1
|
Paranitharan N, Kataria S, Arumugam VA, Hsieh HL, Muthukrishnan S, Velayuthaprabhu S. Integrin α1 upregulation by TF:FVIIa complex promotes cervical cancer migration through PAR2-dependent MEK1/2 activation. Biochem Biophys Res Commun 2025; 742:151151. [PMID: 39657349 DOI: 10.1016/j.bbrc.2024.151151] [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: 11/24/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Tissue factor (TF) and protease-activated receptor 2 (PAR2) have been associated with the progression of cancer, while integrins are essential for the adhesion and migration of cancer cells. This study aimed to explore the cross-talk between the TF:FVIIa complex, PAR2 signaling, and the expression of integrin α1 in cervical cancer cells. Utilizing data from The Cancer Genome Atlas (TCGA), the research examined the relationship between the TF and PAR2 genes and the integrin α1 gene (ITGA1) in reproductive cancers, revealing a positive correlation between integrin α1 expression and both TF and PAR2 genes. Analyses through Western blotting and RT-PCR demonstrated that TF:FVIIa complex transactivates PAR2, which significantly increases the phosphorylation of MEK1/2 and subsequently elevates integrin α1 expression. Inhibition of either PAR2 or MEK1/2 resulted in a decrease in the FVIIa-induced increase in integrin α1 expression. Additionally, cell migration studies indicated that elevated expression of integrin α1, mediated by the TF:FVIIa/PAR2 pathway, was linked to enhanced cell migration, which could be inhibited by blocking integrin α1. This investigation uncovers a novel signaling pathway in HeLa cells, highlighting the significance of the TF:FVIIa:PAR2 axis in modulating integrins that are vital for cancer progression, thereby offering insights for potential targeted therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
| | - Shivangi Kataria
- Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Vijaya Anand Arumugam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, India.
| | - Hsi-Lung Hsieh
- Research Center for Chinese Herbal Medicine, Department of Nursing, Division of Basic Medical Sciences, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, And Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | | |
Collapse
|
2
|
Mafi A, Hedayati N, Kahkesh S, Khoshayand S, Alimohammadi M, Farahani N, Hushmandi K. The landscape of circRNAs in gliomas temozolomide resistance: Insights into molecular pathways. Noncoding RNA Res 2024; 9:1178-1189. [PMID: 39022676 PMCID: PMC11250881 DOI: 10.1016/j.ncrna.2024.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 07/20/2024] Open
Abstract
As the deadliest type of primary brain tumor, gliomas represent a significant worldwide health concern. Circular RNA (circRNA), a unique non-coding RNA molecule, seems to be one of the most alluring target molecules involved in the pathophysiology of many kinds of cancers. CircRNAs have been identified as prospective targets and biomarkers for the diagnosis and treatment of numerous disorders, particularly malignancies. Recent research has established a clinical link between temozolomide (TMZ) resistance and certain circRNA dysregulations in glioma tumors. CircRNAs may play a therapeutic role in controlling or overcoming TMZ resistance in gliomas and may provide guidance for a novel kind of individualized glioma therapy. To address the biological characteristics of circRNAs and their potential to induce resistance to TMZ, this review has highlighted and summarized the possible roles that circRNAs may play in molecular pathways of drug resistance, including the Ras/Raf/ERK PI3K/Akt signaling pathway and metabolic processes in gliomas.
Collapse
Affiliation(s)
- Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sara Khoshayand
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
3
|
Rong S, Dai B, Yang C, Lan Z, Wang L, Xu L, Chen W, Chen J, Wu Z. HNRNPC modulates PKM alternative splicing via m6A methylation, upregulating PKM2 expression to promote aerobic glycolysis in papillary thyroid carcinoma and drive malignant progression. J Transl Med 2024; 22:914. [PMID: 39380010 PMCID: PMC11459990 DOI: 10.1186/s12967-024-05668-9] [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: 03/19/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
The heterogeneous nuclear ribonucleoprotein C (HNRNPC) plays a crucial role in tumorigenesis, yet its role in papillary thyroid carcinoma (PTC) remains elusive. Herein, we elucidated the function and molecular mechanism of HNRNPC in PTC tumorigenesis and progression. Our study unveiled a significant upregulation of HNRNPC in PTC, and knockdown of HNRNPC markedly inhibited the proliferation, invasion, and metastasis of BCPAP cells. Furthermore, HNRNPC modulated PKM alternative splicing in BCPAP cells primarily through m6A modification. Additionally, by upregulating PKM2 expression, HNRNPC promoted aerobic glycolysis in BCPAP cells, thereby facilitating malignant progression in PTC. In summary, our findings demonstrate that HNRNPC regulates PKM alternative splicing through m6A methylation modification and promotes the proliferation, invasion and metastasis of PTC through glucose metabolism pathways mediated by PKM2. These discoveries provide new biomarkers for screening and diagnosing PTC patients and offer novel therapeutic targets for personalized treatment strategies.
Collapse
Affiliation(s)
- Shikuo Rong
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Thyroid Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Bao Dai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunrong Yang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ziteng Lan
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Linhe Wang
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Weijian Chen
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Chen
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zeyu Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Sharma S, Rani H, Mahesh Y, Jolly MK, Dixit J, Mahadevan V. Loss of p53 epigenetically modulates epithelial to mesenchymal transition in colorectal cancer. Transl Oncol 2024; 43:101848. [PMID: 38412660 PMCID: PMC10907866 DOI: 10.1016/j.tranon.2023.101848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 02/29/2024] Open
Abstract
Epithelial to Mesenchymal transition (EMT) drives cancer metastasis and is governed by genetic and epigenetic alterations at multiple levels of regulation. It is well established that loss/mutation of p53 confers oncogenic function to cancer cells and promotes metastasis. Though transcription factors like ZEB1, SLUG, SNAIL and TWIST have been implied in EMT signalling, p53 mediated alterations in the epigenetic machinery accompanying EMT are not clearly understood. This work attempts to explore epigenetic signalling during EMT in colorectal cancer (CRC) cells with varying status of p53. Towards this, we have induced EMT using TGFβ on CRC cell lines with wild type, null and mutant p53 and have assayed epigenetic alterations after EMT induction. Transcriptomic profiling of the four CRC cell lines revealed that the loss of p53 confers more mesenchymal phenotype with EMT induction than its mutant counterparts. This was also accompanied by upregulation of epigenetic writer and eraser machinery suggesting an epigenetic signalling cascade triggered by TGFβ signalling in CRC. Significant agonist and antagonistic relationships observed between EMT factor SNAI1 and SNAI2 with epigenetic enzymes KDM6A/6B and the chromatin organiser SATB1 in p53 null CRC cells suggest a crosstalk between epigenetic and EMT factors. The observed epigenetic regulation of EMT factor SNAI1 correlates with poor clinical outcomes in 270 colorectal cancer patients taken from TCGA-COAD. This unique p53 dependent interplay between epigenetic enzymes and EMT factors in CRC cells may be exploited for development of synergistic therapies for CRC patients presenting to the clinic with loss of p53.
Collapse
Affiliation(s)
- Shreya Sharma
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bangalore, India
| | - Harsha Rani
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Bangalore, India
| | | | | | | | | |
Collapse
|
5
|
Su M, Hou Y, Cai S, Li W, Wei Y, Wang R, Wu M, Liu M, Chang J, Yang K, Yiu K, Chen C. Elevated ITGA1 levels in type 2 diabetes: implications for cardiac function impairment. Diabetologia 2024; 67:850-863. [PMID: 38413438 PMCID: PMC10954979 DOI: 10.1007/s00125-024-06109-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 02/29/2024]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes mellitus is known to contribute to the development of heart failure with preserved ejection fraction (HFpEF). However, identifying HFpEF in individuals with type 2 diabetes early on is often challenging due to a limited array of biomarkers. This study aims to investigate specific biomarkers associated with the progression of HFpEF in individuals with type 2 diabetes, for the purpose of enabling early detection and more effective management strategies. METHODS Blood samples were collected from individuals with type 2 diabetes, both with and without HFpEF, for proteomic analysis. Plasma integrin α1 (ITGA1) levels were measured and compared between the two groups. Participants were further categorised based on ITGA1 levels and underwent detailed transthoracic echocardiography at baseline and during a median follow-up period of 30 months. Multivariable linear and Cox regression analyses were conducted separately to assess the associations between plasma ITGA1 levels and changes in echocardiography indicators and re-hospitalisation risk. Additionally, proteomic data for the individuals' left ventricles, from ProteomeXchange database, were analysed to uncover mechanisms underlying the change in ITGA1 levels in HFpEF. RESULTS Individuals with type 2 diabetes and HFpEF showed significantly higher plasma ITGA1 levels than the individuals with type 2 diabetes without HFpEF. These elevated ITGA1 levels were associated with left ventricular remodelling and impaired diastolic function. Furthermore, during a median follow-up of 30 months, multivariable analysis revealed that elevated ITGA1 levels independently correlated with deterioration of both diastolic and systolic cardiac functions. Additionally, higher baseline plasma ITGA1 levels independently predicted re-hospitalisation risk (HR 2.331 [95% CI 1.387, 3.917], p=0.001). Proteomic analysis of left ventricular myocardial tissue provided insights into the impact of increased ITGA1 levels on cardiac fibrosis-related pathways and the contribution made by these changes to the development and progression of HFpEF. CONCLUSIONS/INTERPRETATION ITGA1 serves as a biomarker for monitoring cardiac structural and functional damage, can be used to accurately diagnose the presence of HFpEF, and can be used to predict potential deterioration in cardiac structure and function as well as re-hospitalisation for individuals with type 2 diabetes. Its measurement holds promise for facilitating risk stratification and early intervention to mitigate the adverse cardiovascular effects associated with diabetes. DATA AVAILABILITY The proteomic data of left ventricular myocardial tissue from individuals with type 2 diabetes, encompassing both those with and without HFpEF, is available from the ProteomeXchange database at http://proteomecentral.proteomexchange.org .
