1
|
Ge Y, Ma S, Zhou Q, Xiong Z, Wang Y, Li L, Chao Z, Zhang J, Li T, Wu Z, Gao Y, Qu G, Xi Z, Liu B, Wu X, Wang Z. Oncogene goosecoid is transcriptionally regulated by E2F1 and correlates with disease progression in prostate cancer. Chin Med J (Engl) 2024; 137:1844-1856. [PMID: 37997674 PMCID: PMC12077560 DOI: 10.1097/cm9.0000000000002865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Although some well-established oncogenes are involved in cancer initiation and progression such as prostate cancer (PCa), the long tail of cancer genes remains to be defined. Goosecoid ( GSC ) has been implicated in cancer development. However, the comprehensive biological role of GSC in pan-cancer, specifically in PCa, remains unexplored. The aim of this study was to investigate the role of GSC in PCa development. METHODS We performed a systematic bioinformatics exploration of GSC using datasets from The Cancer Genome Atlas, Genotype-Tissue Expression, Gene Expression Omnibus, German Cancer Research Center, and our in-house cohorts. First, we evaluated the expression of GSC and its association with patient prognosis, and identified GSC -relevant genetic alterations in cancers. Further, we focused on the clinical characterization and prognostic analysis of GSC in PCa. To understand the transcriptional regulation of GSC by E2F transcription factor 1 ( E2F1 ), we performed chromatin immunoprecipitation quantitative polymerase chain reaction (qPCR). Functional experiments were conducted to validate the effect of GSC on the tumor cellular phenotype and sensitivity to trametinib. RESULTS GSC expression was elevated in various tumors and significantly correlated with patient prognosis. The alterations of GSC contribute to the progression of various tumors especially in PCa. Patients with PCa and high GSC expression exhibited worse progression-free survival and biochemical recurrence outcomes. Further, GSC upregulation in patients with PCa was mostly accompanied with higher Gleason score, advanced tumor stage, lymph node metastasis, and elevated prostate-specific antigen (PSA) levels. Mechanistically, the transcription factor, E2F1 , stimulates GSC by binding to its promoter region. Detailed experiments further demonstrated that GSC acted as an oncogene and influenced the response of PCa cells to trametinib treatment. CONCLUSIONS GSC was highly overexpressed and strongly correlated with patient prognosis in PCa. We found that GSC , regulated by E2F1 , acted as an oncogene and impeded the therapeutic efficacy of trametinib in PCa.
Collapse
Affiliation(s)
- Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, Qinghai 810001, China
| | - Zezhong Xiong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tengfei Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zixi Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuan Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guanyu Qu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zirui Xi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xi Wu
- Department of Urology, First Hospital of Laohekou City, Xiangyang, Hubei 441800, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
2
|
Wang W, Guo H, Wu S, Xian S, Zhang W, Zhang R, Chen Z, Su K, Zhang Y, Zhu Y, Chu D, Zhao M, Tang Z, Zheng C, Huang Z, Ma Q, Guo R. Construction of Metastasis-Specific Regulation Network in Ovarian Cancer Based on Prognostic Stemness-Related Signatures. Reprod Sci 2023; 30:2634-2654. [PMID: 36940084 DOI: 10.1007/s43032-022-01134-3] [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: 03/12/2022] [Accepted: 11/14/2022] [Indexed: 03/21/2023]
Abstract
WE aimed to reveal the correlation between ovarian cancer (OV) metastasis and cancer stemness in OV. RNA-seq data and clinical information of 591 OV samples (551 without metastasis and 40 with metastasis) were obtained from TCGA. The edgeR method was used to determine differentially expressed genes (DEGs) and transcription factors (DETFs). Then, mRNA expression-based stemness index was calculated using one-class logistic regression (OCLR). Weighted gene co-expression network analysis (WGCNA) was used to define stemness-related genes (SRGs). Univariate and multivariate Cox proportional hazard regression were conducted to identify the prognostic SRGs (PSRGs). PSRGs, DETFs, and 50 hallmark pathways quantified by gene set variation analysis (GSVA) were integrated into Pearson co-expression analysis. Significant co-expression interactions were utilized to construct an OV metastasis-specific regulation network. Cell communication analysis was carried out based on single cell RNA sequencing data to explore the molecular regulation mechanism of OV. Eventually, assay for targeting accessible-chromatin with high throughout sequencing (ATAC), chromatin immunoprecipitation sequencing (ChIP-seq) validation, and multiple data sets were used to validate the expression levels and prognostic values of key stemness-related signatures. Moreover, connectivity map (CMap) was used to identify potential inhibitors of stemness-related signatures. Based on edgeR, WGCNA, and Cox proportional hazard regression, 22 PSRGs were defined to construct a prognostic prediction model for metastatic OV. In the metastasis-specific regulation network, key TF-PSRS interaction pair was NR4A1-EGR3 (correlation coefficient = 0.81, p < 0.05, positive), and key PSRG-hallmark pathway interaction pair was EGR3-TNFα signaling via NFκB (correlation coefficient = 0.44, p < 0.05, positive), which were validated in multi-omics databases. Thioridazine was postulated to be the most significant compound in treatment of OV metastasis. PSRGs played critical roles in OV metastasis. Specifically, EGR3 was the most significant PSRG, which was positively regulated by DETF NR4A1, inducing metastasis via TNFα signaling.
