1
|
Liu L, Zhang C, Yang B, Wang M. MARCH8 ubiquitinates and degrades CEMIP to induce colorectal cancer cell ferroptosis through inactivating PI3K/AKT pathway. Pathol Res Pract 2025; 269:155909. [PMID: 40107011 DOI: 10.1016/j.prp.2025.155909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/23/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Cell migration-inducing and hyaluronan-binding protein (CEMIP) is found to act as an oncogene in colorectal cancer (CRC) progression, but the underlying molecular mechanisms need to be further elucidated. METHODS The mRNA and protein levels of CEMIP and membrane-associated ring-CH-type finger 8 (MARCH8) were examined by qRT-PCR and western blot. Cell functions were detected by CCK8 assay, colony formation assay and transwell assay. The levels of ROS, Fe2 +, GSH, and MDA were examined to evaluate cell ferroptosis. The interaction between MARCH8 and CEMIP was assessed by Co-IP assay and ubiquitination assay. The protein levels of ferroptosis-related markers (ACSL4, GPX4 and FTH1) and PI3K/AKT-related markers were tested using western bolt. The anti-tumor effect of MARCH8 was further confirmed by constructing xenograft tumor models. RESULTS CEMIP expression was higher in CRC tissues and cells. CEMIP knockdown could suppress CRC cell proliferation, migration, invasion, and enhance ferroptosis. MARCH8 ubiquitinated CEMIP to decrease its expression, thus inhibiting CRC cell proliferation, metastasis and inducing ferroptosis. And CEMIP overexpression could abolish the anti-proliferation, anti-metastasis and pro-ferroptosis effect of MARCH8. Also, MARCH8 overexpression repressed the activity of PI3K/AKT pathway, and CEMIP upregulation partially reversed this effect. In vivo experiments suggested that MARCH8 reduced CRC tumorigenesis by inducing ferroptosis. CONCLUSION MARCH8 promoted CRC cell ferroptosis via inhibiting PI3K/AKT pathway by enhancing the ubiquitination and degradation of CEMIP.
Collapse
Affiliation(s)
- Lintao Liu
- Department of Colorectal and Anal Surgery, Jilin Province People's Hospital, Changchun, China
| | - Cheng Zhang
- Department of Colorectal and Anal Surgery, Jilin Province People's Hospital, Changchun, China
| | - Bo Yang
- Department of Colorectal and Anal Surgery, Jilin Province People's Hospital, Changchun, China
| | - Maonan Wang
- Department of Abdominal Tumor Surgery, Jilin Cancer Hospital, Changchun, China.
| |
Collapse
|
2
|
Yue Y, Zhou FF, Zhang JR, Xu F. Expression and significance of CEMIP and CYP11B2 in serum in women with foetal growth restriction. J OBSTET GYNAECOL 2024; 44:2389169. [PMID: 39218008 DOI: 10.1080/01443615.2024.2389169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Foetal growth restriction (FGR) occurs when a foetus fails to reach its growth potential. This observational study assessed the expression and significance of cell migration-including protein (CEMIP) and aldosterone synthase (CYP11B2) in the serum of pregnant women with FGR. METHODS 40 singleton FGR-suffered pregnant women, as well as 40 normal singleton pregnant women, were enrolled. The expression of CEMIP and CYP11B2 in serum was detected in early pregnancy. The correlations between parameters were evaluated. The predictive variables for FGR were determined. The diagnostic value of CEMIP and CYP11B2 for FGR was analysed. RESULTS CEMIP and CYP11B2 mRNA expression in the serum of pregnant women with FGR decreased (both P < 0.001). CEMIP (95%CI: 0.802-0.921, P < 0.001) and CYP11B2 (95%CI: 0.795-0.907, P < 0.001) mRNA expression in serum and soluble fms like tyrosine kinase-1 (sFLT1)/placental growth factor (PlGF) ratio (95%CI: 0.866-0.974, P < 0.001) were independent predictors of FGR, and CEMIP (r = -0.578, P = 0.001) and CYP11B2 (r = -0.602, P < 0.001) mRNA expression in serum were negatively correlated with sFLT1/PlGF ratio. CEMIP (AUC = 0.741) and CYP11B2 (AUC = 0.764) mRNA expression in serum had good diagnostic value for FGR. CONCLUSION The expression of CEMIP and CYP11B2 is reduced in the serum of pregnant women with FGR and may become new diagnostic markers for FGR.
