1
|
Lei Y, Dong R, Sun C, Hu Y, Yan Y, Song G, Wang Y. The Role of CXCL13 in GC-1 Cell Cycle Arrest Induced by Titanium Dioxide Nanoparticles Through JAK2/STAT3 Signaling Pathway. J Appl Toxicol 2025. [PMID: 39777719 DOI: 10.1002/jat.4747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/02/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) can induce the cell cycle arrest in spermatogonia, and the JAK2/STAT3 signaling pathway plays a pivotal role in cell cycle progression, but the specific upstream regulatory mechanisms are not completely clarified. The purpose of this study was to investigate whether CXCL13 regulated the JAK2/STAT3 signaling pathway to participate in cell cycle arrest after mouse spermatogonia cell line (GC-1) exposure to TiO2 NPs. The GC-1 cells were treated with TiO2 NPs at different concentrations (0, 10, 20, 30, and 40 μg/mL) for 24 h to detect cell viability, cell cycle distribution, CXCL13 protein, JAK2/STAT3 pathway-related proteins, and cell cycle-related proteins. The CXCL13 recombinant protein was used to verify the role of CXCL13 in cell cycle and JAK2/STAT3 signaling pathway. TiO2 NPs inhibited cell viability; regulated cell cycle-related proteins including remarkably decreased Cyclin D1, CDK4, Cyclin E1, and CDK2 as well as increased p21; and induced cell cycle arrest at the G0/G1 phase. TiO2 NPs inhibited the levels of CXCL13 protein and weakened the activation of the JAK2/STAT3 signaling pathway by reducing the levels of p-JAK2/JAK2 and p-STAT3/STAT3 proteins. Furthermore, CXCL13 mitigated the suppression of the JAK2/STAT3 signaling pathway and the G0/G1 cell cycle arrest caused by TiO2 NPs. Taken together, TiO2 NPs downregulated the expression of CXCL13 to inhibit the activation of downstream JAK2/STAT3 signaling pathway, eventually inducing cell cycle arrest at the G0/G1 phase. These results provide a novel insight for complemented understanding of the mechanisms of TiO2 NPs-induced cell cycle arrest in GC-1 cells.
Collapse
Affiliation(s)
- Yuzhu Lei
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Ruoyun Dong
- Xi'an Center for Disease Control and Prevention, Xi'an, China
| | - Chenhao Sun
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yunhua Hu
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yizhong Yan
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Guanling Song
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yan Wang
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
2
|
Tuncer SÇ, Gur C, Kucukler S, Akarsu SA, Kandemir FM. Effects of zingerone on rat induced testicular toxicity by sodium arsenite via oxidative stress, endoplasmic reticulum stress, inflammation, apoptosis, and autophagy pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:603-610. [PMID: 38629098 PMCID: PMC11017849 DOI: 10.22038/ijbms.2024.73342.15934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 12/05/2023] [Indexed: 04/19/2024]
Abstract
Objectives This study aimed to investigate the effects of zingerone (ZNG) treatment on testicular toxicity in rats induced by sodium arsenite (SA). Materials and Methods In the study, five groups were formed (n=7) and the experimental groups were designated as follows; Vehicle group, ZNG group, SA group, SA+ZNG 25 group, and SA+ZNG 50 group. While SA was administered orally to rats at 10 mg/kg/bw, ZNG was given to rats orally at 25 and 50 mg/kg/bw doses for 14 days. Results As a result of the presented study, an increase was observed in the MDA contents of the testicular tissue of the rats administered SA, while significant decreases were observed in GSH levels, SOD, CAT, and GPx activities. The mRNA transcript levels of the pro-inflammatory genes NF-κB, TNF-α, IL-1β, and IL-6 were triggered after SA administration. Additionally, SA administration caused inflammation by increasing RAGE, NLRP3, and JAK-2/STAT3 gene expression. Moreover, endoplasmic reticulum (ER) stress occurred in the testicular tissues of SA-treated rats and thus ATF-6, PERK, IRE1, and GRP78 genes were up-regulated. SA caused apoptosis by up-regulating Bax and Caspase-3 expressions and inhibiting Bcl-2 expression in testicles. SA caused histological irregularities in the testicles, resulting in decreased sperm quality. Conclusion ZNG treatment reduced SA-induced oxidative stress, ER stress, inflammation, apoptosis, and histological irregularities in the testicles while increasing sperm quality. As a result, it was observed that ZNG could alleviate the toxicity caused by SA in the testicles.
