1
|
Fan Y, Wu L, Qiu X, Shi H, Wu L, Lin J, Lin J, Teng T. Single-cell RNA-seq analysis reveals microenvironmental infiltration of myeloid cells and pancreatic prognostic markers in PDAC. Discov Oncol 2025; 16:81. [PMID: 39847195 PMCID: PMC11757659 DOI: 10.1007/s12672-025-01830-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a heterogeneous make-up of myeloid cells that influences the therapeutic response and prognosis. However, understanding the myeloid cell at both a genetic and cellular level remains a significant challenge. METHODS Single-cell RNA sequencing (scRNA-seq) data were downloaded from t the Tumor Immune Single-cell Hub and gene expression data were retrieved from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database. Gene set variation analysis (GSVA) was used to estimate the relative proportions of each cell type based on the signatures identified by scRNA-seq or previous literature. Cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) was performed to evaluate the abundance of immune infiltrating cells. For further analysis, LASSO and Cox analyses were used to construct a risk model using univariate Cox regression. RESULTS Using the scRNA-seq dataset, we identified 7 clusters of myeloid cells, and these clusters were assigned a cell type based on their marker genes. In addition, the results of the CellChat analysis and SCENIC analysis indicate that TAM-spp1 cells may promote the migration of pancreatic tumor cells on different levels. Moreover, the TAM-spp1 cell is most closely related to poor prognoses. An 8-gene risk model was constructed by using univariate Cox and LASSO analyses. In the GEO cohorts, this risk model demonstrated excellent predictive abilities for prognosis. Further, patients with high-risk scores had a lower likelihood of benefiting from immunotherapy. CONCLUSION Using bulk RNA-seq and single-cell RNA-seq, we analyzed myeloid heterogeneity at the single-cell level, and we developed an 8-gene model that predicts survival outcomes and immunotherapy response in PADC.
Collapse
Affiliation(s)
- Yanying Fan
- Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China
| | - Lili Wu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
- Department of Surgical Nursing, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Xinyu Qiu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Han Shi
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Longhang Wu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China
| | - Juan Lin
- Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China
| | - Jie Lin
- Fuzhou First General Hospital Affiliated With Fujian Medical University, Fuzhou, China.
| | - Tianhong Teng
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, People's Republic of China.
| |
Collapse
|
2
|
Wang SY, Wang YX, Guan LS, Shen A, Huang RJ, Yuan SQ, Xiao YL, Wang LS, Lei D, Zhao Y, Lin C, Wang CP, Yuan ZP. Construction of a prognostic model for gastric cancer based on immune infiltration and microenvironment, and exploration of MEF2C gene function. BMC Med Genomics 2025; 18:13. [PMID: 39810215 PMCID: PMC11734330 DOI: 10.1186/s12920-024-02082-4] [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: 11/10/2023] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Advanced gastric cancer (GC) exhibits a high recurrence rate and a dismal prognosis. Myocyte enhancer factor 2c (MEF2C) was found to contribute to the development of various types of cancer. Therefore, our aim is to develop a prognostic model that predicts the prognosis of GC patients and initially explore the role of MEF2C in immunotherapy for GC. METHODS Transcriptome sequence data of GC was obtained from The Cancer Genome Atlas (TCGA), the Gene Expression Omnibus (GEO) and PRJEB25780 cohort for subsequent immune infiltration analysis, immune microenvironment analysis, consensus clustering analysis and feature selection for definition and classification of gene M and N. Principal component analysis (PCA) modeling was performed based on gene M and N for the calculation of immune checkpoint inhibitor (ICI) Score. Then, a Nomogram was constructed and evaluated for predicting the prognosis of GC patients, based on univariate and multivariate Cox regression. Functional enrichment analysis was performed to initially investigate the potential biological mechanisms. Through Genomics of Drug Sensitivity in Cancer (GDSC) dataset, the estimated IC50 values of several chemotherapeutic drugs were calculated. Tumor-related transcription factors (TFs) were retrieved from the Cistrome Cancer database and utilized our model to screen these TFs, and weighted correlation network analysis (WGCNA) was performed to identify transcription factors strongly associated with immunotherapy in GC. Finally, 10 patients with advanced GC were enrolled from Sun Yat-sen University Cancer Center, including paired tumor tissues, paracancerous tissues and peritoneal metastases, for preparing sequencing library, in order to perform external validation. RESULTS Lower ICI Score was correlated with improved prognosis in both the training and validation cohorts. First, lower mutant-allele tumor heterogeneity (MATH) was associated with lower ICI Score, and those GC patients with lower MATH and lower ICI Score had the best prognosis. Second, regardless of the T or N staging, the low ICI Score group had significantly higher overall survival (OS) compared to the high ICI Score group. For its mechanisms, consistently, for Camptothecin, Doxorubicin, Mitomycin, Docetaxel, Cisplatin, Vinblastine, Sorafenib and Paclitaxel, all of the IC50 values were significantly lower in the low ICI Score group compared to the high ICI Score group. As a result, based on univariate and multivariate Cox regression, ICI Score was considered to be an independent prognostic factor for GC. And our Nomogram showed good agreement between predicted and actual probabilities. Based on CIBERSORT deconvolution analysis, there was difference of immune cell composition found between high and low ICI Score groups, probably affecting the efficacy of immunotherapy. Then, MEF2C, a tumor-related transcription factor, was screened out by WGCNA analysis. Higher MEF2C expression is significantly correlated with a worse OS. Moreover, its higher expression is also negatively correlated with tumor mutation burden (TMB) and microsatellite instability (MSI), but positively correlated with several immunosuppressive molecules, indicating MEF2C may exert its influence on tumor development by upregulating immunosuppressive molecules. Finally, based on transcriptome sequencing data on 10 paired tumor tissues from Sun Yat-sen University Cancer Center, MEF2C expression was significantly lower in paracancerous tissues compared to tumor tissues and peritoneal metastases, and it was also lower in tumor tissues compared to peritoneal metastases, indicating a potential positive association between MEF2C expression and tumor invasiveness. CONCLUSIONS Our prognostic model can effectively predict outcomes and facilitate stratification GC patients, offering valuable insights for clinical decision-making. The identified transcription factor MEF2C can serve as a biomarker for assessing the efficacy of immunotherapy for GC.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Yu-Xin Wang
- The First Hospital of Jilin University, Changchun, 130000, China
| | - Lu-Shun Guan
- China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Ao Shen
- Departments of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Run-Jie Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yu-Long Xiao
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Li-Shuai Wang
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Dan Lei
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Yin Zhao
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Chuan Lin
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Chang-Ping Wang
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Zhi-Ping Yuan
- Department of Oncology, The First People's Hospital of Yibin, No.65, Wenxing Street, Cuiping District, Yibin, 644000, China.
| |
Collapse
|
3
|
Xia L, Nie T, Lu F, Huang L, Shi X, Ren D, Lu J, Li X, Xu T, Cui B, Wang Q, Gao G, Yang Q. Direct regulation of FNIP1 and FNIP2 by MEF2 sustains MTORC1 activation and tumor progression in pancreatic cancer. Autophagy 2024; 20:505-524. [PMID: 37772772 PMCID: PMC10936626 DOI: 10.1080/15548627.2023.2259735] [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: 01/12/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) orchestrates diverse environmental signals to facilitate cell growth and is frequently activated in cancer. Translocation of MTORC1 from the cytosol to the lysosomal surface by the RRAG GTPases is the key step in MTORC1 activation. Here, we demonstrated that transcription factors MEF2A and MEF2D synergistically regulated MTORC1 activation via modulating its cyto-lysosome shutting. Mechanically, MEF2A and MEF2D controlled the transcription of FNIP1 and FNIP2, the components of the FLCN-FNIP1 or FNIP2 complex that acts as a RRAGC-RRAGD GTPase-activating element to promote the recruitment of MTORC1 to lysosome and its activation. Furthermore, we determined that the pro-oncogenic protein kinase SRC/c-Src directly phosphorylated MEF2D at three conserved tyrosine residues. The tyrosine phosphorylation enhanced MEF2D transcriptional activity and was indispensable for MTORC1 activation. Finally, both the protein and tyrosine phosphorylation levels of MEF2D are elevated in human pancreatic cancers, positively correlating with MTORC1 activity. Depletion of both MEF2A and MEF2D or expressing the unphosphorylatable MEF2D mutant suppressed tumor cell growth. Thus, our study revealed a transcriptional regulatory mechanism of MTORC1 that promoted cell anabolism and proliferation and uncovered its critical role in pancreatic cancer progression.Abbreviation: ACTB: actin beta; ChIP: chromatin immunoprecipitation; EGF: epidermal growth factor; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; FLCN: folliculin; FNIP1: folliculin interacting protein 1; FNIP2: folliculin interacting protein 2; GAP: GTPase activator protein; GEF: guanine nucleotide exchange factors; GTPase: guanosine triphosphatase; LAMP2: lysosomal associated membrane protein 2; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MEF2: myocyte enhancer factor 2; MEF2A: myocyte enhancer factor 2A; MEF2D: myocyte enhancer factor 2D; MEF2D-3YF: Y131F, Y333F, Y337F mutant; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; NR4A1: nuclear receptor subfamily 4 group A member 1; RPTOR: regulatory associated protein of MTOR complex 1; RHEB: Ras homolog, mTORC1 binding; RPS6KB1: ribosomal protein S6 kinase B1; RRAG: Ras related GTP binding; RT-qPCR: real time-quantitative PCR; SRC: SRC proto-oncogene, non-receptor tyrosine kinase; TMEM192: transmembrane protein 192; WT: wild-type.