Collapse
Affiliation(s)
- Yang Yue
- Department of Obstetrics, Longhua District Maternity and Child Health Hospital, Shenzhen City, Guangdong Province, China
| | - Fang Fang Zhou
- Department of Obstetrics, Longhua District Maternity and Child Health Hospital, Shenzhen City, Guangdong Province, China
| | - Jia Rong Zhang
- Department of Obstetrics, Longhua District Maternity and Child Health Hospital, Shenzhen City, Guangdong Province, China
| | - Fu Xu
- Department of Anesthesiology, Longhua District People's Hospital, Shenzhen City, Guangdong Province, China
| |
Collapse
|
3
|
Liu T, Ma Y, Han S, Sun P. Genome-wide investigation of lncRNAs revealed their tight association with gastric cancer. J Cancer Res Clin Oncol 2024; 150:261. [PMID: 38761291 PMCID: PMC11102383 DOI: 10.1007/s00432-024-05790-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a significant health issue globally, ranking as the fifth most common cancer with over 10,000 new cases reported annually. Long non-coding RNA (lncRNA) has emerged as a critical player in cellular functions, influencing GC's development, growth, metastasis, and prognosis. However, our understanding of lncRNA's role in the pathogenesis of GC remains limited. Therefore, it is particularly important to explore the relationship between lncRNA and gastric cancer. METHODS we conducted a comprehensive analysis of RNA sequencing data from the GEO database and stomach adenocarcinoma (STAD) data from the TCGA database to identify lncRNAs that exhibit altered expression levels in GC and the mechanisms underlying lncRNA-mediated transcription and post-transcriptional regulation were explored. RESULTS This study uncovered 94 lncRNAs with differential expression and, through co-expression analysis, linked these to 1508 differentially expressed genes (DEGs). GO functional enrichment analysis highlighted that these DEGs are involved in critical pathways, such as cell adhesion and the positive regulation of cell migration. By establishing a lncRNA-miRNA-mRNA regulatory network, we found that the ceRNA mechanism, particularly involving RP11-357H14.17 and CTD-2377D24.4, could play a role in GC progression. Experimental validation of selected differentially expressed lncRNAs and mRNAs (including RP11-357H14.17-CLDN1, BBOX1, TRPM2-AS, CLDN1, PLAU, HOXB7) confirmed the RNA-seq results. CONCLUSIONS Overall, our findings highlight the critical role of the lncRNA-mRNA regulatory network in the development and progression of GC, offering potential biomarkers for diagnosis and targets for innovative treatment strategies.
Collapse
Affiliation(s)
- Tong Liu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Yuedong Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Shuo Han
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Pengda Sun
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
4
|
Sun Y, Xie L, Ren X, Ran L, He H, Kong F, Yang S, Zhang M. miR-148a-3p regulates proliferation and apoptosis of idiopathic gingival fibroma by targeting NPTX1. Oral Dis 2024; 30:2136-2149. [PMID: 37357360 DOI: 10.1111/odi.14655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/18/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023]
Abstract
OBJECTIVE Idiopathic gingival fibromatosis (IGF) is a rare heterogeneous disease that results in the progressive and diffuse hyperplasia of gingival tissues. MicroRNAs are implicated in the development and progression of various tumors. The present study aimed to explore the potential roles and mechanisms of miR-148a-3p in IGF. METHODS Gingival fibroblasts (GFs) were transfected with miR-148a-3p mimics, miR-148a-3p inhibitors, or siNPTX1, and then, the proliferation and apoptosis of GFs and the expression of related genes were evaluated using Cell Counting Kit-8 assays, 5-ethynyl-2'-deoxyuridine assays, flow cytometry, reverse transcription-quantitative polymerase chain reaction, and western blot analysis, respectively. RESULTS miR-148a-3p was highly expressed in GFs of IGF (IGF-GFs) as compared with normal GFs (N-GFs). Overexpression of miR-148a-3p promoted the proliferation and inhibited the apoptosis of N-GFs, whereas downregulation of miR-148a-3p had the opposite effect in IGF-GFs. Knockdown of NPTX1 reversed miR-148a-3p-mediated effects in IGF-GFs. Dual-luciferase reporter assay confirmed that NPTX1 is a direct target of miR-148a-3p. CONCLUSION These findings identify that miR-148a-3p could regulate cell proliferation and apoptosis by targeting NPTX1, providing new insights for the further study of the molecular mechanism and treatment of IGF.