Collapse
Affiliation(s)
- Mengqi Su
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yilin Hou
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sidong Cai
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wenpeng Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yinxia Wei
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Run Wang
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Min Wu
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Mingya Liu
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kelaier Yang
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen, China
| | - Kaihang Yiu
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Cardiology, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| | - Cong Chen
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
6
|
Min Y, Deng W, Yuan H, Zhu D, Zhao R, Zhang P, Xue J, Yuan Z, Zhang T, Jiang Y, Xu K, Wu D, Cai Y, Suo C, Chen X. Single extracellular vesicle surface protein-based blood assay identifies potential biomarkers for detection and screening of five cancers. Mol Oncol 2024; 18:743-761. [PMID: 38194998 PMCID: PMC10920081 DOI: 10.1002/1878-0261.13586] [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: 05/15/2023] [Revised: 11/21/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Extracellular vesicles (EVs) and EV proteins are promising biomarkers for cancer liquid biopsy. Herein, we designed a case-control study involving 100 controls and 100 patients with esophageal, stomach, colorectal, liver, or lung cancer to identify common and type-specific biomarkers of plasma-derived EV surface proteins for the five cancers. EV surface proteins were profiled using a sequencing-based proximity barcoding assay. In this study, five differentially expressed proteins (DEPs) and eight differentially expressed protein combinations (DEPCs) showed promising performance (area under curve, AUC > 0.900) in pan-cancer identification [e.g., TENM2 (AUC = 0.982), CD36 (AUC = 0.974), and CD36-ITGA1 (AUC = 0.971)]. Our classification model could properly discriminate between cancer patients and controls using DEPs (AUC = 0.981) or DEPCs (AUC = 0.965). When distinguishing one cancer from the other four, the accuracy of the classification model using DEPCs (85-92%) was higher than that using DEPs (78-84%). We validated the performance in an additional 14 cancer patients and 14 controls, and achieved an AUC value of 0.786 for DEPs and 0.622 for DEPCs, highlighting the necessity to recruit a larger cohort for further validation. When clustering EVs into subpopulations, we detected cluster-specific proteins highly expressed in immune-related tissues. In the context of colorectal cancer, we identified heterogeneous EV clusters enriched in cancer patients, correlating with tumor initiation and progression. These findings provide epidemiological and molecular evidence for the clinical application of EV proteins in cancer prediction, while also illuminating their functional roles in cancer physiopathology.
Collapse
Affiliation(s)
- Yuxin Min
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
| | - Wenjiang Deng
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, School of Life ScienceHuman Phenome Institute, Fudan UniversityShanghaiChina
| | - Dongliang Zhu
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
| | - Renjia Zhao
- State Key Laboratory of Genetic Engineering, School of Life ScienceHuman Phenome Institute, Fudan UniversityShanghaiChina
| | - Pengyan Zhang
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
| | - Jiangli Xue
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
| | - Ziyu Yuan
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
| | - Tiejun Zhang
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
- Yiwu Research Institute of Fudan UniversityChina
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, School of Life ScienceHuman Phenome Institute, Fudan UniversityShanghaiChina
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
| | - Kelin Xu
- Department of Biostatistics, School of Public HealthFudan UniversityShanghaiChina
| | - Di Wu
- Vesicode ABStockholmSweden
| | - Yanling Cai
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of UrologyThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen Institute of Translational MedicineShenzhenChina
| | - Chen Suo
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
- Shanghai Institute of Infectious Disease and BiosecurityShanghaiChina
| | - Xingdong Chen
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
- Yiwu Research Institute of Fudan UniversityChina
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Li Z, Xu T, Li X, Wang T, Tang G, Zhao H, Zhao Y, Ye K, Gao P. Viral integration promotes SV40T-induced immortalization by disturbing the expression of DNA/chromosome- and ECM-associated functional genes. Gene 2024; 896:148060. [PMID: 38048968 DOI: 10.1016/j.gene.2023.148060] [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: 10/18/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023]
Abstract
Lentivirus containing simian virus 40 large T antigen (SV40T) is routinely used to induce cell immortalization. However, the roles of viral integration itself in this progress is still controversial. Here, we transformed primary mouse embryonic fibroblasts (MEFs) with SV40T lentivirus and studied the roles of viral integration in the immortalization using RNA sequencing (RNA-seq) and whole genome sequencing (WGS). During the immortalization, differentially expressed genes (DGEs) are enriched in viral infection and several diverse activities. However, DEGs between immortalized and aging cells are significantly enriched in DNA/chromosome- and extracellular matrix (ECM)-associated activities. Gene regulatory network (GRN) analysis shows that although p53 is a key regulatory factor, many other transcription factors also play critical roles in the process, like STAT1. Of these DEGs, 32 genes have viral integration in their coding and/or regulatory regions. Our findings suggest that viral integration may promote SV40T-mediated immortalization by disturbing the expression of DNA/chromosome- and ECM-associated genes.
Collapse
Affiliation(s)
- Zihang Li
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tun Xu
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiujuan Li
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tingjie Wang
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Guangbo Tang
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Huanhuan Zhao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuanli Zhao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kai Ye
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China; Faculty of Science, Leiden University, Leiden, the Netherlands.
| | - Peng Gao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
8
|
Ma YY, Zhang GJ, Liu PF, Liu Y, Ding JC, Xu H, Hao L, Pan D, Wang HL, Wang JK, Xu P, Shi ZD, Pang K. Comprehensive Genomic Analysis of Puerarin in Inhibiting Bladder Urothelial Carcinoma Cell Proliferation and Migration. Recent Pat Anticancer Drug Discov 2024; 19:516-529. [PMID: 37694778 PMCID: PMC11348475 DOI: 10.2174/1574892819666230908110107] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Bladder urothelial carcinoma (BUC) ranks second in the incidence of urogenital system tumors, and the treatment of BUC needs to be improved. Puerarin, a traditional Chinese medicine (TCM), has been shown to have various effects such as anti-cancer effects, the promotion of angiogenesis, and anti-inflammation. This study investigates the effects of puerarin on BUC and its molecular mechanisms. METHODS Through GeneChip experiments, we obtained differentially expressed genes (DEGs) and analyzed these DEGs using the Ingenuity® Pathway Analysis (IPA®), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathway enrichment analyses. The Cell Counting Kit 8 (CCK8) assay was used to verify the inhibitory effect of puerarin on the proliferation of BUC T24 cells. String combined with Cytoscape® was used to create the Protein-Protein Interaction (PPI) network, and the MCC algorithm in cytoHubba plugin was used to screen key genes. Gene Set Enrichment Analysis (GSEA®) was used to verify the correlation between key genes and cell proliferation. RESULTS A total of 1617 DEGs were obtained by GeneChip. Based on the DEGs, the IPA® and pathway enrichment analysis showed they were mainly enriched in cancer cell proliferation and migration. CCK8 experiments proved that puerarin inhibited the proliferation of BUC T24 cells, and its IC50 at 48 hours was 218μmol/L. Through PPI and related algorithms, 7 key genes were obtained: ITGA1, LAMA3, LAMB3, LAMA4, PAK2, DMD, and UTRN. GSEA showed that these key genes were highly correlated with BUC cell proliferation. Survival curves showed that ITGA1 upregulation was associated with poor prognosis of BUC patients. CONCLUSION Our findings support the potential antitumor activity of puerarin in BUC. To the best of our knowledge, bioinformatics investigation suggests that puerarin demonstrates anticancer mechanisms via the upregulation of ITGA1, LAMA3 and 4, LAMB3, PAK2, DMD, and UTRN, all of which are involved in the proliferation and migration of bladder urothelial cancer cells.
