1
|
Kozole SL, Beningo KA. Myosin Light Chains in the Progression of Cancer. Cells 2024; 13:2081. [PMID: 39768172 PMCID: PMC11674124 DOI: 10.3390/cells13242081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The myosin light chains (MLCs) of non-muscle myosin II are known to regulate cellular architecture and generate cellular forces; they also have an increasingly emerging role in the progression of cancer. The phosphorylation state of the myosin light chains controls the activity of myosins that are implicated in invasion and proliferation. In cancers, when proliferation is greatly increased, cytokinesis relies on phosphorylated light chains to activate the contractile forces used to separate the cells. Likewise, during metastasis, kinase pathways culminate in aligning MLC structures for enhanced cell motility through stress fiber contraction and the accumulation of myosin filaments at the leading edge. This review summarizes the myosin light chain family members known to promote cancer progression and evidence of how their altered activities change the behavior of cells involving the mechanical-based processes of proliferation and cell movements during metastasis. In addition, myosin light chains impact the immune response to cancers and currently serve as biomarkers in staging this disease; a brief summary of these topics is provided at the end of the review.
Collapse
Affiliation(s)
| | - Karen A. Beningo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA;
| |
Collapse
|
2
|
Sun R, Tan L, Ding X, A J, Xue Z, Cai X, Li S, Guo T. A pathway activity-based proteomic classifier stratifies prostate tumors into two subtypes. Clin Proteomics 2023; 20:50. [PMID: 37950160 PMCID: PMC10638831 DOI: 10.1186/s12014-023-09441-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Prostate cancer (PCa) is the second most common cancer in males worldwide. The risk stratification of PCa is mainly based on morphological examination. Here we analyzed the proteome of 667 tumor samples from 487 Chinese PCa patients and characterized 9576 protein groups by PulseDIA mass spectrometry. Then we developed a pathway activity-based classifier concerning 13 proteins from seven pathways, and dichotomized the PCa patients into two subtypes, namely PPS1 and PPS2. PPS1 is featured with enhanced innate immunity, while PPS2 with suppressed innate immunity. This classifier exhibited a correlation with PCa progression in our cohort and was further validated by two published transcriptome datasets. Notably, PPS2 was significantly correlated with poor biochemical recurrence (BCR)/metastasis-free survival (log-rank P-value < 0.05). The PPS2 was also featured with cell proliferation activation. Together, our study presents a novel pathway activity-based stratification scheme for PCa.
Collapse
Affiliation(s)
- Rui Sun
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China.
| | - Lingling Tan
- Westlake Omics (Hangzhou) Biotechnology Co., Ltd., Hangzhou, 310024, China
| | - Xuan Ding
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China
| | - Jun A
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China
| | - Zhangzhi Xue
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China
| | - Xue Cai
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China
| | - Sainan Li
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China
| | - Tiannan Guo
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, 310024, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, 310024, China.
| |
Collapse
|
3
|
Luo L, Wu A, Shu X, Liu L, Feng Z, Zeng Q, Wang Z, Hu T, Cao Y, Tu Y, Li Z. Hub gene identification and molecular subtype construction for Helicobacter pylori in gastric cancer via machine learning methods and NMF algorithm. Aging (Albany NY) 2023; 15:11782-11810. [PMID: 37768204 PMCID: PMC10683617 DOI: 10.18632/aging.205053] [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: 02/28/2023] [Accepted: 07/19/2023] [Indexed: 09/29/2023]
Abstract
Helicobacter pylori (HP) is a gram-negative and spiral-shaped bacterium colonizing the human stomach and has been recognized as the risk factor of gastritis, peptic ulcer disease, and gastric cancer (GC). Moreover, it was recently identified as a class I carcinogen, which affects the occurrence and progression of GC via inducing various oncogenic pathways. Therefore, identifying the HP-related key genes is crucial for understanding the oncogenic mechanisms and improving the outcomes of GC patients. We retrieved the list of HP-related gene sets from the Molecular Signatures Database. Based on the HP-related genes, unsupervised non-negative matrix factorization (NMF) clustering method was conducted to stratify TCGA-STAD, GSE15459, GSE84433 samples into two clusters with distinct clinical outcomes and immune infiltration characterization. Subsequently, two machine learning (ML) strategies, including support vector machine-recursive feature elimination (SVM-RFE) and random forest (RF), were employed to determine twelve hub HP-related genes. Beyond that, receiver operating characteristic and Kaplan-Meier curves further confirmed the diagnostic value and prognostic significance of hub genes. Finally, expression of HP-related hub genes was tested by qRT-PCR array and immunohistochemical images. Additionally, functional pathway enrichment analysis indicated that these hub genes were implicated in the genesis and progression of GC by activating or inhibiting the classical cancer-associated pathways, such as epithelial-mesenchymal transition, cell cycle, apoptosis, RAS/MAPK, etc. In the present study, we constructed a novel HP-related tumor classification in different datasets, and screened out twelve hub genes via performing the ML algorithms, which may contribute to the molecular diagnosis and personalized therapy of GC.