Collapse
Affiliation(s)
- Yuyang Sun
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
- Department of Stomatology, Taihe Affiliated Hospital of Hubei University of Medicine, Shiyan, China
| | - Liangkun Xie
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
| | - Xiaobin Ren
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
| | - Liquan Ran
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
| | - Hongbing He
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
| | - Fanying Kong
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Shuran Yang
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
- Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Mingzhu Zhang
- Kunming Medical University Affiliated Stomatology Hospital, Kunming, China
| |
Collapse
|
5
|
Guo S, Guo Y, Chen Y, Cui S, Zhang C, Chen D. The role of CEMIP in cancers and its transcriptional and post-transcriptional regulation. PeerJ 2024; 12:e16930. [PMID: 38390387 PMCID: PMC10883155 DOI: 10.7717/peerj.16930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
CEMIP is a protein known for inducing cell migration and binding to hyaluronic acid. Functioning as a hyaluronidase, CEMIP primarily facilitates the breakdown of the extracellular matrix component, hyaluronic acid, thereby regulating various signaling pathways. Recent evidence has highlighted the significant role of CEMIP in different cancers, associating it with diverse pathological states. While identified as a biomarker for several diseases, CEMIP's mechanism in cancer seems distinct. Accumulating data suggests that CEMIP expression is triggered by chemical modifications to itself and other influencing factors. Transcriptionally, chemical alterations to the CEMIP promoter and involvement of transcription factors such as AP-1, HIF, and NF-κB regulate CEMIP levels. Similarly, specific miRNAs have been found to post-transcriptionally regulate CEMIP. This review provides a comprehensive summary of CEMIP's role in various cancers and explores how both transcriptional and post-transcriptional mechanisms control its expression.
Collapse
Affiliation(s)
- Song Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yunfei Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yuanyuan Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Shuaishuai Cui
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Chunmei Zhang
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Dahu Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| |
Collapse
|
6
|
Liu Y, Hu G, Li Y, Kong X, Yang K, Li Z, Lao W, Li J, Zhong J, Zhang S, Leng Y, Bi C, Zhai A. Research on the biological mechanism and potential application of CEMIP. Front Immunol 2023; 14:1222425. [PMID: 37662915 PMCID: PMC10471826 DOI: 10.3389/fimmu.2023.1222425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
Cell migration-inducing protein (CEMIP), also known as KIAA1199 and hyaluronan-binding protein involved in hyaluronan depolymerization, is a new member of the hyaluronidase family that degrades hyaluronic acid (HA) and remodels the extracellular matrix. In recent years, some studies have reported that CEMIP can promote the proliferation, invasion, and adhesion of various tumor cells and can play an important role in bacterial infection and arthritis. This review focuses on the pathological mechanism of CEMIP in a variety of diseases and expounds the function of CEMIP from the aspects of inhibiting cell apoptosis, promoting HA degradation, inducing inflammatory responses and related phosphorylation, adjusting cellular microenvironment, and regulating tissue fibrosis. The diagnosis and treatment strategies targeting CEMIP are also summarized. The various functions of CEMIP show its great potential application value.
