therapeutics, López de Hoyos 42, 28006, Madrid, Spain
| |
Collapse
|
6
|
Bond KH, Sims-Lucas S, Oxburgh L. Targets for Renal Carcinoma Growth Control Identified by Screening FOXD1 Cell Proliferation Pathways. Cancers (Basel) 2022; 14:cancers14163958. [PMID: 36010951 PMCID: PMC9406217 DOI: 10.3390/cancers14163958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary FOXD1 regulates the proliferation of clear cell renal cell carcinoma (ccRCC) cells, and ccRCC cells in which FOXD1 has been inactivated do not form tumors efficiently in an animal model. Reproducing growth inhibition in tumor cells by inhibiting FOXD1 pathways presents a possible therapeutic approach for ccRCC and other cancers. We have established an analysis strategy to identify FOXD1-regulated target pathways that may be therapeutically tractable, and compounds that modulate these pathways were selected for testing. Targets in three pathways were identified: FOXM1, PME1, and TMEM167A, which were inhibited by compounds FDI-6, AMZ-30, and silibinin, respectively. The effects of these compounds on the growth of tumor cells from patients cultured in a novel 3D tumor-replica culture environment revealed that FDI-6 and silibinin had strong growth inhibitory effects. This investigation informs new therapeutic targets to control ccRCC tumor growth, and provides a strategy to compare the responsiveness of individual patient tumor replicas to growth-inhibitory compounds. Abstract Clinical association studies suggest that FOXD1 is a determinant of patient outcome in clear cell renal cell carcinoma (ccRCC), and laboratory investigations have defined a role for this transcription factor in controlling the growth of tumors through regulation of the G2/M cell cycle transition. We hypothesized that the identification of pathways downstream of FOXD1 may define candidates for pharmacological modulation to suppress the G2/M transition in ccRCC. We developed an analysis pipeline that utilizes RNA sequencing, transcription factor binding site analysis, and phenotype validation to identify candidate effectors downstream from FOXD1. Compounds that modulate candidate pathways were tested for their ability to cause growth delay at G2/M. Three targets were identified: FOXM1, PME1, and TMEM167A, which were targeted by compounds FDI-6, AMZ-30, and silibinin, respectively. A 3D ccRCC tumor replica model was used to investigate the effects of these compounds on the growth of primary cells from five patients. While silibinin reduced 3D growth in a subset of tumor replicas, FDI-6 reduced growth in all. This study identifies tractable pathways to target G2/M transition and inhibit ccRCC growth, demonstrates the applicability of these strategies across patient tumor replicas, and provides a platform for individualized patient testing of compounds that inhibit tumor growth.
Collapse
Affiliation(s)
- Kyle H. Bond
- Rogosin Institute, Room 2-43, 310 East 67th St., New York, NY 10065, USA
| | - Sunder Sims-Lucas
- Children’s Hospital of Pittsburgh, Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Leif Oxburgh
- Rogosin Institute, Room 2-43, 310 East 67th St., New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
7
|
Salah RA, Nasr MA, El-Derby AM, Abd Elkodous M, Mohamed RH, El-Ekiaby N, Osama A, Elshenawy SE, Hamad MHM, Magdeldin S, Gabr MM, Abdelaziz AI, El-Badri NS. Hepatocellular carcinoma cell line-microenvironment induced cancer-associated phenotype, genotype and functionality in mesenchymal stem cells. Life Sci 2022; 288:120168. [PMID: 34826437 DOI: 10.1016/j.lfs.2021.120168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 12/21/2022]
Abstract
Mesenchymal stromal cells (MSCs) have shown promise in liver cancer treatment. However, when MSCs are recruited to hepatic site of injury, they acquire cancerous promoting phenotype. AIMS To assess the influence of Hepatocellular carcinoma (HCC) microenvironment on human adipose MSCs (hA-MSCs) and predict hA-MSCs intracellular miRNAs role. MATERIALS AND METHODS After indirect co-culturing with Huh-7 cells, hA-MSCs were characterized via cell cycle profile, proliferation and migration potentials by MTT and scratch assays respectively. Functional enrichment analysis of deregulated proteins and miRNA targets was also analyzed. KEY FINDINGS Co-cultured hA-MSCs could acquire a cancer-associated phenotype as shown by upregulation of CAF, cancer markers, and downregulation of differentiation markers. Migration of these cancer-associated cells was increased concomitantly with upregulation of adhesion molecules, but not epithelial to mesenchymal transition markers. Co-cultured cells showed increased proliferation confirmed by downregulation in cell percentage in G0/G1, G2/M and upregulation in S phases of cell cycle. Upregulation of miR-17-5p and 615-5p in co-cultured hA-MSCs was also observed. Functional enrichment analysis of dysregulated proteins in co-cultured hA-MSCs, including our selected miRNAs targets, showed their involvement in development of cancer-associated characteristics. SIGNIFICANCE This study suggests an interaction between tumor cells and surrounding stromal components to generate cancer associated phenotype of some CAF-like characteristics, known to favor cancer progression. This sheds the light on the use of hA-MSCs in HCC therapy. hA-MSCs modulation may be partially achieved via dysregulation of intracellular miR17-5P and 615-5p expression, suggesting an important role for miRNAs in HCC pathogenesis, and as a possible therapeutic candidate.
