1
|
杨 晶, 殷 丽, 段 婷, 牛 民, 何 震, 陈 心, 张 小, 李 静, 耿 志, 左 芦. [High expression of ATP5A1 in gastric carcinoma is correlated with a poor prognosis and enhanced glucose metabolism in tumor cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:974-980. [PMID: 38862456 PMCID: PMC11166711 DOI: 10.12122/j.issn.1673-4254.2024.05.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To analyze the expression level of ATP5A1 in gastric carcinoma and its influence on the prognosis of the patients and glucose metabolism in the tumor cells. METHODS We retrospectively analyzed the data of 115 patients undergoing radical resection of gastric carcinoma in our hospital from February, 2013 to November, 2016. ATP5A1 expression in the surgical specimens were detected using immunohistochemistry, and the long-term prognosis of the patients with high (n=58) and low ATP5A1 expression (n=57) were analyzed. In gastric carcinoma MGC803 cells, the effects of lentivirus-mediated ATP5A1 knockdown or overexpression on glucose metabolism were investigated. We also observed the growth and glucose metabolism of xenografts derived from MGC803 cells with ATP5A1 knockdown or overexpression in nude mice. RESULTS ATP5A1 was significantly overexpressed in gastric carcinoma tissues in close correlation with blood CEA and CA19-9 levels, pathological grade, T stage and N stage (P < 0.05). ATP5A1 overexpression was an independent risk factor for a significantly lowered 5-year survival rate of patients with gastric carcinoma (P < 0.05). ROC curve analysis demonstrated the predictive value of high ATP5A1 expression for the patients'prognosis (P < 0.001). In MGC803 cells, ATP5A1 overexpression significantly upregulated cellular glucose uptake and lactate production and increased the protein levels of HK2, PFK1, and LDHA (P < 0.05), while ATP5A1 knockdown produced the opposite changes (P < 0.05). In the tumor-bearing mice, overexpression of ATP5A1 increased glucose metabolism of the tumor cells and promoted tumor growth (P < 0.05). Overexpression of ATP5A1 promoted the expressions of p-JNK and p-JUN in MGC803 cells (P < 0.05), and the JNK inhibitor SP600125 significantly inhibited the enhancement of cellular glucose metabolism induced by ATP5A1 overexpression (P < 0.05). CONCLUSION High ATP5A1 expression in gastric cancer is associated a poor long-term prognosis of the patients, and its effect is mediated at least partly by promoting glucose metabolism of the cells through the JNK/JUN pathway.
Collapse
|
2
|
Patrad E, Khalighfard S, Amiriani T, Khori V, Alizadeh AM. Molecular mechanisms underlying the action of carcinogens in gastric cancer with a glimpse into targeted therapy. Cell Oncol 2022; 45:1073-1117. [PMID: 36149600 DOI: 10.1007/s13402-022-00715-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer imposes a substantial global health burden despite its overall incidence decrease. A broad spectrum of inherited, environmental and infectious factors contributes to the development of gastric cancer. A profound understanding of the molecular underpinnings of gastric cancer has lagged compared to several other tumors with similar incidence and morbidity rates, owing to our limited knowledge of the role of carcinogens in this malignancy. The International Agency for Research on Cancer (IARC) has classified gastric carcinogenic agents into four groups based on scientific evidence from human and experimental animal studies. This review aims to explore the potential comprehensive molecular and biological impacts of carcinogens on gastric cancer development and their interactions and interferences with various cellular signaling pathways. CONCLUSIONS In this review, we highlight recent clinical trial data reported in the literature dealing with different ways to target various carcinogens in gastric cancer. Moreover, we touch upon other multidisciplinary therapeutic approaches such as surgery, adjuvant and neoadjuvant chemotherapy. Rational clinical trials focusing on identifying suitable patient populations are imperative to the success of single-agent therapeutics. Novel insights regarding signaling pathways that regulate gastric cancer can potentially