1
|
Xin S, Zhang Y, Zhang Z, Li Z, Sun X, Liu X, Jin L, Li W, Tang C, Mei W, Cao Q, Wang H, Wei Z, Zhou Z, Li R, Wen X, Yang G, Chen W, Zheng J, Ye L. ScRNA-seq revealed the tumor microenvironment heterogeneity related to the occurrence and metastasis in upper urinary tract urothelial carcinoma. Cancer Gene Ther 2024; 31:1201-1220. [PMID: 38877164 DOI: 10.1038/s41417-024-00779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 06/16/2024]
Abstract
Metastasis is the greatest clinical challenge for UTUCs, which may have distinct molecular and cellular characteristics from earlier cancers. Herein, we provide single-cell transcriptome profiles of UTUC para cancer normal tissue, primary tumor lesions, and lymphatic metastases to explore possible mechanisms associated with UTUC occurrence and metastasis. From 28,315 cells obtained from normal and tumor tissues of 3 high-grade UTUC patients, we revealed the origin of UTUC tumor cells and the homology between metastatic and primary tumor cells. Unlike the immunomicroenvironment suppression of other tumors, we found no immunosuppression in the tumor microenvironment of UTUC. Moreover, it is imperative to note that stromal cells are pivotal in the advancement of UTUC. This comprehensive single-cell exploration enhances our comprehension of the molecular and cellular dynamics of metastatic UTUCs and discloses promising diagnostic and therapeutic targets in cancer-microenvironment interactions.
Collapse
Affiliation(s)
- Shiyong Xin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Urology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, China
| | - Yanwei Zhang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhenhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ziyao Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xianchao Sun
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiang Liu
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Liang Jin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Weiyi Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Chaozhi Tang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wangli Mei
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qiong Cao
- Department of Pathology, The Third Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Haojie Wang
- Department of Central Laboratory, Zhengzhou University, Luoyang Central Hospital, Luoyang, 471003, China
| | - Zhihao Wei
- Department of Pathology, Yiluo Hospital of Luoyang, The Teaching Hospital of Henan University of Science and Technology, Luoyang, China
| | - Zhen Zhou
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongbing Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiaofei Wen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Weihua Chen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Lin Ye
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
2
|
Zhang Y, Chang L, Huang P, Cao M, Hong R, Zhao X, He X, Xu L. Loss of PTPRS elicits potent metastatic capability and resistance to temozolomide in glioblastoma. Mol Carcinog 2024; 63:1235-1247. [PMID: 38517048 DOI: 10.1002/mc.23720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/24/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor type with worse clinical outcome due to the hallmarks of strong invasiveness, high rate of recurrence, and therapeutic resistance to temozolomide (TMZ), the first-line drug for GBM, representing a major challenge for successful GBM therapeutics. Understanding the underlying mechanisms that drive GBM progression will shed novel insight into therapeutic strategies. Receptor-type tyrosine-protein phosphatase S (PTPRS) is a frequently mutated gene in human cancers, including GBM. Its role in GBM has not yet been clarified. Here, inactivating PTPRS mutation or deficiency was frequently found in GBM, and deficiency in PTPRS significantly induced defects in the G2M checkpoint and limited GBM cells proliferation, leading to potent resistance to TMZ treatment in vitro and in vivo. Surprisingly, loss of PTPRS triggered an unexpected mesenchymal phenotype that markedly enhances the migratory capabilities of GBM cells through upregulating numerous matrix metalloproteinases via MAPK-MEK-ERK signaling. Therefore, this work provides a therapeutic window for precisely excluding PTPRS-mutated patients who do not respond to TMZ.
Collapse
Affiliation(s)
- Yihua Zhang
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Liugang Chang
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Ping Huang
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Min Cao
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Rujun Hong
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Xinhu Zhao
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Xuzhi He
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, The Army Medical University, Chongqing, China
| |
Collapse
|
3
|
Fu H, Wu S, Shen H, Luo K, Huang Z, Lu N, Li Y, Lan Q, Xian Y. Glutamine Metabolism Heterogeneity in Glioblastoma Unveils an Innovative Combination Therapy Strategy. J Mol Neurosci 2024; 74:52. [PMID: 38724832 DOI: 10.1007/s12031-024-02201-x] [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: 08/07/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Treatment of glioblastoma multiforme (GBM) remains challenging. Unraveling the orchestration of glutamine metabolism may provide a novel viewpoint on GBM therapy. The study presented a full and comprehensive comprehending of the glutamine metabolism atlas and heterogeneity in GBM for facilitating the development of a more effective therapeutic choice. Transcriptome data from large GBM cohorts were integrated in this study. A glutamine metabolism-based classification was established through consensus clustering approach, and a classifier by LASSO analysis was defined for differentiating the classification. Prognosis, signaling pathway activity, tumor microenvironment, and responses to immune checkpoint blockade (ICB) and small molecular drugs were characterized in each cluster. A combinational therapy of glutaminase inhibitor CB839 with dihydroartemisinin (DHA) was proposed, and the influence on glutamine metabolism, apoptosis, reactive oxygen species (ROS), and migration was measured in U251 and U373 cells. We discovered that GBM presented heterogeneous glutamine metabolism-based clusters, with unique survival outcomes, activity of signaling pathways, tumor microenvironment, and responses to ICB and small molecular compounds. In addition, the classifier could accurately differentiate the two clusters. Strikingly, the combinational therapy of CB839 with DHA synergistically attenuated glutamine metabolism, triggered apoptosis and ROS accumulation, and impaired migrative capacity in GBM cells, demonstrating the excellent preclinical efficacy. Altogether, our findings unveil the glutamine metabolism heterogeneity in GBM and propose an innovative combination therapy of CB839 with DHA for this malignant disease.
Collapse
Affiliation(s)
- Huangde Fu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China.
- Department of Neurosurgery, The Third Nanning People's Hospital, Nanning, Guangxi, 530005, China.
| | - Shengtian Wu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Hechun Shen
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Kai Luo
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Zhongxiang Huang
- Department of Pathology, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Nankun Lu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Yaolin Li
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Qian Lan
- Department of Laboratory, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Yishun Xian
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| |
Collapse
|
4
|
Yu H, Wang M, Wang X, Jiang X. Immune-related matrisomes are potential biomarkers to predict the prognosis and immune microenvironment of glioma patients. FEBS Open Bio 2022; 13:307-322. [PMID: 36560848 PMCID: PMC9900094 DOI: 10.1002/2211-5463.13541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 11/11/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) plays a vital role in the progression and metastasis of glioma and is an important part of the tumor microenvironment. The matrisome is composed of ECM components and related proteins. There have been several studies on the effects of matrisomes on the glioma immune microenvironment, but most of these studies were performed on individual glioma immune-related matrisomes rather than integral analysis. Hence, an overall analysis of all potential immune-related matrisomes in gliomas is needed. Here, we divided 667 glioma patients in The Cancer Genome Atlas (TCGA) database into low, moderate, and high immune infiltration groups. Immune-related matrisomes differentially expressed among the three groups were analyzed, and a risk signature was established. Eight immune-related matrisomes were screened, namely, LIF, LOX, MMP9, S100A4, SRPX2, SLIT1, SMOC1, and TIMP1. Kaplan-Meier analysis, operating characteristic curve analysis, and nomogram were constructed to analyze the relationships between risk signatures and the prognosis of glioma patients. The risk signature was significantly correlated with the overall survival of glioma patients. Both high- and low-risk signatures were also associated with some immune checkpoints. In addition, analysis of somatic mutations and anti-PD1/L1 immunotherapy responses in the high- and low-risk groups showed that the high-risk group had worse prognosis and a higher response to anti-PD1/L1 immunotherapy. Our analysis of immune-related matrisomes may improve understanding of the characteristics of the glioma immune microenvironment and provide direction for glioma immunotherapy development in the future.