Collapse
Affiliation(s)
- Lianghua Luo
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ahao Wu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xufeng Shu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Li Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zongfeng Feng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qingwen Zeng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhonghao Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Tengcheng Hu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Cao
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhengrong Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
4
|
Chan TC, Pan CT, Hsieh HY, Vejvisithsakul PP, Wei RJ, Yeh BW, Wu WJ, Chen LR, Shiao MS, Li CF, Shiue YL. The autocrine glycosylated-GREM1 interacts with TGFB1 to suppress TGFβ/BMP/SMAD-mediated EMT partially by inhibiting MYL9 transactivation in urinary carcinoma. Cell Oncol (Dordr) 2023; 46:933-951. [PMID: 36920729 DOI: 10.1007/s13402-023-00788-8] [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] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
PURPOSE Urothelial carcinoma (UC) is a common disease in developed counties. This study aimed to identify autocrine roles and signaling pathways of gremlin 1, DAN family BMP antagonist (GREM1), which inhibits tumor growth and epithelial-mesenchymal transition (EMT) in UC. METHODS Systematic in vitro and in vivo studies using genetic engineering, different urinary bladder urothelial carcinoma (UBUC)-derived cell lines, and mouse models were performed, respectively. Further, primary upper tract urothelial carcinoma (UTUC) and UBUC specimens were evaluated by immunohistochemistry. RESULTS GREM1 protein levels conferred better disease-specific and metastasis-free survival rates and played an independent prognostic factor in UTUC and UBUC. Hypermethylation is the primary cause of low GREM1 levels. In different UBUC-derived cell lines, the autocrine/secreted and glycosylated GREM1 interacted with transforming growth factor beta 1 (TGFB1) and inhibited TGFβ/BMP/SMAD signaling and myosin light chain 9 (MYL9) transactivation, subsequently cell proliferation and epithelial-mesenchymal transition (EMT). Secreted and glycosylated GREM1 also suppressed tumor growth, metastasis, and MYL9 levels in the mouse model. Instead, cytosolic GREM1 promoted cell proliferation and EMT by activating the tumor necrosis factor (TNF)/AKT/nuclear factor kappa B (NFκB) axis. CONCLUSIONS Clinical associations, animal models, and in vitro indications provided solid evidence to show that the epithelial autocrine GREM1 is a novel tumor suppressor in UCs. The glycosylated-GREM1 hampered cell proliferation, migration, invasion, and in vitro angiogenesis through interaction with TGFB1 to inactivate TGFβ/BMP/SMAD-mediated EMT in an autocrine manner.
Collapse
Affiliation(s)
- Ti-Chun Chan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 71004, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 71004, Taiwan
| | - Cheng-Tang Pan
- Institute of Precision Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Department of Mechanical and Electro-Mechanical Engineering, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Institute of Advanced Semiconductor Packaging and Testing, College of Semiconductor and Advanced Technology Research, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Hsin-Yu Hsieh
- Institute of Precision Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Pichpisith Pierre Vejvisithsakul
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Ren-Jie Wei
- Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan
- Institute of Medical Science and Technology, School of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, 83102, Taiwan
| | - Bi-Wen Yeh
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Wen-Jeng Wu
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Lih-Ren Chen
- Division of Physiology, Livestock Research Institute, Tainan, 71246, Taiwan
| | - Meng-Shin Shiao
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Chien-Feng Li
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 71004, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 71004, Taiwan.
- Institute of Precision Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
| | - Yow-Ling Shiue
- Institute of Precision Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
- Institute of Biomedical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
5
|
Matellan C, Lachowski D, Cortes E, Chiam KN, Krstic A, Thorpe SD, Del Río Hernández AE. Retinoic acid receptor β modulates mechanosensing and invasion in pancreatic cancer cells via myosin light chain 2. Oncogenesis 2023; 12:23. [PMID: 37130839 PMCID: PMC10154384 DOI: 10.1038/s41389-023-00467-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 05/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common and lethal form of pancreatic cancer, characterised by stromal remodelling, elevated matrix stiffness and high metastatic rate. Retinoids, compounds derived from vitamin A, have a history of clinical use in cancer for their anti-proliferative and differentiation effects, and more recently have been explored as anti-stromal therapies in PDAC for their ability to induce mechanical quiescence in cancer associated fibroblasts. Here, we demonstrate that retinoic acid receptor β (RAR-β) transcriptionally represses myosin light chain 2 (MLC-2) expression in pancreatic cancer cells. As a key regulatory component of the contractile actomyosin machinery, MLC-2 downregulation results in decreased cytoskeletal stiffness and traction force generation, impaired response to mechanical stimuli via mechanosensing and reduced ability to invade through the basement membrane. This work highlights the potential of retinoids to target the mechanical drivers of pancreatic cancer.
Collapse
Affiliation(s)
- Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Ernesto Cortes
- Department of Physiology, School of Medicine, Autonomous University of Madrid, 28029, Madrid, Spain
| | - Kai Ning Chiam
- UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Aleksandar Krstic
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Stephen D Thorpe
- UCD School of Medicine, University College Dublin, Dublin, Ireland.
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland.
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland.
| | - Armando E Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
6
|
MYL5 as a Novel Prognostic Marker is Associated with Immune Infiltrating in Breast Cancer: A Preliminary Study. Breast J 2023; 2023:9508632. [PMID: 36846347 PMCID: PMC9957649 DOI: 10.1155/2023/9508632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/19/2023]
Abstract
Background Myosin light chain plays a vital regulatory function in a large-scale cellular physiological procedure, however, the role of myosin light chain 5 (MYL5) in breast cancer has not been reported. In this study, we aimed to elucidate the effects of MYL5 on clinical prognosis and immune cell infiltration, and further explore the potential mechanism in breast cancer patients. Methods In this study, we first explored the expression pattern and prognostic value of MYL5 in breast cancer across multiple databases, including Oncomine, TCGA, GTEx, GEPIA2, PrognoScan, and Kaplan-Meier Plotter. The correlations of MYL5 expression with immune cell infiltration and associational gene markers in breast cancer were analyzed by using the TIMER, TIMER2.0, and TISIDB databases. The enrichment and prognosis analysis of MYL5-related genes were implemented by using LinkOmics datasets. Results We found that there was a low expression of MYL5 in breast cancer than in corresponding normal tissue by analyzing the data from Oncomine and TCGA datasets. Furthermore, research showed the prognosis of the MYL5 high-expression group was better than the low-expression group in breast cancer patients. Furthermore, MYL5 expression is markedly related to the tumor-infiltrating immune cells (TIICs), including cancer-associated fibroblast, B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, and dendritic cell, and related to immune molecules as well as the associated gene markers of TIICs. Conclusion MYL5 can serve as a prognostic signature in breast cancer and is associated with immune infiltration. This study first offers a relatively comprehensive understanding of the oncogenic roles of MYL5 for breast cancer.