Collapse
Affiliation(s)
- Yang Liu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Gang Hu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuetong Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinyi Kong
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Kaming Yang
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhenlin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wanwen Lao
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiaxin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jianhua Zhong
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shitong Zhang
- Department of General Practice, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuxin Leng
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Changlong Bi
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Aixia Zhai
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
7
|
Han X, Yin M, Gong C, Zhang C, Zhu G, Hu M, Tan K, Jiang L, Wang G, Li L. A1BG-AS1 promotes the biological functions of osteosarcoma cells via regulating the microRNA-148a-3p/USP22 axis and stabilizing the expression of SIRT1 through deubiquitinase function. Expert Opin Ther Targets 2023; 27:1017-1029. [PMID: 37747800 DOI: 10.1080/14728222.2023.2263908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 09/24/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND The study aims to explore the role of A1BG antisense RNA 1 (A1BG-AS1), microRNA (miR)-148a-3p and ubiquitin-specific protease 22 (USP22) on osteosarcoma (OS) cell growth. RESEARCH DESIGN & METHODS A1BG-AS1, miR-148a-3p, USP22, and silent information regulator 2 homolog 1 (SIRT1) levels in OS tissues and cells were determined. The effects of A1BG-AS1, miR-148a-3p, and USP22 on the biological functions of OS cells were examined by functional assays. In vivo assay was conducted to observe the effect of A1BG-AS1 on OS growth in vitro. The relationship of A1BG-AS1, miR-148a-3p, and USP22 was analyzed by bioinformatics analysis, RNA-fluorescence in situ hybridization, luciferase activity, and RNA binding protein immunoprecipitation assays. The relation between USP22 and SIRT1 was evaluated by immunoprecipitation. RESULTS A1BG-AS1 and USP22 were highly expressed, and miR-148a-3p was lowly expressed in OS tissues and cells. Down-regulation of A1BG-AS1 and USP22 or up-regulation of miR-148a-3p impaired the malignant behaviors of OS cells. A1BG-AS1 sponged miR-148a-3p, and miR-148a-3p targeted USP22, thereby inhibiting USP22 expression. Up-regulating USP22 reversed the A1BG-AS1 suppression-induced phenotypic inhibition of OS cells. USP22 affected the biological functions of OS cells by deubiquitinating SIRT1. CONCLUSION A1BG-AS1 facilitates the biological functions of OS cells via mediating the miR-148a-3p/USP22 axis.
Collapse
Affiliation(s)
- Xiuxin Han
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Mengfan Yin
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
- Department of Orthopedic Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Chen Gong
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Genbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Mengxue Hu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - La Jiang
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| |
Collapse
|
8
|
Mo X, Shen X, Mo X, Yu F, Tan W, Deng Z, He J, Luo Z, Chen Z, Yang J. CEMIP promotes small cell lung cancer proliferation by activation of glutamine metabolism via FBXW7/c-Myc-dependent axis. Biochem Pharmacol 2023; 209:115446. [PMID: 36746261 DOI: 10.1016/j.bcp.2023.115446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Small cell lung cancer (SCLC) is the most malignant lung cancer with rapid growth and early metastasis, but still lacks effective targeted therapies to improve the prognosis. Here, we demonstrated that a novel oncogenic protein, cell migration inducing hyaluronic binding protein (CEMIP), was robustly overexpressed in SCLC tissues than that in noncancerous tissues and high expression of CEMIP predicted poor outcomes in clinical specimens and in large sample size cohorts from public databases (GEPIA 2 and CPTAC). Liquid chromatography mass spectrometry (LC-MS) and in vitro/in vivo functional assays indicated that CEMIP contributed to the proliferation by increasing glutamine consumption and their metabolites (glutamate and glutathione) levels in SCLC cells. Moreover, the addition of a GLS1 inhibitor CB-839 dramatically reduced CEMIP-induced SCLC cell proliferation. Mechanistically, beyond as a scaffold protein, CEMIP facilitates glutamine-dependent cell proliferation through inhibiting c-Myc ubiquitination and increasing c-Myc stabilization and nuclear accumulation via hindering the interaction between FBXW7 (a E3 ubiquitin ligase) and its target substrate c-Myc. Taken together, our findings reveal a novel oncogenic role of CEMIP in sustaining SCLC growth via FBXW7/c-Myc-dependent axis, and provide new evidence that inhibition of CEMIP might be a potential therapeutic strategy for the treatment of SCLC.