Collapse
Affiliation(s)
- Hao Yu
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Minjie Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeWuhanChina
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeWuhanChina
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
5
|
Boot J, Rosser G, Kancheva D, Vinel C, Lim YM, Pomella N, Zhang X, Guglielmi L, Sheer D, Barnes M, Brandner S, Nelander S, Movahedi K, Marino S. Global hypo-methylation in a proportion of glioblastoma enriched for an astrocytic signature is associated with increased invasion and altered immune landscape. eLife 2022; 11:e77335. [PMID: 36412091 PMCID: PMC9681209 DOI: 10.7554/elife.77335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
We describe a subset of glioblastoma, the most prevalent malignant adult brain tumour, harbouring a bias towards hypomethylation at defined differentially methylated regions. This epigenetic signature correlates with an enrichment for an astrocytic gene signature, which together with the identification of enriched predicted binding sites of transcription factors known to cause demethylation and to be involved in astrocytic/glial lineage specification, point to a shared ontogeny between these glioblastomas and astroglial progenitors. At functional level, increased invasiveness, at least in part mediated by SRPX2, and macrophage infiltration characterise this subset of glioblastoma.
Collapse
Affiliation(s)
- James Boot
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Gabriel Rosser
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Dailya Kancheva
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit BrusselBrusselsBelgium
| | - Claire Vinel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Yau Mun Lim
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, and Department of Neurodegenerative Disease, Queen Square, Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Nicola Pomella
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Xinyu Zhang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Loredana Guglielmi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Denise Sheer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Michael Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Sebastian Brandner
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, and Department of Neurodegenerative Disease, Queen Square, Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Sven Nelander
- Department of Immunology Genetics and Pathology, Uppsala UniversityUppsalaSweden
| | - Kiavash Movahedi
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit BrusselBrusselsBelgium
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| |
Collapse
|
6
|
Sun Z, Gao X. SRPX2 attenuated oxygen–glucose deprivation and reperfusion-induced injury in cardiomyocytes via alleviating endoplasmic reticulum stress-induced apoptosis through targeting PI3K/Akt/mTOR axis. Open Life Sci 2022; 17:1497-1504. [DOI: 10.1515/biol-2022-0513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/06/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Myocardial infraction (MI) is the leading cause of high morbidity and mortality worldwide. It was still urgently needed to find new and effective drugs for MI treatment by the use of myocardial ischemia/reperfusion (I/R) model. Sushi repeats contain the protein X-Linked 2 (SRPX2), which regulates a variety of important cell functions. However, its possible role in myocardial I/R and the progression of MI is still unclear. In this study, we investigated the role of SRPX2 in myocardial I/R. SRPX2 showed low expression in IR rats and H9C2 cells induced by oxygen–glucose deprivation/reperfusion (OGD/R). SRPX2 could increase OGD/R-induced H9C2 cell survival. In addition, SRPX2 suppressed the apoptosis of OGD/R-induced H9C2 cells. Furthermore, we found that SRPX2 could inhibit ER stress induced by OGD/R in H9C2 cells. Mechanically, we found that SRPX2 suppressed the PI3K/Akt/mTOR pathway, thus attenuating OGD/R -induced injury in H9C2 cells. Therefore, SRPX2 has the potential to serve as a target for MI treatment.
Collapse
Affiliation(s)
- Zhiyuan Sun
- Department of Cardiovascular, Tianjin Fifth Central Hospital , Tianjin 300450 , China
| | - Xin Gao
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine , No. 100, Cross Street, Hongshan Road , Nanjing City , Jiangsu Province 210028 , China
| |
Collapse
|
7
|
Sushi-Repeat-Containing Protein X-Linked 2: A Potential Therapeutic Target for Inflammation and Cancer Therapy. J Immunol Res 2022; 2022:2931214. [PMID: 35935582 PMCID: PMC9352485 DOI: 10.1155/2022/2931214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Accumulating evidence has showed that sushi-repeat-containing protein X-linked 2 (SRPX2) is an abnormal expression in a variety of cancers and involved in cancer carcinogenesis, chemosensitivity, and prognosis, which mainly promote cancer cell metastasis, invasion, and migration by regulating the uPAR/integrins/FAK signaling pathway, epithelial-mesenchymal transition (EMT), angiogenesis, and glycosylation. Inflammation has been regarded as a key role in regulating cancer initiation, progression, EMT, and therapeutics. Furthermore, SRPX2 exhibited excellent antifibrosis effect via the TGFβR1/SMAD3/SRPX2/AP1/SMAD7 signaling pathway. Therefore, this review provides compelling evidence that SRPX2 might be a therapeutic target for inflammation and cancer-related inflammation for future cancer therapeutics.
Collapse
|
8
|
Seyfrid M, Maich WT, Shaikh VM, Tatari N, Upreti D, Piyasena D, Subapanditha M, Savage N, McKenna D, Mikolajewicz N, Han H, Chokshi C, Kuhlmann L, Khoo A, Salim SK, Archibong-Bassey B, Gwynne W, Brown K, Murtaza N, Bakhshinyan D, Vora P, Venugopal C, Moffat J, Kislinger T, Singh S. CD70 as an actionable immunotherapeutic target in recurrent glioblastoma and its microenvironment. J Immunother Cancer 2022; 10:e003289. [PMID: 35017149 PMCID: PMC8753449 DOI: 10.1136/jitc-2021-003289] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Glioblastoma (GBM) patients suffer from a dismal prognosis, with standard of care therapy inevitably leading to therapy-resistant recurrent tumors. The presence of cancer stem cells (CSCs) drives the extensive heterogeneity seen in GBM, prompting the need for novel therapies specifically targeting this subset of tumor-driving cells. Here, we identify CD70 as a potential therapeutic target for recurrent GBM CSCs. EXPERIMENTAL DESIGN In the current study, we identified the relevance and functional influence of CD70 on primary and recurrent GBM cells, and further define its function using established stem cell assays. We use CD70 knockdown studies, subsequent RNAseq pathway analysis, and in vivo xenotransplantation to validate CD70's role in GBM. Next, we developed and tested an anti-CD70 chimeric antigen receptor (CAR)-T therapy, which we validated in vitro and in vivo using our established preclinical model of human GBM. Lastly, we explored the importance of CD70 in the tumor immune microenvironment (TIME) by assessing the presence of its receptor, CD27, in immune infiltrates derived from freshly resected GBM tumor samples. RESULTS CD70 expression is elevated in recurrent GBM and CD70 knockdown reduces tumorigenicity in vitro and in vivo. CD70 CAR-T therapy significantly improves prognosis in vivo. We also found CD27 to be present on the cell surface of multiple relevant GBM TIME cell populations, notably putative M1 macrophages and CD4 T cells. CONCLUSION CD70 plays a key role in recurrent GBM cell aggressiveness and maintenance. Immunotherapeutic targeting of CD70 significantly improves survival in animal models and the CD70/CD27 axis may be a viable polytherapeutic avenue to co-target both GBM and its TIME.
Collapse
Affiliation(s)
- Mathieu Seyfrid
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - William Thomas Maich
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Nazanin Tatari
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Deepak Upreti
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Deween Piyasena
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Minomi Subapanditha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Dillon McKenna
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Nicholas Mikolajewicz
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hong Han
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Chirayu Chokshi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Laura Kuhlmann
- Department of Medical Biophysics, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Amanda Khoo
- Department of Medical Biophysics, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Sabra Khalid Salim
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - William Gwynne
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Kevin Brown
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Nadeem Murtaza
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - David Bakhshinyan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Parvez Vora
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Chitra Venugopal
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Jason Moffat
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Thomas Kislinger
- Department of Medical Biophysics, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Sheila Singh
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
9
|
Arya KR, Bharath Chand RP, Abhinand CS, Nair AS, Oommen OV, Sudhakaran PR. Identification of Hub Genes and Key Pathways Associated with Anti- VEGF Resistant Glioblastoma Using Gene Expression Data Analysis. Biomolecules 2021; 11:biom11030403. [PMID: 33803224 PMCID: PMC8000064 DOI: 10.3390/biom11030403] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/02/2021] [Indexed: 12/30/2022] Open
Abstract
Anti-VEGF therapy is considered to be a useful therapeutic approach in many tumors, but the low efficacy and drug resistance limit its therapeutic potential and promote tumor growth through alternative mechanisms. We reanalyzed the gene expression data of xenografts of tumors of bevacizumab-resistant glioblastoma multiforme (GBM) patients, using bioinformatics tools, to understand the molecular mechanisms of this resistance. An analysis of the gene set data from three generations of xenografts, identified as 646, 873 and 1220, differentially expressed genes (DEGs) in the first, fourth and ninth generations, respectively, of the anti-VEGF-resistant GBM cells. Gene Ontology (GO) and pathway enrichment analyses demonstrated that the DEGs were significantly enriched in biological processes such as angiogenesis, cell proliferation, cell migration, and apoptosis. The protein–protein interaction network and module analysis revealed 21 hub genes, which were enriched in cancer pathways, the cell cycle, the HIF1 signaling pathway, and microRNAs in cancer. The VEGF pathway analysis revealed nine upregulated (IL6, EGFR, VEGFA, SRC, CXCL8, PTGS2, IDH1, APP, and SQSTM1) and five downregulated hub genes (POLR2H, RPS3, UBA52, CCNB1, and UBE2C) linked with several of the VEGF signaling pathway components. The survival analysis showed that three upregulated hub genes (CXCL8, VEGFA, and IDH1) were associated with poor survival. The results predict that these hub genes associated with the GBM resistance to bevacizumab may be potential therapeutic targets or can be biomarkers of the anti-VEGF resistance of GBM.