Collapse
Affiliation(s)
- Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | - Mohamed A Nasr
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | - M Abd Elkodous
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | - Rania Hassan Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nada El-Ekiaby
- School of Medicine NewGiza University (NGU), Cairo, Egypt
| | - Aya Osama
- Proteomics and metabolomics Research Program, Basic Research Department, Children Cancer Hospital Egypt, 57357 Cairo, Egypt
| | - Shimaa E Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt
| | | | - Sameh Magdeldin
- Proteomics and metabolomics Research Program, Basic Research Department, Children Cancer Hospital Egypt, 57357 Cairo, Egypt; Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Mahmoud M Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | | | - Nagwa S El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Egypt.
| |
Collapse
|
8
|
Mass Spectrometry and Computer Simulation Predict the Interactions of AGPS and HNRNPK in Glioma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6181936. [PMID: 34621897 PMCID: PMC8492241 DOI: 10.1155/2021/6181936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/11/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022]
Abstract
Ether lipids are overexpressed in malignant tumor and play an important role in tumor process. Glioma is the most common malignant central nervous system tumor, and the content of ether lipids is higher than that of normal tissues. Alkylglycerone phosphate synthase (AGPS) is a key enzyme in the synthesis of ether esters and plays a vital role in maintaining the morphology and pathogenic properties of tumor cells. The cell proliferation and the content of tumor-related lipid such as monoalkylglycerol ether (MAGe), lysophosphatidic acid ether (LPAe), lysophosphatidylcholine ether (LPCe), lysophosphatidylethanolamine ether (LPEe), phosphatidyl inositol (PI), phosphatidylcholine (PC), and phosphatidylserine (PS) were suppressed after AGPS silencing in U251, H4, and TJ905 cells; however, heterogeneous nuclear ribonucleoprotein K (HNRNPK) could reverse the above phenomenon such as cellar proliferation and ether lipid secretion. We found that HNRNPK was the target protein of AGPS by coimmunoprecipitation and mass spectrometry assay and verified by western blot assay in U251 cells. It confirmed that AGPS and HNRNPK are coexpressed in the cellular nucleus by a confocal laser microscope. The main protein-protein interaction mechanism between AGPS and HNRNPK is hydrogen bond, conjugation bond, hydrophobic bond, and electrostatic force by computer simulation prediction.
Collapse
|
9
|
Abstract
RNAs are involved in an enormous range of cellular processes, including gene regulation, protein synthesis, and cell differentiation, and dysfunctional RNAs are associated with disorders such as cancers, neurodegenerative diseases, and viral infections. Thus, the identification of compounds with the ability to bind RNAs and modulate their functions is an exciting approach for developing next-generation therapies. Numerous RNA-binding agents have been reported over the past decade, but the design of synthetic molecules with selectivity for specific RNA sequences is still in its infancy. In this perspective, we highlight recent advances in targeting RNAs with synthetic molecules, and we discuss the potential value of this approach for the development of innovative therapeutic agents.
Collapse
Affiliation(s)
- Farzad Zamani
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan
| | - Takayoshi Suzuki
- The Institute of Scientific and Industrial Research, Osaka University, Mihogaoka 8-1, Ibaraki, Osaka 567-0047, Japan
| |
Collapse
|
10
|
Deb R, Joshi N, Nagotu S. Peroxisomes of the Brain: Distribution, Functions, and Associated Diseases. Neurotox Res 2021; 39:986-1006. [PMID: 33400183 DOI: 10.1007/s12640-020-00323-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
Peroxisomes are versatile cell organelles that exhibit a repertoire of organism and cell-type dependent functions. The presence of oxidases and antioxidant enzymes is a characteristic feature of these organelles. The role of peroxisomes in various cell types in human health and disease is under investigation. Defects in the biogenesis of the organelle and its function lead to severe debilitating disorders. In this manuscript, we discuss the distribution and functions of peroxisomes in the nervous system and especially in the brain cells. The important peroxisomal functions in these cells and their role in the pathology of associated disorders such as neurodegeneration are highlighted in recent studies. Although the cause of the pathogenesis of these disorders is still not clearly understood, emerging evidence supports a crucial role of peroxisomes. In this review, we discuss research highlighting the role of peroxisomes in brain development and its function. We also provide an overview of the major findings in recent years that highlight the role of peroxisome dysfunction in various associated diseases.