Collapse
Affiliation(s)
- Xiaoxiang Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China; Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Xiaoju Shen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiaocheng Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Fei Yu
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Weidan Tan
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zhihua Deng
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University for Nationalities, Baise, 533000 Guangxi, China
| | - Jingchuan He
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zhuo Luo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zhiquan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Jie Yang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
9
|
Chen H, Wang Q, Liu J, Chen Y, Zhang Q, Chai L, Wang Y, Li D, Qiu Y, Li M. CEMIP as a prognostic biomarker for cancers: a meta- and bioinformatic analysis. Expert Rev Mol Diagn 2022; 22:1107-1115. [PMID: 36631437 DOI: 10.1080/14737159.2022.2168191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Cell migration-inducing and hyaluronan-binding protein (CEMIP) is overexpressed in several cancers and is related to prognosis in cancer patients. Here, we conducted a meta-analysis to explore the prognostic effects of CEMIP in cancer patients. METHODS Relevant published studies were systematically searched in four databases. The role of CEMIP was evaluated using pooled hazard ratios (HRs), odd ratios (ORs), and 95% confidence intervals (95% CIs). The Cancer Genome Atlas (TCGA) was used to investigate the prognostic value of CEMIP in various cancers. RESULTS 11 literatures with 1355 patients were included in this meta-analysis. The results showed that overexpression of CEMIP was significantly associated with poor OS (HR = 3.03; 95% CI: 2.00-4.59; p < 0.001), DFS (HR = 3.38; 95% CI: 2.41-4.74; p < 0.001). Elevated CEMIP expression is associated with advanced clinical stage, lymph node metastasis, and poor histological grade. In addition, TCGA datasets were used to verify that CEMIP was found highly expressed in multiple cancers and was associated with poorer survival. CONCLUSION The results demonstrated that CEMIP could be a novel prognostic biomarker for cancer patients. However, because the included studies mainly focused on Asian populations, further research is needed to verify its applicability.
Collapse
Affiliation(s)
- Huan Chen
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qingting Wang
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jin Liu
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuqian Chen
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qianqian Zhang
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Limin Chai
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yan Wang
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Danyang Li
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuanjie Qiu
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Manxiang Li
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Shen X, Mo X, Tan W, Mo X, Li L, Yu F, He J, Deng Z, Xing S, Chen Z, Yang J. KIAA1199 Correlates With Tumor Microenvironment and Immune Infiltration in Lung Adenocarcinoma as a Potential Prognostic Biomarker. Pathol Oncol Res 2022; 28:1610754. [PMID: 36419650 PMCID: PMC9676226 DOI: 10.3389/pore.2022.1610754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/25/2022] [Indexed: 09/05/2023]
Abstract
Background: KIAA1199 has been considered a key regulator of carcinogenesis. However, the relationship between KIAA1199 and immune infiltrates, as well as its prognostic value in lung adenocarcinoma (LUAD) remains unclear. Methods: The expression of KIAA1199 and its influence on tumor prognosis were analyzed using a series of databases, comprising TIMER, GEPIA, UALCAN, LCE, Prognoscan and Kaplan-Meier Plotter. Further, immunohistochemistry (IHC), western blot (WB) and receiver operating characteristic (ROC) curve analyses were performed to verify our findings. The cBioPortal was used to investigate the genomic alterations of KIAA1199. Prediction of candidate microRNA (miRNAs) and transcription factor (TF) targeting KIAA1199, as well as GO and KEGG analyses, were performed based on LinkedOmics. TIMER and TISIDB databases were used to explore the relationship between KIAA1199 and tumor immune infiltration. Results: High expression of KIAA1199 was identified in LUAD and Lung squamous cell carcinoma (LUSC) patients. High expression of KIAA1199 indicated a worse prognosis in LUAD patients. The results of IHC and WB analyses showed that the expression level of KIAA1199 in tumor tissues was higher than that in adjacent tissues. GO and KEGG analyses indicated KIAA1199 was mainly involved in extracellular matrix (ECM)-receptor interaction and extracellular matrix structure constituent. KIAA1199 was positively correlated with infiltrating levels of CD4+ T cells, macrophages, neutrophil cells, dendritic cells, and showed positive relationship with immune marker subsets expression of a variety of immunosuppressive cells. Conclusion: High expression of KIAA1199 predicts a poor prognosis of LUAD patients. KIAA1199 might exert its carcinogenic role in the tumor microenvironment via participating in the extracellular matrix formation and regulating the infiltration of immune cells in LUAD. The results indicate that KIAA1199 might be a novel biomarker for evaluating prognosis and immune cell infiltration in LUAD.