Collapse
|
10
|
Liu Y, Chen T, Guo M, Li Y, Zhang Q, Tan G, Yu L, Tan Y. FOXA2-Interacting FOXP2 Prevents Epithelial-Mesenchymal Transition of Breast Cancer Cells by Stimulating E-Cadherin and PHF2 Transcription. Front Oncol 2021; 11:605025. [PMID: 33718155 PMCID: PMC7947682 DOI: 10.3389/fonc.2021.605025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
FOXP2, a member of forkhead box transcription factor family, was first identified as a language-related gene that played an important role in language learning and facial movement. In addition, FOXP2 was also suggested regulating the progression of cancer cells. In previous studies, we found that FOXA2 inhibited epithelial-mesenchymal transition (EMT) in breast cancer cells. In this study, by identifying FOXA2-interacting proteins from FOXA2-pull-down cell lysates with Mass Spectrometry Analysis, we found that FOXP2 interacted with FOXA2. After confirming the interaction between FOXP2 and FOXA2 through Co-IP and immunofluorescence assays, we showed a correlated expression of FOXP2 and FOXA2 existing in clinical breast cancer samples. The overexpression of FOXP2 attenuated the mesenchymal phenotype whereas the stable knockdown of FOXP2 promoted EMT in breast cancer cells. Even though FOXP2 was believed to act as a transcriptional repressor in most cases, we found that FOXP2 could activate the expression of tumor suppressor PHF2. Meanwhile, we also found that FOXP2 could endogenously bind to the promoter of E-cadherin and activate its transcription. This transcriptional activity of FOXP2 relied on its interaction with FOXA2. Furthermore, the stable knockdown of FOXP2 enhanced the metastatic capacity of breast cancer cells in vivo. Together, the results suggested that FOXP2 could inhibit EMT by activating the transcription of certain genes, such as E-cadherin and PHF2, in concert with FOXA2 in breast cancer cells.
Collapse
Affiliation(s)
- Yuxiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Taolin Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Mingyue Guo
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Yu Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Qian Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Guixiang Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Li Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| | - Yongjun Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, China
| |
Collapse
|
11
|
Chen H, Zeng Y, Shao M, Zhao H, Fang Z, Gu J, Liao B, Jin Y. Calcineurin A gamma and NFATc3/SRPX2 axis contribute to human embryonic stem cell differentiation. J Cell Physiol 2021; 236:5698-5714. [PMID: 33393109 DOI: 10.1002/jcp.30255] [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: 08/24/2020] [Revised: 12/09/2020] [Accepted: 12/21/2020] [Indexed: 12/29/2022]
Abstract
Our understanding of signaling pathways regulating the cell fate of human embryonic stem cells (hESCs) is limited. Calcineurin-NFAT signaling is associated with a wide range of biological processes and diseases. However, its role in controlling hESC fate remains unclear. Here, we report that calcineurin A gamma and the NFATc3/SRPX2 axis control the expression of lineage and epithelial-mesenchymal transition (EMT) markers in hESCs. Knockdown of PPP3CC, the gene encoding calcineurin A gamma, or NFATC3, downregulates certain markers both at the self-renewal state and during differentiation of hESCs. Furthermore, NFATc3 interacts with c-JUN and regulates the expression of SRPX2, the gene encoding a secreted glycoprotein known as a ligand of uPAR. We show that SRPX2 is a downstream target of NFATc3. Both SRPX2 and uPAR participate in controlling expression of lineage and EMT markers. Importantly, SRPX2 knockdown diminishes the upregulation of multiple lineage and EMT markers induced by co-overexpression of NFATc3 and c-JUN in hESCs. Together, this study uncovers a previously unknown role of calcineurin A gamma and the NFATc3/SRPX2 axis in modulating the fate determination of hESCs.
Collapse
Affiliation(s)
- Hao Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanwu Zeng
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Min Shao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hanzhi Zhao
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhuoqing Fang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Junjie Gu
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Bing Liao
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Basic Clinical Research Center, Renji Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ying Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Basic Clinical Research Center, Renji Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Zheng H, Li C, Li Z, Zhu K, Bao H, Xiong J, Liang P. HOXB9 enhances the ability of lung cancer cells to penetrate the blood-brain barrier. Aging (Albany NY) 2020; 13:4999-5019. [PMID: 33411683 PMCID: PMC7950248 DOI: 10.18632/aging.202324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 12/21/2022]
Abstract
Even after multimodal therapy, the prognosis is dismal for patients with brain metastases from non-small cell lung cancer (NSCLC). Although the blood-brain barrier (BBB) limits tumor cell penetration into the brain parenchyma, some nevertheless colonize brain tissue through mechanisms that are not fully clear. Here we show that homeobox B9 (HOXB9), which is commonly overexpressed in NSCLC, promotes epithelial-to-mesenchymal transition (EMT) and tumor migration and invasion. Animal experiments showed that HOXB9 expression correlates positively with the brain metastatic potential of human NSCLC cells, while brain metastatic cells derived through in vivo selection showed greater HOXB9 expression than their cells of origin. Comparable results were obtained after immunohistochemical analysis of clinical primary NSCLC and matched brain metastasis samples obtained after surgery. Using an in vitro BBB model, knockdown and overexpression experiments showed that HOXB9-dependent expression of MMP9 in NSCLC cells leads to reduced expression of junctional proteins in cultured human vascular endothelial cells and enhanced transmigration of tumor cells. These data indicate that HOXB9 enables NSCLC cells to break away from the primary tumor by inducing EMT, and promotes brain metastasis by driving MMP9 production and degradation of intercellular adhesion proteins in endothelial cells comprising the BBB.
Collapse
Affiliation(s)
- HongShan Zheng
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| | - ChenLong Li
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| | - ZhenZhe Li
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| | - KaiBin Zhu
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| | - HongBo Bao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| | - JinSheng Xiong
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| | - Peng Liang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin 150001, Heilongjiang, P.R. China
| |
Collapse
|
13
|
Gao Z, Wu J, Wu X, Zheng J, Ou Y. SRPX2 boosts pancreatic cancer chemoresistance by activating PI3K/AKT axis. Open Med (Wars) 2020; 15:1072-1082. [PMID: 33336063 PMCID: PMC7718643 DOI: 10.1515/med-2020-0157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/02/2020] [Accepted: 08/07/2020] [Indexed: 12/28/2022] Open
Abstract
Background and aim This investigation was aimed at disclosing whether SRPX2 affected pancreatic cancer (PC) chemoresistance by regulating PI3K/Akt/mTOR signaling. Methods Totally 243 PC patients were recruited, and they were incorporated into partial remission (PR) group, stable disease (SD) group and progressive disease (PD) group in accordance with their chemotherapeutic response. PC cell lines (i.e. AsPC1, Capan2, VFPAC-1, HPAC, PANC-1, BxPC-3 and SW1990) and human pancreatic ductal epithelial cell lines (hTERT-HPNE) were also collected. Results PC patients of SD + PD group were associated with higher post-chemotherapeutic SRPX2 level than PR group, and their post-chemotherapeutic SRPX2 level was above the pretherapeutic SRPX2 level (P < 0.05). PR population showed lower SRPX2 level after chemotherapy than before chemotherapy (P < 0.05). Besides high serum SRPX2 level and SRPX2 level change before and after chemotherapy were independent predictors of poor PC prognosis. Additionally, si-SRPX2 enhanced chemosensitivity of PC cell lines, and expressions of p-PI3K, p-AKT and p-mTOR were suppressed by si-SRPX2 (P < 0.05). IGF-1 treatment could changeover the impact of si-SRPX2 on proliferation, migration, invasion and chemoresistance of PC cells (P < 0.05). Conclusion The SRPX2-PI3K/AKT/mTOR axis could play a role in modifying progression and chemoresistance of PC cells, which might help to improve PC prognosis.