Collapse
Affiliation(s)
- Sibel Çiğdem Tuncer
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Cihan Gur
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Serkan Ali Akarsu
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| |
Collapse
|
3
|
Park MN, Jeon HW, Rahman MA, Park SS, Jeong SY, Kim KH, Kim SH, Kim W, Kim B. Daemonorops draco Blume Induces Apoptosis Against Acute Myeloid Leukemia Cells via Regulation of the miR-216b/c-Jun. Front Oncol 2022; 12:808174. [PMID: 35356209 PMCID: PMC8959842 DOI: 10.3389/fonc.2022.808174] [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: 11/03/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Daemonorops draco Blume (DD), also called dragon’s blood, has been used as a traditional Korean medicine, especially for relieving pain caused by wound infection. Recently, it has been described that DD has antibacterial and analgesic effects. In this study, the underlying anticancer effect of DD associated with apoptosis was investigated in acute myeloid leukemia cell lines U937 and THP-1. DD exhibited cytotoxic effects and induced apoptosis in U937 and THP-1 cells. Moreover, DD treatment significantly reduced mitochondrial membrane potential (ΔΨ). The protein expression of cleaved poly(ADP-ribose) polymerase, cleaved caspase-3, p-H2A.X, CCAAT/enhancer-binding protein (CHOP), and activating transcription factor 4 was upregulated by DD treatment. Consistently, DD-treated cells had increased reactive oxygen species (ROS) level in a concentration-dependent manner via miR-216b activation in association with c-Jun inhibition. N-acetyl-L-cysteine pretreatment reversed the cytotoxic effect of DD treatment as well as prevented ROS accumulation. Collectively, the results of this study suggest that the anticancer effect of DD in AML was mediated by CHOP-dependent apoptosis along with ROS accumulation and included upregulation of miR-216b followed by a decrease in c-Jun.
Collapse
Affiliation(s)
- Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea.,Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hee Won Jeon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Md Ataur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea.,Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Se Sun Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Se Yun Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sung-Hoon Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Woojin Kim
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea.,Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
4
|
Rah B, Rather RA, Bhat GR, Baba AB, Mushtaq I, Farooq M, Yousuf T, Dar SB, Parveen S, Hassan R, Mohammad F, Qassim I, Bhat A, Ali S, Zargar MH, Afroze D. JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies. Front Pharmacol 2022; 13:821344. [PMID: 35401182 PMCID: PMC8987160 DOI: 10.3389/fphar.2022.821344] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
JAK/STAT signaling pathway is one of the important regulatory signaling cascades for the myriad of cellular processes initiated by various types of ligands such as growth factors, hormones, and cytokines. The physiological processes regulated by JAK/STAT signaling are immune regulation, cell proliferation, cell survival, apoptosis and hematopoiesis of myeloid and non-myeloid cells. Dysregulation of JAK/STAT signaling is reported in various immunological disorders, hematological and other solid malignancies through various oncogenic activation mutations in receptors, downstream mediators, and associated transcriptional factors such as STATs. STATs typically have a dual role when explored in the context of cancer. While several members of the STAT family are involved in malignancies, however, a few members which include STAT3 and STAT5 are linked to tumor initiation and progression. Other STAT members such as STAT1 and STAT2 are pivotal for antitumor defense and maintenance of an effective and long-term immune response through evolutionarily conserved programs. The effects of JAK/STAT signaling and the persistent activation of STATs in tumor cell survival; proliferation and invasion have made the JAK/STAT pathway an ideal target for drug development and cancer therapy. Therefore, understanding the intricate JAK/STAT signaling in the pathogenesis of solid malignancies needs extensive research. A better understanding of the functionally redundant roles of JAKs and STATs may provide a rationale for improving existing cancer therapies which have deleterious effects on normal cells and to identifying novel targets for therapeutic intervention in solid malignancies.