Collapse
Affiliation(s)
- Rachayeeta Deb
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Neha Joshi
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Shirisha Nagotu
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
11
|
Chen L, Zhang W, He L, Jin L, Qian L, Zhu Y. Effect of alkylglycerone phosphate synthase on the expression levels of lncRNAs in glioma cells and its functional prediction. Oncol Lett 2020; 20:66. [PMID: 32863899 PMCID: PMC7436103 DOI: 10.3892/ol.2020.11927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/18/2020] [Indexed: 01/01/2023] Open
Abstract
Alkylglycerone phosphate synthase (AGPS) is a key enzyme for ether ester synthesis and acts as an oncogene in malignant tumors. The present study aimed to investigate the effect of AGPS silencing on the expression levels of long non-coding RNAs (lncRNAs) and the co-expression with mRNAs in glioma U251 cells using microarray analysis. Furthermore, the underlying biological functions of crucial lncRNAs identified were investigated. It was discovered that in vitro U251 cell proliferation was suppressed following the genetic silencing of AGPS. Differentially expressed lncRNAs and mRNAs in U251 cells were sequenced following AGPS silencing. The results from the Gene Ontology analysis identified that the co-expressed mRNAs were mainly involved in biological processes, such as 'cellular response to hypoxia', 'extracellular matrix organization' and 'PERK-mediated unfolded protein response'. In addition, Kyoto Encyclopedia of Genes and Genomes signaling pathway enrichment analysis revealed that the co-expressed mRNAs were the most enriched in the 'AGE/RAGE signaling pathway in diabetic conditions'. Additionally, the PI3K/Akt and epidermal growth factor receptor signaling pathways serve important roles in tumor processes, for example carcinogenesis and angiogenesis. Furthermore, it was identified that the lncRNA AK093732 served a vital role in the regulatory network and the core pathway in this network regulated by this lncRNA was discovered to be the 'Cytokine-cytokine receptor interaction'. In conclusion, the findings of the present study suggested that AGPS may affect cell proliferation and the degree of malignancy. In addition, the identified lncRNAs and their co-expressed mRNAs screened using microarrays may have significant biological effects in the occurrence, development and metastasis of glioma, and thus may be novel markers of glioma.
Collapse
Affiliation(s)
- Lei Chen
- Department of Otolaryngology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Weijian Zhang
- Postgraduate School of Tianjin Medical University, Tianjin 300070, P.R. China
| | - Lihua He
- Postgraduate School of Tianjin Medical University, Tianjin 300070, P.R. China
- Department of Clinical Laboratory, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei 056002, P.R. China
| | - Li Jin
- Integrated Chinese and Western Medicine School of Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Liyu Qian
- Department of Tumor Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Yu Zhu
- Department of Clinical Laboratory, Tianjin Haihe Hospital, Tianjin 300350, P.R. China
| |
Collapse
|
12
|
Yu L, Gui S, Liu Y, Qiu X, Qiu B, Zhang X, Pan J, Fan J, Qi S, Zhang G. Long intergenic non-protein coding RNA 00475 silencing acts as a tumor suppressor in glioma under hypoxic condition by impairing microRNA-449b-5p-dependent AGAP2 up-regulation. Ther Adv Med Oncol 2020; 12:1758835920940936. [PMID: 32849915 PMCID: PMC7425262 DOI: 10.1177/1758835920940936] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/12/2020] [Indexed: 01/03/2023] Open
Abstract
Objective Long non-coding RNAs have been demonstrated to be involved in the progression of a variety of cancers, including glioma. Through microarray analyses, long intergenic non-protein coding RNA 00475 (LINC00475) was identified in the glioma development. However, its potential role remains incompletely understood. This study aimed to elucidate the effect of LINC00475 on the development of glioma under hypoxic conditions. Methods Glioma cells underwent hypoxic treatment and were collected. The functional role of LINC00475 and AGAP2 in glioma was determined using ectopic expression, depletion, and reporter assay experiments. Then, the expression of LINC00475, microRNA (miR)-449b-5p, AGAP2, FAK, and HIF-1α was determined. In addition, cell migration and invasion were examined. Finally, a tumor xenograft was carried out in nude mice to explore the role of LINC00475 on oxidation in vivo. Results LINC00475 was identified to be overexpressed in hypoxic glioma samples, which was further observed to bind to and down-regulate miR-449b-5p, and negatively targeted AGAP2. Moreover, we also revealed a positive correlation between LINC00475 and AGAP2 expression in glioma. In addition, silencing of LINC00475 decreased the extent of FAK phosphorylation and reduced the expression of HIF-1α and AGAP2. It was also observed that LINC00475 silencing suppressed glioma cell proliferation, migration, and invasion, and promoted cell apoptosis. Moreover, oxidation of nude mice was promoted by LINC00475 silencing. Conclusion Taken together, LINC00475 silencing exerted an inhibitory effect on glioma under hypoxic conditions by down-regulating AGAP2 via up-regulation of miR-449b-5p.