Collapse
Affiliation(s)
- Wenwen Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Hongjun Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Shengyu Wu
- Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200092, China
| | - Shuyuan Xian
- Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200092, China
| | - Weiwei Zhang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Ruitao Zhang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Zhihua Chen
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Ke Su
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Ying Zhang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Ying Zhu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Danxia Chu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Mengling Zhao
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Zhihua Tang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Chunlan Zheng
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China.
| | - Qian Ma
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China.
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450052, China.
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Henan Province, Henan, 450052, China.
| |
Collapse
|
3
|
Yao H, Zhang S, Xie H, Fan Y, Miao M, Zhu R, Yuan L, Gu M, You Y, You B. RCN2 promotes Nasopharyngeal carcinoma progression by curbing Calcium flow and Mitochondrial apoptosis. Cell Oncol (Dordr) 2023; 46:1031-1048. [PMID: 36952101 PMCID: PMC10356900 DOI: 10.1007/s13402-023-00796-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 03/24/2023] Open
Abstract
OBJECTIVE Evidence suggests that calcium release from the endoplasmic reticulum (ER) can be induced to cause calcium overload, which in turn can trigger mitochondrial-dependent apoptosis. Dysregulation of systemic calcium homeostasis and changing levels of calcium-binding proteins have been shown to be associated with the malignant behavior of tumors. However, the precise molecular mechanism underlying Nasopharyngeal carcinoma (NPC) remains uncertain. METHODS Reticulocalbin (RCN2) expression in NPC was assessed using GEO database, western blot analysis and qRT-PCR. Apoptosis was assessed using flow cytometric analysis and the expression levels of apoptosis-related proteins were determined using western blot analysis. Intracellular calcium ion concentrations were measured using fluorescence imaging. The findings from these analyses were validated in vitro using nude mice models. Luciferase and ChIP assays were used to measure transcriptional regulation. Clinical significance was evaluated using tissue microarray analysis (n=150). RESULTS Our results showed that RCN2 promotes malignancy by causing Ca2+ flow imbalance, which leads to the initiation of the stress-mediated mitochondrial apoptosis pathway. We demonstrate that calreticulin (CALR) resides primarily in the endoplasmic reticulum and interacts with RCN2. Moreover, the transcription factors YY1 and homeobox protein goosecoid (GSC) both contribute to the initiation of RCN2 transcription by directly binding to the predicted promoter region of RCN2. Finally, high expression of RCN2 combined with high expression of GSC and YY1 may serve as an important clinical biomarker of poor prognosis in patients with NPC. CONCLUSION YY1 and GSC are upstream regulators of RCN2, involved in mitochondrial calcium overload and stress-induced mitochondrial apoptosis. Thus, they can play significant role in the malignant development of NPCs.
Collapse
Affiliation(s)
- Hui Yao
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
- Changhai Hospital of Shanghai, No. 168 Changhai Road, Shanghai, 200433, China
| | - Siyu Zhang
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Haijing Xie
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Yue Fan
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Mengyu Miao
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Rui Zhu
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Ling Yuan
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Miao Gu
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Medical College of Nantong University, Nantong, 226019, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Medical College of Nantong University, Nantong, 226019, China.
| | - Bo You
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Institute of Otolaryngology head and neck surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
- Medical College of Nantong University, Nantong, 226019, China.