Collapse
|
7
|
Chen J, Meng Y, Huang X, Liao X, Tang X, Xu Y, Li J. Potential effective diagnostic biomarker in patients with primary and metastatic small intestinal neuroendocrine tumors. Front Genet 2023; 14:1110396. [PMID: 37091799 PMCID: PMC10119396 DOI: 10.3389/fgene.2023.1110396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Background: Small intestinal neuroendocrine tumors (SI-NETs) are the most common malignant tumors of the small intestine, with many patients presenting with metastases and their incidence increasing. We aimed to find effective diagnostic biomarkers for patients with primary and metastatic SI-NETs that could be applied for clinical diagnosis. Methods: We downloaded GSE65286 (training set) and GSE98894 (test set) from the GEO database and performed differential gene expression analysis to obtain differentially expressed genes (DEGs) and differentially expressed long non-coding RNAs (DElncRNAs). The functions and pathways involved in these genes were further explored by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. In addition, a global regulatory network involving dysregulated genes in SI-NETs was constructed based on RNAInter and TRRUST v2 databases, and the diagnostic power of hub genes was identified by receiver operating characteristic curve (ROC). Results: A total of 2,969 DEGs and DElncRNAs were obtained in the training set. Enrichment analysis revealed that biological processes (BPs) and KEGG pathways were mainly associated with cancer. Based on gene set enrichment analysis (GSEA), we obtained five BPs (cytokinesis, iron ion homeostasis, mucopolysaccharide metabolic process, platelet degranulation and triglyceride metabolic process) and one KEGG pathway (ppar signaling pathway). In addition, the core set of dysregulated genes obtained included MYL9, ITGV8, FGF2, FZD7, and FLNC. The hub genes were upregulated in patients with primary SI-NETs compared to patients with metastatic SI-NETs, which is consistent with the training set. Significantly, the results of ROC analysis showed that the diagnostic power of the hub genes was strong in both the training and test sets. Conclusion: In summary, we constructed a global regulatory network in SI-NETs. In addition, we obtained the hub genes including MYL9, ITGV8, FGF2, FZD7, and FLNC, which may be useful for the diagnosis of patients with primary and metastatic SI-NETs.
Collapse
|
8
|
Zhang Y, Li Y, Zuo Z, Li T, An Y, Zhang W. An epithelial-mesenchymal transition-related mRNA signature associated with the prognosis, immune infiltration and therapeutic response of colon adenocarcinoma. Pathol Oncol Res 2023; 29:1611016. [PMID: 36910014 PMCID: PMC9998511 DOI: 10.3389/pore.2023.1611016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/14/2023] [Indexed: 03/14/2023]
Abstract
Background: Epithelial-mesenchymal transition (EMT) is closely associated with cancer cell metastasis. Colon adenocarcinoma (COAD) is one of the most common malignancies in the world, and its metastasis leading to poor prognosis remains a challenge for clinicians. The purpose of this study was to explore the prognostic value of EMT-related genes (EMTRGs) by bioinformatics analysis and to develop a new EMTRGs prognostic signature for COAD. Methods: The TCGA-COAD dataset was downloaded from the TCGA portal as the training cohort, and the GSE17538 and GSE29621 datasets were obtained from the GEO database as the validation cohort. The best EMTRGs prognostic signature was constructed by differential expression analysis, Cox, and LASSO regression analysis. Gene set enrichment analysis (GSEA) is used to reveal pathways that are enriched in high-risk and low-risk groups. Differences in tumor immune cell levels were analyzed using microenvironmental cell population counter and single sample gene set enrichment analysis. Subclass mapping analysis and Genomics of Drug Sensitivity in Cancer were applied for prediction of immunotherapy response and chemotherapy response, respectively. Results: A total of 77 differentially expressed EMTRGs were identified in the TCGA-COAD cohort, and they were significantly associated with functions and pathways related to cancer cell metastasis, proliferation, and apoptosis. We constructed EMTRGs prognostic signature with COMP, MYL9, PCOLCE2, SCG2, and TIMP1 as new COAD prognostic biomarkers. The high-risk group had a poorer prognosis with enhanced immune cell infiltration. The GSEA demonstrated that the high-risk group was involved in "ECM Receptor Interaction," "WNT Signaling Pathway" and "Colorectal Cancer." Furthermore, patients with high risk scores may respond to anti-CTLA4 therapy and may be more resistant to targeted therapy agents BI 2536 and ABT-888. Conclusion: Together, we developed a new EMTRGs prognostic signature that can be an independent prognostic factor for COAD. This study has guiding implications for individualized counseling and treatment of COAD patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Yan Li
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Zan Zuo
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Ting Li
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Ying An
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Wenjing Zhang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.,Department of Medical Oncology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
9
|
Matsushita K, Kobayashi S, Akita H, Konno M, Asai A, Noda T, Iwagami Y, Asaoka T, Gotoh K, Mori M, Doki Y, Eguchi H, Ishii H. Clinicopathological significance of MYL9 expression in pancreatic ductal adenocarcinoma. Cancer Rep (Hoboken) 2022; 5:e1582. [PMID: 34821071 PMCID: PMC9575502 DOI: 10.1002/cnr2.1582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma is one of the most aggressive malignancies, and often involves invasion and distant metastasis from the early tumor stages. Myosin II reportedly plays a key role in regulating tumor progression and metastasis. AIMS We examined whether myosin regulatory light polypeptide 9 (MYL9) regulates cancer cell proliferation. METHODS AND RESULTS To investigate the expression pattern and clinical significance of MYL9 in pancreatic ductal adenocarcinoma, we performed immunohistochemical analysis of samples collected from 101 patients with pancreatic ductal adenocarcinoma. The expression of MYL9 was investigated to evaluate its functional role and contribution to proliferation and apoptosis in pancreatic ductal adenocarcinoma cells in vitro. The results showed that MYL9 was predominantly expressed in the cytoplasm and membrane of pancreatic ductal adenocarcinoma cells. Multivariate analysis indicated that MYL9 acted as an independent prognostic factor for overall survival and distant metastasis-free survival. MYL9 expression was strongly associated with malignancy in in vitro analyses, including proliferation and anti-apoptotic activities. CONCLUSIONS Our findings suggest that MYL9 is an independent prognostic factor of pancreatic ductal adenocarcinoma. MYL9 is a crucial biomarker and potential therapeutic target for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Katsunori Matsushita
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Shogo Kobayashi
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Hirofumi Akita