Collapse
Affiliation(s)
- Yu-Yang Ma
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, No.199, South Jiefang Road, Xuzhou, Jiangsu, China
- Department of Urology, Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu City, Anhui Province, China
| | - Ge-jin Zhang
- Department of Urology, Suqian Zhongwu Hospital. No. 3786, Development Avenue, Suqian Economic and Technological Development Zone, Suqian, China
| | - Peng-fei Liu
- Jiangsu Provincial Key Laboratory of Educational Big Data Science and Engineering, Jiangsu Normal University, 101 Shanghai Road, Tongshan, Xuzhou, 221116, China
- School of Mathematics and Statistics and Research Institute of Mathematical Sciences (RIMS), Jiangsu Normal University, 101 Shanghai Road, Tongshan, Xuzhou, 221116, China
| | - Ying Liu
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College. No.199, South Jiefang Road, Xuzhou, Jiangsu, China
| | - Ji-cun Ding
- Department of Burn and Plastic Surgery, Xuzhou First People's Hospital, No. 269, Daxue Road, Tongshan District, Xuzhou, Jiangsu, China
| | - Hao Xu
- Department of Urology, Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu City, Anhui Province, China
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, No.199, South Jiefang Road, Xuzhou, Jiangsu, China
| | - Deng Pan
- Department of Urology, Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu City, Anhui Province, China
| | - Hai-luo Wang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, No.199, South Jiefang Road, Xuzhou, Jiangsu, China
| | - Jing-kai Wang
- Graduate School, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Peng Xu
- Graduate School, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Zhen-Duo Shi
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, No.199, South Jiefang Road, Xuzhou, Jiangsu, China
- Department of Urology, Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu City, Anhui Province, China
| | - Kun Pang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, No.199, South Jiefang Road, Xuzhou, Jiangsu, China
- Department of Urology, Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu City, Anhui Province, China
| |
Collapse
|
9
|
Pan YB, Xu WJ, Huang MS, Lu YD, Zhou YJ, Teng Y, Gong JB, Fu XY, Mao XL, Li SW. Anoikis-related signature identifies tumor microenvironment landscape and predicts prognosis and drug sensitivity in colorectal cancer. J Cancer 2024; 15:841-857. [PMID: 38213716 PMCID: PMC10777033 DOI: 10.7150/jca.91627] [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: 10/28/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024] Open
Abstract
Background: Anoikis, a mechanism of programmed apoptosis, plays an important role in growth and metastasis of tumors. However, there are still few available comprehensive reports on the impact of anoikis on colorectal cancer. Method: A clustering analysis was done on 133 anoikis-related genes in GSE39582, and we compared clinical features between clusters, the tumor microenvironment was analyzed with algorithms such as "Cibersort" and "ssGSEA". We investigated risk scores of clinical feature groups and anoikis-associated gene mutations after creating a predictive model. We incorporated clinical traits to build a nomogram. Additionally, the quantitative real-time PCR was employed to investigate the mRNA expression of selected anoikis-associated genes. Result: We identified two anoikis-related clusters with distinct prognoses, clinical characteristics, and biological functions. One of the clusters was associated with anoikis resistance, which activated multiple pathways encouraging tumor metastasis. In our prognostic model, oxaliplatin may be a sensitive drug for low-risk patients. The nomogram showed good ability to predict survival time. And SIRT3, PIK3CA, ITGA3, DAPK1, and CASP3 increased in CRC group through the PCR assay. Conclusion: Our study identified two distinct modes of anoikis in colorectal cancer, with active metastasis-promoting pathways inducing an anti-anoikis subtype, which has a stronger propensity for metastasis and a worse prognosis than an anoikis-activated subtype. Massive immune cell infiltration may be an indicator of anoikis resistance. Anoikis' role in the colorectal cancer remains to be investigated.
Collapse
Affiliation(s)
- Yu-Biao Pan
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, China
| | - Wang-jin Xu
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Meng-sha Huang
- Hospital of Huangyan affiliated to Wenzhou Medical University, Huangyan, Zhejiang, China
| | - Yan-di Lu
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
| | - Yi-jing Zhou
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
| | - Ya Teng
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Jian-bin Gong
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, China
| | - Xin-yu Fu
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
| | - Xin-li Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Shao-wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
10
|
Yu S, Wang S, Xiong B, Peng C. Gut microbiota: key facilitator in metastasis of colorectal cancer. Front Oncol 2023; 13:1270991. [PMID: 38023192 PMCID: PMC10643165 DOI: 10.3389/fonc.2023.1270991] [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: 08/08/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Colorectal cancer (CRC) ranks third in terms of incidence among all kinds of cancer. The main cause of death is metastasis. Recent studies have shown that the gut microbiota could facilitate cancer metastasis by promoting cancer cells proliferation, invasion, dissemination, and survival. Multiple mechanisms have been implicated, such as RNA-mediated targeting effects, activation of tumor signaling cascades, secretion of microbiota-derived functional substances, regulation of mRNA methylation, facilitated immune evasion, increased intravasation of cancer cells, and remodeling of tumor microenvironment (TME). The understanding of CRC metastasis was further deepened by the mechanisms mentioned above. In this review, the mechanisms by which the gut microbiota participates in the process of CRC metastasis were reviewed as followed based on recent studies.
Collapse
Affiliation(s)
- Siyi Yu
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Shuyi Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Chunwei Peng
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
11
|
Chang PK, Yen IC, Tsai WC, Lee SY. Polygonum barbatum extract reduces colorectal cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition via YAP and β-catenin pathway regulation. Sci Rep 2023; 13:18368. [PMID: 37884620 PMCID: PMC10603200 DOI: 10.1038/s41598-023-45630-1] [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: 06/26/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide with novel therapeutic developmental challenges. Polygonum barbatum has anticancer potential, but its mechanism(s) are unclear. This study investigates the inhibitory effect of P. barbatum on human CRC cells. Polygonum barbatum extract (PBE) and quercetin standard HPLC fingerprints were determined using analytical RP-HPLC and evaluations were completed using the human colon cancer cell line HCT-116 (KRASG13D mutation) and HT-29 (BRAF mutation) cells. Post-PBE treatment, cell viability, colony formation, migration, invasion, and apoptosis, as well as changes in the whole-transcriptome of cells were analyzed. PBE significantly reduced CRC cell growth, migration, and invasion, and the genes responsible for extracellular matrix (ECM) organization, cell motility, and cell growth were suppressed by PBE. The differentially expressed genes revealed that PBE treatment exerted a significant effect on the ECM interaction and focal adhesion pathways. Epithelial-to-mesenchymal transition markers, N-cadherin, vimentin, SLUG, and SNAIL, were shown to be regulated by PBE. These effects were associated with blockade of the Yes-associated protein and the GSK3β/β-catenin axis. PBE exerts a significant inhibitory effect on CRC cells and may be applicable in clinical trials.
Collapse
Affiliation(s)
- Pi-Kai Chang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - I-Chuan Yen
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Cheng Tsai
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Lee
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
12
|
Whitehead CA, Fang H, Su H, Morokoff AP, Kaye AH, Hanssen E, Nowell CJ, Drummond KJ, Greening DW, Vella LJ, Mantamadiotis T, Stylli SS. Small extracellular vesicles promote invadopodia activity in glioblastoma cells in a therapy-dependent manner. Cell Oncol (Dordr) 2023; 46:909-931. [PMID: 37014551 PMCID: PMC10356899 DOI: 10.1007/s13402-023-00786-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
PURPOSE The therapeutic efficacy of radiotherapy/temozolomide treatment for glioblastoma (GBM) is limited by the augmented invasiveness mediated by invadopodia activity of surviving GBM cells. As yet, however the underlying mechanisms remain poorly understood. Due to their ability to transport oncogenic material between cells, small extracellular vesicles (sEVs) have emerged as key mediators of tumour progression. We hypothesize that the sustained growth and invasion of cancer cells depends on bidirectional sEV-mediated cell-cell communication. METHODS Invadopodia assays and zymography gels were used to examine the invadopodia activity capacity of GBM cells. Differential ultracentrifugation was utilized to isolate sEVs from conditioned medium and proteomic analyses were conducted on both GBM cell lines and their sEVs to determine the cargo present within the sEVs. In addition, the impact of radiotherapy and temozolomide treatment of GBM cells was studied. RESULTS We found that GBM cells form active invadopodia and secrete sEVs containing the matrix metalloproteinase MMP-2. Subsequent proteomic studies revealed the presence of an invadopodia-related protein sEV cargo and that sEVs from highly invadopodia active GBM cells (LN229) increase invadopodia activity in sEV recipient GBM cells. We also found that GBM cells displayed increases in invadopodia activity and sEV secretion post radiation/temozolomide treatment. Together, these data reveal a relationship between invadopodia and sEV composition/secretion/uptake in promoting the invasiveness of GBM cells. CONCLUSIONS Our data indicate that sEVs secreted by GBM cells can facilitate tumour invasion by promoting invadopodia activity in recipient cells, which may be enhanced by treatment with radio-chemotherapy. The transfer of pro-invasive cargos may yield important insights into the functional capacity of sEVs in invadopodia.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Huaqi Su
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew P Morokoff
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - Andrew H Kaye
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Eric Hanssen
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Advanced Microscopy Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, 3052, Australia
| | - Katharine J Drummond
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Laura J Vella
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia.
| |
Collapse
|
13
|
Bech JM, Terkelsen T, Bartels AS, Coscia F, Doll S, Zhao S, Zhang Z, Brünner N, Lindebjerg J, Madsen GI, Fang X, Mann M, Afonso Moreira JM. Proteomic Profiling of Colorectal Adenomas Identifies a Predictive Risk Signature for Development of Metachronous Advanced Colorectal Neoplasia. Gastroenterology 2023; 165:121-132.e5. [PMID: 36966943 DOI: 10.1053/j.gastro.2023.03.208] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND & AIMS Colonic adenomatous polyps, or adenomas, are frequent precancerous lesions and the origin of most cases of colorectal adenocarcinoma. However, we know from epidemiologic studies that although most colorectal cancers (CRCs) originate from adenomas, only a small fraction of adenomas (3%-5%) ever progress to cancer. At present, there are no molecular markers to guide follow-up surveillance programs. METHODS We profiled, by mass spectrometry-based proteomics combined with machine learning analysis, a selected cohort of formalin-fixed, paraffin-embedded high-grade (HG) adenomas with long clinical follow-up, collected as part of the Danish national screening program. We grouped subjects in the cohort according to their subsequent history of findings: a nonmetachronous advanced neoplasia group (G0), with no new HG adenomas or CRCs up to 10 years after polypectomy, and a metachronous advanced neoplasia group (G1) where individuals developed a new HG adenoma or CRC within 5 years of diagnosis. RESULTS We generated a proteome dataset from 98 selected HG adenoma samples, including 20 technical replicates, of which 45 samples belonged to the nonmetachronous advanced neoplasia group and 53 to the metachronous advanced neoplasia group. The clear distinction of these 2 groups seen in a uniform manifold approximation and projection plot indicated that the information contained within the abundance of the ∼5000 proteins was sufficient to predict the future occurrence of HG adenomas or development of CRC. CONCLUSIONS We performed an in-depth analysis of quantitative proteomic data from 98 resected adenoma samples using various novel algorithms and statistical packages and found that their proteome can predict development of metachronous advanced lesions and progression several years in advance.