| |
Collapse
|
4
|
Conteduca G, Cangelosi D, Coco S, Malacarne M, Baldo C, Arado A, Pinto R, Testa B, Coviello DA. NSD1 Mutations in Sotos Syndrome Induce Differential Expression of Long Noncoding RNAs, miR646 and Genes Controlling the G2/M Checkpoint. Life (Basel) 2022; 12:life12070988. [PMID: 35888078 PMCID: PMC9324496 DOI: 10.3390/life12070988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/16/2022] Open
Abstract
An increasing amount of evidence indicates the critical role of the NSD1 gene in Sotos syndrome (SoS), a rare genetic disease, and in tumors. Molecular mechanisms affected by NSD1 mutations are largely uncharacterized. In order to assess the impact of NSD1 haploinsufficiency in the pathogenesis of SoS, we analyzed the gene expression profile of fibroblasts isolated from the skin samples of 15 SoS patients and of 5 healthy parents. We identified seven differentially expressed genes and five differentially expressed noncoding RNAs. The most upregulated mRNA was stratifin (SFN) (fold change, 3.9, Benjamini−Hochberg corrected p < 0.05), and the most downregulated mRNA was goosecoid homeobox (GSC) (fold change, 3.9, Benjamini−Hochberg corrected p < 0.05). The most upregulated lncRNA was lnc-C2orf84-1 (fold change, 4.28, Benjamini−Hochberg corrected p < 0.001), and the most downregulated lncRNA was Inc-C15orf57 (fold change, −0.7, Benjamini−Hochberg corrected p < 0.05). A gene set enrichment analysis reported the enrichment of genes involved in the KRAS and E2F signaling pathways, splicing regulation and cell cycle G2/M checkpoints. Our results suggest that NSD1 is involved in cell cycle regulation and that its mutation can induce the down-expression of genes involved in tumoral and neoplastic differentiation. The results contribute to defining the role of NSD1 in fibroblasts for the prevention, diagnosis and control of SoS.
Collapse
Affiliation(s)
- Giuseppina Conteduca
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
| | - Davide Cangelosi
- Clinical Bioinformatics Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| | - Michela Malacarne
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
| | - Chiara Baldo
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
| | - Alessia Arado
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
| | - Rute Pinto
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
| | - Barbara Testa
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
| | - Domenico A. Coviello
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (G.C.); (M.M.); (C.B.); (A.A.); (R.P.); (B.T.)
- Correspondence: ; Tel.: +39-010-5636-3977
| |
Collapse
|
5
|
Ertekin Ö, Monavari M, Krüger R, Fuentes-Chandía M, Parma B, Letort G, Tripal P, Boccaccini AR, Bosserhoff AK, Ceppi P, Kappelmann-Fenzl M, Leal-Egaña A. 3D hydrogel-based microcapsules as an in vitro model to study tumorigenicity, cell migration and drug resistance. Acta Biomater 2022; 142:208-220. [PMID: 35167953 DOI: 10.1016/j.actbio.2022.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/06/2022] [Accepted: 02/09/2022] [Indexed: 02/06/2023]
Abstract
In this work, we analyzed the reliability of alginate-gelatin microcapsules as artificial tumor model. These tumor-like scaffolds are characterized by their composition and stiffness (∼25 kPa), and their capability to restrict -but not hinder- cell migration, proliferation and release from confinement. Hydrogel-based microcapsules were initially utilized to detect differences in mechano-sensitivity between MCF7 and MDA-MB-231 breast cancer cells, and the endothelial cell line EA.hy926. Additionally, we used RNA-seq and transcriptomic methods to determine how the culture strategy (i.e. 2D v/s 3D) may pre-set the expression of genes involved in multidrug resistance, being then validated by performing cytotoxicological tests and assays of cell morphology. Our results show that both breast cancer cells can generate elongated multicellular spheroids inside the microcapsules, prior being released (mimicking intravasation stages), a behavior which was not observed in endothelial cells. Further, we demonstrate that cells isolated from 3D scaffolds show resistance to cisplatin, a process which seems to be strongly influenced by mechanical stress, instead of hypoxia. We finally discuss the role played by aneuploidy in malignancy and resistance to anticancer drugs, based on the increased number of polynucleated cells found within these microcapsules. Overall, our outcomes demonstrate that alginate-gelatin microcapsules represent a simple, yet very accurate tumor-like model, enabling us to mimic the most relevant malignant hints described in vivo, suggesting that confinement and mechanical stress need to be considered when studying pathogenicity and drug resistance of cancer cells in vitro. STATEMENT OF SIGNIFICANCE: In this work, we analyzed the reliability of alginate-gelatin microcapsules as an artificial tumor model. These scaffolds are characterized by their composition, elastic properties, and their ability to restrict cell migration, proliferation, and release from confinement. Our results demonstrate four novel outcomes: (i) studying cell migration and proliferation in 3D enabled discrimination between malignant and non-pathogenic cells, (ii) studying the cell morphology of cancer aggregates entrapped in alginate-gelatin microcapsules enabled determination of malignancy degree in vitro, (iii) determination that confinement and mechanical stress, instead of hypoxia, are required to generate clones resistant to anticancer drugs (i.e. cisplatin), and (iv) evidence that resistance to anticancer drugs could be due to the presence of polynucleated cells localized inside polymer-based artificial tumors.