improve treatment responses to targeted therapy alone or in combination with other/conventional treatments. Preventive strategies such as control of H. pylori infection through eradication or immunization as well as dietary habit and lifestyle changes may reduce the incidence of this multifactorial disease, especially in high prevalence areas. Further in-depth understanding of the molecular mechanisms involved in the role of carcinogenic agents in gastric cancer development may offer valuable information and update state-of-the-art resources for physicians and researchers to explore novel ways to combat this disease, from bench to bedside. A schematic outlining of the interaction between gastric carcinogenic agents and intracellular pathways in gastric cancer H. pylori stimulates multiple intracellular pathways, including PI3K/AKT, NF-κB, Wnt, Shh, Ras/Raf, c-MET, and JAK/STAT, leading to epithelial cell proliferation and differentiation, apoptosis, survival, motility, and inflammatory cytokine release. EBV can stimulate intracellular pathways such as the PI3K/Akt, RAS/RAF, JAK/STAT, Notch, TGF-β, and NF-κB, leading to cell survival and motility, proliferation, invasion, metastasis, and the transcription of anti-apoptotic genes and pro-inflammatory cytokines. Nicotine and alcohol can lead to angiogenesis, metastasis, survival, proliferation, pro-inflammatory, migration, and chemotactic by stimulating various intracellular signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, ROS, and JAK/STAT. Processed meat contains numerous carcinogenic compounds that affect multiple intracellular pathways such as sGC/cGMP, p38 MAPK, ERK, and PI3K/AKT, leading to anti-apoptosis, angiogenesis, metastasis, inflammatory responses, proliferation, and invasion. Lead compounds may interact with multiple signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, DNA methylation-dependent, and epigenetic-dependent, leading to tumorigenesis, carcinogenesis, malignancy, angiogenesis, DNA hypermethylation, cell survival, and cell proliferation. Stimulating signaling pathways such as PI3K/Akt, RAS/RAF, JAK/STAT, WNT, TGF-β, EGF, FGFR2, and E-cadherin through UV ionizing radiation leads to cell survival, proliferation, and immortalization in gastric cancer. The consequence of PI3K/AKT, NF-κB, Ras/Raf, ROS, JAK/STAT, and WNT signaling stimulation by the carcinogenic component of Pickled vegetables and salted fish is the Warburg effect, tumorigenesis, angiogenesis, proliferation, inflammatory response, and migration.
Collapse
Affiliation(s)
- Elham Patrad
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalighfard
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
The FOXO family of transcription factors: key molecular players in gastric cancer. J Mol Med (Berl) 2022; 100:997-1015. [PMID: 35680690 DOI: 10.1007/s00109-022-02219-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
Gastric cancer (GC) is the fifth most frequently diagnosed cancer worldwide and the third leading cause of cancer-related death with an oncological origin. Despite its decline in incidence and mortality in recent years, GC remains a global public problem that seriously threatens patients' health and lives. The forkhead box O proteins (FOXOs) are a family of evolutionarily conserved transcription factors (TFs) with crucial roles in cell fate decisions. In mammals, the FOXO family consists of four members FOXO1, 3a, 4, and 6. FOXOs play crucial roles in a variety of biological processes, such as development, metabolism, and stem cell maintenance, by regulating the expression of their target genes in space and time. An accumulating amount of evidence has shown that the dysregulation of FOXOs is involved in GC progression by affecting multiple cellular processes, including proliferation, apoptosis, invasion, metastasis, cell cycle progression, carcinogenesis, and resistance to chemotherapeutic drugs. In this review, we systematically summarize the recent findings on the regulatory mechanisms of FOXO family expression and activity and elucidate its roles in GC progression. Moreover, we also highlight the clinical implications of FOXOs in GC treatment.