Collapse
|
5
|
Zhang HJ, Chen G, Chen SW, Fu ZW, Zhou HF, Feng ZB, Mo JX, Li CB, Liu J. Overexpression of cyclin-dependent kinase 1 in esophageal squamous cell carcinoma and its clinical significance. FEBS Open Bio 2021; 11:3126-3141. [PMID: 34586751 PMCID: PMC8564100 DOI: 10.1002/2211-5463.13306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/04/2021] [Accepted: 09/28/2021] [Indexed: 12/25/2022] Open
Abstract
Cyclin‐dependent kinase 1 (CDK1) plays a significant role in certain malignancies. However, it remains unclear whether CDK1 plays a role in esophageal squamous cell carcinoma (ESCC). The aim of this study was to analyze the expression and clinical value of CDK1 in ESCC. CDK1 protein in 151 ESCC tissues and 138 normal esophageal tissues was detected by immunohistochemistry. RNA‐seq of eight pairs of ESCC and adjacent esophageal specimens was performed to evaluate the levels of CDK1 mRNA. Microarray and external RNA‐seq data from 664 cases of ESCC and 1733 cases of control tissues were used to verify the difference in CDK1 expression between the two groups. A comprehensive analysis of all data was performed to evaluate the difference in CDK1 between ESCC tissues and control tissues. Further, functional enrichment analyses were performed based on differentially expressed genes (DEGs) of ESCC and co‐expressed genes (CEGs) of CDK1. In addition, a lncRNA‐miRNA‐CDK1 network was constructed. The expression of CDK1 protein was obviously increased in ESCC tissues (3.540 ± 2.923 vs. 1.040 ± 1.632, P < 0.001). RNA‐seq indicated that the mRNA level of CDK1 was also highly expressed in ESCC tissues (5.261 ± 0.703 vs. 2.229 ± 1.161, P < 0.0001). Comprehensive analysis revealed consistent up‐regulation of CDK1 (SMD = 1.41; 95% CI 1.00–1.83). Further, functional enrichment analyses revealed that the functions of these genes were mainly concentrated in the cell cycle. A triple regulatory network of PVT1‐hsa‐miR‐145‐5p/hsa‐miR‐30c‐5p‐CDK1 was constructed using in silico analysis. In summary, overexpression of CDK1 is closely related to ESCC tumorigenesis.
Collapse
Affiliation(s)
- Han-Jie Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shang-Wei Chen
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zong-Wang Fu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hua-Fu Zhou
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhen-Bo Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Xian Mo
- Department of Cardio-Thoracic Surgery, The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou, China.,Wuzhou Gongren Hospital, Wuzhou, China
| | - Chang-Bo Li
- Department of Cardio-Thoracic Surgery, The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou, China.,Wuzhou Gongren Hospital, Wuzhou, China
| | - Jun Liu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Jia Z, Xie Y, Wu H, Wang Z, Li A, Li Z, Yang Z, Zhang Z, Xing Z, Zhang X. Phlorizin from sweet tea inhibits the progress of esophageal cancer by antagonizing the JAK2/STAT3 signaling pathway. Oncol Rep 2021; 46:137. [PMID: 34036398 PMCID: PMC8165578 DOI: 10.3892/or.2021.8088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 04/15/2021] [Indexed: 12/24/2022] Open
Abstract
Phlorizin, an important member of the dihydrochalcone family, has been widely used as a Chinese Traditional Medicine for treatment of numerous diseases. The present study aimed to investigate the potential therapeutic effects of phlorizin on esophageal cancer. Phlorizin, extracted from sweet tea, was used to treat esophageal cancer cells. Cell proliferation, migration and invasion were determined using Cell Counting Kit-8 and colony formation assays, and wound healing and Transwell assays, respectively. RNA sequencing and bioinformatics analysis was used to investigate the potential mechanism of phlorizin in the development of esophageal cancer. Fluorescent staining and flow cytometry was used to measure the level of apoptosis. The expression level of the proteins, P62/SQSTM1 and LC3 І/II, and the effect of phlorizin on the JAK2/STAT3 signaling pathway was detected using western blot analysis. The results demonstrated that phlorizin could inhibit cell proliferation, migration and invasion. Bioinformatics analysis showed that phlorizin might be involved in pleiotropic effects, such as the ‘JAK/STAT signaling pathway’ (hsa04630), ‘MAPK signaling pathway’(hsa04010) and ‘apoptosis’ (hsa04210). It was also confirmed that phlorizin promoted apoptosis and inhibited autophagy in the esophageal cancer cells. Notably, phlorizin might inhibit the proteins in the JAK/STAT signaling pathway, which would affect cancer cells. Taken together, the present data showed that phlorizin inhibited the progression of esophageal cancer by antagonizing the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhenxian Jia