Collapse
Affiliation(s)
- Özlem Ertekin
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Diagno Biotechnology, Marmara Technopark, Gebze, Kocaeli, Turkey
| | - Mahshid Monavari
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - René Krüger
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, and University Clinics Erlangen, Erlangen 91054, Germany
| | - Miguel Fuentes-Chandía
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH, USA
| | - Beatrice Parma
- Interdisciplinary Center for Clinical Research (IZKF), Friedrich-Alexander Universität Erlangen-Nürnberg Glueckstrasse 6, Erlangen 91054, Germany
| | - Gaelle Letort
- Center for Interdisciplinary Research in Biology, Collège de France UMR7241/U1050, 11, Place Marcelin Berthelot, Paris 75231 CEDEX 05, France
| | - Philipp Tripal
- Optical Imaging Centre Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 3, Erlangen 91058, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Anja K Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Fahrstraße 17, Erlangen 91054, Germany
| | - Paolo Ceppi
- Interdisciplinary Center for Clinical Research (IZKF), Friedrich-Alexander Universität Erlangen-Nürnberg Glueckstrasse 6, Erlangen 91054, Germany; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense DK-5230, Denmark
| | - Melanie Kappelmann-Fenzl
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Fahrstraße 17, Erlangen 91054, Germany; Faculty of Applied Informatics, University of Applied Science Deggendorf, Deggendorf 94469, Germany
| | - Aldo Leal-Egaña
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Institute for Molecular Systems Engineering, University of Heidelberg. INF 253, Heidelberg 69120, Germany.
| |
Collapse
|
6
|
Ding X, Liu H, Yuan Y, Zhong Q, Zhong X. Roles of GFPT2 Expression Levels on the Prognosis and Tumor Microenvironment of Colon Cancer. Front Oncol 2022; 12:811559. [PMID: 35330716 PMCID: PMC8940194 DOI: 10.3389/fonc.2022.811559] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/16/2022] [Indexed: 01/02/2023] Open
Abstract
Background Recently, increasing evidence has suggested that Glutamine-fructose-6-phosphate transaminase 2 (GFPT2) is related to carcinogenesis. However, the potential roles of GFPT2 in colon cancer still need to be fully investigated. Methods We examined the protein levels of GFPT2 by immunohistochemistry (IHC) in tissues collected from 83 patients with colon cancer. We further detected GFBPT2 protein levels by Western Blot assay. We checked the relationship between GFPT2 expression levels and overall survival (OS), stromal and immune scores and immune components from The Cancer Gene Atlas (TCGA) database. GFBP2-related pathways were validated in the Cancer Cell Line Encyclopedia (CCLE) database. Expression of GFPT2 in single cell subpopulations was calculated from The Tumor Immune Single Cell Center (TISCH). The levels of GFPT2 and drug sensitivity data were performed from CellMiner dataset. Results GFPT2 was highly expressed and correlated with poor pathological features in 83 colon cancer patients. Moreover, increased GFPT2 expression was significantly associated with poorer OS in 329 colon adenocarcinoma (COAD) patients. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed the differentially expressed genes of GFPT2 were mostly enriched in focal adhesion, ECM receptor interaction, JAK/STAT signaling pathway and immune related pathways. In addition, GFPT2 expression was correlated with the tumor microenvironment (TME). GFPT2 expression was linked to cancer-associated fibroblasts (CAFs)-associated factors and epithelial-mesenchymal transition (EMT)-related factors. GFPT2 was positively correlated with immunosuppressive cells and regulated immunosuppressive factors and T-cell exhaustion. Finally, our data suggested that the expression of GFPT2 may be a judgment of the sensitivity of a certain class of drugs. Conclusions Our work reveals the roles of GFPT2 in tumorigenesis, particularly in immune response, TME and drug resistance, which are crucial for the development of customized cancer therapies.