Collapse
|
4
|
Bae WJ, Woo KJ, Ahn JM, Yang CM, Kim YS, Kim S, Lee D. miR-4742-5p promotes invasiveness of gastric cancer via targeting Rab43: An in vitro study. Biochem Biophys Res Commun 2022; 613:180-186. [PMID: 35597125 DOI: 10.1016/j.bbrc.2022.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/12/2022] [Indexed: 11/02/2022]
Abstract
miRNA (miR)-4742-5p is a recently identified microRNA regarding progression and metastasis in gastric cancer (GC). However, the biological function of this novel miRNA is largely unknown. We identified that the miR-4742-5p expression level was variably increased in GC cell lines. Suppression of miR-4742-5p using miR-inhibitor reduced the proliferation, migration, and invasion of GC cells with high miR-4742-5p expression, whereas overexpression of miR-4742-5p-mimic enhanced the aforementioned properties in GC cells with low miR-4742-5p expression. miR-4742-5p expression induced the decreases of Zo-1 and E-cadherin expression as well as the increases of vimentin and N-cadherin expression, leading to epithelial-mesenchymal transition (EMT) of cancer cells. RNA sequencing results indicated Ras-related GTP-binding protein 43 (Rab43) as a potential target gene. We identified that the expression of Rab43 is associated with activation of AKT and nuclear factor-kappa B (NF-κB) which are key oncogenic pathways in cancer cells. Our results demonstrate a new component in GC progression, promising a potential therapeutic strategy.
Collapse
Affiliation(s)
- Won Jung Bae
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Kyoung-Jin Woo
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Ji Mi Ahn
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Chan-Mo Yang
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - You-Sun Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea; Department of Biochemistry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea.
| | - Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
5
|
Chen YH, Li CL, Chen WJ, Liu J, Wu HT. Diverse roles of FOXO family members in gastric cancer. World J Gastrointest Oncol 2021; 13:1367-1382. [PMID: 34721771 PMCID: PMC8529928 DOI: 10.4251/wjgo.v13.i10.1367] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/06/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is the fifth most diagnosed cancer and the third leading cause of cancer-related death worldwide. Although progress has been made in diagnosis, surgical resection, systemic chemotherapy, and immunotherapy, patients with GC still have a poor prognosis. The overall 5-year survival rate in patients with advanced GC is less than 5%. The FOXO subfamily, of the forkhead box family of transcription factors, consists of four members, FOXO1, FOXO3, FOXO4, and FOXO6. This subfamily plays an important role in many cellular processes, such as cell cycle, cell growth, apoptosis, autophagy, stress resistance, protection from aggregate toxicity, DNA repair, tumor suppression, and metabolism, in both normal tissue and malignant tumors. Various studies support a role for FOXOs as tumor suppressors based on their ability to inhibit angiogenesis and metastasis, and promote apoptosis, yet several other studies have shown that FOXOs might also promote tumor progression in certain circumstances. To elucidate the diverse roles of FOXOs in GC, this article systematically reviews the cellular functions of FOXOs in GC to determine potential therapeutic targets and treatment strategies for patients with GC.
Collapse
Affiliation(s)
- Yu-Han Chen
- Department of Clinical Medicine, Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Changjiang Scholar's Laboratory, Department of Physiology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Chun-Lan Li
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Changjiang Scholar's Laboratory, Department of Physiology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Wen-Jia Chen
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Changjiang Scholar's Laboratory, Department of Physiology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jing Liu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Changjiang Scholar's Laboratory, Department of Physiology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Hua-Tao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
6
|