Collapse
Affiliation(s)
- Lei Yu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou 510515, Guangdong Province, P. R. China
| | - Si Gui
- Department of Radiology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, P. R. China
| | - Yawei Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xiaoyu Qiu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Binghui Qiu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xi'an Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Jun Pan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Jun Fan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Guozhong Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou 510515, Guangdong Province, P. R. China
| |
Collapse
|
13
|
Abstract
Peroxisomes are metabolic organelles involved in lipid metabolism and cellular redox balance. Peroxisomal function is central to fatty acid oxidation, ether phospholipid synthesis, bile acid synthesis, and reactive oxygen species homeostasis. Human disorders caused by genetic mutations in peroxisome genes have led to extensive studies on peroxisome biology. Peroxisomal defects are linked to metabolic dysregulation in diverse human diseases, such as neurodegeneration and age-related disorders, revealing the significance of peroxisome metabolism in human health. Cancer is a disease with metabolic aberrations. Despite the critical role of peroxisomes in cell metabolism, the functional effects of peroxisomes in cancer are not as well recognized as those of other metabolic organelles, such as mitochondria. In addition, the significance of peroxisomes in cancer is less appreciated than it is in degenerative diseases. In this review, I summarize the metabolic pathways in peroxisomes and the dysregulation of peroxisome metabolism in cancer. In addition, I discuss the potential of inactivating peroxisomes to target cancer metabolism, which may pave the way for more effective cancer treatment.
Collapse
|
14
|
Fontaine D, Figiel S, Félix R, Kouba S, Fromont G, Mahéo K, Potier-Cartereau M, Chantôme A, Vandier C. Roles of endogenous ether lipids and associated PUFAs in the regulation of ion channels and their relevance for disease. J Lipid Res 2020; 61:840-858. [PMID: 32265321 PMCID: PMC7269763 DOI: 10.1194/jlr.ra120000634] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Indexed: 12/16/2022] Open
Abstract
Ether lipids (ELs) are lipids characterized by the presence of either an ether linkage (alkyl lipids) or a vinyl ether linkage [i.e., plasmalogens (Pls)] at the sn1 position of the glycerol backbone, and they are enriched in PUFAs at the sn2 position. In this review, we highlight that ELs have various biological functions, act as a reservoir for second messengers (such as PUFAs) and have roles in many diseases. Some of the biological effects of ELs may be associated with their ability to regulate ion channels that control excitation-contraction/secretion/mobility coupling and therefore cell physiology. These channels are embedded in lipid membranes, and lipids can regulate their activities directly or indirectly as second messengers or by incorporating into membranes. Interestingly, ELs and EL-derived PUFAs have been reported to play a key role in several pathologies, including neurological disorders, cardiovascular diseases, and cancers. Investigations leading to a better understanding of their mechanisms of action in pathologies have opened a new field in cancer research. In summary, newly identified lipid regulators of ion channels, such as ELs and PUFAs, may represent valuable targets to improve disease diagnosis and advance the development of new therapeutic strategies for managing a range of diseases and conditions.
Collapse
Affiliation(s)
- Delphine Fontaine
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sandy Figiel
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Romain Félix
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sana Kouba
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Gaëlle Fromont
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Department of Pathology, CHRU Bretonneau, F-37044 Tours CEDEX 9, France
| | - Karine Mahéo
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | | | - Aurélie Chantôme
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | - Christophe Vandier
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France. mailto:
| |
Collapse
|
15
|
Yan T, Zhuang J, He L. Carboxamide derivatives induce apoptosis in the U251 glioma cell line. Oncol Lett 2019; 18:1409-1414. [PMID: 31423205 DOI: 10.3892/ol.2019.10434] [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: 03/29/2018] [Accepted: 04/08/2019] [Indexed: 12/11/2022] Open
Abstract
Glioma is a malignant tumor that is frequently treated using chemotherapy. The aim of the present study was to examine the antitumor activity of two novel carboxamide derivatives in glioma, and investigate the underlying mechanisms. Two previously designed and synthesized carboxamide derivatives were selected and their effects on glioma cells were evaluated. Specifically, assays to evaluate proliferation, apoptosis, oxidation, caspase-3, -8 and -9 activity, and the expression of Bcl-2 and surviving in glioma cells were conducted. The carboxamide derivatives were revealed to inhibit proliferation, as well as to induce apoptosis and oxidative damage in glioma U251 cells. In addition, the carboxamide derivatives increased the activity of caspase-3, -8 and -9, and suppressed the expression of Bcl-2 and survivin. These findings demonstrate that the carboxamide derivatives displayed antitumor activity against glioma in vitro, which may have been mediated via the induction of oxidative damage and apoptosis.