Collapse
Affiliation(s)
- Zhenyuan Gao
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Anhui, China
| | - Jisong Wu
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Anhui, China
| | - Xiao Wu
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Anhui, China
| | - Jialei Zheng
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Anhui, China
| | - Yimei Ou
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Anhui, China
| |
Collapse
|
14
|
Sasahira T, Kurihara-Shimomura M, Nishiguchi Y, Shimomura H, Kirita T. Sushi Repeat Containing Protein X-linked 2 Is a Downstream Signal of LEM Domain Containing 1 and Acts as a Tumor-Promoting Factor in Oral Squamous Cell Carcinoma. Int J Mol Sci 2020; 21:ijms21103655. [PMID: 32455867 PMCID: PMC7279144 DOI: 10.3390/ijms21103655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 01/22/2023] Open
Abstract
Because oral squamous cell carcinomas (OSCCs) have a high potential for locoregional invasion and nodal metastasis, early detection and treatment are essential. A LAP2, emerin, MAN1 (LEM) domain containing 1 (LEMD1) is associated with local progression, clinical stage, nodal metastasis, poor prognosis, angiogenesis, and lymphangiogenesis in OSCC. Although LEMD is a cancer-testis antigen, the cancer-related signals related to LEMD1 remain unknown. In this study, we used a microarray analysis of OSCC cells to identify sushi repeat containing protein X-linked 2 (SRPX2) as a LEMD1-related downstream signal. LEMD1 expression was correlated with lymph node metastasis of OSCC according to the immunohistochemistry analysis. Furthermore, patients expressing SRPX2 had a significantly worse prognosis than those without SRPX2 expression. The concentration of SRPX2 in OSCC was positively correlated with the concentrations of LEMD1, urokinase plasminogen activator receptor (uPAR), and hepatocyte growth factor (HGF). In OSCC cells, SRPX2 secretion levels were elevated by interactions with uPAR and HGF. We also found that SRPX2 promotes endothelial cell proliferation and adhesion between endothelial cells and OSCC cells. These results suggest that SRPX2 might be a useful tumor marker for OSCC.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/secondary
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Drug Resistance, Neoplasm/genetics
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Female
- Gene Expression Regulation, Neoplastic/genetics
- Hepatocyte Growth Factor/metabolism
- Humans
- Lymphatic Metastasis
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Middle Aged
- Mouth Neoplasms/genetics
- Mouth Neoplasms/metabolism
- Mouth Neoplasms/mortality
- Mouth Neoplasms/pathology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neovascularization, Pathologic/genetics
- Oligonucleotide Array Sequence Analysis
- Prognosis
- RNA, Small Interfering
- Receptors, Urokinase Plasminogen Activator/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Tomonori Sasahira
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan; (M.K.-S.); (Y.N.)
- Correspondence: ; Tel.: +81-744-29-8849; Fax: +81-744-25-7308
| | - Miyako Kurihara-Shimomura
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan; (M.K.-S.); (Y.N.)
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan; (H.S.); (T.K.)
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan; (M.K.-S.); (Y.N.)
| | - Hiroyuki Shimomura
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan; (H.S.); (T.K.)
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan; (H.S.); (T.K.)
| |
Collapse
|
15
|
Wu Z, Wang C, Chen Y, Sun Z, Yan W. SRPX2 Promotes Cell Proliferation and Invasion in Osteosarcoma Through Regulating Hippo Signaling Pathway. Onco Targets Ther 2020; 13:1737-1749. [PMID: 32161469 PMCID: PMC7049857 DOI: 10.2147/ott.s225602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background/Purpose Osteosarcoma (OS), a primary bone malignancy, is characterized by a high rate of metastasis. It has been found that Sushi repeat containing protein X-linked 2 (SRPX2) is involved in tumor cell proliferation, adhesion, invasion and migration. The current work aimed to explore the effect of SRPX2 on OS cell invasion and proliferation. Methods Immunohistochemistry (IHC), Western blotting and reverse transcription-polymerase chain reaction (RT-PCR) were used to detect the expression of the associated protein in OS tissues and cell lines. Cell counting kit-8 (CCK8), transwell and colony formation assays were used to determine cell viability, invasion, and proliferation, respectively. The in vivo tumorigenic ability of SRPX2 gene was determined using nude mouse tumorigenesis test. Results SRPX2 knockdown suppressed the viability, while SRPX2 overexpression increased the invasion and colony formation ability of the cells in vitro. In vivo experiments demonstrated that SRPX2 knockdown inhibited tumor growth and invasion as evidenced by decreased Ki67 and N-cadherin levels, and increased E-cadherin level. Downregulation of SRPX2 increased YAP phosphorylation resulting in reduced nuclear translocation to activate Hippo signaling pathway. The promotion of cell viability, colony-forming ability, and invasion, and the inhibition of CTGF, Cyr61, and Birc5 levels promoted by SRPX2 overexpression were reversed by YAP inhibition. Conclusion SRPX2 increased cell proliferation and invasion in osteosarcoma by activating Hippo signaling pathway.
Collapse
Affiliation(s)
- Zhiqiang Wu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Chunmeng Wang
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yong Chen
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Zhengwang Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
16
|
Casella G, Munk R, Kim KM, Piao Y, De S, Abdelmohsen K, Gorospe M. Transcriptome signature of cellular senescence. Nucleic Acids Res 2019; 47:7294-7305. [PMID: 31251810 DOI: 10.1093/nar/gkz555] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
Cellular senescence, an integral component of aging and cancer, arises in response to diverse triggers, including telomere attrition, macromolecular damage and signaling from activated oncogenes. At present, senescent cells are identified by the combined presence of multiple traits, such as senescence-associated protein expression and secretion, DNA damage and β-galactosidase activity; unfortunately, these traits are neither exclusively nor universally present in senescent cells. To identify robust shared markers of senescence, we have performed RNA-sequencing analysis across eight diverse models of senescence triggered in human diploid fibroblasts (WI-38, IMR-90) and endothelial cells (HUVEC, HAEC) by replicative exhaustion, exposure to ionizing radiation or doxorubicin, and expression of the oncogene HRASG12V. The intersection of the altered transcriptomes revealed 50 RNAs consistently elevated and 18 RNAs consistently reduced across all senescence models, including many protein-coding mRNAs and some non-coding RNAs. We propose that these shared transcriptome profiles will enable the identification of senescent cells in vivo, the investigation of their roles in aging and malignancy and the development of strategies to target senescent cells therapeutically.