Collapse
Affiliation(s)
- Jacob Mathias Bech
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Sino-Danish Center for Education and Research, Aarhus University, Aarhus, Denmark
| | - Thilde Terkelsen
- Center for Health Data Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Fabian Coscia
- Spatial Proteomics Group, Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Siqi Zhao
- Beijing Institute of Genomics, China National Center for Bioinformation, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Zhaojun Zhang
- Beijing Institute of Genomics, China National Center for Bioinformation, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Nils Brünner
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jan Lindebjerg
- Department of Pathology, Lillebaelt Hospital, Vejle Hospital, Vejle, Denmark
| | | | - Xiangdong Fang
- Beijing Institute of Genomics, China National Center for Bioinformation, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Matthias Mann
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
14
|
Wang J, Chen G, Yu X, Zhou X, Zhang Y, Wu Y, Tong J. Transcriptome analyses reveal differentially expressed genes associated with development of the palatal organ in bighead carp (Hypophthalmichthys nobilis). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY PART D: GENOMICS AND PROTEOMICS 2023; 46:101072. [PMID: 36990038 DOI: 10.1016/j.cbd.2023.101072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/12/2023] [Accepted: 03/11/2023] [Indexed: 03/28/2023]
Abstract
The palatal organ is a filter-feeding related organ and occupies a considerable proportion of the head of bighead carp (Hypophthalmichthys nobilis), a large cyprinid fish intensive aquaculture in Asia. In this study, we performed RNA-seq of the palatal organ during growth periods of two (M2), six (M6) and 15 (M15) months of age after hatching. The numbers of differentially expressed genes (DEGs) were 1384, 481 and 1837 for M2 VS M6, M6 VS M15 and M2 VS M15 respectively. The following signaling pathways of energy metabolism and cytoskeleton function were enriched, including ECM-receptor interaction, Cardiac muscle contraction, Steroid biosynthesis and PPAR signaling pathway. Several members of collagen family (col1a1, col2a1, col6a2, col6a3, col9a2), Laminin gamma 1 (lamc1), integrin alpha 1 (itga1), Fatty acid binding protein 2 (fads2) and lipoprotein lipase (lpl), and Protein tyrosine kinase 7 (Ptk7) are candidate genes for growth and development of basic tissues of the palatal organ. Furthermore, taste-related genes such as fgfrl1, fgf8a, fsta and notch1a were also identified, which may be involved in the development of taste buds of the palatal organ. The transcriptome data obtained in this study provide insights into the understanding functions and development mechanisms of palatal organ, and potential candidate genes that may be related to the genetic modulation of head size of bighead carp.
Collapse
Affiliation(s)
- Junru Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Geng Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiaomu Yu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiaoyu Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yifan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yanhong Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingou Tong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| |
Collapse
|
15
|
Single-cell RNA sequencing in orthopedic research. Bone Res 2023; 11:10. [PMID: 36828839 PMCID: PMC9958119 DOI: 10.1038/s41413-023-00245-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 02/26/2023] Open
Abstract
Although previous RNA sequencing methods have been widely used in orthopedic research and have provided ideas for therapeutic strategies, the specific mechanisms of some orthopedic disorders, including osteoarthritis, lumbar disc herniation, rheumatoid arthritis, fractures, tendon injuries, spinal cord injury, heterotopic ossification, and osteosarcoma, require further elucidation. The emergence of the single-cell RNA sequencing (scRNA-seq) technique has introduced a new era of research on these topics, as this method provides information regarding cellular heterogeneity, new cell subtypes, functions of novel subclusters, potential molecular mechanisms, cell-fate transitions, and cell‒cell interactions that are involved in the development of orthopedic diseases. Here, we summarize the cell subpopulations, genes, and underlying mechanisms involved in the development of orthopedic diseases identified by scRNA-seq, improving our understanding of the pathology of these diseases and providing new insights into therapeutic approaches.
Collapse
|
16
|
Amentoflavone Exerts Anti-Neuroinflammatory Effects by Inhibiting TLR4/MyD88/NF- κB and Activating Nrf2/HO-1 Pathway in Lipopolysaccharide-Induced BV2 Microglia. Mediators Inflamm 2022; 2022:5184721. [PMID: 36523959 PMCID: PMC9747320 DOI: 10.1155/2022/5184721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 12/12/2022] Open
Abstract
Background Amentoflavone, a natural biflavone, exerts anti-inflammation, antioxidation, and antiapoptosis effects on many diseases. However, the mechanism of amentoflavone on neuroinflammation-related diseases has not been comprehensively examined clearly. Methods BV2 microglial cells were treated with amentoflavone (10 μM), followed by lipopolysaccharide (LPS). Microglial activation and migration ability and the expression of proinflammatory cytokines and other signaling proteins were determined using immunohistochemistry, immunofluorescence, quantitative real-time polymerase chain reaction, Western blotting, enzyme-linked immunosorbent assay, and wound-healing assays. Results Amentoflavone restored LPS-induced microglia activation, migration, and inflammation response which depends on regulating toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor kappa B (NF-κB) pathway. In addition, amentoflavone also enhanced nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) levels in LPS-treated BV2 microglial cells. Conclusions Amentoflavone ameliorated LPS-induced neuroinflammatory response and oxidative stress in BV2 microglia. These data provide new insight into the mechanism of amentoflavone in the treatment of neuroinflammation-related diseases. Therefore, amentoflavone may be a potential therapeutic option for neurological disorders.
Collapse
|
17
|
Li X, Wang N, Leng H, Yuan H, Xu L. Hsa_circ_0043949 reinforces temozolomide resistance via upregulating oncogene ITGA1 axis in glioblastoma. Metab Brain Dis 2022; 37:2979-2993. [PMID: 36301458 DOI: 10.1007/s11011-022-01069-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/08/2022] [Indexed: 10/31/2022]
Abstract
Temozolomide (TMZ) resistance limits its use in glioblastoma (GBM). Exosomes can carry circular RNAs (circRNAs) to regulate chemoresistance. To date, the role of exosomal hsa_circ_0043949 (circ_0043949) in GBM resistance to TMZ is unclear. Relative expression of circ_0043949 in clinical samples, GBM cell lines, and exosomes was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The half-maximal inhibitory concentration (IC50) of TMZ, cell proliferation, apoptosis, invasion, and migration were analyzed via MTT, EdU, flow cytometry, transwell, and wound-healing assays. Relative protein levels were evaluated by western blotting. Target relationship was predicted by bioinformatics analysis and validated by dual-luciferase reporter and RNA pull-down assays. Exosomes were isolated by ultracentrifugation and verified by transmission electron microscopy, nanoparticle tracking analysis (NTA), and western blotting. The effect of exosomal circ_0043949 on TMZ resistance was validated by xenograft assay. Higher expression of circ_0043949 was gained in TMZ-resistant GBM samples and cells. Inhibition of circ_0043949 reduced TMZ resistance via decreasing IC50 of TMZ, repressing proliferation, invasion, migration, and inducing apoptosis in TMZ-resistant GBM cells. Circ_0043949 mediated integrinalpha1 (ITGA1) expression via function as a miR-876-3p sponge. Circ_0043949 was also upregulated in TMZ-resistant GBM cells-derived exosomes, and exosomal circ_0043949 increased the resistance of TMZ-resistant GBM cells to TMZ in xenograft models. TMZ-resistant GBM cells-derived exosomal circ_0043949 promoted TMZ resistance via upregulating ITGA1 expression via sequestering miR-876-3p, offering a potential target for the treatment of TMZ resistance in GBM.
Collapse
Affiliation(s)
- Xuzhao Li
- Department of Neurosurgery, The First People's Hospital of Changde City, Changde City, 415000, Hunan, China
| | - Nianhua Wang
- Department of Neurosurgery, The First People's Hospital of Changde City, Changde City, 415000, Hunan, China
| | - Haibin Leng
- Department of Neurosurgery, The First People's Hospital of Changde City, Changde City, 415000, Hunan, China
| | - Huichun Yuan
- Department of Neurosurgery, The First People's Hospital of Changde City, Changde City, 415000, Hunan, China
| | - Lixin Xu
- Department of Neurosurgery, The First People's Hospital of Changde City, Changde City, 415000, Hunan, China.