Collapse
Affiliation(s)
- Li Xia
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Tiejian Nie
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Fangfang Lu
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Lu Huang
- Department of Anesthesiology, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiaolong Shi
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Dongni Ren
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jianjun Lu
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiaobin Li
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Tuo Xu
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Bozhou Cui
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Qing Wang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Qian Yang
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
4
|
Jiang B, Zhou Y, Liu Y, He S, Liao B, Peng T, Yao L, Qi L. Research Progress on the Role and Mechanism of IL-37 in Liver Diseases. Semin Liver Dis 2023; 43:336-350. [PMID: 37582401 PMCID: PMC10620037 DOI: 10.1055/a-2153-8836] [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] [Indexed: 08/17/2023]
Abstract
Cytokines are important components of the immune system that can predict or influence the development of liver diseases. IL-37, a new member of the IL-1 cytokine family, exerts potent anti-inflammatory and immunosuppressive effects inside and outside cells. IL-37 expression differs before and after liver lesions, suggesting that it is associated with liver disease; however, its mechanism of action remains unclear. This article mainly reviews the biological characteristics of IL-37, which inhibits hepatitis, liver injury, and liver fibrosis by inhibiting inflammation, and inhibits the development of hepatocellular carcinoma (HCC) by regulating the immune microenvironment. Based on additional evidence, combining IL-37 with liver disease markers for diagnosis and treatment can achieve more significant effects, suggesting that IL-37 can be developed into a powerful tool for the clinical adjuvant treatment of liver diseases, especially HCC.
Collapse
Affiliation(s)
- Baoyi Jiang
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Yulin Zhou
- Department of Clinical Laboratory, Shunde New Rongqi Hospital, Foshan, China
| | - Yanting Liu
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Siqi He
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Baojian Liao
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Tieli Peng
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Leyi Yao
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Ling Qi
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| |
Collapse
|
5
|
Das BK, Kannan A, Velasco GJ, Kunika MD, Lambrecht N, Nguyen Q, Zhao H, Wu J, Gao L. Single-cell dissection of Merkel cell carcinoma heterogeneity unveils transcriptomic plasticity and therapeutic vulnerabilities. Cell Rep Med 2023; 4:101101. [PMID: 37421947 PMCID: PMC10394170 DOI: 10.1016/j.xcrm.2023.101101] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/23/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023]
Abstract
Merkel cell carcinoma (MCC), a rare but aggressive skin cancer, remains a challenge in the era of precision medicine. Immune checkpoint inhibitors (ICIs), the only approved therapy for advanced MCC, are impeded by high primary and acquired resistance. Hence, we dissect transcriptomic heterogeneity at single-cell resolution in a panel of patient tumors, revealing phenotypic plasticity in a subset of treatment-naive MCC. The tumor cells in a "mesenchymal-like" state are endowed with an inflamed phenotype that portends a better ICI response. This observation is also validated in the largest whole transcriptomic dataset available from MCC patient tumors. In contrast, ICI-resistant tumors predominantly express neuroepithelial markers in a well-differentiated state with "immune-cold" landscape. Importantly, a subtle shift to "mesenchymal-like" state reverts copanlisib resistance in primary MCC cells, highlighting potential strategies in patient stratification for therapeutics to harness tumor cell plasticity, augment treatment efficacy, and avert resistance.
Collapse
Affiliation(s)
- Bhaba K Das
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Aarthi Kannan
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA; Department of Dermatology, University of California-Irvine, Irvine, CA 92697, USA
| | - Graham J Velasco
- Pathology Department, Tibor Rubin VA Medical Center, VA Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Mikaela D Kunika
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Nils Lambrecht
- Pathology Department, Tibor Rubin VA Medical Center, VA Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Quy Nguyen
- Genomics Research and Technology Hub, Department of Biological Chemistry, University of California-Irvine, Irvine, CA 92697, USA
| | - Haibo Zhao
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA
| | - Jie Wu
- Genomics Research and Technology Hub, Department of Biological Chemistry, University of California-Irvine, Irvine, CA 92697, USA
| | - Ling Gao
- Southern California Institute for Research and Education, Long Beach, CA 90822, USA; Department of Dermatology, University of California-Irvine, Irvine, CA 92697, USA; Dermatology Section, Tibor Rubin VA Medical Center, VA Long Beach Healthcare System, Long Beach, CA 90822, USA.
| |
Collapse
|
6
|
Ang HL, Mohan CD, Shanmugam MK, Leong HC, Makvandi P, Rangappa KS, Bishayee A, Kumar AP, Sethi G. Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med Res Rev 2023. [PMID: 36929669 DOI: 10.1002/med.21948] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a complex process with a primordial role in cellular transformation whereby an epithelial cell transforms and acquires a mesenchymal phenotype. This transformation plays a pivotal role in tumor progression and self-renewal, and exacerbates resistance to apoptosis and chemotherapy. EMT can be initiated and promoted by deregulated oncogenic signaling pathways, hypoxia, and cells in the tumor microenvironment, resulting in a loss-of-epithelial cell polarity, cell-cell adhesion, and enhanced invasive/migratory properties. Numerous transcriptional regulators, such as Snail, Slug, Twist, and ZEB1/ZEB2 induce EMT through the downregulation of epithelial markers and gain-of-expression of the mesenchymal markers. Additionally, signaling cascades such as Wnt/β-catenin, Notch, Sonic hedgehog, nuclear factor kappa B, receptor tyrosine kinases, PI3K/AKT/mTOR, Hippo, and transforming growth factor-β pathways regulate EMT whereas they are often deregulated in cancers leading to aberrant EMT. Furthermore, noncoding RNAs, tumor-derived exosomes, and epigenetic alterations are also involved in the modulation of EMT. Therefore, the regulation of EMT is a vital strategy to control the aggressive metastatic characteristics of tumor cells. Despite the vast amount of preclinical data on EMT in cancer progression, there is a lack of clinical translation at the therapeutic level. In this review, we have discussed thoroughly the role of the aforementioned transcription factors, noncoding RNAs (microRNAs, long noncoding RNA, circular RNA), signaling pathways, epigenetic modifications, and tumor-derived exosomes in the regulation of EMT in cancers. We have also emphasized the contribution of EMT to drug resistance and possible therapeutic interventions using plant-derived natural products, their semi-synthetic derivatives, and nano-formulations that are described as promising EMT blockers.
Collapse
Affiliation(s)
- Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hin Chong Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia Centre for Materials Interface, Pontedera, Pisa, Italy
| | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
7
|
Trefzer TB, Schneider MA, Jechow K, Chua RL, Muley T, Winter H, Kriegsmann M, Meister M, Eils R, Conrad C. Intratumoral Heterogeneity and Immune Modulation in Lung Adenocarcinoma in Female Smokers and Never Smokers. Cancer Res 2022; 82:3116-3129. [PMID: 35819252 PMCID: PMC9437562 DOI: 10.1158/0008-5472.can-21-3836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/13/2022] [Accepted: 07/08/2022] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE Single-cell analysis of healthy lung tissue and lung cancer reveals distinct tumor cell populations, including cells with differential immune modulating capacity between smokers and never smokers, which could guide future therapeutic strategies.
Collapse
Affiliation(s)
- Timo B. Trefzer
- Charité - Universitätsmedizin Berlin and Berlin Institute of Health, Digital Health Center, Berlin, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc A. Schneider
- Translational Research Unit, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Katharina Jechow
- Charité - Universitätsmedizin Berlin and Berlin Institute of Health, Digital Health Center, Berlin, Germany
| | - Robert Lorenz Chua
- Charité - Universitätsmedizin Berlin and Berlin Institute of Health, Digital Health Center, Berlin, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Mark Kriegsmann
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Meister
- Translational Research Unit, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Roland Eils
- Charité - Universitätsmedizin Berlin and Berlin Institute of Health, Digital Health Center, Berlin, Germany
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, Heidelberg, Germany
| | - Christian Conrad
- Charité - Universitätsmedizin Berlin and Berlin Institute of Health, Digital Health Center, Berlin, Germany
| |
Collapse
|
8
|
Karimzadeh M, Hoffman MM. Virtual ChIP-seq: predicting transcription factor binding by learning from the transcriptome. Genome Biol 2022; 23:126. [PMID: 35681170 PMCID: PMC9185870 DOI: 10.1186/s13059-022-02690-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
Existing methods for computational prediction of transcription factor (TF) binding sites evaluate genomic regions with similarity to known TF sequence preferences. Most TF binding sites, however, do not resemble known TF sequence motifs, and many TFs are not sequence-specific. We developed Virtual ChIP-seq, which predicts binding of individual TFs in new cell types, integrating learned associations with gene expression and binding, TF binding sites from other cell types, and chromatin accessibility data in the new cell type. This approach outperforms methods that predict TF binding solely based on sequence preference, predicting binding for 36 TFs (MCC>0.3).