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Yuning Xie
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Hongjiao Wu
- College of Life Science, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Zhuo Wang
- School of Pharmacy, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Ze Li
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Zhenbang Yang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Zhi Zhang
- Department of Oncology, Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, Hebei 063000, P.R. China
| | - Zhaobin Xing
- College of Life Science, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| |
Collapse
|
7
|
Song Y, Wang X, Wang F, Peng X, Li P, Liu S, Zhang D. Identification of four genes and biological characteristics of esophageal squamous cell carcinoma by integrated bioinformatics analysis. Cancer Cell Int 2021; 21:123. [PMID: 33602210 PMCID: PMC7890804 DOI: 10.1186/s12935-021-01814-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) has become one of the most serious diseases affecting populations worldwide and is the primary subtype of esophageal cancer (EC). However, the molecular mechanisms governing the development of ESCC have not been fully elucidated. METHODS The robust rank aggregation method was performed to identify the differentially expressed genes (DEGs) in six datasets (GSE17351, GSE20347, GSE23400, GSE26886, GSE38129 and GSE77861) from the Gene Expression Omnibus (GEO). The Search Tool for the Retrieval of Interacting Genes (STRING) database was utilized to extract four hub genes from the protein-protein interaction (PPI) network. Module analysis and disease free survival analysis of the four hub genes were performed by Cytoscape and GEPIA. The expression of hub genes was analyzed by GEPIA and the Oncomine database and verified by real-time quantitative PCR (qRT-PCR). RESULTS In total, 720 DEGs were identified in the present study; these genes consisted of 302 upregulated genes and 418 downregulated genes that were significantly enriched in the cellular component of the extracellular matrix part followed by the biological process of the cell cycle phase and nuclear division. The primary enriched pathways were hsa04110:Cell cycle and hsa03030:DNA replication. Four hub genes were screened out, namely, SPP1, MMP12, COL10A1 and COL5A2. These hub genes all exhibited notably increased expression in ESCC samples compared with normal samples, and ESCC patients with upregulation of all four hub genes exhibited worse disease free survival. CONCLUSIONS SPP1, MMP12, COL10A1 and COL5A2 may participate in the tumorigenesis of ESCC and demonstrate the potential to serve as molecular biomarkers in the early diagnosis of ESCC. This study may help to elucidate the molecular mechanisms governing ESCC and facilitate the selection of targets for early treatment and diagnosis.
Collapse
Affiliation(s)
- Yexun Song
- Department of Otolaryngology-Head Neck Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China.,Department of Otolaryngology-Head Neck Surgery, The Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Xianyao Wang
- Department of Otolaryngology-Head Neck Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China.,Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Fengjun Wang
- Department of Otolaryngology-Head Neck Surgery, The Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Xiaowei Peng
- Department of Oncology Plastic Surgery, Hunan Province Cancer Hospital, Changsha, 410007, Hunan, China
| | - Peiyu Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China.,Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, 410013, Hunan, China
| | - Shaojun Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China.,Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, 410013, Hunan, China
| | - Decai Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China. .,Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, 410013, Hunan, China.