Collapse
Affiliation(s)
- Xiaoju Shen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaocheng Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Weidan Tan
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiaoxiang Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Li Li
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, China
| | - Fei Yu
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Jingchuan He
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhihua Deng
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Shangping Xing
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiquan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Jie Yang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
Guan J, Liu X, Wang K, Jia Y, Yang B. Identification of a novel necroptosis-associated miRNA signature for predicting the prognosis in head and neck squamous cell carcinoma. Open Med (Wars) 2022; 17:1682-1698. [PMID: 36349193 PMCID: PMC9601379 DOI: 10.1515/med-2022-0575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 09/23/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive malignancies that have a poor prognosis. Necroptosis has been demonstrated in recent years to be a form of inflammatory cell death occurring in multicellular organism, which plays complex roles in cancer. However, the expression of necroptosis-related miRNAs and genes in HNSCC and their correlations with prognosis remain unclear. In this study, R software was used to screen differentially expressed miRNAs downloaded from The Cancer Genome Atlas. A prognostic model containing six necroptosis-related miRNAs (miR-141-3p, miR-148a-3p, miR-331-3p, miR-543, miR-425-5p, and miR-7-5p) was generated, whose risk score was validated as an independent prognostic factor for HNSCC. Target genes of the key miRNAs were obtained from TargetScan, miRDB, and miRTarBase, and 193 genes in the intersection of the three databases were defined as consensus genes. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses indicated that the composition of the tumor microenvironment as well as specific pathways may be closely related to necroptosis in HNSCC. Nine key genes were also obtained by the MCODE and cytoHubba plug-ins of Cytoscape: PIK3CD, NRAS, PTK2, IRS2, IRS1, PARP1, KLF4, SMAD2, and DNMT1. A prognostic model formed by the key gene was also established, which can efficiently predict the overall survival of HNSCC patients. In conclusion, necroptosis-related miRNAs and genes play important roles in tumor development and metastasis and can be used to predict the prognosis of HNSCC.
Collapse
Affiliation(s)
- Jiezhong Guan
- Department of Prosthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Xinyu Liu
- Department of Prosthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Kang Wang
- Department of Prosthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Yiqun Jia
- Stomatology Center, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Bo Yang
- Department of Prosthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Domanegg K, Sleeman JP, Schmaus A. CEMIP, a Promising Biomarker That Promotes the Progression and Metastasis of Colorectal and Other Types of Cancer. Cancers (Basel) 2022; 14:cancers14205093. [PMID: 36291875 PMCID: PMC9600181 DOI: 10.3390/cancers14205093] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary CEMIP (cell migration-inducing and hyaluronan-binding protein) has been implicated in the pathogenesis of numerous diseases, including colorectal and other forms of cancer. The molecular functions of CEMIP are currently under investigation and include the degradation of the extracellular matrix component hyaluronic acid (HA), as well as the regulation of a number of signaling pathways. In this review, we survey our current understanding of how CEMIP contributes to tumor growth and metastasis, focusing particularly on colorectal cancer, for which it serves as a promising biomarker. Abstract Originally discovered as a hypothetical protein with unknown function, CEMIP (cell migration-inducing and hyaluronan-binding protein) has been implicated in the pathogenesis of numerous diseases, including deafness, arthritis, atherosclerosis, idiopathic pulmonary fibrosis, and cancer. Although a comprehensive definition of its molecular functions is still in progress, major functions ascribed to CEMIP include the depolymerization of the extracellular matrix component hyaluronic acid (HA) and the regulation of a number of signaling pathways. CEMIP is a promising biomarker for colorectal cancer. Its expression is associated with poor prognosis