Collapse
Affiliation(s)
- Tao Yan
- Department of Pharmacy, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Junxue Zhuang
- Department of Pharmacy, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin 301800, P.R. China
| | - Lu He
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|
16
|
Development of alkyl glycerone phosphate synthase inhibitors: Structure-activity relationship and effects on ether lipids and epithelial-mesenchymal transition in cancer cells. Eur J Med Chem 2018; 163:722-735. [PMID: 30576903 DOI: 10.1016/j.ejmech.2018.11.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/06/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023]
Abstract
In aggressive tumors, alkylglyceronephosphate synthase (AGPS) controls cellular ether phospholipid utilization and metabolism to promote cancer cell proliferation and motility. SAR studies on the first-in-class AGPS inhibitor 1, discovered by our group, led to the 2,6-difluoro analog 2i which showed higher binding affinity than 1in vitro. In 231MFP cancer cells, 2i reduced ether lipids levels and cell migration rate. When tested in PC-3 and MDA-MB-231 cancer cells, 2i specifically impaired epithelial to mesenchymal transition (EMT) by modulating E-cadherin, Snail and MMP2 expression levels. Moreover, the combination of siRNAs against AGPS and 2i provided no additive effect, confirming that the modulation of 2i on EMT specifically relies on AGPS inhibition. Finally, this compound also affected cancer cell proliferation especially in MDA-MB-231 cells expressing higher AGPS level, whereas it provided negligible effects on MeT5A, a non-tumorigenic cell line, thus showing cancer specificity.
Collapse
|
17
|
Qian L, Zhu Y. Computer-aided drug design and inhibitive effect of a novel nitrogenous heterocyclic compound and its mechanism on glioma U251 cells and breast cancer MCF-7 cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1931-1939. [PMID: 29983547 PMCID: PMC6027699 DOI: 10.2147/dddt.s168130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Glioma and breast cancer are severe malignant cancerous tumors that highlight the importance of developing new anti-cancer drugs. The aim of this study was to explore the effects of a novel nitrogenous heterocyclic compound on glioma and breast cancer cells and to determine its mechanism of action. Methods We designed and synthesized a novel nitrogenous heterocyclic compound, 3-(4-amino-1H-benzo[d]imidazole-2-carboxamido)-4-oxo-3,4-dihydroimidazo[5,1-d][1,2,3,5] tetrazine-8-carboxamide, based on alkylglycerone phosphate synthase (AGPS) using computer-aided drug design (CADD), and we measured its effect on the proliferation, invasion, cell cycle and apoptosis of U251 glioma and MCF-7 breast cancer cells. In addition, the compound’s effect on the expression of tumor-related mRNA, circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs) was explored. Results It was found that the nitrogenous heterocyclic compound could induce cell cycle arrest at the G2/M phase of U251/MCF-7 cells and activate apoptosis. Real-time PCR showed that the expression levels of tumor-related mRNA, circRNAs and lncRNAs were impacted. Conclusion We concluded that the nitrogenous heterocyclic compound inhibits the proliferation and invasion of U251 glioma and MCF-7 breast cancer cells through the induction of apoptosis and cell cycle arrest by regulating tumor-related genes.
Collapse
Affiliation(s)
- Liyu Qian
- Department of Tumor Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China,
| | - Yu Zhu
- Tianjin Key Laboratory of Cerebral Vessels and Neuraldegenerative Disease, Department of Clinical Laboratory, Tianjin Huanhu Hospital, Tianjin 300350, China,
| |
Collapse
|
18
|
Zhu Y, Han Y, Ma Y, Yang P. ADME/toxicity prediction and antitumor activity of novel nitrogenous heterocyclic compounds designed by computer targeting of alkylglycerone phosphate synthase. Oncol Lett 2018; 16:1431-1438. [PMID: 30008821 PMCID: PMC6036461 DOI: 10.3892/ol.2018.8873] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/15/2018] [Indexed: 12/24/2022] Open
Abstract
Alkylglycerone phosphate synthase (AGPS) is an oncogene and can be considered as an antitumor drug target. The aim of the present study was to design novel nitrogenous heterocyclic compound improving targetability by computer-aided drug design technology targeting AGPS. A total of 12 nitrogenous heterocyclic compounds were designed and predicted the absorption, distribution, metabolism and excretion parameters/toxicity. Their activity in terms of proliferation inhibition, cell cycle arrest and apoptosis induction was then measured using an MTS assay and a high-content screening system in U251 cells. The results showed that anti-glioma activity was present in compounds N4, N5, N6, N7, N8 and N12, which was in accordance with the computer prediction. Therefore, these compounds may be suitable for the development of a novel glioma therapeutic drug.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Yuan Han
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Ping Yang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vessels and Neural Degeneration, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| |
Collapse
|
19
|
Hou S, Tan J, Yang B, He L, Zhu Y. Effect of alkylglycerone phosphate synthase on the expression profile of circRNAs in the human thyroid cancer cell line FRO. Oncol Lett 2018; 15:7889-7899. [PMID: 29731907 PMCID: PMC5920571 DOI: 10.3892/ol.2018.8356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 02/27/2018] [Indexed: 12/17/2022] Open
Abstract
Thyroid cancer is a common primary tumor in China. Therefore, it is important to investigate the underlying molecular mechanism of thyroid cancer in order to achieve effective individualized treatments. In our previous study, a positive correlation between the expression of alkylglycerone phosphate synthase (AGPS) and the malignant phenotype of thyroid cancer cell lines was identified. The inactivation of AGPS was able to decrease the malignancy of cancer, and inhibit tumor growth and invasion. However, the function of AGPS on thyroid cancer was unclear. In the present study, it was revealed that AGPS was able to regulate the expression of circular RNAs (circRNAs), which may be the mechanism of its anticancer activity. Therefore, the effects of AGPS silencing and knockout on circRNA expression in the thyroid cancer cell line FRO were investigated using circRNAs microarray, and Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed in order to investigate the underlying molecular mechanism of AGPS for the regulation of thyroid cancer through circRNAs.