Collapse
Affiliation(s)
- Mehran Karimzadeh
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Princess Margaret Cancer Centre, Toronto, ON, Canada.,Vector Institute, Toronto, ON, Canada
| | - Michael M Hoffman
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Princess Margaret Cancer Centre, Toronto, ON, Canada. .,Vector Institute, Toronto, ON, Canada. .,Department of Computer Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Bhatt S, Singh P, Sharma A, Rai A, Dohare R, Sankhwar S, Sharma A, Syed MA. Deciphering Key Genes and miRNAs Associated With Hepatocellular Carcinoma via Network-Based Approach. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:843-853. [PMID: 32795971 DOI: 10.1109/tcbb.2020.3016781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hepatocellular carcinoma (HCC)is a common type of liver cancer and has a high mortality world-widely. The diagnosis, prognoses, and therapeutics are very poor due to the unclear molecular mechanism of progression of the disease. To unveil the molecular mechanism of progression of HCC, we extract a large sample of mRNA expression levels from the GEO database where a total of 167 samples were used for study, and out of them, 115 samples were from HCC tumor tissue. This study aims to investigate the module of differentially expressed genes (DEGs)which are co-expressed only in HCC sample data but not in normal tissue samples. Thereafter, we identified the highly significant module of significant co-expressed genes and formed a PPI network for these genes. There were only six genes (namely, MSH3, DMC1, ALPP, IL10, ZNF223, and HSD17B7)obtained after analysis of the PPI network. Out of six only MSH3, DMC1, HSD17B7, and IL10 were found enriched in GO Term & Pathway enrichment analysis and these candidate genes were mainly involved in cellular process, metabolic and catalytic activity, which promote the development & progression of HCC. Lastly, the composite 3-node FFL reveals the driver miRNAs and TFs associated with our key genes.
Collapse
|
10
|
Acidic Tumor Microenvironment Promotes Pancreatic Cancer through miR-451a/MEF2D Axis. JOURNAL OF ONCOLOGY 2022; 2022:3966386. [PMID: 35069734 PMCID: PMC8769849 DOI: 10.1155/2022/3966386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer (PC), as a highly malignant and aggressive solid tumor, is common in the digestive system. The acidic microenvironment is one of the critical markers of cancer. Nonetheless, there are few studies on how the acidic microenvironment affects the development of PC. This study focused on investigating the specific molecular mechanisms of the acidic microenvironment in PC. In our study, qRT-PCR was conducted for examining microRNA (miR)-451a and myocyte enhancer factor 2D (MEF2D) expressions in PANC-1 cells. Then, detailed functional effects of an acidic environment on miR-451a and MEF2D in PANC-1 cells were detected by CCK-8, colony formation, flow cytometry, wound healing, transwell, mitochondrial functionality measurement, JC-1 staining, DCFH-DA staining, and sphere formation assays. The relationship between miR-451a and MEF2D was confirmed by luciferase reporter analysis. Under acidic conditions, the increase of proliferation, migration, and invasion of PANC-1 cells was observed. Moreover, the mitochondrial oxidative respiration-related gene miR-451a was reduced in acidic conditions. In addition, we found that, in PANC-1 cells under an acidic environment, miR-451a overexpression enhanced oxygen consumption, mitochondrial membrane potential (MMP) loss, and ROS generation and inhibited proliferation, migration, invasion, and stemness via sponging MEF2D. In a word, our results revealed that the acidic microenvironment regulated PC progression by affecting the miR-451a/MEF2D axis, indicating a novel avenue for the future treatment of PC.
Collapse
|
11
|
Groth T, Gunawan R, Neelamegham S. A systems-based framework to computationally describe putative transcription factors and signaling pathways regulating glycan biosynthesis. Beilstein J Org Chem 2021; 17:1712-1724. [PMID: 34367349 PMCID: PMC8313979 DOI: 10.3762/bjoc.17.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 07/12/2021] [Indexed: 01/05/2023] Open
Abstract
Glycosylation is a common posttranslational modification, and glycan biosynthesis is regulated by a set of glycogenes. The role of transcription factors (TFs) in regulating the glycogenes and related glycosylation pathways is largely unknown. In this work, we performed data mining of TF–glycogene relationships from the Cistrome Cancer database (DB), which integrates chromatin immunoprecipitation sequencing (ChIP-Seq) and RNA-Seq data to constitute regulatory relationships. In total, we observed 22,654 potentially significant TF–glycogene relationships, which include interactions involving 526 unique TFs and 341 glycogenes that span 29 the Cancer Genome Atlas (TCGA) cancer types. Here, TF–glycogene interactions appeared in clusters or so-called communities, suggesting that changes in single TF expression during both health and disease may affect multiple carbohydrate structures. Upon applying the Fisher’s exact test along with glycogene pathway classification, we identified TFs that may specifically regulate the biosynthesis of individual glycan types. Integration with Reactome DB knowledge provided an avenue to relate cell-signaling pathways to TFs and cellular glycosylation state. Whereas analysis results are presented for all 29 cancer types, specific focus is placed on human luminal and basal breast cancer disease progression. Overall, the article presents a computational approach to describe TF–glycogene relationships, the starting point for experimental system-wide validation.
Collapse
Affiliation(s)
- Theodore Groth
- Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Rudiyanto Gunawan
- Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Sriram Neelamegham
- Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA.,Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA.,Medicine, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
12
|
A cytokine in turmoil: Transforming growth factor beta in cancer. Biomed Pharmacother 2021; 139:111657. [PMID: 34243626 DOI: 10.1016/j.biopha.2021.111657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer remains one of the debilitating health threats to mankind in view of its incurable nature. Many factors are complicit in the initiation, progression and establishment of cancers. Early detection of cancer is the only window of hope that allows for appreciable management and possible limited survival. However, understanding of cancer biology and knowledge of the key factors that interplay at multi-level in the initiation and progression of cancer may hold possible avenues for cancer treatment and management. In particular, dysregulation of growth factor signaling such as that of transforming growth factor beta (TGF-β) and its downstream mediators play key roles in various cancer subtypes. Expanded understanding of the context/cell type-dependent roles of TGF-β and its downstream signaling mediators in cancer may provide leads for cancer pharmacotherapy. Reliable information contained in original articles, reviews, mini-reviews and expert opinions on TGF-β, cancer and the specific roles of TGF-β signaling in various cancer subtypes were retrieved from major scientific data bases including PubMed, Scopus, Medline, Web of Science core collections just to mention but a sample by using the following search terms: TGF-β in cancer, TGF-β and colorectal cancer, TGF-β and brain cancer, TGF-β in cancer initiation, TGF-β and cell proliferation, TGF-β and cell invasion, and TGF-β-based cancer therapy. Retrieved information and reports were carefully examined, contextualized and synchronized into a coherent scientific content to highlight the multiple roles of TGF-β signaling in normal and cancerous cells. From a conceptual standpoint, development of pharmacologically active agents that exert non-specific inhibitory effects on TGF-β signaling on various cell types will undoubtedly lead to a plethora of serious side effects in view of the multi-functionality and pleiotropic nature of TGF-β. Such non-specific targeting of TGF-β could derail any beneficial therapeutic intention associated with TGF-β-based therapy. However, development of pharmacologically active agents designed specifically to target TGF-β signaling in cancer cells may improve cancer pharmacotherapy. Similarly, specific targeting of downstream mediators of TGF-β such as TGF-β type 1 and II receptors (TβRI and TβRII), receptor-mediated Smads, mitogen activated protein kinase (MAPK) and importing proteins in cancer cells may be crucial for cancer pharmacotherapy.
Collapse
|
13
|
Myocyte Enhancer Factor 2C as a New Player in Human Breast Cancer Brain Metastases. Cells 2021; 10:cells10020378. [PMID: 33673112 PMCID: PMC7917785 DOI: 10.3390/cells10020378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/28/2021] [Accepted: 02/07/2021] [Indexed: 12/24/2022] Open
Abstract
Myocyte enhancer factor 2C (MEF2C) is increasingly expressed in mice along with breast cancer brain metastases (BCBM) development. We aim to ascertain MEF2C expression in human BCBM, establish the relationship with disease severity, disclose the involvement of vascular endothelial growth factor receptor-2 (VEGFR-2) and β-catenin, also known as KDR and CTNNB1, respectively, and investigate if matched primary tumors express the protein. We studied resected BCBM for the expression of MEF2C, VEGFR-2, and ß-catenin, as well as proliferation (Ki-67) and epithelial (pan Cytokeratin) markers, and related experimental and clinical data. MEF2C expression was further assessed in matched primary tumors and non-BCBM samples used as controls. MEF2C expression was observed in BCBM, but not in controls, and was categorized into three phenotypes (P): P1, with extranuclear location; P2, with extranuclear and nuclear staining, and P3, with nuclear location. Nuclear translocation increased with metastases extension and Ki-67-positive cells number. P1 was associated with higher VEFGR-2 plasma membrane immunoreactivity, whereas P2 and P3 were accompanied by protein dislocation. P1 was accompanied by β-catenin membrane expression, while P2 and P3 exhibited β-catenin nuclear translocation. Primary BC samples expressed MEF2C in mammary ducts and scattered cells in the parenchyma. MEF2C emerges as a player in BCBM associated with disease severity and VEGFR-2 and β-catenin signaling.
Collapse
|
14
|
Lucendo-Villarin B, Nell P, Hellwig B, Filis P, Feuerborn D, O'Shaughnessy PJ, Godoy P, Rahnenführer J, Hengstler JG, Cherianidou A, Sachinidis A, Fowler PA, Hay DC. Genome-wide expression changes induced by bisphenol A, F and S in human stem cell derived hepatocyte-like cells. EXCLI JOURNAL 2020; 19:1459-1476. [PMID: 33312107 PMCID: PMC7726493 DOI: 10.17179/excli2020-2934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
The debate about possible adverse effects of bisphenol A (BPA) has been ongoing for decades. Bisphenol F (BPF) and S (BPS) have been suggested as “safer” alternatives. In the present study we used hepatocyte-like cells (HLCs) derived from the human embryonic stem cell lines Man12 and H9 to compare the three bisphenol derivatives. Stem cell-derived progenitors were produced using an established system and were exposed to BPA, BPF and BPS for 8 days during their transition to HLCs. Subsequently, we examined cell viability, inhibition of cytochrome P450 (CYP) activity, and genome-wide RNA profiles. Sub-cytotoxic, inhibitory concentrations (IC50) of CYP3A were 20, 9.5 and 25 µM for BPA, BPF and BPS in Man12 derived HLCs, respectively. The corresponding concentrations for H9-derived HLCs were 19, 29 and 31 µM. These IC50 concentrations were used to study global expression changes in this in vitro study and are higher than unconjugated BPA in serum of the general population. A large overlap of up- as well as downregulated genes induced by the three bisphenol derivatives was seen. This is at least 28-fold higher compared to randomly expected gene expression changes. Moreover, highly significant correlations of expression changes induced by the three bisphenol derivatives were obtained in pairwise comparisons. Dysregulated genes were associated with reduced metabolic function, cellular differentiation, embryonic development, cell survival and apoptosis. In conclusion, no major differences in cytochrome inhibitory activities of BPA, BPF and BPS were observed and gene expression changes showed a high degree of similarity.