for patients suffering from colorectal and other types of cancer and functionally contributes to tumor progression and metastasis. Here, we review our current understanding of how CEMIP is able to foster the process of tumor growth and metastasis, focusing particularly on colorectal cancer. Studies in cancer cells suggest that CEMIP exerts its pro-tumorigenic and pro-metastatic activities through stimulating migration and invasion, suppressing cell death and promoting survival, degrading HA, regulating pro-metastatic signaling pathways, inducing the epithelial–mesenchymal transition (EMT) program, and contributing to the metabolic reprogramming and pre-metastatic conditioning of future metastatic microenvironments. There is also increasing evidence indicating that CEMIP may be expressed in cells within the tumor microenvironment that promote tumorigenesis and metastasis formation, although this remains in an early stage of investigation. CEMIP expression and activity can be therapeutically targeted at a number of levels, and preliminary findings in animal models show encouraging results in terms of reduced tumor growth and metastasis, as well as combating therapy resistance. Taken together, CEMIP represents an exciting new player in the progression of colorectal and other types of cancer that holds promise as a therapeutic target and biomarker.
Collapse
Affiliation(s)
- Kevin Domanegg
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leopoldshafen, Germany
- Correspondence:
| | - Anja Schmaus
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
13
|
Chen Y, Liu W, Li D, Cao Y, Wang W, Li C, An R. MiRNA-148a inhibits cell growth and drug resistance by regulating WNT10a expression in renal cell carcinoma. Transl Androl Urol 2022; 11:996-1006. [PMID: 35958896 PMCID: PMC9360522 DOI: 10.21037/tau-22-464] [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: 07/01/2022] [Accepted: 07/14/2022] [Indexed: 12/05/2022] Open
Abstract
Background We aimed to explore miR-148a exerts a tumor suppressor effect and arsenic trioxide (As2O3) sensitivity on renal cell carcinoma (RCC). Methods We performed polymerase chain reaction (PCR) on 42 pairs of tumor and paracancerous samples collected from RCC patients to investigate the miR-148a expression; meanwhile, we analyzed the interplay between clinical indicators and miR-148a expression of RCC. Then, the influence of miR-148a overexpression on the functions of RCC cells were analyze using transwell migration assay, Cell Counting Kit-8 (CCK-8), and cell wound healing assay. Furthermore, the ability of miR-148a to sensitize Caki-1 cells treated with As2O3 were detected using flow cytometry. Finally, the relevant mechanism of miR-148a on the downstream gene Wnt family member 10A (WNT10a) was explored by cell reverse method. Results The results from RCC patients indicated a significantly lower miR-148a level than adjacent tissues. The low miR-148a expression increased prevalence of distant metastasis and decreased survival rate compared to those with high expression in patients. In the RCC cell lines, the proliferation and metastasis ability of the miR-148a mimic group was remarkably lower than the miR-NC group. At the same time, it was verified that WNT10a was remarkably higher cell lines and RCC tissues; and negatively related to miR-148a expression. In addition, miR-148a mimics were found to remarkably reduce the protein expression of WNT10a. In the cell reverse experiment, overexpression of WNT10a was confirmed to offset the miR-148a mimics effect on metastasis and proliferation of RCC cells. In addition, an increase in relative apoptosis was detected in As2O3 treated with/without miR-148a mimics for 48 hours, and apoptosis was significantly reduced after transfection with WNT10a in the Caki-1 cell line and significantly reduced after combined treatment. Conclusions The study revealed that miR-148a is associated with distant metastases and leads to poor prognosis in RCC patients. Moreover, miR-148a inhibit the malignant progression and increase the sensitivity of RCC cells to As2O3 by regulating WNT10a.
Collapse
Affiliation(s)
- Yongsheng Chen
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenhua Liu
- Intensive Care Unit (ICU) Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dechao Li
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yan Cao
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wentao Wang
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Changfu Li
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ruihua An
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|