Collapse
Affiliation(s)
- Xiaorong Ding
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hua Liu
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Ying Yuan
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Qin Zhong
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Xiaomin Zhong
- Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
7
|
Bose M, Grover P, Sanders AJ, Zhou R, Ahmad M, Shwartz S, Lala P, Nath S, Yazdanifar M, Brouwer C, Mukherjee P. Overexpression of MUC1 Induces Non-Canonical TGF-β Signaling in Pancreatic Ductal Adenocarcinoma. Front Cell Dev Biol 2022; 10:821875. [PMID: 35237602 PMCID: PMC8883581 DOI: 10.3389/fcell.2022.821875] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal human cancers. Transforming Growth Factor Beta (TGF-β) is a cytokine that switches from a tumor-suppressor at early stages to a tumor promoter in the late stages of tumor development, by yet unknown mechanisms. Tumor associated MUC1 is aberrantly glycosylated and overexpressed in >80% of PDAs and is associated with poor prognosis. MUC1 expression is found in the early stages of PDA development with subsequent increase in later stages. Analysis of human PDA samples from TCGA database showed significant differences in gene expression and survival profiles between low and high MUC1 samples. Further, high MUC1 expression was found to positively correlate to TGF-βRII expression and negatively correlate to TGF-βRI expression in PDA cell lines. We hypothesized that MUC1 overexpression induces TGF-β mediated non-canonical signaling pathways which is known to be associated with poor prognosis. In this study, we report that MUC1 overexpression in PDA cells directly activates the JNK pathway in response to TGF-β, and leads to increased cell viability via up-regulation and stabilization of c-Myc. Conversely, in low MUC1 expressing PDA cells, TGF-β preserves its tumor-suppressive function and inhibits phosphorylation of JNK and stabilization of c-Myc. Knockdown of MUC1 in PDA cells also results in decreased phosphorylation of JNK and c-Myc in response to TGF-β treatment. Taken together, the results indicate that overexpression of MUC1 plays a significant role in switching the TGF-β function from a tumor-suppressor to a tumor promoter by directly activating JNK. Lastly, we report that high-MUC1 PDA tumors respond to TGF-β neutralizing antibody in vivo showing significantly reduced tumor growth while low-MUC1 tumors do not respond to TGF-β neutralizing antibody further confirming our hypothesis.
Collapse
Affiliation(s)
- Mukulika Bose
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | - Priyanka Grover
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | - Alexa J. Sanders
- Department of Bioinformatics, UNC Charlotte, Charlotte, NC, United States
| | - Ru Zhou
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | - Mohammad Ahmad
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | - Sophia Shwartz
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | - Priyanka Lala
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | - Sritama Nath
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| | | | - Cory Brouwer
- Department of Bioinformatics, UNC Charlotte, Charlotte, NC, United States
| | - Pinku Mukherjee
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States
| |
Collapse
|
8
|
Lou T, Liu C, Qu H, Zhang Z, Wang S, Zhuang H. FOXA1 can be modulated by HDAC3 in the progression of epithelial ovarian carcinoma. J Transl Med 2022; 20:19. [PMID: 34991620 PMCID: PMC8740004 DOI: 10.1186/s12967-021-03224-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022] Open
Abstract
FOXA1 is associated with malignant tumors, but the function of FOXA1 in EOC is unclear. HDAC3 can influence the proliferation, migration and invasion ability of EOC. In this study, we wanted to explore the function of FOXA1 in ovarian cancer and the relationship between HDAC3 and FOXA1.The expression of HDAC3 and FOXA1 was detected by immunohistochemical staining of primary lesions from 127 epithelial ovarian carcinoma patients. A proliferation assay, a Transwell assay, an apoptosis assay and animal experiments were used to assess the proliferation, invasion and apoptosis abilities of ovarian cancer cells before and after transfection with FOXA1. The relevance of the in vitro findings was confirmed in xenografts. The H-scores for FOXA1 and HDAC3 staining in FIGO stage III-IV were noticeably higher and predicted adverse clinical outcomes in patients with ovarian cancer. The expression level of HDAC3 was significantly correlated with the expression level of FOXA1. Invasion, proliferation and apoptosis capacity and tumor formation were decreased in the FOXA1-knockdown cells. Experiments in xenografts confirmed that HDAC3 mediated tumor formation. In conclusion, FOXA1 can be modulated by HDAC3 through the Wnt/β-catenin signaling pathway, and FOXA1 plays essential roles in the proliferation, apoptosis and invasion of EOC cell lines and xenograft experiments.
Collapse
Affiliation(s)
- Tong Lou
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Chongdong Liu
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Hong Qu
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Zhiqiang Zhang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China
| | - Huiyu Zhuang
- Department of Obstetrics and Gynecology, Beijing Chaoyang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
9
|
Pretzsch E, Lampert C, Bazhin AV, Link H, Jacob S, Guba M, Werner J, Neumann J, Angele MK, Bösch F. EMT-related genes are unlikely to be involved in extracapsular growth of lymph node metastases in gastric cancer. Pathol Res Pract 2021; 229:153688. [PMID: 34872022 DOI: 10.1016/j.prp.2021.153688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND In gastric cancer (GC), extracapsular growth (ECG) pattern of lymph node metastases is associated with decreased overall survival rates compared to intracapsular lymph node metastases (ICG). Epithelial-to-mesenchymal transition (EMT) plays a pivotal role in hematogenous metastatic spread. Aim of the present study was to analyze if EMT related genes are involved in the growth pattern of lymph node metastases in GC. METHODS Out of our prospective database with 529 patients who underwent surgical resection for GC between 2002 and 2014 forty lymph node positive patients were identified (20 ECG, 20 ICG). The expression of 84 EMT-associated genes were analyzed by RT2 Profiler PCR Array (n = 20). Results were validated by Real-Time PCR (n = 20). RESULTS GC with ECG showed differently expressed EMT related genes. GC leading to ECG showed an upregulation of three and downregulation of eleven genes. Those differences, however, could not be confirmed in PCR analysis. CONCLUSIONS This study demonstrates that EMT related genes are not responsible for the different growth patterns of lymph node metastases in GC. Further studies are required to evaluate the underlying mechanisms of ECG in GC as it might provide a potential therapeutic target for this subgroup of more aggressive tumors in the future.
