Yingcun Liu, Yuqing Fu, Xin Xue, Gang Tang, Liangyi Si
{"title":"BRD2 protects the rat H9C2 cardiomyocytes from hypoxia‑reoxygenation injury by targeting Nrf2/HO‑1 signaling pathway.","authors":"Yingcun Liu, Yuqing Fu, Xin Xue, Gang Tang, Liangyi Si","doi":"10.3892/etm.2023.12241","DOIUrl":"10.3892/etm.2023.12241","url":null,"abstract":"<p><p>Myocardial ischemia-reperfusion (I/R) injury is a common complication of acute myocardial infarction following percutaneous coronary intervention, but there are currently no effective pharmacological targets for adjuvant therapy due to a lack of knowledge of I/R injury mechanisms in cardiomyocytes. To evaluate the effects of hypoxia-reoxygenation on the plasma proteome of cardiomyocytes and prospective therapeutic targets, five sets of H9C2 cardiomyocytes from rats were cultured under various hypoxic circumstances. Using Cell Counting Kit-8 (CCK8) and lactose dehydrogenase (LDH) release assays, the cell viability and LDH release of H9C2 cells were analyzed. Proteome sequencing was then performed on cardiomyocytes to show the quantitative protein changes during the I/R injury process. After hypoxia/reoxygenation, bromodomain-containing protein 2 (BRD2) expression was evaluated. After administering the BRD2 inhibitor dBET1, the expression of nuclear factor erythroid 2-related factor 2/haem oxygenase-1 (Nrf2/HO-1) was identified. The results showed that in the group exposed to 4 h of hypoxia followed by 4 h of reoxygenation (H/R4), the cell survival rate was dramatically reduced, although the apoptotic rate and LDH were much higher than in the normal oxygen group. In addition, the expressions of 2,325 proteins differed considerably between these two groups, with 128 upregulated and 122 downregulated proteins being discovered in the H/R4 group. After 4 h of reoxygenation, the BRD2 expression was increased. Following the addition of dBET1 to suppress BRD2, the expression of Nrf2/HO-1 was reduced, but the rate of apoptosis increased. In conclusion, through the Nrf2/HO-1 signaling pathway, BRD2 protects cardiomyocytes from damage caused by hypoxia/reoxygenation. This may have implications for novel treatment targets to minimize I/R damage to the myocardium.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"542"},"PeriodicalIF":0.0,"publicationDate":"2023-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/71/03/etm-26-05-12241.PMC10587885.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Function of GSK‑3 signaling in spinal cord injury (Review).","authors":"Xiong Dong, Hongxiang Hong, Zhiming Cui","doi":"10.3892/etm.2023.12240","DOIUrl":"10.3892/etm.2023.12240","url":null,"abstract":"<p><p>Spinal cord injury (SCI) is a major social problem with a heavy burden on patient physiology and psychology. Glial scar formation and irreversible neuron loss are the two key points during SCI progression. During the acute phase of spinal cord injury, glial scars form, limiting the progression of inflammation. However, in the subacute or chronic phase, glial scarring inhibits axon regeneration. Following spinal cord injury, irreversible loss of neurons leads to further aggravation of spinal cord injury. Several therapies have been developed to improve either glial scar or neuron loss; however, few therapies reach the stage of clinical trials and there are no mainstream therapies for SCI. Exploring the key mechanism of SCI is crucial for finding further treatments. Glycogen synthase kinase-3 (GSK-3) is a widely expressed kinase with important physiological and pathophysiological functions <i>in vivo</i>. Dysfunction of the GSK-3 signaling pathway during SCI has been widely discussed for controlling neurite growth <i>in vitro</i> and <i>in vivo</i>, improving the proliferation and neuronal differentiation of endogenous neural stem cells and functional recovery from spinal cord injury. SCI can decrease the phosphorylated (p)/total (t)-GSK-3β ratio, which leads to an increase in apoptosis, whereas treatment with GSK-3 inhibitors can promote neurogenesis. In addition, several therapies for the treatment of SCI involve signaling pathways associated with GSK-3. Furthermore, signaling pathways associated with GSK-3 also participate in the pathological process of neuropathic pain that remains following SCI. The present review summarized the roles of GSK-3 signaling in SCI to aid in the understanding of GSK-3 signaling during the pathological processes of SCI and to provide evidence for the development of comprehensive treatments.