Collapse
Affiliation(s)
- Gabriel Casella
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| | - Kyoung Mi Kim
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Biomedical Research Center, Baltimore, Maryland 21224, USA
| |
Collapse
|
17
|
Anwer M, Bolkvadze T, Puhakka N, Ndode-Ekane XE, Pitkänen A. Genotype and Injury Effect on the Expression of a Novel Hypothalamic Protein Sushi Repeat-Containing Protein X-Linked 2 (SRPX2). Neuroscience 2019; 415:184-200. [DOI: 10.1016/j.neuroscience.2019.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/04/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022]
|
18
|
Jiang X, Tan J, Wen Y, Liu W, Wu S, Wang L, Wangou S, Liu D, Du C, Zhu B, Xie D, Ren C. MSI2-TGF-β/TGF-β R1/SMAD3 positive feedback regulation in glioblastoma. Cancer Chemother Pharmacol 2019; 84:415-425. [PMID: 31250154 DOI: 10.1007/s00280-019-03892-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/07/2019] [Indexed: 01/09/2023]
Abstract
PURPOSE Glioblastoma is the most malignant glioma tumors with inevitable relapse and resistance to chemotherapy; however, the mechanisms driving chemoresistance remain to be fully elucidated. This study is to explore the molecular and cellular mechanisms involving in the chemoresistance of glioblastoma. METHODS The expression of musashi (MSI) RNA-binding protein in the tumor tissues and cells of glioblastoma was measured. The effects of MSI2 in epithelial-to-mesenchymal transition (EMT), resistance to temozolomide (TMZ), tumor cell invasion, migration, and proliferation and associated signaling were evaluated. RESULTS High MSI2 expression was observed in the glioblastoma tissues. Silencing or overexpression of MSI2 significantly affected tumor cells invasion, migration, and proliferation. Silencing of MSI2 expression significantly inhibited O6-methylguanine-DNA methyltransferase (MGMT) expression and tumor growth, and reversed resistance to TMZ in xenograft tumor models. MSI2 expression regulated EMT through activating the transcription factors Snail and the TGFβ R1/SMAD3 signaling. CONCLUSIONS Our study demonstrated a positive feedback loop of MSI2-TGFβ/SMAD3 signaling which activates the EMT and MGMT which may contribute to chemoresistance in glioblastoma. This study also highlights that MSI2 could be a new target for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Xingjun Jiang
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China.
| | - Jun Tan
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Yin Wen
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Weidong Liu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Shuyu Wu
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Lei Wang
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Siyi Wangou
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Dingyang Liu
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Can Du
- Department of Neurosurgery, Collaborative Innovation Center for Cancer Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, People's Republic of China
| | - Bin Zhu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
| | - Caiping Ren
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, The Key Laboratory for Carcinogenesis of Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, School of Basic Medical Science, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
19
|
Li H, Zhang SR, Xu HX, Wang WQ, Li S, Li TJ, Ni QX, Yu XJ, Liu L, Wu CT. SRPX2 and RAB31 are effective prognostic biomarkers in pancreatic cancer. J Cancer 2019; 10:2670-2678. [PMID: 31258775 PMCID: PMC6584922 DOI: 10.7150/jca.32072] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/01/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: SRPX2 and RAB31 play important roles in tumorigenesis and metastasis; however, their prognostic value in pancreatic cancer remains unclear. This study aimed to investigate the potential interactions and effects of SRPX2 and RAB31 on the diagnosis and prognosis of pancreatic cancer. Methods: The expression of SRPX2 and RAB31 in pancreatic tumor tissues and cells was evaluated through database mining of the Oncomine, Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, and validated the results through immunohistochemistry (IHC) and Western blot in our clinical database. Protein-protein interactions were explored by immunofluorescence and Co-immunoprecipitation (Co-IP). Two hundred tissue microarray specimens from patients (79 training and 121 validation), who underwent curative pancreatectomy for pancreatic ductal adenocarcinoma (PDAC) were used. Additionally, the association between the SRPX2 and RAB31 and prognosis of PDAC patients after surgery was analyzed. Results: The expression of SRPX2 and RAB31 was highly increased in pancreatic cancer, and there was a significant positive correlation between these two proteins. Co-IP showed the direct interaction between SRPX2 and RAB31. Kaplan-Meier analysis showed that positive expression of SRPX2 and RAB31 was associated with reduced disease-free survival (DFS) and overall survival (OS) of PDAC patients in the training set and the validation sets. Furthermore, multivariate analysis indicated that the 8th edition TNM stage and combination of SRPX2 and RAB31 were independent prognostic factors that associated with OS and DFS in the training, and the validation sets, respectively. Conclusions: The combination of SRPX2 and RAB31 can be important markers for the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Hao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Shi-Rong Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Hua-Xiang Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Shuo Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Tian-Jiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Quan-Xing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Chun-Tao Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 20032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.,Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Alshabi AM, Vastrad B, Shaikh IA, Vastrad C. Identification of Crucial Candidate Genes and Pathways in Glioblastoma Multiform by Bioinformatics Analysis. Biomolecules 2019; 9:biom9050201. [PMID: 31137733 PMCID: PMC6571969 DOI: 10.3390/biom9050201] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to investigate the molecular mechanisms underlying glioblastoma multiform (GBM) and its biomarkers. The differentially expressed genes (DEGs) were diagnosed using the limma software package. The ToppGene (ToppFun) was used to perform pathway and Gene Ontology (GO) enrichment analysis of the DEGs. Protein-protein interaction (PPI) networks, extracted modules, miRNA-target genes regulatory network and TF-target genes regulatory network were used to obtain insight into the actions of DEGs. Survival analysis for DEGs was carried out. A total of 590 DEGs, including 243 up regulated and 347 down regulated genes, were diagnosed between scrambled shRNA expression and Lin7A knock down. The up-regulated genes were enriched in ribosome, mitochondrial translation termination, translation, and peptide biosynthetic process. The down-regulated genes were enriched in focal adhesion, VEGFR3 signaling in lymphatic endothelium, extracellular matrix organization, and extracellular matrix. The current study screened the genes in the PPI network, extracted modules, miRNA-target genes regulatory network, and TF-target genes regulatory network with higher degrees as hub genes, which included NPM1, CUL4A, YIPF1, SHC1, AKT1, VLDLR, RPL14, P3H2, DTNA, FAM126B, RPL34, and MYL5. Survival analysis indicated that the high expression of RPL36A and MRPL35 were predicting longer survival of GBM, while high expression of AP1S1 and AKAP12 were predicting shorter survival of GBM. High expression of RPL36A and AP1S1 were associated with pathogenesis of GBM, while low expression of ALPL was associated with pathogenesis of GBM. In conclusion, the current study diagnosed DEGs between scrambled shRNA expression and Lin7A knock down samples, which could improve our understanding of the molecular mechanisms in the progression of GBM, and these crucial as well as new diagnostic markers might be used as therapeutic targets for GBM.
Collapse
Affiliation(s)
- Ali Mohamed Alshabi
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran 61441, Saudi Arabia.
| | - Basavaraj Vastrad
- Department of Pharmaceutics, SET`S College of Pharmacy, Dharwad, Karnataka 580002, India.
| | - Ibrahim Ahmed Shaikh
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 61441, Saudi Arabia.
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India.