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Masamitsu Konno
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Ayumu Asai
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Artificial Intelligence Research CenterThe Institute of Scientific and Industrial Research, Osaka UniversityIbarakiOsakaJapan
- Institute of Scientific and Industrial ResearchOsaka UniversityIbarakiOsakaJapan
| | - Takehiro Noda
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Yoshifumi Iwagami
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Tadafumi Asaoka
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Kunihito Gotoh
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Masaki Mori
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Department of Surgery and ScienceGraduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Hideshi Ishii
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| |
Collapse
|
10
|
Integrative Analysis of Bulk RNA-Seq and Single-Cell RNA-Seq Unveils the Characteristics of the Immune Microenvironment and Prognosis Signature in Prostate Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6768139. [PMID: 35909899 PMCID: PMC9325591 DOI: 10.1155/2022/6768139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022]
Abstract
The immune microenvironment is a culmination of the collaborative effort of immune cells and is important in cancer development. The underlying mechanisms of the tumor immune microenvironment in regulating prostate cancer (PRAD) are unclear. In the current study, 144 natural killer cell-related genes were identified using differential expression, single-sample gene set enrichment analysis, and weighted gene coexpression network analysis. Furthermore, VCL, ACTA2, MYL9, MYLK, MYH11, TPM1, ACTG2, TAGLN, and FLNC were selected as hub genes via the protein-protein interaction network. Based on the expression patterns of the hub genes, endothelial, epithelial, and tissue stem cells were identified as key cell subpopulations, which could regulate PRAD via immune response, extracellular signaling, and protein formation. Moreover, 27 genes were identified as prognostic signatures and used to construct the risk score model. Receiver operating characteristic curves revealed the good performance of the risk score model in both the training and testing datasets. Different chemotherapeutic responses were observed between the low- and high-risk groups. Additionally, a nomogram based on the risk score and other clinical features was established to predict the 1-, 3-, and 5-year progression-free interval of patients with PRAD. This study provides novel insights into the molecular mechanisms of the immune microenvironment and its role in the pathogenesis of PARD. The identification of key cell subpopulations has a potential therapeutic and prognostic use in PRAD.
Collapse
|
11
|
Koyaman-Nasu R, Wang Y, Hasegawa I, Endo Y, Nakayama T, Kimura MY. The cellular and molecular basis of CD69 function in anti-tumor immunity. Int Immunol 2022; 34:555-561. [PMID: 35689672 DOI: 10.1093/intimm/dxac024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy utilizes our immune system to attack cancer cells and is an extremely promising strategy for cancer treatment. Although immune-checkpoint blockade, such as anti-PD-1 antibody (Ab), has demonstrated significant enhancement of anti-tumor immunity and has induced notable clinical outcomes, its response rates remain low, and adverse effects are always a matter of concern; therefore, new targets for cancer immunotherapy are always desired. In this situation, new concepts are needed to fuel the investigation of new target molecules for cancer immunotherapy. We propose that CD69 is one such target molecule. CD69 is known to be an activation marker of leukocytes and is also considered a crucial regulator of various immune responses through its interacting proteins. CD69 promotes T cell retention in lymphoid tissues via sphingosine-1-phosphate receptor 1 (S1P1) internalization and also plays roles in the pathogenesis of inflammatory disorders through interacting with its functional ligands Myl9/12 (myosin light chains 9, 12a and 12b). In anti-tumor immunity, CD69 is known to be expressed on T cells in the tumor microenvironment (TME) and tumor-draining lymph nodes (TDLNs). We revealed that CD69 negatively regulates the effector function of intratumoral T cells and importantly controls the 'exhaustion' of CD8 T cells. In addition, we and others showed that either CD69 deficiency or the administration of anti-CD69 monoclonal antibody enhances anti-tumor immunity. Thus, CD69 is an attractive target for cancer immunotherapy.
Collapse
Affiliation(s)
- Ryo Koyaman-Nasu
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yangsong Wang
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Ichita Hasegawa
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yukihiro Endo
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Motoko Y Kimura
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
12
|
Feng M, Dong N, Zhou X, Ma L, Xiang R. Myosin light chain 9 promotes the proliferation, invasion, migration and angiogenesis of colorectal cancer cells by binding to Yes-associated protein 1 and regulating Hippo signaling. Bioengineered 2022; 13:96-106. [PMID: 34974798 PMCID: PMC8805887 DOI: 10.1080/21655979.2021.2008641] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Colorectal cancer is a common type of cancer with high incidence and poor prognosis. Increased expression of myosin light chain 9 (MYL9) has been reported in early-stage and recurrent colorectal cancer tissues. This study aimed to investigate the precise role of MYL9 on the progression of colorectal cancer. MYL9 expression in several colorectal cancer cell lines was detected by Western blotting and RT-qPCR. Following MYL9 overexpression or knockdown, MYL9 expression was determined via RT-qPCR. Cell proliferation was detected with Cell Counting Kit-8 assay. Cell invasion, migration and angiogenesis were, respectively, examined with transwell, wound healing and tube formation assays. The binding between MYL9 and Yes-associated protein 1 (YAP1) was verified by a co-immunoprecipitation assay. The expression of YAP1, connective tissue growth factor and cysteine-rich angiogenic inducer 61 was examined by Western blotting. Subsequently, YAP1 silencing or Hippo antagonist was performed to clarify the regulatory mechanisms of MYL9 in colorectal cancer progression. Experimental results showed that MYL9 expression was elevated in colorectal cancer cell lines. MYL9 overexpression promoted cell proliferation, invasion, migration and angiogenesis, while silencing of MYL9 exerted the opposite effects. Results of co-immunoprecipitation assay indicated that MYL9 could bind to YAP1. Further experiments revealed that MYL9 affected the expression of YAP1 and its downstream signaling proteins. Afterward, YAP1 knockdown or the addition of Hippo antagonist inhibited the proliferation, invasion, migration and angiogenesis of colorectal cancer cells. Overall, MYL9 promotes the proliferation, invasion, migration and angiogenesis of colorectal cancer cells by binding to YAP1 and thereby activating Hippo signaling.