Collapse
Affiliation(s)
- Elise Pretzsch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christopher Lampert
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Helena Link
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sven Jacob
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Markus Guba
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Neumann
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin K Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Florian Bösch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
10
|
Dratwa M, Wysoczańska B, Łacina P, Kubik T, Bogunia-Kubik K. TERT-Regulation and Roles in Cancer Formation. Front Immunol 2020; 11:589929. [PMID: 33329574 PMCID: PMC7717964 DOI: 10.3389/fimmu.2020.589929] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is a catalytic subunit of telomerase. Telomerase complex plays a key role in cancer formation by telomere dependent or independent mechanisms. Telomere maintenance mechanisms include complex TERT changes such as gene amplifications, TERT structural variants, TERT promoter germline and somatic mutations, TERT epigenetic changes, and alternative lengthening of telomere. All of them are cancer specific at tissue histotype and at single cell level. TERT expression is regulated in tumors via multiple genetic and epigenetic alterations which affect telomerase activity. Telomerase activity via TERT expression has an impact on telomere length and can be a useful marker in diagnosis and prognosis of various cancers and a new therapy approach. In this review we want to highlight the main roles of TERT in different mechanisms of cancer development and regulation.
Collapse
Affiliation(s)
- Marta Dratwa
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Barbara Wysoczańska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Tomasz Kubik
- Department of Computer Engineering, Faculty of Electronics, Wrocław University of Science and Technology, Wroclaw, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
11
|
Shen C, Li J, Chang S, Che G. [Advancement of E2F1 in Common Tumors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:921-926. [PMID: 33070516 PMCID: PMC7583875 DOI: 10.3779/j.issn.1009-3419.2020.101.32] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
细胞周期相关转录因子E2F1(E2F transcription factor 1)是细胞周期相关转录因子E2F家族成员之一,主要参与包括细胞周期进展、DNA修复、DNA复制、细胞分化,增殖和凋亡等多种细胞过程。E2F1在全身多种肿瘤组织和细胞中呈高表达,起着促癌基因的作用,E2F1表达上调与肿瘤的发生、发展、转移及预后密切相关。因此,E2F1有望成为肿瘤治疗的新靶点。本文就E2F1在目前常见肿瘤中的最新研究进展做一综述。
Collapse
Affiliation(s)
- Cheng Shen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jue Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuai Chang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Yan B, Xiong C, Huang F, Zhang M, Mo Y, Bai H. Big data-based identification of methylated genes associated with drug resistance and prognosis in ovarian cancer. Medicine (Baltimore) 2020; 99:e20802. [PMID: 32629664 PMCID: PMC7337574 DOI: 10.1097/md.0000000000020802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
It is imperative to further the understanding of the drug resistance mechanisms of ovarian cancer (OC) and to identify useful biological markers for prognosis prediction.Cormine, cBioportal, and The Cancer Genome Atlas databases were used to search microarray data of gene methylation related to OC, drug resistance in OC, and prognosis, and to analyze methylated genes potentially inducing the drug resistance in OC. Fifty-five DNA-methylated genes significantly associated with drug resistance in OC were screened, and the regulatory mechanisms underlying changes in methylation levels of these genes were systematically integrated.Enrichment and annotation of biological processes indicated that most of the above DNA-methylated genes were significantly associated with cell proliferation and cell cycle. In addition, pathway enrichment demonstrated that the above DNA-methylated genes were significantly associated with PI3K-AKT and P53 signaling pathways. Among the 55 genes, 4 were significantly associated with OC prognostic disease-free survival, namely bromodomain containing 4, PDZ domain containing 1 (PDZK1), phosphatase and tensin homolog, and TNF receptor superfamily member 10c; 5 were significantly related to overall survival, namely bromodomain containing 4, PDZK1, PIK3C2B, Rh associated glycoprotein, and DYRK; among them, the degree of methylation of TNF receptor superfamily member 10c, PDZK1, and Rh associated glycoprotein genes was significantly correlated with mRNA expression. Furthermore, PDZK1, Rh associated glycoprotein, and TNF receptor superfamily member 10c genes showed significant hypomethylation in drug-resistance tissues of OC, and their mRNAs had significantly high expression.The association between the methylation of these 55 genes and OC and drug resistance in OC, in addition to bioinformatics analyses clarify the important mechanisms of gene methylation in the development, progression, and drug resistance of OC.