Collapse
Affiliation(s)
- B Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - P Nell
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - B Hellwig
- Department of Statistics, Technical University Dortmund, Dortmund, Germany
| | - P Filis
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - D Feuerborn
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - P J O'Shaughnessy
- Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, UK
| | - P Godoy
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - J Rahnenführer
- Department of Statistics, Technical University Dortmund, Dortmund, Germany
| | - J G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, Dortmund, Germany
| | - A Cherianidou
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany
| | - A Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Cologne, Germany
| | - P A Fowler
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - D C Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
15
|
Abstract
Epithelial–mesenchymal plasticity contributes to many biological processes, including tumor progression. Various epithelial–mesenchymal transition (EMT) responses have been reported and no common, EMT-defining gene expression program has been identified. Here, we have performed a comparative analysis of the EMT response, leveraging highly multiplexed single-cell RNA sequencing (scRNA-seq) to measure expression profiles of 103,999 cells from 960 samples, comprising 12 EMT time course experiments and independent kinase inhibitor screens for each. We demonstrate that the EMT is vastly context specific, with an average of only 22% of response genes being shared between any two conditions, and over half of all response genes were restricted to 1–2 time course experiments. Further, kinase inhibitor screens revealed signaling dependencies and modularity of these responses. These findings suggest that the EMT is not simply a single, linear process, but is highly variable and modular, warranting quantitative frameworks for understanding nuances of the transition. It is unclear if a common EMT expression program exists. Here, the authors perform multiplexed single-cell RNA sequencing across 12 EMT time courses and 16 kinase inhibitor screens, and find that EMT transcriptional responses are context specific and EMT is not a single, linear transition.
Collapse
|
16
|
Contextual Regulation of TGF-β Signaling in Liver Cancer. Cells 2019; 8:cells8101235. [PMID: 31614569 PMCID: PMC6829617 DOI: 10.3390/cells8101235] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer is one of the leading causes for cancer-related death worldwide. Transforming growth factor beta (TGF-β) is a pleiotropic cytokine that signals through membrane receptors and intracellular Smad proteins, which enter the nucleus upon receptor activation and act as transcription factors. TGF-β inhibits liver tumorigenesis in the early stage by inducing cytostasis and apoptosis, but promotes malignant progression in more advanced stages by enhancing cancer cell survival, EMT, migration, invasion and finally metastasis. Understanding the molecular mechanisms underpinning the multi-faceted roles of TGF-β in liver cancer has become a persistent pursuit during the last two decades. Contextual regulation fine-tunes the robustness, duration and plasticity of TGF-β signaling, yielding versatile albeit specific responses. This involves multiple feedback and feed-forward regulatory loops and also the interplay between Smad signaling and non-Smad pathways. This review summarizes the known regulatory mechanisms of TGF-β signaling in liver cancer, and how they channel, skew and even switch the actions of TGF-β during cancer progression.
Collapse
|
17
|
Li K, Xu C, Du Y, Junaid M, Kaushik AC, Wei DQ. Comprehensive epigenetic analyses reveal master regulators driving lung metastasis of breast cancer. J Cell Mol Med 2019; 23:5415-5431. [PMID: 31215771 PMCID: PMC6653217 DOI: 10.1111/jcmm.14424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/30/2019] [Accepted: 05/12/2019] [Indexed: 01/02/2023] Open
Abstract
The lung metastasis of breast cancer involves complicated regulatory changes driven by chromatin remodelling. However, the epigenetic reprogramming and regulatory mechanisms in lung metastasis of breast cancer remain unclear. Here, we generated and analysed genome‐wide profiles of multiple histone modifications (H3K4me3, H3K27ac, H3K27me3, H3K4me1 and H3K9me3), as well as transcriptome data in lung‐metastatic and non‐lung‐metastatic breast cancer cells. Our results showed that the expression changes were correlated with the enrichment of specific histone modifications in promoters and enhancers. Promoter and enhancer reprogramming regulated gene expression in a synergetic way, and involved in multiple important biological processes and pathways. In addition, lots of gained super‐enhancers were identified in lung‐metastatic cells. We also identified master regulators driving differential gene expression during lung metastasis of breast cancer. We found that the cooperations between regulators were much closer in lung‐metastatic cells. Moreover, regulators such as TFAP2C, GTF2I and LMO4 were found to have potential prognostic value for lung metastasis free (LMF) survival of breast cancer. Functional studies motivated by our data analyses uncovered an important role of LMO4 in regulating metastasis. This study provided comprehensive insights into regulatory mechanisms, as well as potential prognostic markers for lung metastasis of breast cancer.
Collapse
Affiliation(s)
- Kening Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Congling Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Yuxin Du
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai JiaoTong University, Shanghai, China
| | - Muhammad Junaid
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aman-Chandra Kaushik
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
MEF-2 isoforms' (A-D) roles in development and tumorigenesis. Oncotarget 2019; 10:2755-2787. [PMID: 31105874 PMCID: PMC6505634 DOI: 10.18632/oncotarget.26763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/01/2019] [Indexed: 12/29/2022] Open
Abstract
Myocyte enhancer factor (MEF)-2 plays a critical role in proliferation, differentiation, and development of various cell types in a tissue specific manner. Four isoforms of MEF-2 (A-D) differentially participate in controlling the cell fate during the developmental phases of cardiac, muscle, vascular, immune and skeletal systems. Through their associations with various cellular factors MEF-2 isoforms can trigger alterations in complex protein networks and modulate various stages of cellular differentiation, proliferation, survival and apoptosis. The role of the MEF-2 family of transcription factors in the development has been investigated in various cell types, and the evolving alterations in this family of transcription factors have resulted in a diverse and wide spectrum of disease phenotypes, ranging from cancer to infection. This review provides a comprehensive account on MEF-2 isoforms (A-D) from their respective localization, signaling, role in development and tumorigenesis as well as their association with histone deacetylases (HDACs), which can be exploited for therapeutic intervention.
Collapse
|
19
|
Tong H, Liu X, Li T, Qiu W, Peng C, Shen B, Zhu Z. INTS8 accelerates the epithelial-to-mesenchymal transition in hepatocellular carcinoma by upregulating the TGF-β signaling pathway. Cancer Manag Res 2019; 11:1869-1879. [PMID: 30881114 PMCID: PMC6396674 DOI: 10.2147/cmar.s184392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the third leading cause of death by malignancy worldwide. HCC has a poor prognosis due to tumor invasiveness and metastasis. There is substantial evidence that the epithelial-to-mesenchymal transition (EMT) plays a central role in cancer metastasis. In a previous study, a possible association between integrator complex 8 (INTS8) and the progression and development of HCC was discovered. However, its role and the molecular mechanisms in HCC are poorly understood. Methods The PROGgeneV2 platform database and Kaplan–Meier plotter analysis were used to analyze the potential effects of INTS8 in HCC. Moreover, we performed migration, transwell, and metastasis assays to investigate the effects of INTS8 on HCC cells. In addition, relevant signaling pathways were examined by western blot and RT-qPCR assays. Results We used the PROGgeneV2 platform database and Kaplan–Meier plotter analysis, which indicated that increased expression of INTS8 is associated with poor overall survival of HCC. Moreover, INTS8 expression was higher in HCC tissues than in adjacent noncancerous tissues. INTS8 depletion reduced the invasion and migration of HCC cell lines. Downregulation of INTS8 in vivo resulted in fewer observed metastatic nodules in lungs. Moreover, INTS8 knockdown also increased the expression of epithelial markers (E-cadherin) and decreased the expression of mesenchymal markers (N-cadherin and vimentin) following the downregulation of SMAD4. In addition, pretreatment with TGF-β1 could partly prevent the decrease in the expression of SMAD4 and EMT markers induced by INTS8 knockdown. Conclusion Overall, these findings suggest that INTS8 accelerates the EMT in HCC by upregulating the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Hui Tong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, ;
| | - Xiaohui Liu
- France National Research Center International Joint Laboratory (CNRS-LIAI), Sino-French Research Center for Life Sciences and Genomics, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tao Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, ;
| | - Weihua Qiu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, ;
| | - Chenghong Peng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, ;
| | - Baiyong Shen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, ;
| | - Zhecheng Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, ;
| |
Collapse
|
20
|
Zhou Z, Lin Z, He Y, Pang X, Wang Y, Ponnusamy M, Ao X, Shan P, Tariq MA, Li P, Wang J. The Long Noncoding RNA D63785 Regulates Chemotherapy Sensitivity in Human Gastric Cancer by Targeting miR-422a. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:405-419. [PMID: 30195778 PMCID: PMC6036868 DOI: 10.1016/j.omtn.2018.05.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/24/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023]
Abstract
Gastric cancer is one of the most prevalent tumor types in the world. Chemotherapy is the most common choice for cancer treatment. However, chemotherapy resistance and adverse side effects limit its clinical applications. Aberrant expression of long noncoding RNAs (lncRNAs) has been found in various stages of gastric cancer development and progression. In this study, we identified that an oncogenic lncRNA, long intergenic non-protein-coding RNA D63785 (lncR-D63785), is highly expressed in gastric cancer tissues and cells. Silencing of lncR-D63785 inhibited cell proliferation, cell migration and invasion in gastric cancer cell lines and reduced tumor volume and size in mice. We found that the expression of lncR-D63785 was inversely correlated with microRNA 422a (miR-422a) expression, which was involved in the downregulation of expression of myocyte enhancer factor-2D (MEF2D) and drug sensitivity. Knockdown of lncR-D63785 increased the expression of miR-422a and the sensitivity of gastric cancer cells to apoptosis induced by the anticancer drug doxorubicin (DOX). This indicates that lncR-D63785 acts as a competitive endogenous RNA (ceRNA) of miR-422a and promotes chemoresistance by blocking miR-422-dependent suppression of MEF2D. Together, our results suggest that the therapeutic suppression of lncR-D63785 alone or in combination with chemotherapeutic agents may be a promising strategy for treating gastric cancer.