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"541"},"PeriodicalIF":0.0,"publicationDate":"2023-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/97/4e/etm-26-05-12240.PMC10587879.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Trehalose delays postmenopausal osteoporosis by enhancing AKT/TFEB pathway‑dependent autophagy flow in rats.","authors":"Yongli Wang, Xingcun Li, Hongliang Gao, Qian Lu","doi":"10.3892/etm.2023.12237","DOIUrl":"10.3892/etm.2023.12237","url":null,"abstract":"<p><p>Osteoporosis is a systemic bone metabolic disorder that plagues the health and quality of life of the elderly. Autophagy plays an important role in bone formation while maintaining the homeostasis of the body. Trehalose is a mTOR-independent autophagy inducer, but to the best of our knowledge, there is no rat model of postmenopausal osteoporosis. The present study found that trehalose can delay postmenopausal osteoporosis in rats, which may be achieved by inducing and enhancing AKT/transcription factor EB pathway-dependent autophagy flow. The specific mechanism of its occurrence needs to be further studied. Trehalose-containing drugs are promising for delaying postmenopausal osteoporosis. Hematoxylin and eosin (H&E) staining, western blotting, micro computerized tomography (CT) scanning and Transmission electron microscopy were used to investigate the role of trehalose in postmenopausal osteoporosis rat model at protein, cell and histology aspects. According to the H&E staining results, the bone trabecular histological structure of the trehalose group was superior to that of the model group. The Micro CT scanning indicated the imaging structure of bone trabeculae in the trehalose group was superior to than that in the model group. Western blotting indicated the activation of autophagic flow in trehalose group, the autophagy degree of the trehalose group is greater than that of the model group; Transmission electron microscopy indicated the autophagy degree of the Trehalose group was greater than that of the model group under electron microscopy. Trehalose can delay postmenopausal osteoporosis in rats, which may be achieved by inducing and enhancing Akt/TFEB pathway-dependent autophagy flow.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"538"},"PeriodicalIF":0.0,"publicationDate":"2023-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/9a/a6/etm-26-05-12237.PMC10587861.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695348","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yufeng Zhang, Yingzhen Niu, Yonggang Peng, Xueyang Pan, Fei Wang
{"title":"COL3A1, COL5A1 and COL6A2 serve as potential molecular biomarkers for osteoarthritis based on weighted gene co‑expression network analysis bioinformatics analysis.","authors":"Yufeng Zhang, Yingzhen Niu, Yonggang Peng, Xueyang Pan, Fei Wang","doi":"10.3892/etm.2023.12239","DOIUrl":"10.3892/etm.2023.12239","url":null,"abstract":"<p><p>Osteoarthritis (OA) is a non-inflammatory degenerative joint disease, characterized by joint pain and stiffness. The prevalence of OA increases with age. However, the relationship between biomarkers [collagen type III α1 (COL3A1), COL5A1, COL6A2, COL12A1] and OA remains unclear. The OA subchondral bone dataset GSE51588 was downloaded from the GEO database, and the differentially expressed genes (DEGs) were screened. Weighted gene co-expression network analysis was performed, and a protein-protein interaction network was constructed and further analyzed using Cytoscape and STRING. Functional enrichment analysis was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and then Gene Set Enrichment Analysis (GSEA) was used to formulate the molecular functions and pathways based on the results of GO and KEGG analyses. Comparative Toxicogenomics Database and TargetScan were used to identify the hub-gene-related diseases and the microRNAs that regulated the central hub genes. Immunohistochemical staining was performed to confirm the expression of related proteins in OA and non-OA tissue samples. A total of 1,679 DEGs were identified. GO analysis showed that the DEGs were primarily enriched in the process of 'immune system', 'extracellular region', 'secretory granule', 