Collapse
|
13
|
Extracellular Matrix Alterations in Metastatic Processes. Int J Mol Sci 2019; 20:ijms20194947. [PMID: 31591367 PMCID: PMC6802000 DOI: 10.3390/ijms20194947] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of extracellular-secreted macromolecules, such as collagen, enzymes and glycoproteins, whose main functions deal with structural scaffolding and biochemical support of cells and tissues. ECM homeostasis is essential for organ development and functioning under physiological conditions, while its sustained modification or dysregulation can result in pathological conditions. During cancer progression, epithelial tumor cells may undergo epithelial-to-mesenchymal transition (EMT), a morphological and functional remodeling, that deeply alters tumor cell features, leading to loss of epithelial markers (i.e., E-cadherin), changes in cell polarity and intercellular junctions and increase of mesenchymal markers (i.e., N-cadherin, fibronectin and vimentin). This process enhances cancer cell detachment from the original tumor mass and invasiveness, which are necessary for metastasis onset, thus allowing cancer cells to enter the bloodstream or lymphatic flow and colonize distant sites. The mechanisms that lead to development of metastases in specific sites are still largely obscure but modifications occurring in target tissue ECM are being intensively studied. Matrix metalloproteases and several adhesion receptors, among which integrins play a key role, are involved in metastasis-linked ECM modifications. In addition, cells involved in the metastatic niche formation, like cancer associated fibroblasts (CAF) and tumor associated macrophages (TAM), have been found to play crucial roles in ECM alterations aimed at promoting cancer cells adhesion and growth. In this review we focus on molecular mechanisms of ECM modifications occurring during cancer progression and metastatic dissemination to distant sites, with special attention to lung, liver and bone. Moreover, the functional role of cells forming the tumor niche will also be reviewed in light of the most recent findings.
Collapse
|
14
|
Galaine J, Turco C, Vauchy C, Royer B, Mercier-Letondal P, Queiroz L, Loyon R, Mouget V, Boidot R, Laheurte C, Lakkis Z, Jary M, Adotévi O, Borg C, Godet Y. CD4 T cells target colorectal cancer antigens upregulated by oxaliplatin. Int J Cancer 2019; 145:3112-3125. [PMID: 31396953 DOI: 10.1002/ijc.32620] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Immune checkpoint blockade has proven its efficacy in hypermutated subtypes of metastatic colorectal cancers (mCRC). Immunogenic potential can also be observed with conventional chemotherapies, but this property has never been explored thoroughly in CRC patients. The CRC therapeutic arsenal includes oxaliplatin, a well-characterized platinum drug already described as immunogenic. Here, we investigated the impact of the oxaliplatin-based treatment on mCRC immunopeptidome. We demonstrated that oxaliplatin-resistant CRC cell lines overexpressed telomerase reverse transcriptase (TERT), colorectal-associated-tumor antigen-1 (COA-1) and mesothelin tumor-associated antigens. We identified new HLA class-II-restricted and promiscuous peptides derived from COA-1 and mesothelin. The two naturally processed peptides COA-1331-345 and Meso366-380 appear to be the most immunogenic in mCRC patients. A prospective cohort of 162 mCRC patients enabled us to explore the impact of oxaliplatin exposure on the antitumor-specific immune response. Interestingly, chemotherapy-naive mCRC patients present high immune CD4 T-cell responses directed against TERT, COA-1 and mesothelin-derived peptides. These antitumor T-cell responses were maintained after 3 months of oxaliplatin-based treatment. Altogether, these findings highlight the interest of immunostimulatory agents to improve the management of chemoresistant mCRC patients. Finally, the high frequency of immune responses targeting the new immunogenic peptides derived from COA-1 and mesothelin support their use in immunomonitoring strategies.