Collapse
Affiliation(s)
- Zhixia Zhou
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Zhijuan Lin
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Military General Hospital, Beijing 100700, China
| | - Xin Pang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Yin Wang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Xiang Ao
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Peipei Shan
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Muhammad Akram Tariq
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China.
| | - Jianxun Wang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
21
|
Wu C, Kan H, Hu M, Liu X, Boye A, Jiang Y, Wu J, Wang J, Yang X, Yang Y. Compound Astragalus and Salvia miltiorrhiza extract inhibits hepatocarcinogenesis via modulating TGF-β/TβR and Imp7/8. Exp Ther Med 2018; 16:1052-1060. [PMID: 30112050 PMCID: PMC6090435 DOI: 10.3892/etm.2018.6292] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/11/2018] [Indexed: 12/15/2022] Open
Abstract
Compound Astragalus and Salvia miltiorrhiza extract (CASE) is a Chinese herbal formula consisting of astragalosides, astragalus polysaccharide and salvianolic acids extracted from Astragalus membranaceus and Salvia miltiorhiza. Previous studies by our group have demonstrated that CASE effectively suppresses diethylinitrosamine (DEN)-induced hepatocellular carcinoma (HCC) in rats via modulating transforming growth factor β/Mothers against decapentaplegic (TGFβ/Smad) signaling. To further elucidate the mechanism of CASE, the effects of CASE on TGF-β1, the serine/threonine kinase receptors of TGF-β [TGF-β receptor type-I (TβRI) and TβRII] and karyopherins [Importin 7 (Imp7) and Imp8], which are crucial for TGF-β/Smad signaling in fibro-hepatocarcinogenesis, were assessed in the present study using in vivo (DEN-induced HCC in rats) and in vitro [TGF-β1-stimulated rat myofibroblasts (MFBs) and HepG2 cells] models of fibro-hepatocarcinogenesis. Hematoxylin and eosin staining revealed that CASE may suppress inflammatory reactions and fibrosis in HCC as well as increasing the differentiation of HCC cells. Positive TGF-β1 staining was increased in HCC nodule areas and in adjacent normal liver tissues in DEN-treated rats, while TβRI staining was increased only in normal adjacent liver tissues. The elevated expression of TGF-β1, TβRI and TβRII was suppressed by CASE. CASE treatment also reduced glutathione S-transferase P 1 and Imp7/8 protein expression in fibro-hepatocarcinogenesis. In vitro experiments confirmed that CASE was able to decrease the expression of TβRI and TβRII in TGF-β1-stimulated MFBs and HepG2 cells. These results indicate that the anti-HCC effect of CASE may be achieved by mediating TGF-β/TβR and Imp7/8 protein expression, suggesting that CASE has multiple targets in HCC treatment.
Collapse
Affiliation(s)
- Chao Wu
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Hongwei Kan
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Min Hu
- Department of Pathology, Anhui University of Chinese Traditional Medicine, Hefei, Anhui 230038, P.R. China
| | - Xin Liu
- Therapeutics Research Centre, University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland 4102, Australia
| | - Alex Boye
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yufeng Jiang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jiajun Wu
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jiyu Wang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xiaochuan Yang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan Yang
- Department of Pharmacology and Institute of Natural Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
22
|
Di Giorgio E, Hancock WW, Brancolini C. MEF2 and the tumorigenic process, hic sunt leones. Biochim Biophys Acta Rev Cancer 2018; 1870:261-273. [PMID: 29879430 DOI: 10.1016/j.bbcan.2018.05.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/25/2018] [Accepted: 05/26/2018] [Indexed: 12/14/2022]
Abstract
While MEF2 transcription factors are well known to cooperate in orchestrating cell fate and adaptive responses during development and adult life, additional studies over the last decade have identified a wide spectrum of genetic alterations of MEF2 in different cancers. The consequences of these alterations, including triggering and maintaining the tumorigenic process, are not entirely clear. A deeper knowledge of the molecular pathways that regulate MEF2 expression and function, as well as the nature and consequences of MEF2 mutations are necessary to fully understand the many roles of MEF2 in malignant cells. This review discusses the current knowledge of MEF2 transcription factors in cancer.
Collapse
Affiliation(s)
- Eros Di Giorgio
- Department of Medicine, Università degli Studi di Udine, P.le Kolbe 4, 33100 Udine, Italy
| | - Wayne W Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Biesecker Center for Pediatric Liver Diseases, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Claudio Brancolini
- Department of Medicine, Università degli Studi di Udine, P.le Kolbe 4, 33100 Udine, Italy.
| |
Collapse
|
23
|
Wang L, Li Q, Ye Z, Qiao B. ZBTB7/miR-137 Autoregulatory Circuit Promotes the Progression of Renal Carcinoma. Oncol Res 2018; 27:1007-1014. [PMID: 29673422 PMCID: PMC7848413 DOI: 10.3727/096504018x15231148037228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Renal carcinoma greatly threatens human health, but the involved molecular mechanisms are far from complete understanding. As a master oncogene driving the initiation of many other cancers, ZBTB7 has not been established to be associated with renal cancer. Our data revealed that ZBTB7 is highly expressed in renal carcinoma specimens and cell lines, compared with normal cells. The silencing of ZBTB7 suppressed the proliferation and invasion of renal cancer cells. ZBTB7 overexpression rendered normal cells with higher proliferation rates and invasiveness. An animal study further confirmed the role of ZBTB7 in the growth of renal carcinoma. Moreover, miR-137 was identified to negatively regulate the expression of ZBTB7, and its abundance is inversely correlated with that of ZBTB7 in renal carcinoma specimens and cell lines. ZBTB7 overexpression may be induced by miR-137 downregulation. Interestingly, ZBTB7 can also suppress miR-137 expression by binding to its recognition site within the miR-137 promoter region. Taken together, we identified an autoregulatory loop consisting of ZBTB7 and miR-137 in gastric cancers, and targeting this pathway may be an effective strategy for renal carcinoma cancer therapy.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Qi Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Zhuo Ye
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Baoping Qiao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
24
|
Yang Y, Liu Q, Li Z, Zhang R, Jia C, Yang Z, Zhao H, Ya S, Mao R, Ailijiang T, Bao Y, Zhang H. GP73 promotes epithelial–mesenchymal transition and invasion partly by activating TGF-β1/Smad2 signaling in hepatocellular carcinoma. Carcinogenesis 2018; 39:900-910. [PMID: 29365054 DOI: 10.1093/carcin/bgy010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 01/12/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- Ying Yang
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qiang Liu
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhipeng Li
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ruili Zhang
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chunli Jia
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhifang Yang
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Huarong Zhao
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Sha Ya
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Rui Mao
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tuerxun Ailijiang
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yongxing Bao
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hua Zhang
- Department of Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
25
|
Chen X, Gao B, Ponnusamy M, Lin Z, Liu J. MEF2 signaling and human diseases. Oncotarget 2017; 8:112152-112165. [PMID: 29340119 PMCID: PMC5762387 DOI: 10.18632/oncotarget.22899] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 09/09/2017] [Indexed: 01/01/2023] Open
Abstract
The members of myocyte Enhancer Factor 2 (MEF2) protein family was previously believed to function in the development of heart and muscle. Recent reports indicate that they are also closely associated with development and progression of many human diseases. Although their role in cancer biology is well established, the molecular mechanisms underlying their action is yet largely unknown. MEF2 family is closely associated with various signaling pathways, including Ca2+ signaling, MAP kinase signaling, Wnt signaling, PI3K/Akt signaling, etc. microRNAs also contribute to regulate the activities of MEF2. In this review, we summarize the known molecular mechanism by which MEF2 family contribute to human diseases.