Zhu L, Chen Y, Liu J, Nie K, Xiao Y, Yu H. MicroRNA-629 promotes the tumorigenesis of non-small-cell lung cancer by targeting FOXO1 and activating PI3K/AKT pathway. Cancer Biomark 2021; 29:347-357. [PMID: 32716350 DOI: 10.3233/cbm-201685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE MicroRNA-629 (miR-629) has been found to play an important role in the pathogenesis of human cancers. However, the function of miR-629 is still unknown in non-small-cell lung cancer (NSCLC). The purpose of this study is to preliminarily elucidate the regulatory mechanism of miR-629 in NSCLC. MATERIALS AND METHODS The mRNA and protein expression was measured by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. The function of miR-629 was investigated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) and Transwell assays. The relationship between miR-629 and FOXO1 was confirmed by dual luciferase assay. RESULTS MiR-629 was upregulated in NSCLC tissues and cells. High expression of miR-629 predicted poor prognosis in patients with NSCLC. Moreover, miR-629 promoted cell proliferation, migration and invasion in NSCLC cells. In addition, FOXO1 was confirmed as a direct target of miR-629 in NSCLC. Furthermore, knockdown of FOXO1 also promoted proliferation, migration and invasion of NSCLC cells. More importantly, overexpression of FOXO1 weakened the carcinogenesis of miR-629 in NSCLC. Besides that, miR-629 promoted EMT and activated the PI3K/AKT pathway in NSCLC. CONCLUSIONS MiR-629 promotes the progression of NSCLC by targeting FOXO1 and regulating EMT/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Radiology, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China.,Department of Radiology, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yinan Chen
- Department of Radiology, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China.,Department of Radiology, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Jing Liu
- Department of Radiology, Dongfang Hospital Affiliated to Shanghai Tongji University, Shanghai, China
| | - Kai Nie
- Department of Radiology, Shanghai Changzheng Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Yongxin Xiao
- Department of Radiology, Shanghai Changzheng Hospital Affiliated to the Second Military Medical University, Shanghai, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
7
|
Zhou J, Wu L, Li W, Xu X, Ju F, Yu S, Guo J, Li G, Shi J, Zhou S. Long Noncoding RNA LINC01485 Promotes Tumor Growth and Migration via Inhibiting EGFR Ubiquitination and Activating EGFR/Akt Signaling in Gastric Cancer. Onco Targets Ther 2020; 13:8413-8425. [PMID: 32904620 PMCID: PMC7457555 DOI: 10.2147/ott.s257151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background Although several long non-coding RNAs (lncRNAs) have been found to be involved in gastric cancer tumorigenesis, the more comprehensive contributions of lncRNAs to gastric cancer require further investigation. Here, we identify a cytoplasmic lncRNA, LINC01485, which promotes tumor growth and migration in gastric cancer. Materials and Methods Microarray and computational analysis were utilized to identify differential expression in LINC01485 and EGFR. Real-time PCR and Western blotting assays were used to confirm the expression of LINC01485 and EGFR in gastric cancer cells. Cell proliferation, wound-healing and transwell assays were performed to measure cell growth, migration and invasion. Immunoprecipitation, RNA pull-down, and RNA fluorescence in situ hybridization (RNA-FISH) assays were used to test the interaction of c-Cbl with LINC01485 and EGFR. Furthermore, tumor xenograft in nude mice was performed to test tumor growth in vivo. Results LINC01485 was upregulated and associated with tumor size, lymphatic metastasis and advanced pathological stage in gastric cancer. LINC01485 promoted gastric cancer cell proliferation, migration and invasion in vitro and in vivo. Furthermore, LINC01485 levels were positively correlated with EGFR expression in gastric cancer tissues and significantly increased the expression and phosphorylation (Tyr1045) of EGFR in gastric cancer cells. Mechanistically, LINC01485 competes with c-Cbl for binding to phosphorylated Tyr1045 site of EGFR, thus interfering with c-Cbl-mediated ubiquitination and subsequent degradation of EGFR. Conclusion LINC01485 promoted EGFR stabilization and activation of EGFR/Akt signaling in gastric cancer. Our findings illustrate the diversity of cytoplasmic lncRNAs in signal transduction and highlight the important roles of lncRNAs in gastric cancer.