| |
Collapse
|
21
|
Liu X, Chen X, Shi L, Shan Q, Cao Q, Yue C, Li H, Li S, Wang J, Gao S, Niu M, Yu R. The third-generation EGFR inhibitor AZD9291 overcomes primary resistance by continuously blocking ERK signaling in glioblastoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:219. [PMID: 31122294 PMCID: PMC6533774 DOI: 10.1186/s13046-019-1235-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022]
Abstract
Background Glioblastoma (GBM) is a fatal brain tumor, lacking effective treatment. Epidermal growth factor receptor (EGFR) is recognized as an attractive target for GBM treatment. However, GBMs have very poor responses to the first- and second-generation EGFR inhibitors. The third-generation EGFR-targeted drug, AZD9291, is a novel and irreversible inhibitor. It is noteworthy that AZD9291 shows excellent blood–brain barrier penetration and has potential for the treatment of brain tumors. Methods In this study, we evaluated the anti-tumor activity and effectiveness of AZD9291 in a preclinical GBM model. Results AZD9291 showed dose-responsive growth inhibitory activity against six GBM cell lines. Importantly, AZD9291 inhibited GBM cell proliferation > 10 times more efficiently than the first-generation EGFR inhibitors. AZD9291 induced GBM cell cycle arrest and significantly inhibited colony formation, migration, and invasion of GBM cells. In an orthotopic GBM model, AZD9291 treatment significantly inhibited tumor survival and prolonged animal survival. The underlying anti-GBM mechanism of AZD9291 was shown to be different from that of the first-generation EGFR inhibitors. In contrast to erlotinib, AZD9291 continuously and efficiently inhibited the EGFR/ERK signaling in GBM cells. Conclusion AZD9291 demonstrated an efficient preclinical activity in GBM in vitro and in vivo models. AZD9291 has been approved for the treatment of lung cancer with good safety and tolerability. Our results support the possibility of conducting clinical trials of anti-GBM therapy using AZD9291. Electronic supplementary material The online version of this article (10.1186/s13046-019-1235-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuejiao Liu
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangyu Chen
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Shi
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qianqian Shan
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiyu Cao
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chenglong Yue
- Surgical Department 9, Xuzhou children's hospital, Xuzhou, Jiangsu, China
| | - Huan Li
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shengsheng Li
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Wang
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shangfeng Gao
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingshan Niu
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Rutong Yu
- Insititute of Nervous System Diseases, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
22
|
Gene-Specific Intron Retention Serves as Molecular Signature that Distinguishes Melanoma from Non-Melanoma Cancer Cells in Greek Patients. Int J Mol Sci 2019; 20:ijms20040937. [PMID: 30795533 PMCID: PMC6412294 DOI: 10.3390/ijms20040937] [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: 01/23/2019] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Skin cancer represents the most common human malignancy, and it includes BCC, SCC, and melanoma. Since melanoma is one of the most aggressive types of cancer, we have herein attempted to develop a gene-specific intron retention signature that can distinguish BCC and SCC from melanoma biopsy tumors. Methods: Intron retention events were examined through RT-sqPCR protocols, using total RNA preparations derived from BCC, SCC, and melanoma Greek biopsy specimens. Intron-hosted miRNA species and their target transcripts were predicted via the miRbase and miRDB bioinformatics platforms, respectively. Ιntronic ORFs were recognized through the ORF Finder application. Generation and visualization of protein interactomes were achieved by the IntAct and Cytoscape softwares, while tertiary protein structures were produced by using the I-TASSER online server. Results: c-MYC and Sestrin-1 genes proved to undergo intron retention specifically in melanoma. Interaction maps of proteins encoded by genes being potentially targeted by retained intron-accommodated miRNAs were generated and SRPX2 was additionally delivered to our melanoma-specific signature. Novel ORFs were identified in MCT4 and Sestrin-1 introns, with potentially critical roles in melanoma development. Conclusions: The property of c-MYC, Sestrin-1, and SRPX2 genes to retain specific introns could be clinically used to molecularly differentiate non-melanoma from melanoma tumors.
Collapse
|
23
|
Freitas D, Campos D, Gomes J, Pinto F, Macedo JA, Matos R, Mereiter S, Pinto MT, Polónia A, Gartner F, Magalhães A, Reis CA. O-glycans truncation modulates gastric cancer cell signaling and transcription leading to a more aggressive phenotype. EBioMedicine 2019; 40:349-362. [PMID: 30662000 PMCID: PMC6413340 DOI: 10.1016/j.ebiom.2019.01.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Changes in glycosylation are known to play critical roles during gastric carcinogenesis. Expression of truncated O-glycans, such as the Sialyl-Tn (STn) antigen, is a common feature shared by many cancers and is associated with cancer aggressiveness and poor-prognosis. METHODS Glycoengineered cell lines were used to evaluate the impact of truncated O-glycans in cancer cell biology using in vitro functional assays, transcriptomic analysis and in vivo models. Tumor patients 'samples and datasets were used for clinical translational significance evaluation. FINDINGS In the present study, we demonstrated that gastric cancer cells expressing truncated O-glycans display major phenotypic alterations associated with higher cell motility and cell invasion. Noteworthy, the glycoengineered cancer cells overexpressing STn resulted in tumor xenografts with less cohesive features which had a critical impact on mice survival. Furthermore, truncation of O-glycans induced activation of EGFR and ErbB2 receptors and a transcriptomic signature switch of gastric cancer cells. The disclosed top activated genes were further validated in gastric tumors, revealing that SRPX2 and RUNX1 are concomitantly overexpressed in gastric carcinomas and its expression is associated with patients' poor-survival, highlighting their prognosis potential in clinical practice. INTERPRETATION This study discloses novel molecular links between O-glycans truncation frequently observed in cancer and key cellular regulators with major impact in tumor progression and patients' clinical outcome.
Collapse
Affiliation(s)
- Daniela Freitas
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, Porto 4050-313, Portugal
| | - Diana Campos
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Joana Gomes
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Filipe Pinto
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Joana A Macedo
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Rita Matos
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Stefan Mereiter
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Marta T Pinto
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - António Polónia
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Fátima Gartner
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, Porto 4050-313, Portugal
| | - Ana Magalhães
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal.
| | - Celso A Reis
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IPATIMUP -Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, Porto 4050-313, Portugal; Faculty of Medicine of the University of Porto, Al. Prof. Hernâni Monteiro, Porto 4200-319, Portugal.
| |
Collapse
|
24
|
SRPX2 knockdown inhibits cell proliferation and metastasis and promotes chemosensitivity in esophageal squamous cell carcinoma. Biomed Pharmacother 2019; 109:671-678. [DOI: 10.1016/j.biopha.2018.10.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022] Open
|
25
|
Hong X, Hong X, Zhao H, He C. Knockdown of SRPX2 inhibits the proliferation, migration, and invasion of prostate cancer cells through the PI3K/Akt/mTOR signaling pathway. J Biochem Mol Toxicol 2018; 33:e22237. [PMID: 30537353 DOI: 10.1002/jbt.22237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 08/05/2018] [Accepted: 08/09/2018] [Indexed: 01/04/2023]
Abstract
Sushi repeat-containing protein X-linked 2 (SRPX2), a novel chondroitin sulfate proteoglycan, is reported to play a critical role in tumorigenesis. However, the expression and functional role of SRPX2 in prostate cancer have not been defined. Thus, the aim of this study was to investigate the expression and functional role of SRPX2 in human prostate cancer. Our results showed that the expression of SRPX2 was obviously increased in human prostate cancer tissues and cell lines. In addition, knockdown of SRPX2 inhibited the proliferation, migration, and invasion of prostate cancer cells, as well as prevented the epithelial-mesenchymal transition process in prostate cancer cells. Mechanically, knockdown of SRPX2 efficiently inhibited the activation of PI3K/Akt/mTOR pathway in prostate cancer cells. Taken together, these data demonstrated that knockdown of SRPX2 inhibits the proliferation and metastasis in human prostate cancer cells, partly through the PI3K/Akt/mTOR signaling pathway. Thus, SRPX2 may be a novel therapeutic target for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xin Hong
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xingyu Hong
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Haomin Zhao
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chengyan He
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Dali R, Verginelli F, Pramatarova A, Sladek R, Stifani S. Characterization of a FOXG1:TLE1 transcriptional network in glioblastoma-initiating cells. Mol Oncol 2018; 12:775-787. [PMID: 29316219 PMCID: PMC5983107 DOI: 10.1002/1878-0261.12168] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/12/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain cancer of glial cell origin, with a median patient survival of less than 20 months. Transcription factors FOXG1 and TLE1 promote GBM propagation by supporting maintenance of brain tumour-initiating cells (BTICs) with stem-like properties. Here, we characterize FOXG1 and TLE1 target genes in GBM patient-derived BTICs using ChIP-Seq and RNA-Seq approaches. These studies identify 150 direct FOXG1 targets, several of which are also TLE1 targets, involved in cell proliferation, differentiation, survival, chemotaxis and angiogenesis. Negative regulators of NOTCH signalling, including CHAC1, are among the transcriptional repression targets of FOXG1:TLE1 complexes, suggesting a crosstalk between FOXG1:TLE1 and NOTCH-mediated pathways in GBM. These results provide previously unavailable insight into the transcriptional programs underlying the tumour-promoting functions of FOXG1:TLE1 in GBM.