| |
Collapse
|
8
|
Hu W, Chen Z, Chen J, Cai D, Chen C, Fang T. LOC441178 Overexpression Inhibits the Proliferation and Migration of Esophageal Carcinoma Cells via Methylation of miR-182. Onco Targets Ther 2020; 13:11253-11263. [PMID: 33173314 PMCID: PMC7648570 DOI: 10.2147/ott.s271711] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/05/2020] [Indexed: 02/05/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have been shown to play an important role in the development and progression of esophageal carcinoma (EC). Recently, lncRNA LOC441178 was shown to be dysregulated in many cancer types; however, the role of LOC441178 in EC remains unclear. Materials and Methods Flow cytometry, transwell and wound healing assays were used to measure the apoptosis and migration in esophageal squamous cell carcinoma (ESCC) cells. RT-qPCR was used to detect the level of miR-182 in LOC441178-overexpressed EC cells. In addition, DNA methylation status of miR-182 promoter in LOC441178-overexpressed ESCC cells was detected by methylation-specific PCR (MSP) and bisulfite sequencing PCR. Results In this study, we found that LOC441178 negatively regulated miR-182 expression in ESCC cells. In addition, overexpression of LOC441178 inhibited the proliferation and migration and induced apoptosis in ESCC cells via downregulation of miR-182. Moreover, overexpression of LOC441178 markedly inhibited the phosphorylation of Akt and phosphorylation FOXO3a and increased the expression of FOXO3a in ESCC cells via downregulation of miR-182. Mechanistically, LOC441178 overexpression epigenetically suppressed miR-182 expression via DNA methylation. In vivo experiments revealed that overexpression of LOC441178 inhibited ESCC tumor growth in mouse xenograft model. Conclusion Collectively, our data suggested that LOC441178 overexpression epigenetically inhibited tumorigenesis of ESCC via DNA methylation of miR-182. These data indicated that the LOC441178/miR-182 axis might represent a novel therapeutic option for the treatment of ESCC.
Collapse
Affiliation(s)
- Weitao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| | - Zongchi Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| | - Jiangmu Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| | - Daxing Cai
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| | - Congjie Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| |
Collapse
|
9
|
Dharmawansa KS, Hoskin DW, Rupasinghe HPV. Chemopreventive Effect of Dietary Anthocyanins against Gastrointestinal Cancers: A Review of Recent Advances and Perspectives. Int J Mol Sci 2020; 21:ijms21186555. [PMID: 32911639 PMCID: PMC7554903 DOI: 10.3390/ijms21186555] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
Abstract
Anthocyanins are a group of dietary polyphenols, abundant mainly in fruits and their products. Dietary interventions of anthocyanins are being studied extensively related to the prevention of gastrointestinal (GI) cancer, among many other chronic disorders. This review summarizes the hereditary and non-hereditary characteristics of GI cancers, chemistry, and bioavailability of anthocyanins, and the most recent findings of anthocyanin in GI cancer prevention through modulating cellular signaling pathways. GI cancer-preventive attributes of anthocyanins are primarily due to their antioxidative, anti-inflammatory, and anti-proliferative properties, and their ability to regulate gene expression and metabolic pathways, as well as induce the apoptosis of cancer cells.
Collapse
Affiliation(s)
- K.V. Surangi Dharmawansa
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada;
| | - David W. Hoskin
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Microbiology and Immunology, and Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - H. P. Vasantha Rupasinghe
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Correspondence: ; Tel.: +1-902-893-6623
| |
Collapse
|
10
|
Zhou C, Fan N, Liu F, Fang N, Plum PS, Thieme R, Gockel I, Gromnitza S, Hillmer AM, Chon SH, Schlösser HA, Bruns CJ, Zhao Y. Linking Cancer Stem Cell Plasticity to Therapeutic Resistance-Mechanism and Novel Therapeutic Strategies in Esophageal Cancer. Cells 2020; 9:cells9061481. [PMID: 32560537 PMCID: PMC7349233 DOI: 10.3390/cells9061481] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Esophageal cancer (EC) is an aggressive form of cancer, including squamous cell carcinoma (ESCC) and adenocarcinoma (EAC) as two predominant histological subtypes. Accumulating evidence supports the existence of cancer stem cells (CSCs) able to initiate and maintain EAC or ESCC. In this review, we aim to collect the current evidence on CSCs in esophageal cancer, including the biomarkers/characterization strategies of CSCs, heterogeneity of CSCs, and the key signaling pathways (Wnt/β-catenin, Notch, Hedgehog, YAP, JAK/STAT3) in modulating CSCs during esophageal cancer progression. Exploring the molecular mechanisms of therapy resistance in EC highlights DNA damage response (DDR), metabolic reprogramming, epithelial mesenchymal transition (EMT), and the role of the crosstalk of CSCs and their niche in the tumor progression. According to these molecular findings, potential therapeutic implications of targeting esophageal CSCs may provide novel strategies for the clinical management of esophageal cancer.