Collapse
Affiliation(s)
- Jianping Zhou
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Lulu Wu
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Weiling Li
- Department of Obstetrics and Gynecology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Xiao Xu
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Feng Ju
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Shao Yu
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Jianfeng Guo
- Department of B-Ultrasound Room, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Gang Li
- Department of B-Ultrasound Room, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Jun Shi
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| | - Sujun Zhou
- Department of Gastrointestinal Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, People's Republic of China
| |
Collapse
|
8
|
Liang CY, Huang ZG, Tang ZQ, Xiao XL, Zeng JJ, Feng ZB. FOXO1 and hsa-microRNA-204-5p affect the biologic behavior of MDA-MB-231 breast cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1146-1158. [PMID: 32509089 PMCID: PMC7270695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
RNA molecules and targeting microRNA (miRNA) have been reported as novel focuses in recent research on breast cancer. This study aimed to probe the expression of FOXO1 in the MDA-MB-231 cell line and to explore the target effects of FOXO1 with hsa-microRNA-204-5p (miR-204) on the biologic behavior of MDA-MB-231 cells. The expression of FOXO1 mRNA and protein in MDA-MB-231 cells were derived and verified from the public databases, literature, and experimental assays, then the downregulation of FOXO1 was confirmed in the MDA-MB-231 cell line. The target binding of FOXO1 and miR-204 was predicted by miRWalk and confirmed by luciferase reporter assays. MiR-204 targeted the 3' untranslated region of FOXO1 and reduced FOXO1 expression in miR-204-transfected cells, resulting in cell growth amplification but inhibition of cell migration and apoptosis, which were assessed using the MTT method, wound healing assays, and flow cytometry, respectively. The protein levels of serine-threonine kinase (AKT), c-jun N-terminal kinase (JNK), extracellular regulatory protein kinase (ERK), and the phosphorylated protein kinases (P-AKT, P-JNK, and P-ERK) were measured by western blot. It was found that AKT, JNK, and ERK remained constant, but P-AKT, P-JNK, and P-ERK were upregulated after miR-204 transfection. In summary, the expression of FOXO1 was downregulated in MDA-MB-231 cells; and the target binding of miR-204 and FOXO1 affected phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK) signal pathways, leading to different alterations of cellular activity in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Chang-Yu Liang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, P. R. China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, P. R. China
| | - Zhong-Qing Tang
- Department of Pathology, Gongren Hospital of WuzhouWuzhou, Guangxi, P. R. China
| | - Xiao-Ling Xiao
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, P. R. China
| | - Jing-Jing Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, P. R. China
| | - Zhen-Bo Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi, P. R. China
| |
Collapse
|
9
|
Huang Q, Zhang J, Peng J, Zhang Y, Wang L, Wu J, Ye L, Fang C. Effect of baicalin on proliferation and apoptosis in pancreatic cancer cells. Am J Transl Res 2019; 11:5645-5654. [PMID: 31632536 PMCID: PMC6789237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVES Pancreatic cancer is one of the most lethal cancer types. Pancreatic cancer is highly malignant and characterized by rapid and uncontrolled growth. This study was designed to investigate the effect of baicalin on proliferation and apoptosis in pancreatic cancer cells. METHODS CCK-8 assay and Clone formation assay were performed to detect the effect of baicalin on proliferation in pancreatic cancer cells. Cell invasion and migration were all assessed with Wound healing assay and Transwell invasion assay. Flow Cytometry Analysis and DAPI staining were performed to detect the effect of baicalin on apoptosis in pancreatic cancer cells. Furthermore, proliferation-associated protein and apoptosis-related protein were detected to evaluate the cell proliferation and apoptosis levels. P-JNK protein, t-JNK protein, Foxo1 protein and BIM protein were examined by western blot to verify whether baicalin could regulate the proliferation and apoptosis via the JNK/Foxo1/BIM signaling pathway in pancreatic cancer cells. RESULTS The cell proliferation level was significantly decreased while the cell apoptosis level was significantly increased in pancreatic cancer SW1990 cells treated with baicalin. As the same, baicalin downregulated the ability of invasion and migration in pancreatic cancer SW1990 cells. CONCLUSION Baicalin might inhibit cell proliferation and promote cell apoptosis via JNK/Foxo1/BIM signaling pathway in pancreatic cancer SW1990 cells.