'collagen-containing extracellular matrix', 'ECM-receptor, glycosaminoglycan binding' and 'systemic lupus erythematosus'. The results of GSEA were similar to those of GO and KEGG enrichment terms for DEGs. A total of 25 important modules were generated, and two core gene clusters and seven core genes were obtained (COL6A2, COL5A2, COL12A1, COL5A1, COL6A1, LUM and COL3A1). Core genes were expressed differentially between OA subchondral bone and normal tissue samples. The expression levels of COL3A1, COL5A1 and COL6A2 in OA subchondral bone tissue were higher compared with those in normal tissues, but COL12A1 expression was not significantly increased; all stained markers were highly expressed in surrounding tissues of immunohistochemical staining. In conclusion, COL3A1, COL5A1 and COL6A2 may be potential molecular biomarkers for OA.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"540"},"PeriodicalIF":0.0,"publicationDate":"2023-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/fa/30/etm-26-05-12239.PMC10587888.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Evaluation of the accuracy of heart dose prediction by machine learning for selecting patients not requiring deep inspiration breath‑hold radiotherapy after breast cancer surgery.","authors":"Ryo Kamizaki, Masahiro Kuroda, Wlla E Al-Hammad, Nouha Tekiki, Hinata Ishizaka, Kazuhiro Kuroda, Kohei Sugimoto, Masataka Oita, Yoshinori Tanabe, Majd Barham, Irfan Sugianto, Yuki Nakamitsu, Masaki Hirano, Yuki Muto, Hiroki Ihara, Soichi Sugiyama","doi":"10.3892/etm.2023.12235","DOIUrl":"10.3892/etm.2023.12235","url":null,"abstract":"<p><p>Increased heart dose during postoperative radiotherapy (RT) for left-sided breast cancer (BC) can cause cardiac injury, which can decrease patient survival. The deep inspiration breath-hold technique (DIBH) is becoming increasingly common for reducing the mean heart dose (MHD) in patients with left-sided BC. However, treatment planning and DIBH for RT are laborious, time-consuming and costly for patients and RT staff. In addition, the proportion of patients with left BC with low MHD is considerably higher among Asian women, mainly due to their smaller breast volume compared with that in Western countries. The present study aimed to determine the optimal machine learning (ML) model for predicting the MHD after RT to pre-select patients with low MHD who will not require DIBH prior to RT planning. In total, 562 patients with BC who received postoperative RT were randomly divided into the trainval (n=449) and external (n=113) test datasets for ML using Python (version 3.8). Imbalanced data were corrected using synthetic minority oversampling with Gaussian noise. Specifically, right-left, tumor site, chest wall thickness, irradiation method, body mass index and separation were the six explanatory variables used for ML, with four supervised ML algorithms used. Using the optimal value of hyperparameter tuning with root mean squared error (RMSE) as an indicator for the internal test data, the model yielding the best F2 score evaluation was selected for final validation using the external test data. The predictive ability of MHD for true MHD after RT was the highest among all algorithms for the deep neural network, with a RMSE of 77.4, F2 score of 0.80 and area under the curve-receiver operating characteristic of 0.88, for a cut-off value of 300 cGy. The present study suggested that ML can be used to pre-select female Asian patients with low MHD who do not require DIBH for the postoperative RT of BC.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"536"},"PeriodicalIF":0.0,"publicationDate":"2023-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10587874/pdf/etm-26-05-12235.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jian-Hua Xi, Nai-Ying Sun, Wen-Jun Guo, Xing-Jie Yang