- Department of Neurosurgery, The First People's Hospital of Changde City, No.818, Renmin Road, Changde, 415000, Hunan, China.
| |
Collapse
|
18
|
Chen JR, Zhao JT, Xie ZZ. Integrin-mediated cancer progression as a specific target in clinical therapy. Biomed Pharmacother 2022; 155:113745. [DOI: 10.1016/j.biopha.2022.113745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/15/2022] Open
|
19
|
Ngalim SH, Yusoff N, Johnson RR, Abdul Razak SR, Chen X, Hobbs JK, Lee YY. A review on mechanobiology of cell adhesion networks in different stages of sporadic colorectal cancer to explain its tumorigenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:63-72. [PMID: 36116549 DOI: 10.1016/j.pbiomolbio.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
Sporadic colorectal cancer (CRC) is strongly linked to extraneous factors, like poor diet and lifestyle, but not to inherent factors like familial genetics. The changes at the epigenomics and signalling pathways are known across the sporadic CRC stages. The catch is that temporal information of the onset, the feedback loop, and the crosstalk of signalling and noise are still unclear. This makes it challenging to diagnose and treat colon cancer effectively with no relapse. Various microbial cells and native cells of the colon, contribute to sporadic CRC development. These cells secrete autocrine and paracrine for their bioenergetics and communications with other cell types. Imbalances of the biochemicals affect the epithelial lining of colon. One side of this epithelial lining is interfacing the dense colon tissue, while the other side is exposed to microbiota and excrement from the lumen. Hence, the epithelial lining is prone to tumorigenesis due to the influence of both biochemical and mechanical cues from its complex surrounding. The role of physical transformations in tumorigenesis have been limitedly discussed. In this context, cellular and tissue structures, and force transductions are heavily regulated by cell adhesion networks. These networks include cell anchoring mechanism to the surrounding, cell structural integrity mechanism, and cell effector molecules. This review will focus on the progression of the sporadic CRC stages that are governed by the underlaying cell adhesion networks within the epithelial cells. Additionally, current and potential technologies and therapeutics that target cell adhesion networks for treatments of sporadic CRC will be incorporated.
Collapse
Affiliation(s)
- Siti Hawa Ngalim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia.
| | - Norwahida Yusoff
- School of Mechanical Engineering, Universiti Sains Malaysia (USM) Engineering Campus, 14300 Nibong Tebal, Penang, Malaysia
| | - Rayzel Renitha Johnson
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Siti Razila Abdul Razak
- Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM) Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Xinyue Chen
- Department of Physics and Astronomy, University of Sheffield, Hounsfield Road, Sheffield, S3 7RH, United Kingdom
| | - Jamie K Hobbs
- Department of Physics and Astronomy, University of Sheffield, Hounsfield Road, Sheffield, S3 7RH, United Kingdom
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia (USM) Kubang Kerian, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
20
|
Brockmueller A, Mueller AL, Shayan P, Shakibaei M. β1-Integrin plays a major role in resveratrol-mediated anti-invasion effects in the CRC microenvironment. Front Pharmacol 2022; 13:978625. [PMID: 36120305 PMCID: PMC9479132 DOI: 10.3389/fphar.2022.978625] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Tumor microenvironment (TME) is one of the most important factors in tumor aggressiveness, with an active exchange between tumor and other TME-associated cells that promotes metastasis. The tumor-inhibitory effect of resveratrol on colorectal cancer (CRC) cells has been frequently reported. However, whether resveratrol can specifically suppress TME-induced CRC invasion via β1-integrin receptors has not been fully elucidated yet. Methods: Two CRC cell lines (HCT116, RKO) were cultured in multicellular, pro-inflammatory 3D-alginate TME cultures (containing fibroblasts, T-lymphocytes) to investigate the role of β1-integrin receptors in the anti-invasive and anti-metastatic effect of resveratrol by antisense oligonucleotides (ASO). Results: Our results show that resveratrol dose-dependently suppressed the migration-promoting adhesion adapter protein paxillin and simultaneously enhanced the expression of E-cadherin associated with the phenotype change of CRC cells, and their invasion. Moreover, resveratrol blocked TME-induced phosphorylation and nuclear translocation of p65-NF-κB, which was associated with changes in the expression pattern of epithelial-mesenchymal-transition-related biomarkers (slug, vimentin, E-cadherin), metastasis-related factors (CXCR4, MMP-9, FAK), and apoptosis (caspase-3). Finally, transient transfection of β1-integrin, in contrast to knockdown of NF-κB, abrogated most anti-invasive, anti-metastatic effects as well as downstream signaling of resveratrol, resulting in a concomitant increase in CRC cell invasion, indicating a central role of β1-integrin receptors in the anti-invasive function of resveratrol. Conclusion: These results demonstrate for the first time that silencing β1-integrins may suppress, at least in part the inhibitory effects of resveratrol on invasion and migration of CRC cells, underscoring the crucial homeostatic role of β1-integrin receptors.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Faculty of Medicine, Institute of Anatomy, Chair of Vegetative Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anna-Lena Mueller
- Musculoskeletal Research Group and Tumor Biology, Faculty of Medicine, Institute of Anatomy, Chair of Vegetative Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Parviz Shayan
- Department of Parasitology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Faculty of Medicine, Institute of Anatomy, Chair of Vegetative Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
- *Correspondence: Mehdi Shakibaei,
| |
Collapse
|
21
|
Xiao S, Yang C, Zhang Y, Lai C. Downregulation of B3GNT6 is a predictor of poor outcomes in patients with colorectal cancer. World J Surg Oncol 2022; 20:110. [PMID: 35387659 PMCID: PMC8988341 DOI: 10.1186/s12957-022-02561-x] [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: 12/07/2021] [Accepted: 03/10/2022] [Indexed: 12/03/2022] Open
Abstract
Background The B3GNT6 protein is a member of the O-GlcNAc transferase (OGT) family and is responsible for the production of the core 3 structure of O-glycans. It is generally expressed in the gastrointestinal (GI) tract; however, its clinical significance in colorectal cancer remains largely unexplored. Methods We obtained mRNA transcriptomic sequencing data from 3 gene expression omnibus (GEO) datasets (GSE37182, GSE39582, GSE103512) and The Cancer Genome Atlas (TCGA) to compare the B3GNT6 mRNA levels between colorectal cancer and normal tissues and further evaluate its value as a prognostic marker in colorectal cancer. We further validated this at the protein level in our cohort using immunohistochemical staining of B3GNT6 as well as the Human Protein Atlas online database. Results B3GNT6 expression was downregulated in colorectal cancer tissues as compared to that in the normal tissues at both mRNA and protein levels. Downregulation of B3GNT6 expression was found to be associated with poor overall survival in patients with colorectal cancer as per the data in GSE39582 and TCGA databases. Low B3GNT6 mRNA levels were significantly associated with chromosome instability (CIN) and KRAS mutations in patients with colorectal cancer. Gene set enrichment analysis (GSEA) revealed that low B3GNT6 expression levels in colorectal cancer were associated with increased proteasome activity. Conclusions The results of this study demonstrate that low expression of B3GNT6 is a potential biomarker for poor outcomes in patients with CRC. Moreover, the low expression of B3GNT6 may indicate more frequent activation of the KRAS/ERK signaling pathway, high CIN, and increased proteasomal activity. These novel findings may prove helpful for molecular diagnosis and provide a new therapeutic target for colorectal cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-022-02561-x.
Collapse
Affiliation(s)
- Shihan Xiao
- Department of General Surgery, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China.,Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China.,International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standardization, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China
| | - Chen Yang
- Department of General Surgery, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China.,Department of Colorectal Surgery, 1st Affiliated Hospital of Zhejiang University, Hangzhou, 310000, Zhejiang Province, China
| | - Yang Zhang
- Department of General Surgery, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China.,Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China.,International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standardization, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China
| | - Chen Lai
- Department of General Surgery, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China. .,Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China. .,International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standardization, Xiangya Hospital of Central South University, Changsha, 410000, Hunan Province, China.