Collapse
Affiliation(s)
- Qin Huang
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Jinshun Zhang
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Jinbang Peng
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Yan Zhang
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Linlin Wang
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Juju Wu
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Liping Ye
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| | - Congcheng Fang
- Department of Gastroenterology, En-ze Medical Group Taizhou Hospital Luqiao, Taizhou 318050, Zhejiang, China
| |
Collapse
|
10
|
Zhang Z, Zhou M, Liu N, Shi Z, Pang Y, Li D, Qi J, Wu H, An R. The protection of New Interacting Motif E shot (NIMoEsh) in mice with collagenase-induced acute stage of intracerebral hemorrhage. Brain Res Bull 2019; 148:70-78. [PMID: 30935978 DOI: 10.1016/j.brainresbull.2019.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/21/2019] [Accepted: 03/27/2019] [Indexed: 12/23/2022]
Abstract
Aberrant c-Jun N terminal kinase (JNK) activation is broadly involved in the pathogenesis of several acute and chronic neurological diseases. However, the mechanism of JNK activation leading to aggravation of injury after ICH remains unclear. In this study, we confirmed that using NIMoEsh to inhibit JNK activation effectively reduced the level of brain injury following ICH. We evaluated brain outcomes by histology, immunofluorescence, Luxol fast blue/Cresyl violet staining and other experimental methods. We found that NIMoEsh could significantly inhibit the activity of JNK and thus improve inflammation, white-matter damage and neuronal cell death after ICH in mice. Our results suggest that JNK activation plays an important role of brain damage after acute stage of ICH and that NIMoEsh may be a potential target drug for the treatment of ICH.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Urology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Min Zhou
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - Nana Liu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - Zhongkai Shi
- Radioimmunoassay Laboratory Department, Heilongjiang Province Hospital, Harbin 150036, China
| | - Yuxin Pang
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - Danyang Li
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - Jiping Qi
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China.
| | - Ruihua An
- Department of Urology, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
11
|
Li Y, Ji S, Fu L, Jiang T, Wu D, Meng F. Over-expression of ARHGAP18 suppressed cell proliferation, migration, invasion, and tumor growth in gastric cancer by restraining over-activation of MAPK signaling pathways. Onco Targets Ther 2018; 11:279-290. [PMID: 29386906 PMCID: PMC5767098 DOI: 10.2147/ott.s130255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Globally, gastric cancer is the second-greatest cause of cancer death. ARHGAP18 belongs to the Rho family of GTPases which is involved in cellular migration, invasion, and growth phases. The aim of the present study was to investigate whether ARHGAP18 could regulate cell proliferation, migration, invasion, and related molecular mechanisms in gastric cancer. Cell Counting Kit-8 (CCK-8) assay results showed that following transfection of a recombinant plasmid, over-expression of ARHGAP18 inhibited cell viability in MGC-803 and BGC823 cells. Using in vitro transwell analysis, migration and invasion abilities were significantly inhibited in cells with high ARHGAP18 expression. Phosphorylation levels of ERK, JNK, and p38 by Western blot analysis significantly declined after transfection of cells with the ARHGAP18 plasmid. Expression levels of ROCK, MTA1, and MMP-2/9 were detected by real-time polymerase chain reaction and Western blotting, and over-expression of ARHGAP18 decreased the expression levels of ROCK, MTA1, and MMP-9. A further in vivo tumor formation study in nude mice indicated that over-expression of ARHGAP18 delayed the progress of tumor formation. These results indicate that ARHGAP18 could act as a tumor suppressor and may serve as a promising therapeutic strategy for gastric cancer.