Collapse
Affiliation(s)
- Xiao Chen
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Bing Gao
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,School of Basic Medicine, Qingdao University, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Zhijuan Lin
- Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Jia Liu
- School of Pharmacy, Qingdao University, Qingdao 266021, China.,School of Basic Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
26
|
Xiang J, Sun H, Su L, Liu L, Shan J, Shen J, Yang Z, Chen J, Zhong X, Ávila MA, Yan X, Liu C, Qian C. Myocyte enhancer factor 2D promotes colorectal cancer angiogenesis downstream of hypoxia-inducible factor 1α. Cancer Lett 2017; 400:117-126. [PMID: 28478181 DOI: 10.1016/j.canlet.2017.04.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/13/2017] [Accepted: 04/25/2017] [Indexed: 12/21/2022]
Abstract
Myocyte enhancer factor 2D (MEF2D) is involved in many aspects of cancer progression, including cell proliferation, invasion, and migration. However, little is known about the role of MEF2D in tumor angiogenesis. Using clinical specimens, colorectal cancer (CRC) cell lines and a mouse model in the present study, we found that MEF2D expression was positively correlated with CD31-positive microvascular density in CRC tissues. MEF2D promoted tumor angiogenesis in vitro and in vivo and induced the expression of proangiogenic cytokines in CRC cells. MEF2D was found to be a downstream effector of hypoxia-inducible factor (HIF)-1α in the induction of tumor angiogenesis. HIF-1α transactivates MEF2D expression by binding to the MEF2D gene promoter. These results demonstrate that the HIF-1α/MEF2D axis can serve as a therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Junyu Xiang
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hui Sun
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Li Su
- Department of Oncology, Chinese Traditional Medicine Hospital, Chongqing, China
| | - Limei Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Juanjuan Shan
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Junjie Shen
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhi Yang
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jun Chen
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xing Zhong
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Matías A Ávila
- Center of Investigation for Applied Medcine, University of Navarra, Pamplona, Spain
| | - Xiaochu Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Chungang Liu
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Cheng Qian
- Center of Biotherapy, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
27
|
Wang Y, Luo Y, Li C, Zhang X, Pi C, Yu L, Wang S, Zhong Z. Optimized formulation of multivesicular liposomes loaded with oleanolic acid enhanced anticancer effect in vitro. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:955-968. [PMID: 28392677 PMCID: PMC5376187 DOI: 10.2147/dddt.s128795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Invasion and metastasis are the main causes leading to the death of patients with hepatocellular carcinoma (HCC). Multivesicular liposomes loaded with oleanolic acid (OA-MVLs) have been well demonstrated to suppress survival, growth and angiogenesis of HCC cells. Emerging evidence demonstrates that OA was able to suppress the invasion of HCC cells by down-regulating myocyte enhancer factor-2. We hypothesized that the optimized OA-MVLs could inhibit the migration and invasion of HCC cells. In this study, we utilized central composite design and response surface methodology to assess the influence of some parameters on particle size and encapsulation efficiency and obtain the optimized formulation of OA-MVLs. Subsequently, the human HCC cell lines SMMC-7721 and HepG2 were treated with different doses of OA-MVLs and OA, respectively. Cellular survival, adhesion, migration and invasion in vitro were evaluated. We found that the optimized OA-MVLs significantly decreased the ability of HCC cells to adhere, migrate and invade in vitro. Furthermore, OA-MVLs significantly inhibited the survival of HCC cells at 160 µmol/L but showed no obvious inhibition effect on the cell vitality of normal liver cells. Our findings indicate that OA-MVLs did inhibit the cell survival, adhesion, invasion and metastasis of HCC cells in vitro. Although the involved mechanisms are still unclear, our findings can contribute to a better development of a preventive and therapeutic strategy for human HCC.
Collapse
Affiliation(s)
- Yunlong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Yuling Luo
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Xiaoqin Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Chao Pi
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Lu Yu
- Department of Chemistry, The Institute of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Shurong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy
| |
Collapse
|
28
|
Zhu HX, Shi L, Zhang Y, Zhu YC, Bai CX, Wang XD, Zhou JB. Myocyte enhancer factor 2D provides a cross-talk between chronic inflammation and lung cancer. J Transl Med 2017; 15:65. [PMID: 28340574 PMCID: PMC5366127 DOI: 10.1186/s12967-017-1168-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/19/2017] [Indexed: 01/11/2023] Open
Abstract
Background Lung cancer is the leading cause of cancer-related morbidity and mortality worldwide. Patients with chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD), are exposed to a higher risk of developing lung cancer. Chronic inflammation may play an important role in the lung carcinogenesis among those patients. The present study aimed at identifying candidate biomarker predicting lung cancer risk among patients with chronic respiratory diseases. Methods We applied clinical bioinformatics tools to analyze different gene profile datasets with a special focus on screening the potential biomarker during chronic inflammation-lung cancer transition. Then we adopted an in vitro model based on LPS-challenged A549 cells to validate the biomarker through RNA-sequencing, quantitative real time polymerase chain reaction, and western blot analysis. Results Bioinformatics analyses of the 16 enrolled GSE datasets from Gene Expression Omnibus online database showed myocyte enhancer factor 2D (MEF2D) level significantly increased in COPD patients coexisting non-small-cell lung carcinoma (NSCLC). Inflammation challenge increased MEF2D expression in NSCLC cell line A549, associated with the severity of inflammation. Extracellular signal-regulated protein kinase inhibition could reverse the up-regulation of MEF2D in inflammation-activated A549. MEF2D played a critical role in NSCLC cell bio-behaviors, including proliferation, differentiation, and movement. Conclusions Inflammatory conditions led to increased MEF2D expression, which might further contribute to the development of lung cancer through influencing cancer microenvironment and cell bio-behaviors. MEF2D might be a potential biomarker during chronic inflammation-lung cancer transition, predicting the risk of lung cancer among patients with chronic respiratory diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1168-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hai-Xing Zhu
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Respiratory Research Institute, Shanghai, China
| | - Lin Shi
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yong Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Respiratory Research Institute, Shanghai, China
| | - Yi-Chun Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chun-Xue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Respiratory Research Institute, Shanghai, China
| | - Xiang-Dong Wang
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.,Fudan University Center for Clinical Bioinformatics, Shanghai, China
| | - Jie-Bai Zhou
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Respiratory Research Institute, Shanghai, China.
| |
Collapse
|
29
|
Midgett M, López CS, David L, Maloyan A, Rugonyi S. Increased Hemodynamic Load in Early Embryonic Stages Alters Endocardial to Mesenchymal Transition. Front Physiol 2017; 8:56. [PMID: 28228731 PMCID: PMC5296359 DOI: 10.3389/fphys.2017.00056] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/23/2017] [Indexed: 12/30/2022] Open
Abstract
Normal blood flow is essential for proper heart formation during embryonic development, as abnormal hemodynamic load (blood pressure and shear stress) results in cardiac defects seen in congenital heart disease. However, the progressive detrimental remodeling processes that relate altered blood flow to cardiac defects remain unclear. Endothelial-mesenchymal cell transition is one of the many complex developmental events involved in transforming the early embryonic outflow tract into the aorta, pulmonary trunk, interventricular septum, and semilunar valves. This study elucidated the effects of increased hemodynamic load on endothelial-mesenchymal transition remodeling of the outflow tract cushions in vivo. Outflow tract banding was used to increase hemodynamic load in the chicken embryo heart between Hamburger and Hamilton stages 18 and 24. Increased hemodynamic load induced increased cell density in outflow tract cushions, fewer cells along the endocardial lining, endocardium junction disruption, and altered periostin expression as measured by confocal microscopy analysis. In addition, 3D focused ion beam scanning electron microscopy analysis determined that a portion of endocardial cells adopted a migratory shape after outflow tract banding that is more irregular, elongated, and with extensive cellular projections compared to normal cells. Proteomic mass-spectrometry analysis quantified altered protein composition after banding that is consistent with a more active stage of endothelial-mesenchymal transition. Outflow tract banding enhances the endothelial-mesenchymal transition phenotype during formation of the outflow tract cushions, suggesting that endothelial-mesenchymal transition is a critical developmental process that when disturbed by altered blood flow gives rise to cardiac malformation and defects.
Collapse
Affiliation(s)
- Madeline Midgett
- Biomedical Engineering, Oregon Health and Science University Portland, OR, USA
| | - Claudia S López
- Biomedical Engineering, Oregon Health and Science UniversityPortland, OR, USA; Multiscale Microscopy Core, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science UniversityPortland, OR, USA
| | - Larry David
- Proteomics Core, Oregon Health and Science University Portland, OR, USA
| | - Alina Maloyan
- Knight Cardiovascular Institute, Oregon Health and Science University Portland, OR, USA
| | - Sandra Rugonyi
- Biomedical Engineering, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
30
|
Wang X, Ding J, Feng Y, Weng L, Zhao G, Xiang J, Zhang M, Xing D. Targeting of growth factors in the treatment of hepatocellular carcinoma: The potentials of polysaccharides. Oncol Lett 2017; 13:1509-1517. [PMID: 28454283 DOI: 10.3892/ol.2017.5602] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/19/2016] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has become a leading cause of cancer-associated mortality worldwide and is thus of great concern. Although various chemotherapeutic drugs are currently used for the treatment of HCC, severe side effects associated with these treatments have prompted interest in novel therapies, including the use of certain biological macromolecules such as polysaccharides. Several studies have shown that polysaccharides have anticancer and antiproliferative effects on HCC. Vascular endothelial growth factor, transforming growth factor β, epidermal growth factor and fibroblast growth factor may be effective targets for polysaccharides and may modulate tumor growth and immunity through increasing the expression levels of cytokines. The present review focuses on the ways in which growth factors contribute to the development of HCC, and on the anti-growth factor activities of natural and synthetic polysaccharides, as well as their effect on proinflammatory cytokines.