Collapse
Affiliation(s)
- Shasha Hou
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Bing Yang
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Lu He
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yu Zhu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vessels and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
| |
Collapse
|
20
|
Lee S, Cheung-See-Kit M, Williams TA, Yamout N, Zufferey R. The glycosomal alkyl-dihydroxyacetonephosphate synthase TbADS is essential for the synthesis of ether glycerophospholipids in procyclic trypanosomes. Exp Parasitol 2018; 185:71-78. [PMID: 29355496 DOI: 10.1016/j.exppara.2018.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/30/2017] [Accepted: 01/14/2018] [Indexed: 01/15/2023]
Abstract
Glycerophospholipids are the main constituents of the biological membranes in Trypanosoma brucei, which causes sleeping sickness in humans. The present work reports the characterization of the alkyl-dihydroxyacetonephosphate synthase TbADS that catalyzes the committed step in ether glycerophospholipid biosynthesis. TbADS localizes to the glycosomal lumen. TbADS complemented a null mutant of Leishmania major lacking alkyl-dihydroxyacetonephosphate synthase activity and restored the formation of normal form of the ether lipid based virulence factor lipophosphoglycan. Despite lacking alkyl-dihydroxyacetonephosphate synthase activity, a null mutant of TbADS in procyclic trypanosomes remained viable and exhibited normal growth. Comprehensive analysis of cellular glycerophospholipids showed that TbADS was involved in the biosynthesis of all ether glycerophospholipid species, primarily found in the PE and PC classes.
Collapse
Affiliation(s)
- Sungsu Lee
- Department of Biological Sciences, St John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA
| | - Melanie Cheung-See-Kit
- Department of Biological Sciences, St John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA
| | - Tyler A Williams
- Department of Biological Sciences, St John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA
| | - Nader Yamout
- Department of Biological Sciences, St John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA
| | - Rachel Zufferey
- Department of Biological Sciences, St John's University, 8000 Utopia Parkway, Jamaica, NY 11439, USA.
| |
Collapse
|
21
|
Tuo H, Shu F, She S, Yang M, Zou XQ, Huang J, Hu HD, Hu P, Ren H, Peng SF, Yang YX. Sorcin induces gastric cancer cell migration and invasion contributing to STAT3 activation. Oncotarget 2017; 8:104258-104271. [PMID: 29262638 PMCID: PMC5732804 DOI: 10.18632/oncotarget.22208] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a globally occurring malignancy that is characterized by a high mortality rate due to a high tendency to metastasize and poor prognoses. Sorcin, as known as SRI, a soluble resistance-related calcium-binding protein, plays a significant role in multidrug resistance. Sorcin is related to the migration and invasion of cancer cells. However, the mechanism remains unclear. Here, we used immunohistochemistry to confirm that the expression of sorcin in cancer tissues is higher than that in the adjacent normal tissues. The wound healing and transwell results indicate that sorcin can induce migration and invasion of GC cells. To explore the role of sorcin in GC metastasis, isobaric tags for relative and absolutely quantitation (iTRAQ) were used to examine cells with and without sorcin knockdown to identify the differentially expressed proteins (DEPs). The results were evaluated via RT-PCR and western blot to confirm the ITRAQ data. Inhibition of sorcin expression can down- regulate the expression of CTSZ, MMP2, MMP9 and p-STAT3 followed by suppression of tumor growth and metastasis. Together, we concluded that sorcin has a oncogenic activity via inducing tumor growth and metastasis, leading to development of therapeutic treatments for GC.
Collapse
Affiliation(s)
- Huan Tuo
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Feng Shu
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Sha She
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiao Qin Zou
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Juan Huang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Huai Dong Hu
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China
| | - Peng Hu
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China.,Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Hong Ren
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China.,Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Shi Fang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Hunan 410008, China.,Department of Health Management Center, Xiangya Hospital, Central South University, Hunan 410008, China
| | - Yi Xuan Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Institute for Viral Hepatitis of Chongqing Medical University, Chongqing 400016, China.,Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
22
|
Cipolla CM, Lodhi IJ. Peroxisomal Dysfunction in Age-Related Diseases. Trends Endocrinol Metab 2017; 28:297-308. [PMID: 28063767 PMCID: PMC5366081 DOI: 10.1016/j.tem.2016.12.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/04/2016] [Accepted: 12/07/2016] [Indexed: 12/21/2022]
Abstract
Peroxisomes carry out many key functions related to lipid and reactive oxygen species (ROS) metabolism. The fundamental importance of peroxisomes for health in humans is underscored by the existence of devastating genetic disorders caused by impaired peroxisomal function or lack of peroxisomes. Emerging studies suggest that peroxisomal function may also be altered with aging and contribute to the pathogenesis of a variety of diseases, including diabetes and its related complications, neurodegenerative disorders, and cancer. With increasing evidence connecting peroxisomal dysfunction to the pathogenesis of these acquired diseases, the possibility of targeting peroxisomal function in disease prevention or treatment becomes intriguing. Here, we review recent developments in understanding the pathophysiological implications of peroxisomal dysfunctions outside the context of inherited peroxisomal disorders.