{"title":"Ectopic gastric mucosa in the submucosa of the stomach: A case report.","authors":"Jian-Hua Xi, Nai-Ying Sun, Wen-Jun Guo, Xing-Jie Yang","doi":"10.3892/etm.2023.12238","DOIUrl":"10.3892/etm.2023.12238","url":null,"abstract":"<p><p>Under normal circumstances, gastric mucosa only exists within the stomach. However, in certain situations, gastric mucosal tissue may undergo ectopia, commonly occurring in the esophagus and intestine, with rare occurrences within the stomach itself. A comprehensive literature review was performed to understand the distinct characteristics of ectopic gastric mucosa (EGM) in the stomach and investigate a rare incident of this disease, providing an in-depth analysis of the clinical, histopathologic, and differential diagnostic findings. The case was a 47-year-old man with acid reflux, heartburn, abdominal distension, and diarrhea (5-10 times daily) for >10 years. A gastroscope indicated a submucosal protuberance lesion in the gastric body that felt hard with biopsy forceps. A well-defined nodule under the mucosal muscle was revealed microscopically, composed of epithelial elements and no atypia. Immunohistochemical staining demonstrated similar EGM expression patterns compared with normal gastric mucosa. The present case report highlights the importance of accurate EGM diagnosis and understanding.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"539"},"PeriodicalIF":0.0,"publicationDate":"2023-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/ad/11/etm-26-05-12238.PMC10587881.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Unraveling the protective mechanisms of Chuanfangyihao against acute lung injury: Insights from experimental validation.","authors":"Hongfang Fu, Xiao Liang, Wanying Tan, Xiaoyu Hu","doi":"10.3892/etm.2023.12234","DOIUrl":"10.3892/etm.2023.12234","url":null,"abstract":"<p><p>Chuanfangyihao (CFYH) is an effective treatment for acute lung injury (ALI) in clinical practice; however, its underlying mechanism of action remains unclear. Therefore, the aim of the present study was to elucidate the pharmacological mechanism of action of CFYH in ALI through experimental validation. First, a rat model of ALI was established using lipopolysaccharide (LPS). Next, the pathological changes in the lungs of the rats and the pathological damage were scored. The wet/dry weight ratios were measured, and ROS content was detected using flow cytometry. ELISA was used to examine IL-6, TNF-α, IL-1β, IL-18, and LDH levels. Immunohistochemistry was used to detect Beclin-1 and NLRP3 expression. Western blotting was performed to analyze the expression of HMGB1, RAGE, TLR4, NF-κB p65, AMPK, p-AMPK, mTOR, p-mTOR, Beclin-1, LC3-II/I, p62, Bcl-2, Bax, Caspase-3, Caspase-1, and GSDMD-NT. The mRNA levels of HMGB1, RAGE, AMPK, mTOR, and HIF-1α were determined using reverse transcription quantitative PCR. CFYH alleviated pulmonary edema and decreased the expression of IL-6, TNF-α, TLR4, NF-κB p65, HMGB1/RAGE, ROS, and HIF-1α. In addition, pretreatment with CFYH reversed ALI-induced programmed cell death. In conclusion, CFYH alleviates LPS-induced ALI, and these findings provide a preliminary clarification of the predominant mechanism of action of CFYH in ALI.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"535"},"PeriodicalIF":0.0,"publicationDate":"2023-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/df/6e/etm-26-05-12234.PMC10587870.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695349","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Transcriptomics and metabolomics study in mouse kidney of the molecular mechanism underlying energy metabolism response to hypoxic stress in highland areas.","authors":"Yujie Gao, Qifu Long, Hui Yang, Ying Hu, Yuzhen Xu, Chaoqun Tang, Cunlin Gu, Sheng Yong","doi":"10.3892/etm.2023.12232","DOIUrl":"10.3892/etm.2023.12232","url":null,"abstract":"<p><p>Exposure to hypoxia disrupts energy metabolism and induces inflammation. However, the pathways and mechanisms underlying energy metabolism disorders caused by hypoxic conditions remain unclear. In the present study, a hypoxic animal model was created and transcriptomic and non-targeted metabolomics techniques were applied to further investigate the pathways and mechanisms of hypoxia exposure that disrupt energy metabolism. Transcriptome results showed that 3,007 genes were significantly differentially expressed under hypoxic exposure, and Gene Ontology annotation analysis and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment analysis showed that the differentially expressed genes (DEGs) were mainly involved in energy metabolism and were significantly enriched in the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) pathway. The DEGs <i>IDH3A</i>, <i>SUCLA2</i>, and <i>MDH2</i> in the TCA cycle and the DEGs <i>NDUFA3</i>, <i>NDUFS7</i>, <i>UQCRC1</i>, <i>CYC1</i> and <i>UQCRFS1</i> in