Collapse
Affiliation(s)
- Min Feng
- Department of Gastroenterology, East Hospital of Zibo Central Hospital, Shandong Province, Zibo City, China
| | - Ningfei Dong
- Department of Gastroenterology, East Hospital of Zibo Central Hospital, Shandong Province, Zibo City, China
| | - Xin Zhou
- Department of Gastroenterology, East Hospital of Zibo Central Hospital, Shandong Province, Zibo City, China
| | - Lihong Ma
- Department of Gastroenterology, West Hospital of Zibo Central Hospital, Zibo, Shandong Province, China
| | - Rui Xiang
- Department of Gastroenterology, West Hospital of Zibo Central Hospital, Zibo, Shandong Province, China
| |
Collapse
|
13
|
Shen W, Wang G, Cooper GR, Jiang Y, Zhou X. The Epithelial and Stromal Immune Microenvironment in Gastric Cancer: A Comprehensive Analysis Reveals Prognostic Factors with Digital Cytometry. Cancers (Basel) 2021; 13:cancers13215382. [PMID: 34771544 PMCID: PMC8582557 DOI: 10.3390/cancers13215382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. Tumor heterogeneity continues to confound researchers' understanding of tumor growth and the development of an effective therapy. Digital cytometry allows interpretation of heterogeneous bulk tissue transcriptomes at the cellular level. We built a novel signature matrix to dissect epithelium and stroma signals using a scRNA-seq data set (GSE134520) for GC and then applied cell mixture deconvolution to estimate diverse epithelial, stromal, and immune cell proportions from bulk transcriptome data in four independent GC cohorts (GSE62254, GSE15459, GSE84437, and TCGA-STAD) from the GEO and TCGA databases. Robust computational methods were applied to identify strong prognostic factors for GC. We identified an EMEC population whose proportions were significantly higher in patients with stage I cancer than other stages, and it was predominantly present in tumor samples but not typically found in normal samples. We found that the ratio of EMECs to stromal cells and the ratio of adaptive T cells to monocytes were the most significant prognostic factors within the non-immune and immune factors, respectively. The STEM score, which unifies these two prognostic factors, was an independent prognostic factor of overall survival (HR = 0.92, 95% CI = 0.89-0.94, p=2.05×10-9). The entire GC cohort was stratified into three risk groups (high-, moderate-, and low-risk), which yielded incremental survival times (p<0.0001). For stage III disease, patients in the moderate- and low-risk groups experienced better survival benefits from radiation therapy ((HR = 0.16, 95% CI = 0.06-0.4, p<0.0001), whereas those in the high-risk group did not (HR = 0.49, 95% CI = 0.14-1.72, p=0.25). We concluded that the STEM score is a promising prognostic factor for gastric cancer.
Collapse
Affiliation(s)
- Wenjun Shen
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China;
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, CA 94035, USA
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou 515041, China
- Correspondence: (W.S.); (Y.J.)
| | - Guoyun Wang
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China;
| | - Georgia R. Cooper
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; (G.R.C.); (X.Z.)
| | - Yuming Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Correspondence: (W.S.); (Y.J.)
| | - Xin Zhou
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; (G.R.C.); (X.Z.)
- Department of Computer Science, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
14
|
Deng Y, Liu L, Feng W, Lin Z, Ning Y, Luo X. High Expression of MYL9 Indicates Poor Clinical Prognosis of Epithelial Ovarian Cancer. Recent Pat Anticancer Drug Discov 2021; 16:533-539. [PMID: 34551701 DOI: 10.2174/1574891x16666210706153740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The prognosis of Epithelial Ovarian Cancer (EOC) is poor, but the prognostic biomarkers are neither sensitive nor specific. Therefore, it is very important to search novel prognostic biomarkers for EOC. OBJECTIVES The present study aimed to investigate Myosin Light Chain 9(MYL9) expression in Epithelial Ovarian Cancer (EOC) tissues (including paraffin-embedded and fresh tissue samples) and its relationship with clinicopathological characteristics, as well as its potential prognostic value in patients with EOC. METHODS Between March 2009 and December 2018, all of 184 paraffin-embedded cancer tissues from patients with EOC and 41 paratumor tissues, pathologically confirmed at the Memorial Hospital of Sun Yat-sen University and Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, were collected for the present study and were assessed for MYL9 protein expression patterns using Immunohistochemistry (IHC). Furthermore, from August 2013 to November 2019, 16 fresh EOC tissues and their paired paratumor tissues, pathologically confirmed at the Integrated Hospital of Traditional Chinese Medicine, Southern Medical University were analyzed using Reverse-Transcription Quantitative PCR (RT-qPCR) to detect MYL9 mRNA expression levels. RESULTS The results showed that MYL9 expression was higher in cancer tissues compared with that in paratumor tissues, and MYL9 overexpression was associated with shorter Recurrence Free Survival (RFS) and Overall Survival (OS) of EOC patients. Furthermore, multivariate Cox model analysis indicated that MYL9 overexpression was an independent poor survival prediction in patients with EOC. CONCLUSION MYL9 is upregulated in EOC and may serve as a useful patent of prognostic biomarker in EOC, and it may demonstrate an important value for the clinical treatment and supervision of patients with EOC.