Collapse
Affiliation(s)
- Cynthia M Cipolla
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
23
|
Wang H, Qian Z, Zhao H, Zhang X, Che S, Zhang H, Shang H, Bao J, Hao C, Liu J, Li Z. CSN5 silencing reverses sorafenib resistance of human hepatocellular carcinoma HepG2 cells. Mol Med Rep 2015; 12:3902-3908. [PMID: 26035694 DOI: 10.3892/mmr.2015.3871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 03/18/2015] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common tumor types, and is the third leading cause of cancer mortalities worldwide. A large number of patients with HCC are diagnosed at a late stage when the curative treatment of surgical resection and liver transplantation are no longer applicable. Sorafenib has been proved to improve overall survival in advanced HCC; however, drug resistance is common. The present study reported that the CSN5 is correlated with sorafenib resistance of the HCC cell line HepG2/S. Following silencing of CSN5, resistance to sorafenib was reversed, and multi-drug‑resistance proteins, including as adenosine triphosphate binding cassette (ABC)B1, ABCC2 and ABCG2 as well as CDK6, cyclin D1 and B‑cell lymphoma 2 were downregulated. In addition, it was demonstrated that the integrin beta-1, transforming growth factor‑β1 and nuclear factor‑κB pathways were modified by CSN5.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis
- COP9 Signalosome Complex
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Cyclin-Dependent Kinase 6/genetics
- Cyclin-Dependent Kinase 6/metabolism
- Drug Resistance, Neoplasm
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Hep G2 Cells
- Humans
- Integrin beta1/genetics
- Integrin beta1/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Multidrug Resistance-Associated Protein 2
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- NF-kappa B/physiology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Peptide Hydrolases/genetics
- Peptide Hydrolases/metabolism
- Phenylurea Compounds/pharmacology
- RNA, Small Interfering/genetics
- Signal Transduction
- Sorafenib
- Transforming Growth Factor beta1/physiology
Collapse
Affiliation(s)
- Haibo Wang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Zhengyao Qian
- Department of Cardiology, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Hui Zhao
- Tianjin Entry‑Exit Inspection and Quarantine Bureau, International Travel Healthcare Center, Tianjin 300456, P.R. China
| | - Xibo Zhang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Shuqiang Che
- Department of Nephrology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, P.R. China
| | - Hongtao Zhang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Haitao Shang
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Jianheng Bao
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Chengfei Hao
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Junjian Liu
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Zhonglian Li
- Department of Hepatobiliary and Pancreatic Surgery (2), Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| |
Collapse
|
24
|
Zhao W, Liu S, Dou Q, Li C, DU J, Ren W. The role and mechanism of WEE1 on the cisplatin resistance reversal of the HepG2/DDP human hepatic cancer cell line. Oncol Lett 2015; 10:3081-3086. [PMID: 26722293 DOI: 10.3892/ol.2015.3647] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 05/20/2015] [Indexed: 12/12/2022] Open
Abstract
Drug resistance to cisplatin with continuous drug treatment is one of the most common causes of chemotherapy failure in hepatic carcinoma. Accumulating evidence suggests that WEE1 G2 checkpoint kinase (WEE1) is involved in cisplatin resistance, which has been demonstrated to correlate with cancer initiation and progression. However, the role and molecular mechanism of WEE1 in the drug resistance of hepatic cancer remains unclear. In the present study, using the WEE-knockdown hepatic cancer cell line HepG2/DDP, the role of WEE1 and its molecular mechanism were investigated. It was demonstrated that silencing WEE1 expression resulted in an increased cisplatin sensitivity of HepG2/DDP, in addition to an increased rate of apoptosis and intracellular concentration of rhodamine 123. The expression levels of P-gp, MDR1, MRP1, LRP, BCL-2, survivin and GST in WEE1-silenced HepG2/DDP cells were significantly reduced, and phosphorylation levels of MEK and ERK were significantly downregulated. The results demonstrated that WEE1 negatively regulated the multidrug resistance potential of human hepatic cancer cells by modulating the expression of relevant drug resistance genes and the activity of the MEK/ERK pathway. Therefore, WEE1 may be a monitoring bio-marker for drug resistance, and a therapeutic target in hepatic cancer.