the OXPHOS pathway were validated using mRNA and protein expression, and the results showed downregulation. The results of non-targeted metabolomics showed that 365 significant differential metabolites were identified under plateau hypoxia stress. KEGG enrichment analysis showed that the differential metabolites were mainly enriched in metabolic processes, such as energy, nucleotide and amino acid metabolism. Hypoxia exposure disrupted the TCA cycle and reduced the synthesis of amino acids and nucleotides by decreasing the concentration of cis-aconitate, α-ketoglutarate, NADH, NADPH and that of most amino acids, purines, and pyrimidines. Bioinformatics analysis was used to identify inflammatory genes related to hypoxia exposure and some of them were selected for verification. It was shown that the mRNA and protein expression levels of <i>IL1B</i>, <i>IL12B</i>, <i>S100A8</i> and <i>S100A9</i> in kidney tissues were upregulated under hypoxic exposure. The results suggest that hypoxia exposure inhibits the TCA cycle and the OXPHOS signalling pathway by inhibiting <i>IDH3A</i>, <i>SUCLA2</i>, <i>MDH2</i>, <i>NDUFFA3</i>, <i>NDUFS7</i>, <i>UQCRC1</i>, <i>CYC1</i> and <i>UQCRFS1</i>, thereby suppressing energy metabolism, inducing amino acid and nucleotide deficiency and promoting inflammation, ultimately leading to kidney damage.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"533"},"PeriodicalIF":0.0,"publicationDate":"2023-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/c3/6e/etm-26-05-12232.PMC10587886.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695347","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Integrin β5 is an independent prognostic marker for intrahepatic cholangiocarcinoma in a Chinese population.","authors":"Lixing Ma, Kang Song, Jinfeng Zang","doi":"10.3892/etm.2023.12231","DOIUrl":"10.3892/etm.2023.12231","url":null,"abstract":"<p><p>Intrahepatic cholangiocarcinoma (ICC) is the second most common primary liver tumor and a major cause of cancer mortality worldwide. Integrin β5 (ITGB5) is considered to be involved in the intercellular signal transduction and regulation of tumorigenesis and development. The present study investigated the association between ITGB5 expression levels and the prognosis of ICC, as well as the effects of ITGB5 on the proliferation and invasion of ICC cells. RNA-sequencing transcriptomic profiling data of ICC samples were retrieved from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Tissue specimens from patients with ICC treated at Taizhou People's Hospital were collected and the ITGB5 expression levels were evaluated using immunohistochemical staining. The biological function of ITGB5 in ICC was investigated using Gene Ontology (GO), Gene Set Enrichment Analysis (GSEA) and <i>in vitro</i> experiments using HuCCT1 cells. After knocking down ITGB5 expression, cell proliferation was detected using Cell Counting Kit-8 assay, while cell invasion was assessed using Transwell assays. According to TCGA dataset, ITGB5 was highly expressed in ICC; however, there was no significant difference in prognosis between patients with high and low ITGB5 expression levels. High expression of ITGB5 was present in the tissues of patients with ICC from the GEO database, which was associated with poor prognosis. Survival analyses of the clinical data obtained in the present study revealed that high expression levels of ITGB5 in patients with ICC were associated with a reduced overall survival. GO and GSEA indicated that genes associated with ITGB5 were enriched in the extracellular matrix-receptor interaction and focal adhesion signaling pathways. Silencing ITGB5 inhibited the proliferation and invasion of ICC cells. In conclusion, ITGB5 may act as an essential regulator of ICC development and progression by influencing the proliferation and invasion of ICC cells. However, future studies with larger sample sizes are required to validate the role of ITGB5 in the prognosis of patients with ICC.</p>","PeriodicalId":94002,"journal":{"name":"Experimental and therapeutic medicine","volume":"26 5","pages":"532"},"PeriodicalIF":0.0,"publicationDate":"2023-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/92/ca/etm-26-05-12231.PMC10587877.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49695422","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}