Collapse
Affiliation(s)
- Rola Dali
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
- McGill Center for BioinformaticsMcGill UniversityMontrealCanada
| | - Federica Verginelli
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
- Present address:
Laboratory of Cancer Stem Cell ResearchCandiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | - Albena Pramatarova
- Departments of Human Genetics and MedicineMcGill UniversityMontrealCanada
| | - Robert Sladek
- Departments of Human Genetics and MedicineMcGill UniversityMontrealCanada
| | - Stefano Stifani
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
| |
Collapse
|
27
|
Zhang M, Li X, Fan Z, Zhao J, Liu S, Zhang M, Li H, Goscinski MA, Fan H, Suo Z. High SRPX2 protein expression predicts unfavorable clinical outcome in patients with prostate cancer. Onco Targets Ther 2018; 11:3149-3157. [PMID: 29881288 PMCID: PMC5983007 DOI: 10.2147/ott.s158820] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Sushi repeat-containing protein X-linked 2 (SRPX2) is overexpressed in a variety of different tumor tissues and correlated with poor prognosis in patients. Little research focuses on the role of SRPX2 expression in prostate cancer (PCa), and the clinicopathological significance of the protein expression in this tumor is relatively unknown. However, our previous transcriptome data from those cancer stem-like cells indicated the role of SRPX2 in PCa. Materials and methods In this study, RT-PCR and Western blotting were firstly used to examine the SRPX2 expression in three PCa cell lines including LNCaP, DU145, and PC3, and then SRPX2 protein expression was immunohistochemically investigated and statistically analyzed in a series of 106 paraffin-embedded PCa tissue specimens. Results Significantly lower levels of SRPX2 expression were verified in the LNCaP cells, compared with the expression in the aggressive DU145 and PC3 cells, in both mRNA and protein levels. Immunohistochemically, there were variable SRPX2 protein expressions in the clinical samples. Moreover, high levels of SRPX2 expression in the PCa tissues were significantly associated with Gleason score (P=0.008), lymph node metastasis (P=0.009), and distant metastasis (P=0.021). Furthermore, higher levels of SRPX2 expression in the PCa tissues were significantly associated with shorter overall survival (OS) (P<0.001). Conclusion Our results demonstrate that SRPX2 is highly expressed in aggressive PCa cells in vitro, and its protein expression in PCa is significantly associated with malignant clinical features and shorter OS, strongly indicating its prognostic value in prostate cancers.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaoli Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China.,Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Zhirui Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Shuzheng Liu
- Henan Office for Cancer Research and Control, Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huixiang Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mariusz Adam Goscinski
- Department of Surgery, The Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Huijie Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhenhe Suo
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China.,Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, Norway.,Department of Pathology, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
28
|
Anwer M, Bolkvadze T, Ndode-Ekane XE, Puhakka N, Rauramaa T, Leinonen V, van Vliet EA, Swaab DF, Haapasalo A, Leskelä S, Bister N, Malm T, Carlson S, Aronica E, Pitkänen A. Sushi repeat-containing protein X-linked 2: A novel phylogenetically conserved hypothalamo-pituitary protein. J Comp Neurol 2018; 526:1806-1819. [PMID: 29663392 DOI: 10.1002/cne.24449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022]
Abstract
Sushi repeat-containing protein X-linked 2 (SRPX2) is a novel protein associated with language development, synaptic plasticity, tissue remodeling, and angiogenesis. We investigated the expression and spatial localization of SRPX2 in normal mouse, rat, monkey, and human brain using in situ hybridization and immunohistochemistry. Antibody specificity was determined using in vitro siRNA based silencing of SRPX2. Cell type-specific expression was verified by double-labeling with oxytocin or vasopressin. Western blot was used to detect SRPX2 protein in rat and human plasma and cerebrospinal fluid. Unexpectedly, SRPX2 mRNA expression levels were strikingly higher in the hypothalamus as compared to the cortex. All SRPX2 immunoreactive (ir) neurons were localized in the hypothalamic paraventricular, periventricular, and supraoptic nuclei in mouse, rat, monkey, and human brain. SRPX2 colocalized with vasopressin or oxytocin in paraventricular and supraoptic neurons. Hypothalamic SRPX2-ir positive neurons gave origin to dense projections traveling ventrally and caudally toward the hypophysis. Intense axonal varicosities and terminal arborizations were identified in the rat and human neurohypophysis. SRPX2-ir cells were also found in the adenohypophysis. Light SRPX2-ir projections were observed in the dorsal and ventral raphe, locus coeruleus, and the nucleus of the solitary tract in mouse, rat and monkey. SRPX2 protein was also detected in plasma and CSF. Our data revealed intense phylogenetically conserved expression of SRPX2 protein in distinct hypothalamic nuclei and the hypophysis, suggesting its active role in the hypothalamo-pituitary axis. The presence of SRPX2 protein in the plasma and CSF suggests that some of its functions depend on secretion into body fluids.
Collapse
Affiliation(s)
- Mehwish Anwer
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tamuna Bolkvadze
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Noora Puhakka
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| | - Erwin A van Vliet
- Department of (Neuro) Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, KNAW, Amsterdam, The Netherlands
| | - Annakaisa Haapasalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stina Leskelä
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nea Bister
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Synnöve Carlson
- Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
29
|
Gäbel G, Northoff BH, Weinzierl I, Ludwig S, Hinterseher I, Wilfert W, Teupser D, Doderer SA, Bergert H, Schönleben F, Lindeman JHN, Holdt LM. Molecular Fingerprint for Terminal Abdominal Aortic Aneurysm Disease. J Am Heart Assoc 2017; 6:JAHA.117.006798. [PMID: 29191809 PMCID: PMC5779007 DOI: 10.1161/jaha.117.006798] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Clinical decision making in abdominal aortic aneurysms (AAA) relies completely on diameter. At this point, improved decision tools remain an unmet medical need. Our goal was to identify changes at the molecular level specifically leading up to AAA rupture. Methods and Results Aortic wall tissue specimens were collected during open elective (eAAA; n=31) or emergency repair of ruptured AAA (rAAA; n=17), and gene expression was investigated using microarrays. Identified candidate genes were validated with quantitative real‐time polymerase chain reaction in an independent sample set (eAAA: n=46; rAAA: n=18). Two gene sets were identified, 1 set containing 5 genes linked to terminal progression, that is, positively associated with progression of larger AAA, and with rupture (HILPDA,ANGPTL4,LOX,SRPX2,FCGBP), and a second set containing 5 genes exclusively upregulated in rAAA (ADAMTS9,STC1,GFPT2,GAL3ST4,CCL4L1). Genes in both sets essentially associated with processes related to impaired tissue remodeling, such as angiogenesis and adipogenesis. In gene expression experiments we were able to show that upregulated gene expression for identified candidate genes is unique for AAA. Functionally, the selected upregulated factors converge at processes coordinated by the canonical HIF‐1α signaling pathway and are highly expressed in fibroblasts but not inflammatory cells of the aneurysmatic wall. Histological quantification of angiogenesis and exploration of the HIF‐1α network in rAAA versus eAAA shows enhanced microvessel density but also clear activation of the HIF‐1α network in rAAA. Conclusions Our study shows a specific molecular fingerprint for terminal AAA disease. These changes appear to converge at activation of HIF‐1α signaling in mesenchymal cells. Aspects of this cascade might represent targets for rupture risk assessment.
Collapse
Affiliation(s)
- Gabor Gäbel
- Department of Vascular and Endovascular Surgery, Ludwig-Maximilians-University Munich, Munich, Germany .,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Bernd H Northoff
- Institute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Irina Weinzierl
- Department of Vascular and Endovascular Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Stefan Ludwig
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Irene Hinterseher
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany.,Department of General, Visceral, Vascular and Thoracic Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Wilfert
- Institute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stefan A Doderer
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hendrik Bergert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany.,Vascular and Endovascular Surgery, HELIOS Clinic Erfurt, Erfurt, Germany
| | - Frank Schönleben
- Department of Vascular and Endovascular Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jan H N Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Lesca M Holdt
- Institute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
30
|
Shi C, Uda Y, Dedic C, Azab E, Sun N, Hussein AI, Petty CA, Fulzele K, Mitterberger-Vogt MC, Zwerschke W, Pereira R, Wang K, Pajevic PD. Carbonic anhydrase III protects osteocytes from oxidative stress. FASEB J 2017; 32:440-452. [PMID: 28928248 DOI: 10.1096/fj.201700485rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/05/2017] [Indexed: 12/26/2022]
Abstract
Osteocytes are master orchestrators of bone remodeling; they control osteoblast and osteoclast activities both directly via cell-to-cell communication and indirectly via secreted factors, and they are the main postnatal source of sclerostin and RANKL (receptor activator of NF-kB ligand), two regulators of osteoblast and osteoclast function. Despite progress in understanding osteocyte biology and function, much remains to be elucidated. Recently developed osteocytic cell lines-together with new genome editing tools-has allowed a closer look at the biology and molecular makeup of these cells. By using single-cell cloning, we identified genes that are associated with high Sost/sclerostin expression and analyzed their regulation and function. Unbiased transcriptome analysis of high- vs. low-Sost/sclerostin-expressing cells identified known and novel genes. Dmp1 (dentin matrix protein 1), Dkk1 (Dickkopf WNT signaling pathway inhibitor 1), and Phex were among the most up-regulated known genes, whereas Srpx2, Cd200, and carbonic anhydrase III (CAIII) were identified as novel markers of differentiated osteocytes. Aspn, Enpp2, Robo2, Nov, and Serpina3g were among the transcripts that were most significantly suppressed in high-Sost cells. Considering that CAII was recently identified as being regulated by Sost/sclerostin and capable of controlling mineral homeostasis, we focused our attention on CAIII. Here, we report that CAIII is highly expressed in osteocytes, is regulated by parathyroid hormone both in vitro and in vivo, and protects osteocytes from oxidative stress.-Shi, C., Uda, Y., Dedic, C., Azab, E., Sun, N., Hussein, A. I., Petty, C. A., Fulzele, K., Mitterberger-Vogt, M. C., Zwerschke, W., Pereira, R., Wang, K., Divieti Pajevic, P. Carbonic anhydrase III protects osteocytes from oxidative stress.