| |
Collapse
|
22
|
Qin Z, Luo K, Liu Y, Liao S, He J, He M, Xie T, Jiang X, Li B, Liu H, Huang Q, Tang H, Feng W, Zhan X. ATG16L1 is a Potential Prognostic Biomarker and Immune Signature for Osteosarcoma: A Study Based on Bulk RNA and Single-Cell RNA-Sequencing. Int J Gen Med 2022; 15:1033-1045. [PMID: 35140506 PMCID: PMC8818976 DOI: 10.2147/ijgm.s341879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/10/2022] [Indexed: 11/23/2022] Open
Abstract
Background Osteosarcoma is a common solid malignancy of the bone in children and adolescents, and its metastasis and recurrence are the principal causes of poor treatment outcomes. Methods Autophagy-related genes were used to cluster osteosarcoma patients by consensus clustering analysis using the GSE21257 database. Differentially expressed genes (DEGs) were identified by limma package. Multiple-gene risk signature was constructed using least absolute shrinkage and selection operator (LASSO) analysis and Cox regression analyses. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to determine gene expression levels. Then, single-cell RNA-sequencing dataset GSE152048 were used to identify the correlation between the DEGs and effector molecules expressed in specific tumor-infiltrating immune cells. Results Two clusters were identified in the consensus clustering analysis, which were confirmed by principal component analysis. Limma analysis revealed that 15 genes were related, and 9 genes were screened using protein-protein interaction network and LASSO regression analysis. Cox regression analyses identified 5 genes. Combined with survival analysis, only the autophagy related 16 like 1 gene (ATG16L1) was significant. The results of qRT-PCR showed low expression levels of ATG16L1 in tumor cells group. Immune infiltration analysis revealed significantly lower expression of CD8+ T cells in the high ATG16L1 gene expression group. ScRNA-seq revealed that in the ATG16L1+CD8+ T cell group, the expression of GZMB was lower, whereas the expression of ITGA1 was higher. These results showed that ATG16L1 is an immune-related gene, which is associated with poor prognosis in patients with osteosarcoma. Conclusion ATG16L1 is a potential prognostic biomarker and immune signature and may be a therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Zhaojie Qin
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
- Department of Orthopedic, The People’s Hospital of Hechi, Hechi, 547600, Guangxi, People’s Republic of China
| | - Kai Luo
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Yun Liu
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Shijie Liao
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Juliang He
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, People’s Republic of China
| | - Mingwei He
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Tianyu Xie
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Xiaohong Jiang
- Department of Orthopedic, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Boxiang Li
- Department of Orthopedic, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Huijiang Liu
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
- Department of Orthopedics, The First People’s Hospital of Nanning, Nanning, 530021, Guangxi, People’s Republic of China
| | - Qian Huang
- Department of Orthopedics, The First People’s Hospital of Nanning, Nanning, 530021, Guangxi, People’s Republic of China
| | - Haijun Tang
- Department of Orthopedic, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
| | - Wenyu Feng
- Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
- Correspondence: Xinli Zhan, Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China, Tel +86 771-5350189, Fax +867715350001, Email ; Wenyu Feng, Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China, Tel +86 18277185646, Fax +867715350001, Email
| | - Xinli Zhan
- Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China
- Correspondence: Xinli Zhan, Department of Spinal Bone Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China, Tel +86 771-5350189, Fax +867715350001, Email ; Wenyu Feng, Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People’s Republic of China, Tel +86 18277185646, Fax +867715350001, Email
| |
Collapse
|
23
|
Yu J, Yang K, Zheng J, Sun X, Zhao W. Establishment of a novel prognostic signature based on an identified expression profile of integrin superfamily to predict overall survival of patients with colorectal adenocarcinoma. Gene 2022; 808:145990. [PMID: 34624456 DOI: 10.1016/j.gene.2021.145990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/14/2021] [Accepted: 10/01/2021] [Indexed: 12/02/2022]
Abstract
The abnormal expression of integrin superfamily members commonly related to kinds of malignancies. However, the role of integrins in predicting the prognosis of cancers is still little known, especially for colorectal cancer that is one of the leading causes of cancer-related death. RNA-seq data and clinical features of colorectal adenocarcinoma (COAD) patients were derived from The Cancer Genome Atlas (TCGA), used to analyze the expression pattern and genomic alterations of integrin genes in the COAD cohort. Unsupervised hierarchical clustering divided COAD patients into two clusters (clusters 1 & 2), and we observed that patients in cluster 2 with high expressions of most integrin genes had worse clinical features and shorter overall survival (a median OS: 67.25 months vs 99.93 months, p = 0.012), compared to those in cluster 1. Combined with univariate Cox regression analysis, Pearson Correlation Coefficients (PCC), and Principal Component Analysis (PCA), an integrin-related signature was established, including ITGA1, ITGA5, ITGA7, ITGA11, ITGAX, ITGAM, ITGB1, and ITGB5. And the AUC values for OS at 1, 3, and 5 years was 0.61, 0.59, and 0.56, further demonstrating the predicting capacity of our signature. Furthermore, overexpression of which also significantly correlated with poorer prognosis of colon cancer patients in a separate validation cohort, GSE17536 (p < 0.05). Meanwhile, the AUC values for OS in the validation cohort at 1, 3, and 5 years was 0.62, 0.59, and 0.59. Additionally, enrichment analysis indicated significant differences between cluster 1 and cluster 2 in the biological processes of cell adhesion, signal transduction, extracellular matrix, immune system, and in tumor microenvironment (TME), which were crucial to the progression of tumor. The findings supplied compelling evidence that our signature could be a novel prognostic biomarker for COAD patients, and these genes had the potential to be therapeutic targets.
Collapse
Affiliation(s)
- Junhui Yu
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Kui Yang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Jianbao Zheng
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Wei Zhao
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| |
Collapse
|
24
|
Ren S, Wang J, Xu A, Bao J, Cho WC, Zhu J, Shen J. Integrin α6 overexpression promotes lymphangiogenesis and lymphatic metastasis via activating the NF-κB signaling pathway in lung adenocarcinoma. Cell Oncol (Dordr) 2022; 45:57-67. [PMID: 35025009 DOI: 10.1007/s13402-021-00648-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE It has been reported that tumor-associated lymphangiogenesis plays an important role in lymph node metastasis and contributes to the poor survival of lung adenocarcinoma (LUAD) patients. As yet, however, the molecular mechanism underlying LUAD-associated lymphangiogenesis has remained elusive. METHODS Immunohistochemistry (IHC) was used to determine the expression of integrin subunit alpha 6 (ITGA6) and the lymphatic vessel endothelial hyaluronan receptor 1 (Lyve1) in clinicopathologically characterized LUAD specimens. The effect of ITGA6 overexpression on lymphangiogenesis and lymphatic metastasis was examined by tube formation, scratch wound-healing, and cell migration assays in vitro and a popliteal lymph node metastasis model in vivo. Mechanistically, overexpression of ITGA6 and activation of NF-κB signaling were examined by real-time PCR, ubiquitination and dual-luciferase reporter assays. Finally, high ITGA6 expression in LUAD tissue samples was related to copy number variation (CNV) using the TCGA database. RESULTS We found that ITGA6 overexpression correlated with microlymphatic vessel density in LUAD specimens (p < 0.01). Importantly, by using a popliteal lymph node metastasis model, we found that ITGA6 upregulation significantly enhanced lymphangiogenesis and lymphatic metastasis in vivo (p < 0.05). In addition, we found that ITGA6 overexpression enhanced the capability of A549 and H1299 LUAD cells to induce tube formation and migration in human lymphatic endothelial cells (HLECs). Mechanistically, we found that ITGA6 sustained NF-κB activity via binding and promoting K63 polyubiquitination of TNF receptor-associated factor 2 (TRAF2). Finally, CNV analysis revealed ITGA6 amplification of 27.5% in the LUAD tissue samples in the TCGA database. CONCLUSIONS Taken together, our results uncover a plausible role for ITGA6 in mediating lymphangiogenesis and lymphatic metastasis and may provide a basis for targeting ITGA6 to treat LUAD lymphatic metastasis.
Collapse
Affiliation(s)
- Sijia Ren
- Taizhou Hospital, Zhejiang University, Taizhou, 317000, China
| | - Jing Wang
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Anyi Xu
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Jiaqian Bao
- Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | - Jinrong Zhu
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Jianfei Shen
- Taizhou Hospital, Zhejiang University, Taizhou, 317000, China.
| |
Collapse
|
25
|
Li W, Yang P, Zhong C, Shen X, Shi X, Li X. The circ-PITX1 promotes non-small cell lung cancer development via the miR-30e-5p/ITGA6 axis. Cell Cycle 2022; 21:304-321. [PMID: 35007184 PMCID: PMC8855876 DOI: 10.1080/15384101.2021.2020041] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most prevalent tumors with high incidence and mortality across the globe. Recently, increasing studies have demonstrated that circular RNAs (circRNAs) exert outstanding functions in NSCLC progression. Notwithstanding, we are still in the dark about the function and exact mechanism of circ-PITX1, a newly discovered circRNA. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) confirmed the profile of circ-PITX1 in NSCLC tissues and adjacent normal tissues. Gain- and loss- of function assay verified the impact of circ-PITX1 and miR-30e-5p on the proliferation, invasion, and migration of NSCLC cells (H1975 and A549). Bioinformatics analysis corroborated the downstream mechanisms of circ-PITX1. Dual-luciferase reporter gene assay and RNA immunoprecipitation (RIP) examined the interactions between circ-PITX1 and miR-30e-5p, miR-30e-5p and ITGA6. The protein levels of ITGA6, PI3K, AKT were determined by Western blot. circ-PITX1 was substantially up-regulated in NSCLC tissues and cells, and circ-PITX1 up-regulation was correlated with NSCLC patients’ poor survival. Functionally, circ-PITX1 overexpression or miR-30e-5p inhibition markedly facilitated proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), reduced apoptosis, and enhanced ITGA6/PI3K/AKT expression in NSCLC cells, whereas circ-PITX1 knockdown or miR-30e-5p up-regulation resulted in the opposite results. Mechanistically, circ-PITX1 acted as a sponge of miR-30e-5p, which targeted the 3ʹuntranslated region (UTR) of ITGA6. Knockdown of circ-PITX1 or overexpressing miR-30e-5p reduced ITGA6/PI3K/AKT axis. circ-PITX1 modulates the miR-30e-5p/ITGA6 axis to boost NSCLC progression, hence functioning as an oncogene.