Collapse
Affiliation(s)
- Chenghui Zhou
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
| | - Ningbo Fan
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
| | - Fanyu Liu
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
- Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Nan Fang
- Singleron Biotechnologies, Yaogu Avenue 11, Nanjing 210000, China;
| | - Patrick S. Plum
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (S.G.); (A.M.H.)
| | - René Thieme
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 4107 Leipzig, Germany; (R.T.); (I.G.)
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 4107 Leipzig, Germany; (R.T.); (I.G.)
| | - Sascha Gromnitza
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (S.G.); (A.M.H.)
| | - Axel M. Hillmer
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (S.G.); (A.M.H.)
- Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
| | - Hans A. Schlösser
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
- Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Christiane J. Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
- Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany; (C.Z.); (N.F.); (F.L.); (P.S.P.); (S.-H.C.); (H.A.S.); (C.J.B.)
- Correspondence: ; Tel.: +49-221-4783-0601; Fax: +49-221-4783-0664
| |
Collapse
|
11
|
Jian J, Li S, Liu LZ, Zhen L, Yao L, Gan LH, Huang YQ, Fang N. XPD inhibits cell growth and invasion and enhances chemosensitivity in esophageal squamous cell carcinoma by regulating the PI3K/AKT signaling pathway. Int J Mol Med 2020; 46:201-210. [PMID: 32377720 PMCID: PMC7255471 DOI: 10.3892/ijmm.2020.4593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a lethal disease due to its high aggressiveness. The aim of the present study was to investigate the role of xeroderma pigmentosum complementation group D (XPD) in the growth and invasion of ESCC and to elucidate the potential underlying molecular mechanisms. Western blot analysis and RT-qPCR were used to detect the expression level of XPD in ESCC tissue samples and adjacent normal esophageal tissue samples. The pEGFP-N2/XPD plasmid was transfected into human ESCC cell lines (EC9706 and EC109). The proliferation, apoptosis, migration and invasion of EC9706 or EC109 cells were assessed following transfection with the XPD overexpression plasmid. The chemosensitivity of EC9706 or EC109 cells to cisplatin or fluorouracil was evaluated by CCK-8 assay. The expression levels of phosphoinositide 3-kinase (PI3K)/AKT, nuclear factor (NF)-κB, Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK) signaling pathway-related genes were detected by RT-qPCR and western blot analysis. The results demonstrated that the expression level of XPD was markedly lower in ESCC tissue samples than in adjacent normal esophageal tissue samples. The pEGFP-N2/XPD plasmid was successfully transfected into EC9706 or EC109 cells, inducing XPD overexpression. A High XPD expression markedly suppressed cell proliferation, migration and invasion, and increased the apoptotic rate of EC9706 and EC109 cells. Furthermore, the overexpression of XPD significantly increased the chemosensitivity of EC9706 and EC109 cells to cisplatin or fluorouracil. Following XPD overexpression, the expression levels of PI3K, p-AKT, c-Myc, Cyclin D1, Bcl-2, vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP)-9 were markedly downregulated, while the expression level of p21 was markedly upregulated. On the whole, the findings of the present study demonstrate that XPD inhibits the growth and invasion of EC9706 and EC109 cells, whilst also enhancing the chemosensitivity of EC9706 and EC109 cells to cisplatin or fluorouracil by regulating the PI3K/AKT signaling pathway. XPD may thus be an underlying target for ESCC treatment and drug resistance.