Collapse
Affiliation(s)
- Yuao Deng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Longyang Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13 Shiliugang ST, Guangzhou, 510315, China
| | - Weifeng Feng
- The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Zhongqiu Lin
- Department of Gynecology Oncology, The Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Yingxia Ning
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xin Luo
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| |
Collapse
|
15
|
Xu L, Zhu S, Tang A, Liu W. LncRNA MBLN1-AS1 inhibits the progression of retinoblastoma through targeting miR-338-5p-Wnt/β-catenin signaling pathway. Inflamm Res 2021; 70:217-227. [PMID: 33454803 DOI: 10.1007/s00011-020-01432-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/07/2020] [Accepted: 12/13/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE AND DESIGN Retinoblastoma is the most common primary intraocular malignancy of childhood, which brings a heavy burden to the countries across the world, especially the developing countries. It has been shown that lncRNA muscleblind-like 1 antisense RNA 1 (MBNL1-AS1) exerts anti-tumor effects in various cancers, including bladder cancer, papillary thyroid cancer, and retinoblastoma. In the present study, we hypothesized that MBNL1-AS1 might play a protective role against retinoblastoma. METHODS The expression of MBNL1-AS1 and its potential target miR-338-5p were evaluated in retinoblastoma cell line by real-time quantitative PCR and western blot. The involvement of MBNL1-AS1-miR-338-5p in the cell proliferation was evaluated by cell counting kit-8 (CCK8), and colony formation assay. The cell migration was evaluated by Transwell assay in Y79 cells, a retinoblastoma cell line. The involvement of MBNL1-AS1-miR-338-5p in tumor formation was also evaluated in mice. RESULTS It was found that MBNL1-AS1 overexpression inhibited proliferation and migration in Y79 cells. In addition, the inhibitory effects of MBNL1-AS1 on Y79 cells were significantly reversed in the presence of miR-338-5p mimics, and MBNL1-AS1 overexpression significantly decreased miR-338-5p level in Y79 cells. Furthermore, MBNL1-AS1 overexpression significantly inhibited Wnt/β-catenin signaling pathway, and this inhibitory effect was almost lost in the presence of miR-338-5p mimics. Finally, our in vivo study showed that MBNL1-AS1 overexpression significantly inhibited Y79-induced retinoblastoma in mice, and this inhibitory effect was lost in the presence of miR-338-5p mimics. CONCLUSION Our study shows that MBNL1-AS1 exerts its anti-tumor effect by targeting miR-338-5p, thereby inactivating wnt/β-catenin signaling pathway in retinoblastoma.
Collapse
Affiliation(s)
- Lei Xu
- Department of Ophthalmology, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Zhanggong District, Ganzhou, 341000, Jiangxi Province, China
| | - Shenyu Zhu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi Province, China
| | - Aidong Tang
- Department of Ophthalmology, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Zhanggong District, Ganzhou, 341000, Jiangxi Province, China
| | - Wanrong Liu
- Department of Ophthalmology, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Zhanggong District, Ganzhou, 341000, Jiangxi Province, China.
| |
Collapse
|
16
|
Insulin receptor substrate 1 gene expression is strongly up-regulated by HSPB8 silencing in U87 glioma cells. Endocr Regul 2020; 54:231-243. [PMID: 33885248 DOI: 10.2478/enr-2020-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objective. The aim of the present investigation was to study the expression of genes encoding IRS1 (insulin receptor substrate 1) and some other functionally active proteins in U87 glioma cells under silencing of polyfunctional chaperone HSPB8 for evaluation of the possible significance of this protein in intergenic interactions.Methods. Silencing of HSPB8 mRNA was introduced by HSPB8 specific siRNA. The expression level of HSPB8, IRS1, HK2, GLO1, HOMER3, MYL9, NAMPT, PER2, PERP, GADD45A, and DEK genes was studied in U87 glioma cells by quantitative polymerase chain reaction.Results. It was shown that silencing of HSPB8 mRNA by specific to HSPB8 siRNA led to a strong down-regulation of this mRNA and significant modification of the expression of IRS1 and many other genes in glioma cells: strong up-regulated of HOMER3, GLO1, and PERP and down-regulated of MYL9, NAMPT, PER2, GADD45A, and DEK gene expressions. At the same time, no significant changes were detected in the expression of HK2 gene in glioma cells treated by siRNA, specific to HSPB8. Moreover, the silencing of HSPB8 mRNA enhanced the glioma cells proliferation rate.Conclusions. Results of this investigation demonstrated that silencing of HSPB8 mRNA affected the expression of IRS1 gene as well as many other genes encoding tumor growth related proteins. It is possible that the dysregulation of most of the studied genes in glioma cells after silencing of HSPB8 is reflected by a complex of intergenic interactions and that this polyfunctional chaperone is an important factor for the stability of genome function and regulatory mechanisms contributing to the tumorigenesis control.
Collapse
|
17
|
ERN1 dependent regulation of TMED10, MYL9, SPOCK1, CUL4A and CUL4B genes expression at glucose and glutamine deprivations in U87 glioma cells. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.05.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
18
|
Zhu K, Wang Y, Liu L, Li S, Yu W. Long non-coding RNA MBNL1-AS1 regulates proliferation, migration, and invasion of cancer stem cells in colon cancer by interacting with MYL9 via sponging microRNA-412-3p. Clin Res Hepatol Gastroenterol 2020; 44:101-114. [PMID: 31255531 DOI: 10.1016/j.clinre.2019.05.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/29/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIMS Colon cancer is a common cancer that is a threat to human health. Some long non-coding RNAs (lncRNAs) have been observed to exert roles in colon cancer. Here, the current study is aimed to explore the potential mechanism of lncRNA MBNL1 antisense RNA 1 (MBNL1-AS1) in progression of colon cancer and the associated mechanisms. METHODS Microarray analysis was performed to screen differentially expressed lncRNA and genes associated with colon cancer and its potential mechanism. The functional role of MBNL1-AS1 in colon cancer was analyzed, followed identification of the interaction among MBNL1-AS1, microRNA-412-3p (miR-412-3p), and MYL9. Subsequently, CSC viability, migration, invasion, and apoptosis were detected though a series of in vitro experiments. At last, in vivo experiments were performed to assess tumor formation of colon CSCs. RESULTS MBNL1-AS1 and MYL9 were poorly expressed in colon cancer. MBNL1-AS1 could competitively bind to miR-412-3p so as to promote MYL9 expression. Enhancement of MBNL1-AS1 or inhibition of miR-412-3p was shown to decrease CSC proliferation, migration, and invasion but promote apoptosis. Moreover, MBNL1-AS1 reversed the CSC-like properties as well as xenograft tumor formation in vivo induced by miR-412-3p. CONCLUSION Collectively, the present study suggests an inhibitory role of MBNL1-AS1 in colon cancer by upregulating miR-412-3p-targeted MYL9. Thus, this study provides an enhanced understanding of MBNL1-AS1 along with miR-412-3p and MYL9 as therapeutic targets for colon cancer.