Collapse
Affiliation(s)
- Xuan Wang
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jieyu Ding
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Yuanyuan Feng
- Oncology Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, P.R. China
| | - Lingling Weng
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Guangqiang Zhao
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Jianfeng Xiang
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Minguang Zhang
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Dongwei Xing
- Radiology Department, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
31
|
Pon JR, Marra MA. MEF2 transcription factors: developmental regulators and emerging cancer genes. Oncotarget 2016; 7:2297-312. [PMID: 26506234 PMCID: PMC4823036 DOI: 10.18632/oncotarget.6223] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/14/2015] [Indexed: 12/22/2022] Open
Abstract
The MEF2 transcription factors have roles in muscle, cardiac, skeletal, vascular, neural, blood and immune system cell development through their effects on cell differentiation, proliferation, apoptosis, migration, shape and metabolism. Altered MEF2 activity plays a role in human diseases and has recently been implicated in the development of several cancer types. In particular, MEF2B, the most divergent and least studied protein of the MEF2 family, has a role unique from its paralogs in non-Hodgkin lymphomas. The use of genome-scale technologies has enabled comprehensive MEF2 target gene sets to be identified, contributing to our understanding of MEF2 proteins as nodes in complex regulatory networks. This review surveys the molecular interactions of MEF2 proteins and their effects on cellular and organismal phenotypes. We include a discussion of the emerging roles of MEF2 proteins as oncogenes and tumor suppressors of cancer. Throughout this article we highlight similarities and differences between the MEF2 family proteins, including a focus on functions of MEF2B.
Collapse
Affiliation(s)
- Julia R Pon
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
32
|
Su L, Luo Y, Yang Z, Yang J, Yao C, Cheng F, Shan J, Chen J, Li F, Liu L, Liu C, Xu Y, Jiang L, Guo D, Prieto J, Ávila MA, Shen J, Qian C. MEF2D Transduces Microenvironment Stimuli to ZEB1 to Promote Epithelial-Mesenchymal Transition and Metastasis in Colorectal Cancer. Cancer Res 2016; 76:5054-67. [PMID: 27364559 DOI: 10.1158/0008-5472.can-16-0246] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 06/03/2016] [Indexed: 11/16/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is an essential mechanism of metastasis, including in colorectal cancer. Although EMT processes are often triggered in cancer cells by their surrounding microenvironment, how EMT-relevant genes control these processes is not well understood. In multiple types of cancers, the transcription factor MEF2D has been implicated in cell proliferation, but its contributions to metastasis have not been addressed. Here, we show MEF2D is overexpressed in clinical colorectal cancer tissues where its high expression correlates with metastatic process. Functional investigations showed that MEF2D promoted cancer cell invasion and EMT and that it was essential for certain microenvironment signals to induce EMT and metastasis in vivo Mechanistically, MEF2D directly regulated transcription of the EMT driver gene ZEB1 and facilitated histone acetylation at the ZEB1 promoter. More importantly, MEF2D responded to various tumor microenvironment signals and acted as a central integrator transducing multiple signals to activate ZEB1 transcription. Overall, our results define a critical function for MEF2D in upregulating EMT and the metastatic capacity of colorectal cancer cells. Further, they offer new insights into how microenvironment signals activate EMT-relevant genes and deepen the pathophysiologic significance of MEF2D, with potential implications for the prevention and treatment of metastatic colorectal cancer. Cancer Res; 76(17); 5054-67. ©2016 AACR.
Collapse
Affiliation(s)
- Li Su
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yongli Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhi Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jing Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chao Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Feifei Cheng
- School of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Juanjuan Shan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jun Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Fangfang Li
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Limei Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chungang Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanmin Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lupin Jiang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Deyu Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jesus Prieto
- Center of Investigation for Applied Medicine, University of Navarra, Pamplona, Spain
| | - Matías A Ávila
- Center of Investigation for Applied Medicine, University of Navarra, Pamplona, Spain
| | - Junjie Shen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
33
|
Thillai K, Ross P, Sarker D. Molecularly targeted therapy for advanced hepatocellular carcinoma - a drug development crisis? World J Gastrointest Oncol 2016; 8:173-85. [PMID: 26909132 PMCID: PMC4753168 DOI: 10.4251/wjgo.v8.i2.173] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 11/16/2015] [Accepted: 12/09/2015] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma is the fastest growing cause of cancer related death globally. Sorafenib, a multi-targeted kinase inhibitor, is the only drug proven to improve outcomes in patients with advanced disease offering modest survival benefit. Although comprehensive genomic mapping has improved understanding of the genetic aberrations in hepatocellular cancer (HCC), this knowledge has not yet impacted clinical care. The last few years have seen the failure of several first and second line phase III clinical trials of novel molecularly targeted therapies, warranting a change in the way new therapies are investigated in HCC. Potential reasons for these failures include clinical and molecular heterogeneity, trial design and a lack of biomarkers. This review discusses the current crisis in HCC drug development and how we should learn from recent trial failures to develop a more effective personalised treatment paradigm for patients with HCC.
Collapse
|
34
|
Kong J, Liu X, Li X, Wu J, Wu N, Chen J, Fang F. Pokemon promotes the invasiveness of hepatocellular carcinoma by enhancing MEF2D transcription. Hepatol Int 2016; 10:493-500. [PMID: 26797719 DOI: 10.1007/s12072-015-9697-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 12/11/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Pokemon, a master oncogene crucial for the tumorigenicity and progression of a variety of cancers, has been demonstrated to enhance the proliferation and survival of hepatocellular carcinoma (HCC). However, the contribution of Pokemon to the invasiveness of HCC has not yet been studied. METHODS In this study, we employed HCC cells to investigate the role of Pokemon in the invasion of HCC with multidisciplinary approaches. RESULTS Pokemon overexpression was found to be closely associated with invasion and intrahepatic metastasis of HCC in clinical specimens. Suppression of Pokemon attenuated the invasion of HCC cells by in vitro transwell and wound-healing assays. Myocyte enhancer factor 2D (MEF2D), an oncogene that can promote the invasiveness of HCC, was found to be underexpressed during Pokemon silencing in HCC cells. Restoration of MEF2D abolished the effect of Pokemon downregulation on the migration of HCC cells. Further experiments verified that Pokemon binds two putative recognition sites located within the upstream region of the MEF2D promoter and enhances its transcription. The association between Pokemon and MEF2D was further confirmed in HCC specimens. Animal experiments further confirmed that Pokemon downregulation attenuated the metastasis of HCC cells in mice. CONCLUSION Collectively, Pokemon was found to enhance the migration and invasion of HCC by increasing MEF2D expression. Thus, targeting Pokemon and MEF2D may be an effective strategy to suppress the metastasis of HCC.
Collapse
Affiliation(s)
- Jing Kong
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China.
| | - Xiaoping Liu
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| | - Xiangqian Li
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| | - Jinsheng Wu
- Department of Hepatopancreatobiliary Surgery, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, 223300, Jiangsu, People's Republic of China
| | - Ning Wu
- Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Jun Chen
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| | - Fang Fang
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| |
Collapse
|
35
|
Xu K, Zhao YC. MEF2D/Wnt/β-catenin pathway regulates the proliferation of gastric cancer cells and is regulated by microRNA-19. Tumour Biol 2016; 37:9059-69. [PMID: 26762410 DOI: 10.1007/s13277-015-4766-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/29/2015] [Indexed: 01/06/2023] Open
Abstract
The underlying molecular pathogenesis in gastric cancer remains poorly unknown. The transcription factor myocyte enhancer factor 2D (MEF2D) participates in the initiation and development of many human cancers. However, its potential roles in gastric cancer have surprisingly not been studied. In present study, we first explored MEF2's expression in gastric cancer, finding that only MEF2D rather than MEF2A, 2B, or 2C was elevated in gastric cancer clinical specimens. Furthermore, immunohistochemical analysis on the tissue samples obtained from 260 patients with gastric cancer revealed that MEF2D expression was significantly associated with the clinical stage, vascular invasion, metastasis, and tumor size. Gastric cancer patients with MEF2D expression showed a significantly shorter overall survival time compared with that of patients lacking of MEF2D. Multivariate analysis revealed that MEF2D expression was an independent prognostic factor for overall survival. These results indicated that MEF2D was a prognostic marker for gastric cancer. Notably, MEF2D silencing was able to reduce the proliferation and survival of gastric cancer cells. Further study revealed that MEF2D suppression significantly inactivated the oncogenic Wnt/β-catenin pathway. Downregulation of MEF2D inhibited the tumorigenesis of gastric cancer cells in nude mice. Finally, MEF2D is a direct target of miR-19, which was found to be decreased in gastric cancer clinical specimens. Collectively, we found that miR-19/MEF2D/Wnt/β-catenin regulatory network contributes to the growth of gastric cancer, hinting a new promising target for gastric cancer treatment.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Chao Zhao
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
36
|
Huo Y, Zhao Q, Wang C, Zhao F, Du Y, Sun W. The involvement of myocyte enhancer factor 2D in regulating tumor biology of cardiac myxoma. Tumour Biol 2015; 37:5405-11. [PMID: 26563368 DOI: 10.1007/s13277-015-4386-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 11/04/2015] [Indexed: 11/25/2022] Open
Abstract
The pro-survival transcription factor myocyte enhancer factor 2D (MEF2D) is identified to exhibit pro-tumor effects based on clinical and experimental studies. However, the detailed mechanisms underlying IGF-1-MEF2D pathway-induced tumor biology in cardiac myxoma (CM) was not clear. Here, we investigated the role of MEF2D in CM tissues and cells using RT-PCR, western blot, gene silencing, et al. Our findings revealed MEF2D was significantly increased in CM tissues compared with adjacent normal tissues and closely related to tumor size. In vitro assay demonstrated that IGF-1 enhanced CM cell proliferation in a time-dependent fashion. However, knockdown of MEF2D reversed the IGF-1-induced proliferative effects on CM cells in a time-dependent fashion and further resulted in cell cycle arrest. Based on the molecular level, IGF-1 enhanced the expression of epidermal growth factor receptor (EGFR) and matrix metalloprotein 9 (MMP9) in CM cells, whereas knockdown of MEF2D was able to reduce the expression of EGFR and MMP9 compared with vector control. Furthermore, we found knockdown of MEF2D directly affected G1/S transition in cultured CM cells. In conclusion, MEF2D regulates IGF-1-induced proliferation and apoptosis in CM development, indicating IGF-1-MEF2D pathway may be a useful target for treatment.