Collapse
Affiliation(s)
- Jie Jian
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Shuang Li
- Department of Geriatrics and General Medicine, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Li-Zhen Liu
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Li Zhen
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Ling Yao
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Li-Hong Gan
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Ya-Qing Huang
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Nian Fang
- Department of Gastroenterology, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| |
Collapse
|
12
|
Guo X, Yan Z, Zhang G, Wang X, Pan Y, Huang M. STIP1 Regulates Proliferation and Migration of Lung Adenocarcinoma Through JAK2/STAT3 Signaling Pathway. Cancer Manag Res 2019; 11:10061-10072. [PMID: 31819639 PMCID: PMC6890180 DOI: 10.2147/cmar.s233758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/23/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Recent studies have shown that STIP1 is associated with proliferation and migration in numerous types of tumors; however, the role of STIP1 in lung adenocarcinoma is still poorly understood. Therefore, the aim of this study was to evaluate the role of STIP1 in lung adenocarcinoma, in vitro and in vivo. Methods The expression of STIP1 in lung adenocarcinoma was assessed by immunohistochemistry, RT-qPCR, and Western blot. The effects of STIP1 on the proliferation of lung adenocarcinoma cells were detected by the cell counting kit-8 assay; the effect of STIP1 on adhesion of lung adenocarcinoma cells was detected by Giemsa staining, while the cell scratch and Transwell assays were employed to examine the effect of STIP1 on the migratory ability of lung adenocarcinoma cells. Finally, apoptosis was evaluated by Hoechst staining and flow cytometry. Results The expression level of STIP1 in lung adenocarcinoma tissue was significantly higher than that in adjacent normal tissue (P<0.05). Compared with that in nontransfected controls, cell proliferation, adhesion, and migration, as well as vimentin protein expression and levels of phosphorylated JAK2/STAT3, were significantly decreased (P<0.05) in A549 lung adenocarcinoma cells transfected with STIP1 shRNA, whereas E-cadherin protein expression and rates of apoptosis were significantly increased in these cells (P< 0.05). Conclusion Elevated expression of STIP1 in lung adenocarcinoma may enhance the proliferative, adhesive, and migratory ability, and reduce the apoptosis of lung adenocarcinoma cells through the JAK2/STAT3 signaling pathway and epithelial-mesenchymal transition (EMT), thereby promoting the recurrence and metastatic potential of this cancer. The results indicate that STIP1 may be an effective therapeutic target for the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiangjun Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China.,Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiang su, People's Republic of China
| | - Zhongyi Yan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiang su, People's Republic of China
| | - Gongming Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiang su, People's Republic of China
| | - Xiang Wang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiang su, People's Republic of China
| | - Yun Pan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiang su, People's Republic of China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Wu J, Yang C, Liu J, Chen J, Huang C, Wang J, Liang Z, Wen L, Yi JE, Yuan Z. Betulinic Acid Attenuates T-2-Toxin-Induced Testis Oxidative Damage Through Regulation of the JAK2/STAT3 Signaling Pathway in Mice. Biomolecules 2019; 9:biom9120787. [PMID: 31779213 PMCID: PMC6995557 DOI: 10.3390/biom9120787] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/20/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023] Open
Abstract
T-2 toxin is one of the most toxic type A trichothecene mycotoxins in nature, and it exhibits reproductive toxicity. Betulinic acid (BA) is a natural pentacyclic triterpene compound found in species of Betula, and it has been reported to have antioxidant activity. The aim of the present study was to investigate the protective effect of BA on T-2-toxin-induced testicular injury in mice and explore its molecular mechanism. Sixty adult male mice were randomly divided into groups. The mice were pretreated orally with BA (0.25, 0.5, and 1.0 mg/kg) daily for 14 days, and the T-2 toxin (4 mg/kg body weight) was administered via intraperitoneal injection to induce oxidative stress after the last administration of BA. BA pretreatment significantly increased the secreted levels of testosterone and sperm motility. Moreover, BA pretreatment significantly increased the total antioxidant capacity (T-AOC), the activity of SOD and CAT, and the content of GSH, and it reduced the content of MDA. Furthermore, BA relieved testicular injury and reduced the number of apoptotic cells, and it significantly decreased the protein expression of Janus kinase 2 (JAK2), signal transducers and activators of transcription 3 (STAT3), caspsae-3, and Bcl-2-associated X protein (Bax). BA also increased the expression of B-cell lymphoma-2 (Bcl-2). We suggest that BA reduced the oxidative damage induced by T-2 toxin, and that these protective effects may be partially mediated by the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jing Wu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Chenglin Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Juan Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Jiaxin Chen
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Chao Huang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Ji Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Zengenni Liang
- Department of Hunan Agricultural Product Processing Institute, Changsha 410128, China;
| | - Lixin Wen
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
| | - Jin-e Yi
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
- Correspondence: (J.-e.Y.); (Z.Y.)
| | - Zhihang Yuan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China (C.Y.); (J.L.); (J.C.); (C.H.); (L.W.)
- Correspondence: (J.-e.Y.); (Z.Y.)
| |
Collapse
|