Collapse
Affiliation(s)
- Kongxi Zhu
- Department of Gastroenterology, The Second Hospital of Shandong University, No. 247, Beiyuan Street, 250033 Jinan, Shandon Province, PR China
| | - Yunxia Wang
- Department of Gastroenterology, The Second Hospital of Shandong University, No. 247, Beiyuan Street, 250033 Jinan, Shandon Province, PR China
| | - Lan Liu
- Department of Gastroenterology, The Second Hospital of Shandong University, No. 247, Beiyuan Street, 250033 Jinan, Shandon Province, PR China
| | - Shuai Li
- Department of Gastroenterology, The Second Hospital of Shandong University, No. 247, Beiyuan Street, 250033 Jinan, Shandon Province, PR China
| | - Weihua Yu
- Department of Gastroenterology, The Second Hospital of Shandong University, No. 247, Beiyuan Street, 250033 Jinan, Shandon Province, PR China.
| |
Collapse
|
19
|
Duan R, Ning Y, Wang S, Lindsay BG, Carroll RJ, Chen Y. A fast score test for generalized mixture models. Biometrics 2019; 76:811-820. [PMID: 31863595 DOI: 10.1111/biom.13204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/15/2019] [Accepted: 12/04/2019] [Indexed: 11/30/2022]
Abstract
In biomedical studies, testing for homogeneity between two groups, where one group is modeled by mixture models, is often of great interest. This paper considers the semiparametric exponential family mixture model proposed by Hong et al. (2017) and studies the score test for homogeneity under this model. The score test is nonregular in the sense that nuisance parameters disappear under the null hypothesis. To address this difficulty, we propose a modification of the score test, so that the resulting test enjoys the Wilks phenomenon. In finite samples, we show that with fixed nuisance parameters the score test is locally most powerful. In large samples, we establish the asymptotic power functions under two types of local alternative hypotheses. Our simulation studies illustrate that the proposed score test is powerful and computationally fast. We apply the proposed score test to an UK ovarian cancer DNA methylation data for identification of differentially methylated CpG sites.
Collapse
Affiliation(s)
- Rui Duan
- Department of Biostatistics, Epidemiology, and Informatics, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yang Ning
- Department of Statistical Science, Cornell University, Ithaca, New York
| | - Shuang Wang
- Department of Biostatistics, Columbia University, New York, New York
| | - Bruce G Lindsay
- Department of Statistics, Pennsylvania State University, State College, Pennsylvania
| | - Raymond J Carroll
- Department of Statistics, Texas A&M University, College Station, Texas
| | - Yong Chen
- Department of Biostatistics, Epidemiology, and Informatics, The University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
20
|
Koushki M, Farrokhi Yekta R, Amiri-Dashatan N, Dadpay M, Goshadrou F. Therapeutic effects of hydro-alcoholic extract of Achillea wilhelmsii C. Koch on indomethacin-induced gastric ulcer in rats: a proteomic and metabolomic approach. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:205. [PMID: 31391093 PMCID: PMC6686504 DOI: 10.1186/s12906-019-2623-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/30/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Gastric ulcer is one of the most prevalent diseases worldwide. In Iranian folk medicine, Achillea wilhelmsii (AW) is used as a treatment for gastric ulcer. Previous reports also mentioned Antiulcerogenic properties for this herbal plant. This study investigated the therapeutic effects of Achillea wilhelmsii C. Koch extract on indomethacin-induced gastric lesion in rats, from both proteomic and metabolomic perspectives. METHODS The rats were divided into 4 groups. Gastric ulceration was induced by a single dose of indomethacin (45 mg/kg) by oral gavage. An amount of 800 mg/kg of AW extract was administered orally. Serum and tissue samples were collected for further investigations. The metabolomic study was performed by 1H-NMR CPMG spectrometry. Proteomic analysis was also executed by using two dimensional gel electrophoresis (2DE) followed by liquid chromatography coupled to tandem mass spectrometry (LC-ESI/MS/MS). Real time PCR was used to confirm some of the genes. RESULTS The macroscopic and microscopic investigations confirmed the effectiveness of the AW extract. There was a panel of metabolites which showed alteration during gastric lesion development. The levels of some of these metabolite reversed nearly to their control values after the administration of AW extract. There were also changes in the levels of some proteins including Alb, Fabp5, Hspb1, Tagln, Lgals7, Csta and Myl9 which were reversed after AW administration. CONCLUSIONS Our findings suggested that Achillea wilhelmsii C. Koch extract could be a potential therapy to be used for indomethacin-induced gastric lesion treatment in the future. However, further investigations are needed to confirm the results.
Collapse
Affiliation(s)
- Mehdi Koushki
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nasrin Amiri-Dashatan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Dadpay
- Department of Pathology, AJA University of Medical Sciences, Tehran, Iran
| | - Fatemeh Goshadrou
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Kruthika BS, Sugur H, Nandaki K, Arimappamagan A, Paturu K, Santosh V. Expression pattern and prognostic significance of myosin light chain 9 (MYL9): a novel biomarker in glioblastoma. J Clin Pathol 2019; 72:677-681. [DOI: 10.1136/jclinpath-2019-205834] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/05/2019] [Accepted: 06/07/2019] [Indexed: 11/03/2022]
Abstract
AimsTumour recurrence is inevitable in glioblastoma (GBM) and mostly noted in the peritumoural brain zone (PT). In our previous microarray-based study, we identified Myosin Light Chain 9 (MYL9) as a highly expressed gene in the PT of GBM. Therefore, we aimed to study the expression pattern and clinical significance of MYL9 in GBM.MethodsPatient samples included three retrospective cohorts: 25 GBM cases with differential biopsies of tumour core and PT, 62 retrospective cases of newly diagnosed GBM with survival information and 20 paired samples (newly diagnosed and recurrent GBM). All tumour tissues, archived as formalin fixed paraffin embedded blocks were retrieved and immunohistochemistry for MYL9 and IDH1 R132H was performed. MYL9 expression was correlated with patient prognosis in our cohort and in The Cancer Genome Atlas (TCGA) and Rembrandt cohorts. It was further evaluated in the 20 paired samples of GBM.ResultsMYL9 showed a cytoplasmic membranous staining of tumour cells. The staining pattern was variable and patchy within tumours. Higher MYL9 expression was associated with poor overall and progression-free survival in our and in TCGA and Rembrandt cohorts. The expression of MYL9 was higher in IDH1 R132H immunonegative cases.ConclusionsWe show MYL9 as a novel biomarker, variably expressed in GBM. The association of high MYL9 expression with poor prognosis in newly diagnosed GBM patients and increased expression in recurrent GBM is indicative of its role in conferring tumour aggressiveness.