Collapse
Affiliation(s)
- Wei Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Pan Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Chucheng Zhong
- Department of Thoracic Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, China
| | - Xiaozhen Shen
- Department of Thoracic Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, China
| | - Xingyuan Shi
- Department of Radiology, The Fifth Affiliated Hospital of Guangzhou Medical University, China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
26
|
Bolatkan A, Asada K, Kaneko S, Suvarna K, Ikawa N, Machino H, Komatsu M, Shiina S, Hamamoto R. Downregulation of METTL6 mitigates cell progression, migration, invasion and adhesion in hepatocellular carcinoma by inhibiting cell adhesion molecules. Int J Oncol 2022; 60:4. [PMID: 34913069 PMCID: PMC8698744 DOI: 10.3892/ijo.2021.5294] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
RNA modifications have attracted increasing interest in recent years because they have been frequently implicated in various human diseases, including cancer, highlighting the importance of dynamic post‑transcriptional modifications. Methyltransferase‑like 6 (METTL6) is a member of the RNA methyltransferase family that has been identified in many cancers; however, little is known about its specific role or mechanism of action. In the present study, we aimed to study the expression levels and functional role of METTL6 in hepatocellular carcinoma (HCC), and further investigate the relevant pathways. To this end, we systematically conducted bioinformatics analysis of METTL6 in HCC using gene expression data and clinical information from a publicly available dataset. The mRNA expression levels of METTL6 were significantly upregulated in HCC tumor tissues compared to that in adjacent non‑tumor tissues and strongly associated with poorer survival outcomes in patients with HCC. CRISPR/Cas9‑mediated knockout of METTL6 in HCC cell lines remarkably inhibited colony formation, cell proliferation, cell migration, cell invasion and cell attachment ability. RNA sequencing analysis demonstrated that knockout of METTL6 significantly suppressed the expression of cell adhesion‑related genes. However, chromatin immunoprecipitation sequencing results revealed no significant differences in enhancer activities between cells, which suggests that METTL6 may regulate genes of interest post‑transcriptionally. In addition, it was demonstrated for the first time that METTL6 was localized in the cytosol as detected by immunofluorescence analysis, which indicates the plausible location of RNA modification mediated by METTL6. Our findings provide further insight into the function of RNA modifications in cancer and suggest a possible role of METTL6 as a therapeutic target in HCC.
Collapse
Affiliation(s)
- Amina Bolatkan
- Department of Diagnostic Imaging and Interventional Oncology, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Ken Asada
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Syuzo Kaneko
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Kruthi Suvarna
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Noriko Ikawa
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Hidenori Machino
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Masaaki Komatsu
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Shuichiro Shiina
- Department of Diagnostic Imaging and Interventional Oncology, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
- Department of National Cancer Center Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
27
|
Skrzypek K, Adamek G, Kot M, Badyra B, Majka M. Progression and Differentiation of Alveolar Rhabdomyosarcoma Is Regulated by PAX7 Transcription Factor-Significance of Tumor Subclones. Cells 2021; 10:1870. [PMID: 34440639 PMCID: PMC8391953 DOI: 10.3390/cells10081870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Rhabdomyosarcoma (RMS), is the most frequent soft tissue tumor in children that originates from disturbances in differentiation process. Mechanisms leading to the development of RMS are still poorly understood. Therefore, by analysis of two RMS RH30 cell line subclones, one subclone PAX7 negative, while the second one PAX7 positive, and comparison with other RMS cell lines we aimed at identifying new mechanisms crucial for RMS progression. RH30 subclones were characterized by the same STR profile, but different morphology, rate of proliferation, migration activity and chemotactic abilities in vitro, as well as differences in tumor morphology and growth in vivo. Our analysis indicated a different level of expression of adhesion molecules (e.g., from VLA and ICAM families), myogenic microRNAs, such as miR-206 and transcription factors, such as MYOD, MYOG, SIX1, and ID. Silencing of PAX7 transcription factor with siRNA confirmed the crucial role of PAX7 transcription factor in proliferation, differentiation and migration of RMS cells. To conclude, our results suggest that tumor cell lines with the same STR profile can produce subclones that differ in many features and indicate crucial roles of PAX7 and ID proteins in the development of RMS.
Collapse
Affiliation(s)
| | | | | | | | - Marcin Majka
- Department of Transplantation, Faculty of Medicine, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Krakow, Poland; (G.A.); (M.K.); (B.B.)
| |
Collapse
|
28
|
Mansoori B, Duijf PHG, Mohammadi A, Safarzadeh E, Ditzel HJ, Gjerstorff MF, Cho WCS, Baradaran B. MiR-142-3p targets HMGA2 and suppresses breast cancer malignancy. Life Sci 2021; 276:119431. [PMID: 33785332 DOI: 10.1016/j.lfs.2021.119431] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022]
Abstract
MicroRNAs (miRNAs) have the ability to regulate gene expression programs in cells. Hence, altered expression of miRNAs significantly contributes to breast cancer development and progression. Here, we demonstrate that the miRNA miR-142-3p directly targets the 3' untranslated region of HMGA2, which encodes an onco-embryonic protein that is overexpressed in most cancers, including breast cancer. Down regulation of miR-142-3p predicting poor patient survival in grade 3 breast cancer (P-value = 0.045). MiR-142-3p downregulates HMGA2 mRNA and protein levels. Higher miR-142-3p and lower HMGA2 expressed are found in breast cancer versus normal breast tissue (P-value<0.05), and their levels inversely correlate in breast cancers (P-value = 1.46 × 10-4). We demonstrate that miR-142-3p induces apoptosis and G2/M cell cycle arrest in breast cancer cells. In addition, it inhibits breast cancer stem cell properties and decreases SOX2, NANOG, ALDH and c-Myc expression. MiR-142-3p also decreases cell proliferation through inhibition of the ERK/AKT/STAT3 signaling pathways. Finally, pathway analyses of patient samples suggest that these mechanisms also acting in the tumors of breast cancer patients. Thus, our work identifies HMGA2 as a direct miR-142-3p target and indicates that miR-142-3p is an important suppressor of breast cancer oncogenesis. This identifies miR-142-3p may candidate as a therapeutic molecule for breast cancer treatment.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Aging Research Institute, Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Elham Safarzadeh
- Department of Microbiology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark; Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Li H, Liu Q, Chen Z, Wu M, Zhang C, Su J, Li Y, Zhang C. Hsa_circ_0110757 upregulates ITGA1 to facilitate temozolomide resistance in glioma by suppressing hsa-miR-1298-5p. Cell Death Dis 2021; 12:252. [PMID: 33674567 PMCID: PMC7935991 DOI: 10.1038/s41419-021-03533-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
Temozolomide (TMZ) is the internationally recognized and preferred drug for glioma chemotherapy treatment. However, TMZ resistance in glioma appears after long-term use and is an urgent problem that needs to be solved. Circular RNAs (circRNAs) are noncoding RNAs and play an important role in the pathogenesis and progression of tumors. Hsa_circ_0110757 was identified in TMZ-resistant glioma cells by high-throughput sequencing analysis and was derived from reverse splicing of myeloid cell leukemia-1 (Mcl-1) exons. The role of hsa_circ_0110757 in TMZ-resistant glioma was evaluated both in vitro and in vivo. It was found that hsa_circ_0110757 and ITGA1 are more highly expressed in TMZ-resistant glioma than in TMZ-sensitive glioma. The overexpression of hsa_circ_0110757 in glioma patients treated with TMZ was obviously associated with tumor invasion. This study indicates that hsa_circ_0110757 inhibits glioma cell apoptosis by sponging hsa-miR-1298-5p to promote ITGA1 expression. Thus, hsa_circ_0110757/hsa-miR-1298-5p/ITGA could be a potential therapeutic target for reversing the resistance of glioma to TMZ.
Collapse
Affiliation(s)
- Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Changsha, 410008, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China
| | - Zihua Chen
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Changsha, 410008, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ming Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China
| | - Chao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China
| | - Jun Su
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China
| | - Yue Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China
| | - Chi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, 410008, China.
| |
Collapse
|
30
|
Wang B, Xu T, Li Y, Wang W, Lyu C, Luo D, Yang Q, Ning N, Chen ZJ, Yan J, Chen DB, Li J. Trophoblast H2S Maintains Early Pregnancy via Regulating Maternal-Fetal Interface Immune Hemostasis. J Clin Endocrinol Metab 2020; 105:5854355. [PMID: 32506120 PMCID: PMC7526739 DOI: 10.1210/clinem/dgaa357] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT Dysregulated immune hemostasis occurs in unexplained recurrent spontaneous abortion (URSA). Synthesized by cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE), hydrogen sulfide (H2S) promotes regulatory T-cell differentiation and regulates immune hemostasis; yet, its role in URSA is elusive. OBJECTIVE To determine if H2S plays a role in early pregnancy and if dysregulated H2S signaling results in recurrent spontaneous abortion. DESIGN First trimester placenta villi and decidua were collected from normal and URSA pregnancies. Protein expression was examined by immunohistochemistry and immunoblotting. Human trophoblast HTR8/SVneo and JEG3 cells were treated with H2S donors; HTR8/SVneo cells were transfected with CBS ribonucleic acid interference (RNAi) or complementary deoxyribonucleic acid. Cell migration and invasion were determined by transwell assays; trophoblast transcriptomes were determined by RNA sequencing (RNA-seq). Wild-type, CBS-deficient, and CBA/J × DBA/2 mice were treated with CBS and CSE inhibitors or H2S donors to determine the role of H2S in early pregnancy in vivo. RESULTS CBS and CSE proteins showed cell-specific expressions, but only CBS decreased in the villous cytotrophoblast in URSA versus normal participants. H2S donors promoted migration and invasion and MMP-2 and VEGF expression in human placenta trophoblast cells that contain SV40 viral deoxyribonucleic acid sequences (HTR8/SVneo) and human placenta trophoblast cells (JEG3 cells), similar to forced CBS expression in HTR8/SVneo cells. The CBS-responsive transcriptomes in HTR8/SVneo cells contained differentially regulated genes (ie, interleukin-1 receptor and prostaglandin-endoperoxide synthase 2) that are associated with nuclear factor-κB-mediated inflammatory response. In vivo, dysregulated CBS/H2S signaling significantly increased embryonic resorption and decidual T-helper 1/T-helper 2 imbalance in mice, which was partially rescued by H2S donors. CONCLUSION CBS/H2S signaling maintains early pregnancy, possibly via regulating maternal-fetal interface immune hemostasis, offering opportunities for H2S-based immunotherapies for URSA.