Collapse
Affiliation(s)
- Yan Li
- Department of Biotherapy, Cancer Research Institute, the First Affiliated Hospital of China Medical University, Shenyang City, People's Republic of China
| | - Shan Ji
- Department of Endocrinology, The Fifth People's Hospital of Shenyang City, Shenyang City, People's Republic of China
| | - Liye Fu
- Department of Biotherapy, Cancer Research Institute, the First Affiliated Hospital of China Medical University, Shenyang City, People's Republic of China
| | - Tao Jiang
- Department of Biotherapy, Cancer Research Institute, the First Affiliated Hospital of China Medical University, Shenyang City, People's Republic of China
| | - Di Wu
- Department of Biotherapy, Cancer Research Institute, the First Affiliated Hospital of China Medical University, Shenyang City, People's Republic of China
| | - Fandong Meng
- Department of Biotherapy, Cancer Research Institute, the First Affiliated Hospital of China Medical University, Shenyang City, People's Republic of China
| |
Collapse
|
12
|
Fan S, Li K, Zhang D, Liu F. JNK and NF-κB signaling pathways are involved in cytokine changes in patients with congenital heart disease prior to and after transcatheter closure. Exp Ther Med 2017; 15:1525-1531. [PMID: 29434738 DOI: 10.3892/etm.2017.5595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 08/26/2017] [Indexed: 12/18/2022] Open
Abstract
Congenital heart disease (CHD) is a problem in the structure of the heart that is present at birth. Due to advances in interventional cardiology, CHD may currently be without surgery. The present study aimed to explore the molecular mechanism underlying CHD. A total of 200 cases of CHD treated by transcatheter closure as well as 200 controls were retrospectively assessed. Serum cytokines prior to and after treatment were assessed by reverse-transcription quantitative polymerase chain reaction analysis. Furthermore, the levels of proteins associated with c-Jun N-terminal kinase (JNK) and nuclear factor (NF)-κB were assessed by western blot analysis and immunohistochemistry. Furthermore, an animal model of CHD in young pigs was successfully constructed and treated with inhibitors of JNK and/or NF-κB to investigate the roles of these pathways in CHD. The results revealed that tumor necrosis factor-α, interleukin (IL)-6 and IL-8 were significantly elevated in the experimental group following transcatheter closure treatment, compared with those in the healthy control group, and the serum levels of the anti-inflammatory cytokine IL-10 were significantly reduced. Phosphorylated c-Jun and p65 levels in the experimental group were notably higher in the experimental group compared with the control group, but were restored to normal levels following transcatheter closure treatment. Similar results were also obtained in the pig model. The results of the present study suggested that the CHD-associated changes in cytokines, as well as their recovery following transcatheter closure treatment were associated with the JNK and NF-κB signaling pathways. The present study may provide further understanding of the underlying molecular mechanisms in CHD and propose a potential novel target for the treatment of CHD.
Collapse
Affiliation(s)
- Shunyang Fan
- Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Kefang Li
- Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Deyin Zhang
- Department of Internal Medicine, The Third People's Hospital of Henan Province, Zhengzhou, Henan 450000, P.R. China
| | - Fuyun Liu
- Department of Pediatric Surgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
13
|
Liu DZ, Chang B, Li XD, Zhang QH, Zou YH. MicroRNA-9 promotes the proliferation, migration, and invasion of breast cancer cells via down-regulating FOXO1. Clin Transl Oncol 2017; 19:1133-1140. [PMID: 28397066 DOI: 10.1007/s12094-017-1650-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/13/2017] [Indexed: 02/05/2023]
Abstract
PURPOSE The objective of the study was to investigate the role of microRNA-9 (miR-9) targeting forkhead box O1 (FOXO1) in the proliferation, migration, and invasion of breast cancer cells. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to determine the expressions of miR-9 and FOXO1 mRNA in breast cancer tissues, normal breast tissues, breast cancer cell lines, and normal breast epithelial cells. After the up-regulation of miR-9 expression, qRT-PCR and Western blotting were used to determine the expression of FOXO1. The luciferase reporter gene assay was used to validate the target gene. The CCK-8 assay, scratch-wound healing assay, and Transwell invasion assay were used to investigate the changes in the proliferation, migration, and invasion of breast cancer cells, respectively. RESULTS MicroRNA-9 expression was significantly up-regulated in breast cancer tissues and breast cancer cell lines when compared with normal breast tissues and normal breast epithelial cells (both P < 0.05). FOXO1 mRNA and protein expressions were substantially down-regulated in breast cancer tissues and breast cancer cell lines when compared with normal breast tissues and normal breast epithelial cells (both P < 0.05). There can be a negative correlation between miR-9 and FOXO1 mRNA in breast cancer. Luciferase reporter gene assay indicated that miR-9 can down-regulate FOXO1 expression at a post-transcriptional level through binding specifically to FOXO1 3'UTR. The results of CCK-8 assay, scratch-wound healing assay, and Transwell invasion assay revealed that the inhibition of miR-9 can suppress MCF7 cell proliferation, migration, and invasion. Additionally, the expression of miR-9 increased significantly whilst that of FOXO1 decreased substantially as the disease progressed (P < 0.05). CONCLUSIONS Our study provides evidence that miR-9 can promote the proliferation, migration, and invasion of breast cancer cells via down-regulating FOXO1.