Collapse
Affiliation(s)
- Yufeng Huo
- Department of Cardiac Surgery, Qingdao Branch, Qilu Hospital of Shandong University, No. 758 Hefei Road, Shibei District, Qingdao, Shandong, China
| | - Qingjun Zhao
- Department of Anesthesia, ZhangQiu People's Hospital, No. 1920, Huiquan Road, Zhangqiu, Shandong, China
| | - Cheng Wang
- Department of Cardiothoracic Surgery, JiMo People's Hospital, Qingdao, Shandong, China
| | - Fen Zhao
- Department of Cardiac Surgery, Qingdao Branch, Qilu Hospital of Shandong University, No. 758 Hefei Road, Shibei District, Qingdao, Shandong, China
| | - Yinghai Du
- Department of Cardiac Surgery, Qingdao Branch, Qilu Hospital of Shandong University, No. 758 Hefei Road, Shibei District, Qingdao, Shandong, China
| | - Wenyu Sun
- Department of Cardiac Surgery, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, 250012, Jinan, China.
| |
Collapse
|
37
|
miR-218 suppressed the growth of lung carcinoma by reducing MEF2D expression. Tumour Biol 2015; 37:2891-900. [PMID: 26409449 DOI: 10.1007/s13277-015-4038-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/02/2015] [Indexed: 10/23/2022] Open
Abstract
Lung carcinoma is a deadly malignant disease with poor prognosis and increasing incidence in recent years. However, the molecular mechanism underlying the initiation and progression of lung cancer is still not completely elucidated. Recently, myocyte enhancer factor 2D (MEF2D) has been reported to promote the growth of liver cancer, but its implication in lung cancer is still unknown. This study is aimed to determine the role of MEF2D in lung carcinoma. Quantitative PCR (qPCR) and immunoblot assays showed that MEF2D was overexpressed in lung cancer tissues and cell lines, compared with the matched normal tissues and cell lines. Small interfering RNA (siRNA) suppression of MEF2D was able to reduce the proliferation, survival, and invasion of lung carcinoma cells. The transfection of MEF2D-expressing constructs into normal lung fibroblast cells promoted their proliferation and motility. The role of MEF2D in the growth of lung cancer was also confirmed in mice. Further study revealed that miR-218, which was underexpressed in lung carcinoma, was predicted to bind the 3'-untranslated region (UTR) of MEF2D mRNA. miR-218 was shown to suppress the activity of luciferase with MEF2D 3'-UTR. The changes in miR-218 levels affected the expression of MEF2D in lung cancer cells and normal fibroblast cells. There is also an inverse association between miR-218 abundance and MEF2D levels in the lung carcinoma specimen. Furthermore, the transfection of a plasmid that expressed MEF2D resistance to miR-218 regulation abolished the inhibitory effect of miR-218 on lung cancer cells. Collectively, MEF2D overexpression participated in the growth of lung cancers and its aberrant expression may result from the reduction of tumor suppressor miR-218.
Collapse
|
38
|
Lian JY, Tuo BG. Role of TGFβ signaling pathway in biological behavior of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2015; 23:3874-3881. [DOI: 10.11569/wcjd.v23.i24.3874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with a high fatality rate and strong invasion, and surgical resection is the most effective treatment for early HCC. However, most patients have lost the chance of operation at diagnosis. Additionally, chemotherapy has a poor effect and drug toxicity. In order to improve the prognosis and treatment of HCC, elucidating the cellular and molecular mechanism of HCC and finding new treatment targets are essential. The transforming growth factor β (TGFβ) pathway is involved in hepatocellular carcinogenesis and regulates tumor cell proliferation, apoptosis, migration, invasion and differentiation. TGFβ exerts a protective effect in early HCC. With the progression of HCC, TGFβ promotes the progress of HCC. Studies suggest that Axl/14-3-3zeta signaling is central for the transforming process. Taken into consideration the crucial role of the TGFβ pathway in the development and progression of HCC, it might become an important therapeutic target for HCC in the future.
Collapse
|
39
|
MEF2B mutations in non-Hodgkin lymphoma dysregulate cell migration by decreasing MEF2B target gene activation. Nat Commun 2015; 6:7953. [PMID: 26245647 PMCID: PMC4918335 DOI: 10.1038/ncomms8953] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 06/30/2015] [Indexed: 02/05/2023] Open
Abstract
Myocyte enhancer factor 2B (MEF2B) is a transcription factor with mutation hotspots at K4, Y69 and D83 in diffuse large B-cell lymphoma (DLBCL). To provide insight into the regulatory network of MEF2B, in this study, we analyse global gene expression and DNA-binding patterns. We find that candidate MEF2B direct target genes include RHOB, RHOD, CDH13, ITGA5 and CAV1, and that indirect target genes of MEF2B include MYC, TGFB1, CARD11, MEF2C, NDRG1 and FN1. MEF2B overexpression increases HEK293A cell migration and epithelial-mesenchymal transition, and decreases DLBCL cell chemotaxis. K4E, Y69H and D83V MEF2B mutations decrease the capacity of MEF2B to activate transcription and decrease its' effects on cell migration. The K4E and D83V mutations decrease MEF2B DNA binding. In conclusion, our map of the MEF2B regulome connects MEF2B to drivers of oncogenesis.
Collapse
|
40
|
Kong J, Liu X, Li X, Wu J, Wu N, Chen J, Fang F. miR-125/Pokemon auto-circuit contributes to the progression of hepatocellular carcinoma. Tumour Biol 2015; 37:511-9. [PMID: 26227218 DOI: 10.1007/s13277-015-3596-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 05/19/2015] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of human malignant tumor occurring in hepatic tissues with high mortality. Patients benefit little from current therapeutic modalities, at least partially due to the lack of complete elucidation of molecular network regulating HCC. miR-125 and Pokemon are well-recognized tumor suppressor and oncogenes for HCC, respectively. However, the underlying mechanism by which the two genes exert their functions and the relationship between miR-125 and Pokemon is still unexplored yet. In this study, we found that there is an inverse association between miR-125 and Pokemon expression levels in HCC specimen and cell lines. Online database mining indicated that there are three putative mRNA recognition elements (MREs) of miR-125 within 3' untranslated region (3'UTR) of Pokemon. MREs of miR-125 confer the expression of luciferase with a miR-125-dependent fashion. The alteration in miR-125 abundance regulates the expression of Pokemon at both protein and mRNA levels. Overexpression of Pokemon is able to abrogate the inhibitory effect of miR-125 on HCC progression. Further study showed that Pokemon inhibits the expression of miR-125 by binding of recognition sites within its promoter. In conclusion, we found that there is an auto-regulatory circuit consisting of miR-125 and Pokemon, which promotes the progression of HCC and may be a promising therapeutic target in clinical HCC treatment.
Collapse
Affiliation(s)
- Jing Kong
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China.
| | - Xiaoping Liu
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| | - Xiangqian Li
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| | - Jinsheng Wu
- Department of Hepatopancreatobiliary Surgery, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, 223300, Jiangsu, People's Republic of China
| | - Ning Wu
- Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Jun Chen
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| | - Fang Fang
- Faculty of Life Science and Chemical Engineering, Huaiyin Institute of Technology, 1 Meicheng Road East, Huai'an, 223003, Jiangsu, People's Republic of China
| |
Collapse
|
41
|
Hong X, Hong XY, Li T, He CY. Pokemon and MEF2D co-operationally promote invasion of hepatocellular carcinoma. Tumour Biol 2015; 36:9885-93. [PMID: 26164003 DOI: 10.1007/s13277-015-3744-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 06/30/2015] [Indexed: 11/28/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most deadly human malignancy, and frequent invasion and metastasis is closely associated with its poor prognosis. However, the molecular mechanism underlying HCC invasion is still not completely elucidated. Pokemon is a well-established oncogene for HCC growth, but its contribution to HCC invasion has not been studied yet. In this paper, Pokemon was found to be overexpressed in MHCC-97H HCC cell line, which possesses higher invasiveness. Downregulation of Pokemon abolished the invasion of MHCC-97H HCC cell lines. Pokemon overexpression was able to enhance the invasion of MHCC-97L cells with lower invasiveness. MEF2D, an oncogene promoting the invasion of HCC cells, was further detected to be upregulated and downregulated when Pokemon was overexpressed and silenced, respectively. Online database analysis indicated that one Pokemon recognition site was located within the promoter of MEF2D. Chromatin co-precipitation, luciferase, and qPCR assays all proved that Pokemon can promote the expression of MEF2D in HCC cells. Restoration of MEF2D expression can prevent the impaired invasion of HCC cells with Pokemon silencing, while suppression of MEF2D abolished the effect of Pokemon overexpression on HCC invasion. More interestingly, MEF2D was also found to increase the transcription of Pokemon by binding myocyte enhancer factor 2 (MEF2) sites within its promoter region, implying an auto-regulatory circuit consisting of these two oncogenes that can promote HCC invasion. Our findings can contribute to the understanding of molecular mechanism underlying HCC invasion, and provided evidence that targeting this molecular loop may be a promising strategy for anti-invasion therapy.
Collapse
Affiliation(s)
- Xin Hong
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Chang Chun, 130031, China
| | - Xing-Yu Hong
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Chang Chun, 130031, China
| | - Tao Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Chang Chun, 130031, China
| | - Cheng-Yan He
- Department of Pathology and Pathophysiology, China-Japan Union Hospital of Jilin University, Chang Chun, 130031, China.
| |
Collapse
|