Collapse
|
22
|
Gladilin E, Ohse S, Boerries M, Busch H, Xu C, Schneider M, Meister M, Eils R. TGFβ-induced cytoskeletal remodeling mediates elevation of cell stiffness and invasiveness in NSCLC. Sci Rep 2019; 9:7667. [PMID: 31113982 PMCID: PMC6529472 DOI: 10.1038/s41598-019-43409-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/25/2019] [Indexed: 02/07/2023] Open
Abstract
Importance of growth factor (GF) signaling in cancer progression is widely acknowledged. Transforming growth factor beta (TGFβ) is known to play a key role in epithelial-to-mesenchymal transition (EMT) and metastatic cell transformation that are characterized by alterations in cell mechanical architecture and behavior towards a more robust and motile single cell phenotype. However, mechanisms mediating cancer type specific enhancement of cell mechanical phenotype in response to TGFβ remain poorly understood. Here, we combine high-throughput mechanical cell phenotyping, microarray analysis and gene-silencing to dissect cytoskeletal mediators of TGFβ-induced changes in mechanical properties of on-small-cell lung carcinoma (NSCLC) cells. Our experimental results show that elevation of rigidity and invasiveness of TGFβ-stimulated NSCLC cells correlates with upregulation of several cytoskeletal and motor proteins including vimentin, a canonical marker of EMT, and less-known unconventional myosins. Selective probing of gene-silenced cells lead to identification of unconventional myosin MYH15 as a novel mediator of elevated cell rigidity and invasiveness in TGFβ-stimulated NSCLC cells. Our experimental results provide insights into TGFβ-induced cytoskeletal remodeling of NSCLC cells and suggest that mediators of elevated cell stiffness and migratory activity such as unconventional cytoskeletal and motor proteins may represent promising pharmaceutical targets for restraining invasive spread of lung cancer.
Collapse
Affiliation(s)
- E Gladilin
- German Cancer Research Center, Div. Bioinformatics and Omics Data Analytics, Mathematikon - Berliner Str. 41, 69120, Heidelberg, Germany. .,University Heidelberg, BioQuant, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany. .,Leibniz Institute of Plant Genetics and Crop Plant Research, OT Gatersleben Corrensstrasse 3, 06466, Seeland, Germany.
| | - S Ohse
- University of Freiburg, Institute of Molecular Medicine and Cell Research (IMMZ), Stefan-Meier-Str. 17, 79104, Freiburg, Germany
| | - M Boerries
- University of Freiburg, Institute of Molecular Medicine and Cell Research (IMMZ), Stefan-Meier-Str. 17, 79104, Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department for Biometry, Epidemiology and Medical Bioinformatics and Comprehensive Cancer Center Freiburg (CCCF), University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Breisacherstrasse 153, 79110, Freiburg, Germany
| | - H Busch
- University of Freiburg, Institute of Molecular Medicine and Cell Research (IMMZ), Stefan-Meier-Str. 17, 79104, Freiburg, Germany.,University of Lübeck, Institute of Experimental Dermatology, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - C Xu
- Thoraxklinik at Heidelberg University Hospital, Amalienstr. 5, 69126, Heidelberg, Germany
| | - M Schneider
- Thoraxklinik at Heidelberg University Hospital, Amalienstr. 5, 69126, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - M Meister
- Thoraxklinik at Heidelberg University Hospital, Amalienstr. 5, 69126, Heidelberg, Germany
| | - R Eils
- Center for Digital Health, Berlin Institute of Health, and Charité Universitätsmedizin Berlin, Kapelle-Ufer 2, 10117, Berlin, Germany.,Health Data Science Unit, Heidelberg University Hospital, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| |
Collapse
|
23
|
Gao L, Ye M, Lu X, Huang D. Hybrid Method Based on Information Gain and Support Vector Machine for Gene Selection in Cancer Classification. GENOMICS PROTEOMICS & BIOINFORMATICS 2017; 15:389-395. [PMID: 29246519 PMCID: PMC5828665 DOI: 10.1016/j.gpb.2017.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/25/2017] [Accepted: 08/08/2017] [Indexed: 12/30/2022]
Abstract
It remains a great challenge to achieve sufficient cancer classification accuracy with the entire set of genes, due to the high dimensions, small sample size, and big noise of gene expression data. We thus proposed a hybrid gene selection method, Information Gain-Support Vector Machine (IG-SVM) in this study. IG was initially employed to filter irrelevant and redundant genes. Then, further removal of redundant genes was performed using SVM to eliminate the noise in the datasets more effectively. Finally, the informative genes selected by IG-SVM served as the input for the LIBSVM classifier. Compared to other related algorithms, IG-SVM showed the highest classification accuracy and superior performance as evaluated using five cancer gene expression datasets based on a few selected genes. As an example, IG-SVM achieved a classification accuracy of 90.32% for colon cancer, which is difficult to be accurately classified, only based on three genes including CSRP1, MYL9, and GUCA2B.
Collapse
Affiliation(s)
- Lingyun Gao
- School of Medical Information, Wannan Medical College, Wuhu 241002, China
| | - Mingquan Ye
- School of Medical Information, Wannan Medical College, Wuhu 241002, China.
| | - Xiaojie Lu
- School of Medical Information, Wannan Medical College, Wuhu 241002, China
| | - Daobin Huang
- School of Medical Information, Wannan Medical College, Wuhu 241002, China
| |
Collapse
|