Collapse
Affiliation(s)
- Chao Shi
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Yuhei Uda
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Christopher Dedic
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Ehab Azab
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Ningyuan Sun
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Amira I Hussein
- Department of Orthopedics, School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Christopher A Petty
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | - Keertik Fulzele
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA
| | | | - Werner Zwerschke
- Cell Metabolism and Differentiation Research Group, University of Innsbruck, Innsbruck, Austria
| | - Renata Pereira
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Kunzheng Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China;
| | - Paola Divieti Pajevic
- Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts, USA;
| |
Collapse
|
31
|
Wang L, Su J, Zhao Z, Hou Y, Yin X, Zheng N, Zhou X, Yan J, Xia J, Wang Z. MiR-26b reverses temozolomide resistance via targeting Wee1 in glioma cells. Cell Cycle 2017; 16:1954-1964. [PMID: 28898169 DOI: 10.1080/15384101.2017.1367071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence has demonstrated that microRNAs (miRNA) play a critical role in chemotherapy-induced epithelial-mesenchymal transition (EMT) in glioma. However, the underlying mechanism of chemotherapy-triggered EMT has not been fully understood. In the current study, we determined the role of miR-26b in regulation of EMT in stable temozolomide (TMZ)-resistant (TR) glioma cells, which have displayed mesenchymal features. Our results illustrated that miR-26b was significantly downregulated in TR cells. Moreover, ectopic expression of miR-26b by its mimics reversed the phenotype of EMT in TR cells. Furthermore, we found that miR-26b governed TR-mediate EMT partly due to governing its target Wee1. Notably, overexpression of miR-26b sensitized TR cells to TMZ. These findings suggest that upregulation of miR-26b or targeting Wee1 could serve as novel approaches to reverse chemotherapy resistance in glioma.
Collapse
Affiliation(s)
- Lixia Wang
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Jingna Su
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Zhe Zhao
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Yingying Hou
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Xuyuan Yin
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Nana Zheng
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Xiuxia Zhou
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China
| | - Jingzhe Yan
- b Department of Abdominal Oncosurgery , Jilin Province Cancer Hospital , Changchun , Jilin , China
| | - Jun Xia
- c Department of Biochemistry and Molecular Biology , Bengbu Medical College , Anhui , China
| | - Zhiwei Wang
- a The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology , Soochow University , Suzhou , China.,c Department of Biochemistry and Molecular Biology , Bengbu Medical College , Anhui , China.,d Department of Pathology , Beth Israel Deaconess Medical Center, Harvard Medical School , MA , USA
| |
Collapse
|
32
|
Cheng ZX, Yin WB, Wang ZY. MicroRNA-132 induces temozolomide resistance and promotes the formation of cancer stem cell phenotypes by targeting tumor suppressor candidate 3 in glioblastoma. Int J Mol Med 2017; 40:1307-1314. [PMID: 28901390 PMCID: PMC5627876 DOI: 10.3892/ijmm.2017.3124] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 08/10/2017] [Indexed: 12/01/2022] Open
Abstract
The prognosis of patients suffering from glioblastoma [also referred to as glioblastoma multiforme (GBM)] is dismal despite multimodal therapy. Chemotherapy with temozolomide may suppress tumor growth for a certain period of time (a few months); however, invariable tumor recurrence suggests that glioblastoma initiating cells (GICs) render these tumors persistant. Thus, the understanding of the molecular mechanisms of action of GICs as regards their role in the progression of GBM is important as such knowledge will be helpful in the discovery of novel drug targets, as well as in the design of novel therapeutic strategies for more effective treatment of the disease. In this study, we found that tumor suppressor candidate 3 (TUSC3) was downregulated in temozolomide-resistant U87MG cells (U87MG-res cells) and its restoration sensitized U87MG-res cells to temozolomide. TUSC3 was able to inhibit the formation of GIC phenotypes in the U87MG-res cells. The overexpression of microRNA (miR)-132 inhibited TUSC3 protein expression in the U87MG cells. However, its overexpression did not degrade TUSC3 mRNA expression in the cells. miR-132 was upregulated in the U87MG-res cells and its overexpression induced temozolomide resistance and the formation of cancer stem cell phenotypes in the U87MG cells. Thus, our data indicate that miR-132 induces temozolo-mide resistance and promotes the formation of cancer stem cell phenotypes by targeting TUSC3 in glioblastoma.
Collapse
Affiliation(s)
- Zhen-Xiu Cheng
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Wen-Bo Yin
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Zhong-Yu Wang
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
33
|
Lin X, Chang W, Wang Y, Tian M, Yu Z. SRPX2, an independent prognostic marker, promotes cell migration and invasion in hepatocellular carcinoma. Biomed Pharmacother 2017; 93:398-405. [DOI: 10.1016/j.biopha.2017.06.075] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/18/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
|
34
|
Shang J, Zhang Y, Jiang Y, Li Z, Duan Y, Wang L, Xiao J, Zhao Z. NOD2 promotes endothelial-to-mesenchymal transition of glomerular endothelial cells via MEK/ERK signaling pathway in diabetic nephropathy. Biochem Biophys Res Commun 2017; 484:435-441. [PMID: 28137583 DOI: 10.1016/j.bbrc.2017.01.155] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 01/26/2017] [Indexed: 12/22/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) of glomerular vascular endothelial cells (GEnCs) is now considered to play a critical role in diabetic nephropathy (DN). NOD2 is newly discovered to be closely related to DN renal injury. However, the relationship between NOD2 and EndMT of GEnCs has never been reported. In the present study, we found that NOD2 over-expression was positively correlated with the severity of DN injury in human renal biopsy samples. Immunohistochemical staining of DN renal slices showed gradual absence of endothelial character and gain of mesenchymal character, both of which were associated with NOD2 over-expression. In high glucose stimulated GEnCs, NOD2 was increased. What's more, over-expression and activation of NOD2 could both promote EndMT of GEnCs. On the other hand, silencing of NOD2 markedly attenuated EndMT induced by high glucose. Mechanically, we further found that MEK/ERK signaling pathway was involved in NOD2-regulated EndMT. Collectively, our results indicate that NOD2 has a regulatory role in EndMT via activation of MEK/ERK in high glucose-treated GEnCs. Targeting this pathway is a promising strategy for intervention of DN endothelial dysfunction.
Collapse
Affiliation(s)
- Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yumin Jiang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenzhen Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiqi Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luyao Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Xiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhanzheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
35
|
Liu K, Fan J, Wu J. Sushi repeat-containing protein X-linked 2 promotes angiogenesis through the urokinase-type plasminogen activator receptor dependent integrin αvβ3/focal adhesion kinase pathways. Drug Discov Ther 2017; 11:212-217. [DOI: 10.5582/ddt.2017.01017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Kuiliang Liu
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University
| | - Jianghao Fan
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University
| | - Jing Wu
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University
| |
Collapse
|