Collapse
Affiliation(s)
- Banqin Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tonghui Xu
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Li
- Medical Research Center, Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenfu Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunzi Lyu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dan Luo
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiuhong Yang
- Department of Gynaecology and Obstetrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Obstetrics and Gynecology, Jinan Maternity and Child Care Hospital, Jinan, China
| | - Nannan Ning
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Junhao Yan
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Correspondence and Reprint Requests: Jingxin Li, MD, PhD, Department of Physiology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China. E-mail: . Junhao Yan, MD, PhD, Center for Reproductive Medicine, Shandong University, Jinan, 250021, China. ; Dongbao Chen, Ph.D., Department of Obstetrics & Gynecology, University of California, Irvine, 92697, USA.
| | - Dong-bao Chen
- Department of Obstetrics & Gynecology, University of California, Irvine
- Correspondence and Reprint Requests: Jingxin Li, MD, PhD, Department of Physiology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China. E-mail: . Junhao Yan, MD, PhD, Center for Reproductive Medicine, Shandong University, Jinan, 250021, China. ; Dongbao Chen, Ph.D., Department of Obstetrics & Gynecology, University of California, Irvine, 92697, USA.
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- Correspondence and Reprint Requests: Jingxin Li, MD, PhD, Department of Physiology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China. E-mail: . Junhao Yan, MD, PhD, Center for Reproductive Medicine, Shandong University, Jinan, 250021, China. ; Dongbao Chen, Ph.D., Department of Obstetrics & Gynecology, University of California, Irvine, 92697, USA.
| |
Collapse
|
31
|
Expression and Prognostic Analysis of Integrins in Gastric Cancer. JOURNAL OF ONCOLOGY 2020; 2020:8862228. [PMID: 33335550 PMCID: PMC7722456 DOI: 10.1155/2020/8862228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022]
Abstract
Background Integrins are involved in the biological process of a variety of cancers, but their importance in the diagnosis and prognosis of gastric cancer (GC) is still unclear. Therefore, this study aimed at exploring the significance of ITG gene expression in GC to evaluate its diagnosis and prognosis. Methods GEPIA data were used to evaluate the mRNA expression of ITG genes in GC patients. The prognostic value of these genes was assessed by analyzing their mRNA expression using the Kaplan–Meier curve. The biological function of ITG genes was evaluated by GC tissue sequencing combined with GSEA bioinformatics. Based on the sequencing data, ITGA5 with the largest expression difference was selected for verification, and RT-PCR was used to verify its mRNA expression level in 40 pairs of GC and normal tissues. Results ITG (A2, A3, A4, A5, A6, A11, AE, AL, AM, AV, AX, B1, B2, B4, B5, B6, and B8) was highly expressed in GC tissues, while ITGA8 was low, compared with their expression in normal tissues. RNA-seq data shows that ITG (A2, A5, A11, AV, and B1) expression was associated with poor prognosis and overall survival. In addition, combined with the results of GC tissue mRNA sequencing, it was further found that the differentially expressed genes in the ITGs genes. ITGA5 was highly expressed in GC tissues compared with its expression in normal tissues, as evaluated by qRT–PCR (P < 0.001) and ROC (P < 0.001, AUC (95% CI) = 0.747 (0.641–0.851)), and confirmed that ITGA5 expression was a potential diagnostic marker for GC. Bioinformatics analysis revealed that the signaling pathway involved in ITGA5 was mainly enriched in focal adhesion, ECM-receptor interaction, and PI3K-AKT and was mainly involved in biological processes such as cell adhesion, extracellular matrix, and cell migration. Conclusion This study suggested that ITGs were associated with the diagnosis and prognosis of GC and discovered the prognostic value and biological role of ITGA5 in GC. Thus, ITGA5 might be used as a potential diagnostic marker for GC.
Collapse
|
32
|
Chin PS, Assi SA, Ptasinska A, Imperato MR, Cockerill PN, Bonifer C. RUNX1/ETO and mutant KIT both contribute to programming the transcriptional and chromatin landscape in t(8;21) acute myeloid leukemia. Exp Hematol 2020; 92:62-74. [PMID: 33152396 DOI: 10.1016/j.exphem.2020.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia development occurs in a stepwise fashion whereby an original driver mutation is followed by additional mutations. The first type of mutations tends to be in genes encoding members of the epigenetic/transcription regulatory machinery (i.e., RUNX1, DNMT3A, TET2), while the secondary mutations often involve genes encoding members of signaling pathways that cause uncontrolled growth of such cells such as the growth factor receptors c-KIT of FLT3. Patients usually present with both types of mutations, but it is currently unclear how both mutational events shape the epigenome in developing AML cells. To this end we generated an in vitro model of t(8;21) AML by expressing its driver oncoprotein RUNX1-ETO with or without a mutated (N822K) KIT protein. Expression of N822K-c-KIT strongly increases the self-renewal capacity of RUNX1-ETO-expressing cells. Global analysis of gene expression changes and alterations in the epigenome revealed that N822K-c-KIT expression profoundly influences the open chromatin landscape and transcription factor binding. However, our experiments also revealed that double mutant cells still differ from their patient-derived counterparts, highlighting the importance of studying patient cells to obtain a true picture of how gene regulatory networks have been reprogrammed during tumorigenesis.
Collapse
Affiliation(s)
- Paulynn Suyin Chin
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Salam A Assi
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Anetta Ptasinska
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Maria Rosaria Imperato
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Peter N Cockerill
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Constanze Bonifer
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
33
|
Liu L, Liu H, Luo S, Patz EF, Glass C, Su L, Lin L, Christiani DC, Wei Q. Novel genetic variants of SYK and ITGA1 related lymphangiogenesis signaling pathway predict non-small cell lung cancer survival. Am J Cancer Res 2020; 10:2603-2616. [PMID: 32905494 PMCID: PMC7471352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 06/28/2020] [Indexed: 06/11/2023] Open
Abstract
Although lymphangiogenesis is a vital step in lung cancer metastasis, the association between lymphangiogenesis and non-small cell lung cancer (NSCLC) survival remains unclear. Since single-nucleotide polymorphisms (SNPs) have been reported to predict NSCLC survival, we investigated associations between SNPs in lymphangiogenesis-related pathway genes and NSCLC survival in a discovery genotyping dataset of 1,185 patients from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial and validated the findings in another genotyping dataset of 984 patients from the Harvard Lung Cancer Susceptibility Study. We evaluated associations between 34,509 genetic variants (3252 genotyped and 31,257 imputed) in 247 genes involved in lymphangiogenesis-related pathway and NSCLC survival. After validation, we finally identified two independent SNPs (SYK rs11787670 A>G and ITGA1 rs67715745 T>C) to be significantly associated with NSCLC overall survival (OS), with adjusted hazards ratios of 0.77 and 0.83 (95% confidence interval =0.66-0.90, P=7.20×10-4) and 0.84 (95% confidence interval =0.75-0.92, P=3.50×10-4), respectively. Moreover, an increasing number of combined protective alleles of these two SNPs was significantly associated with an improved NSCLC OS and disease-specific survival (DSS) in the PLCO dataset (P trend=0.011 and 0.006, respectively). Furthermore, the addition of these protective alleles to the prediction model for the 5-year survival increased the time-dependent area under the curve both from 87% to 87.67% for OS (P=0.029) and from 88.54% to 89.06% for DSS (P=0.022). Subsequent expression quantitative trait loci (eQTL) functional analysis revealed that the rs11787670 G allele was significantly associated with an elevated SYK mRNA expression in normal tissues. Additional analyses suggested a suppressor role for both SYK and ITGA1 in NSCLC survival. Collectively, these findings indicated that SYK rs11787670 A>G and ITGA1 rs67715745 T>C may be independent prognostic factors for NSCLC survival once further validated.
Collapse
Affiliation(s)
- Lihua Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical UniversityNanning, Guangxi 530021, China
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC 27710, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of MedicineDurham, NC 27710, USA
| | - Edward F Patz
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Radiology, Pharmacology and Cancer Biology, Duke University School of MedicineDurham, NC 27710, USA
| | - Carolyn Glass
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Pathology, Duke University School of MedicineDurham, NC 27710, USA
| | - Li Su
- Departments of Environmental Health and Epidemiology, Harvard School of Public HealthBoston, MA, 02115 USA
| | - Lijuan Lin
- Departments of Environmental Health and Epidemiology, Harvard School of Public HealthBoston, MA, 02115 USA
| | - David C Christiani
- Departments of Environmental Health and Epidemiology, Harvard School of Public HealthBoston, MA, 02115 USA
- Department of Medicine, Massachusetts General HospitalBoston, MA 02114, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical CenterDurham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of MedicineDurham, NC 27710, USA
- Department of Medicine, Duke University School of MedicineDurham, NC 27710, USA
| |
Collapse
|