Collapse
Affiliation(s)
- Weifeng Zhao
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Shuyuan Liu
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Qian Dou
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Changan Li
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jingpei DU
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Weihua Ren
- Central Laboratory, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| |
Collapse
|
25
|
Xiao J, Liu L, Zhong Z, Xiao C, Zhang J. Mangiferin regulates proliferation and apoptosis in glioma cells by induction of microRNA-15b and inhibition of MMP-9 expression. Oncol Rep 2015; 33:2815-20. [PMID: 25901555 DOI: 10.3892/or.2015.3919] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/02/2015] [Indexed: 11/05/2022] Open
Abstract
Mangiferin, a flavonoid extracted from the leaves of the Anacardiaceae plant, the mango tree, has physiological activity and pharmacological effects in many aspects. The present study aimed to clarify the effect of mangiferin on proliferation and apoptosis of glioma cells and the mechanism of these curative effects of mangiferin. In this experiment, we detected the proliferation using 3-(4,5-dimethylthylthiazol-2-yl)-2,5 diphenyltetrazolium bromide (MTT) assay. Then, cell apoptosis of U87 glioma cells was measured with the Annexin V-FITC/propidium iodide (PI) apoptosis detection kit, DAPI staining assay and the caspase-3 and caspase-9 activity assay kit. Next, quantitative real-time PCR and gelatin zymography were used to analyze the expression of microRNA-15b (miR-15b) and matrix metalloproteinase-9 (MMP-9), respectively. MMP-9 agonist, miR-15b mimics and anti-miR-15b mimics were added to the U87 glioma cells for elucidating the mechanisms involved in the curative effects of mangiferin. In the present study, mangiferin notably restrained the proliferation and increased the apoptosis of the U87 glioma cells. Meanwhile, mangiferin specifically promoted the expression of miR-15b and suppressed the level of MMP-9 in the U87 glioma cells. miR-15b regulated the expression of MMP-9 in the U87 glioma cells. MMP-9 agonist and anti-miR‑15b reduced the curative effects of mangiferin in the U87 glioma cells. In summary, mangiferin regulates proliferation and apoptosis in glioma cells by induction of miR-15b and inhibition of MMP-9 expression.
Collapse
Affiliation(s)
- Jinsong Xiao
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Li Liu
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zian Zhong
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Cheng Xiao
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Junjian Zhang
- Division of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
26
|
Shen C, Yang H, Liu H, Wang X, Zhang Y, Xu R. Inhibitory effect and mechanisms of microRNA-146b-5p on the proliferation and metastatic potential of Caski human cervical cancer cells. Mol Med Rep 2015; 11:3955-61. [PMID: 25572123 DOI: 10.3892/mmr.2015.3151] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 11/19/2014] [Indexed: 11/05/2022] Open
Abstract
Cervical cancer is a common cause of cancer‑associate mortality in females, and metastasis is strongly associated with failure of cervical cancer treatment. Previous studies have indicated that microRNA (miR)‑146b‑5p is involved in the inhibition of proliferation and metastasis of numerous human cancer types. The aim of the present study was to explore the inhibitory effect of miR‑156b‑5p on the proliferation and metastatic potential of Caski human cervical cancer cells, as well as to determine the mechanisms by which it proceeds. The results demonstrated that miR‑146b‑5p was able to inhibit the proliferative, invasive and adhesive potential and block the cell cycle progression of Caski human cervical cancer cells, as determined using MTS and transwell assays as well as flow cytometry. Furthermore, quantitative polymerase chain reaction and western blot analysis revealed that transfection with miR‑146b‑5p decreased the mRNA and protein expression levels of C‑X‑C chemokine receptor type 4, matrix metalloproteinase‑2 and ‑9, c‑Myc, cyclin D1 and human papilloma virus 16. In addition, the secretion levels of transforming growth factor‑β, monocyte chemoattractant protein‑1 and tumor necrosis factor‑α, the telomerase activity, the phosphorylation of c‑Jun N‑terminal protein kinase and protein kinase B and the transcriptional activities of nuclear factor‑κB, signal transducer and activator of transcription‑3 and ‑5 were reduced. However, increased levels of p27 and p53 were detected in the miR‑146b‑5p‑overexpressing Caski cells. These results indicate that miR‑146b‑5p may be a potential therapeutic strategy for the treatment of cervical cancer through regulation of cell chemotaxis and the cell cycle.
Collapse
Affiliation(s)
- Cuiping Shen
- Department of Gynecology and Obstetrics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hui Yang
- Department of Gynecology and Obstetrics, Rizhao People's Hospital, Rizhao, Shandong 276826, P.R. China
| | - Hong Liu
- Department of Gynecology and Obstetrics, Rizhao People's Hospital, Rizhao, Shandong 276826, P.R. China
| | - Xiuqin Wang
- Department of Gynecology and Obstetrics, Rizhao People's Hospital, Rizhao, Shandong 276826, P.R. China
| | - Youzhong Zhang
- Department of Gynecology and Obstetrics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Rui Xu
- Department of Gynecology and Obstetrics, Rizhao People's Hospital, Rizhao, Shandong 276826, P.R. China
| |
Collapse
|