Collapse
Affiliation(s)
- D-Z Liu
- Department of Emmengey, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - B Chang
- Department of Emmengey, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - X-D Li
- Department of Orthopedics, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Q-H Zhang
- Department of Orthopedics, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Y-H Zou
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China.
| |
Collapse
|
14
|
Chaves FN, Bezerra TMM, de Barros Silva PG, Oliveira FAF, Sousa FB, Costa FWG, Alves APNN, Pereira KMA. Evaluation of the p-AKT, p-JNK and FoxO3a function in oral epithelial dysplasia. Oral Dis 2017; 23:367-378. [DOI: 10.1111/odi.12623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/06/2016] [Accepted: 12/02/2016] [Indexed: 12/27/2022]
Affiliation(s)
- FN Chaves
- School of Dentistry; Federal University of Ceara/Sobral; Sobral Ceara Brazil
| | - TMM Bezerra
- Division of Oral Pathology; Department of Dental Clinic; Faculty of Pharmacy, Dentistry and Nursing; Federal University of Ceara; Fortaleza Ceara Brazil
| | - PG de Barros Silva
- Division of Oral Pathology; Department of Dental Clinic; Faculty of Pharmacy, Dentistry and Nursing; Federal University of Ceara; Fortaleza Ceara Brazil
| | - FAF Oliveira
- Division of Oral Pathology; Department of Dental Clinic; Faculty of Pharmacy, Dentistry and Nursing; Federal University of Ceara; Fortaleza Ceara Brazil
| | - FB Sousa
- Division of Oral Pathology; Department of Dental Clinic; Faculty of Pharmacy, Dentistry and Nursing; Federal University of Ceara; Fortaleza Ceara Brazil
| | - FWG Costa
- Division of Oral Pathology; Department of Dental Clinic; Faculty of Pharmacy, Dentistry and Nursing; Federal University of Ceara; Fortaleza Ceara Brazil
| | - APNN Alves
- Division of Oral Pathology; Department of Dental Clinic; Faculty of Pharmacy, Dentistry and Nursing; Federal University of Ceara; Fortaleza Ceara Brazil
| | - KMA Pereira
- School of Dentistry; Federal University of Ceara/Sobral; Sobral Ceara Brazil
| |
Collapse
|
15
|
Choi Y, Ko YS, Park J, Choi Y, Kim Y, Pyo JS, Jang BG, Hwang DH, Kim WH, Lee BL. HER2-induced metastasis is mediated by AKT/JNK/EMT signaling pathway in gastric cancer. World J Gastroenterol 2016; 22:9141-9153. [PMID: 27895401 PMCID: PMC5107595 DOI: 10.3748/wjg.v22.i41.9141] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/12/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigated the relationships between HER2, c-Jun N-terminal kinase (JNK) and protein kinase B (AKT) with respect to metastatic potential of HER2-positive gastric cancer (GC) cells.
METHODS Immunohistochemistry was performed on tissue array slides containing 423 human GC specimens. Using HER2-positve GC cell lines SNU-216 and NCI-N87, HER2 expression was silenced by RNA interference, and the activations of JNK and AKT were suppressed by SP600125 and LY294002, respectively. Transwell assay, Western blot, semi-quantitative reverse transcription-polymerase chain reaction and immunofluorescence staining were used in cell culture experiments.
RESULTS In GC specimens, HER2, JNK, and AKT activations were positively correlated with each other. In vitro analysis revealed a positive regulatory feedback loop between HER2 and JNK in GC cell lines and the role of JNK as a downstream effector of AKT in the HER2/AKT signaling pathway. JNK inhibition suppressed migratory capacity through reversing EMT and dual inhibition of JNK and AKT induced a more profound effect on cancer cell motility.
CONCLUSION HER2, JNK and AKT in human GC specimens are positively associated with each other. JNK and AKT, downstream effectors of HER2, co-operatively contribute to the metastatic potential of HER2-positive GC cells. Thus, targeting of these two molecules in combination with HER2 downregulation may be a good approach to combat HER2-positive GC.
Collapse
|