Collapse
Affiliation(s)
- Jeanne Galaine
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Célia Turco
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Gastrointestinal Surgery, Besançon, France
| | - Charline Vauchy
- INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center un Biotherapy, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | - Bernard Royer
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of pharmacotoxicology, Besançon, France
| | - Patricia Mercier-Letondal
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Lise Queiroz
- INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center un Biotherapy, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | - Romain Loyon
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Virginie Mouget
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Romain Boidot
- Centre Georges-François Leclerc, Platform for Transfer to Cancer Biology, Dijon, France
| | - Caroline Laheurte
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,EFS Bourgogne Franche-Comté, INSERM CIC-1431, CHRU Besançon, Plateforme de BioMonitoring, Besançon, France
| | - Zaher Lakkis
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Gastrointestinal Surgery, Besançon, France
| | - Marine Jary
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Olivier Adotévi
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Christophe Borg
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Yann Godet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| |
Collapse
|
15
|
Avin BA, Wang Y, Gilpatrick T, Workman RE, Lee I, Timp W, Umbricht CB, Zeiger MA. Characterization of human telomerase reverse transcriptase promoter methylation and transcription factor binding in differentiated thyroid cancer cell lines. Genes Chromosomes Cancer 2019; 58:530-540. [PMID: 30664813 PMCID: PMC6621557 DOI: 10.1002/gcc.22735] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/04/2019] [Accepted: 01/14/2019] [Indexed: 12/27/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) activation plays an important role in cancer development by enabling the immortalization of cells. TERT regulation is multifaceted, and its promoter methylation has been implicated in controlling expression through alteration in transcription factor binding. We have characterized TERT promoter methylation, transcription factor binding, and TERT expression levels in five differentiated thyroid cancer (DTC) cell lines and six normal thyroid tissue samples by targeted bisulfite sequencing, ChIP-qPCR, and qRT-PCR. DTC cell lines express varying levels of TERT and exhibit TERT promoter methylation patterns similar to patterns seen in other telomerase positive cancer cell lines. The minimal promoter immediately surrounding the transcription start site is hypomethylated, while further upstream portions show dense methylation. In contrast, the TERT promoter in normal thyroid tissue is largely unmethylated throughout and expresses TERT minimally. Transcription factor binding is also affected by TERT mutation status. The E-twenty-six (ETS) factor GABPA exhibits TERT binding in the TERT mutant DTC cells only, and allele-specific methylation patterns at the minimal promoter were observed as well, which may indicate allele-specific factor recruitment at the minimal promoter. Furthermore, we identified binding sites for activators MYC and GSC in the hypermethylated upstream region, pointing to its possible importance in TERT regulation. Overall, TERT expression and telomerase activity depend on the interplay of multiple regulatory mechanisms including TERT promoter methylation, mutation status, and recruitment of transcription factors. This work explores of the interplay between these regulatory mechanisms and offers insight into cellular control of active telomerase in human cancer.
Collapse
Affiliation(s)
- Brittany A. Avin
- Department of Surgery, Johns Hopkins University, Baltimore, MD, United States 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Yongchun Wang
- Department of Surgery, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Timothy Gilpatrick
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Rachael E. Workman
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Isac Lee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Christopher B. Umbricht
- Department of Surgery, Johns Hopkins University, Baltimore, MD, United States 21287
- Department of Oncology, Johns Hopkins University, Baltimore, MD, United States 21287
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States 21287
| | - Martha A. Zeiger
- Department of Surgery, The University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
16
|
Cui R, Cao G, Bai H, Zhang Z. LPAR1 regulates the development of intratumoral heterogeneity in ovarian serous cystadenocarcinoma by activating the PI3K/AKT signaling pathway. Cancer Cell Int 2019; 19:201. [PMID: 31384176 PMCID: PMC6664705 DOI: 10.1186/s12935-019-0920-0] [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: 03/31/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023] Open
Abstract
Background To explore the role of lysophosphatidic acid receptor 1 (LPAR1) and its correlation with the PI3K/AKT pathway in the development of intratumoral heterogeneity (ITH) in human ovarian serous cystadenocarcinoma (OSC). Methods Immunohistochemical staining was performed to detect LPAR1 expression in matched primary and recurrent lesions from the same patients. Cell models of ITH were established using the limiting dilution methodology and Transwell invasion/migration assays. LPAR1 expression in the ITH cell models was silenced or upregulated with lentiviral particles, and the biological characteristics were evaluated using various in vitro and in vivo assessments of cell function. The levels of phosphorylated PI3K/AKT (p-PI3K/p-AKT) in LPAR1 knockdown and LPAR1-overexpressing cells were detected. Results The H-scores for LPAR1 staining in the lymphatic metastatic and recurrent lesions were noticeably higher than in the primary tumor lesions from the same patients (P = 0.024/0.031). High LPAR1 expression was associated with worse progression-free survival and overall survival (P = 0.017/0.039). Biological functions in vitro, including invasion, migration, and proliferation, and tumor formation in vivo were decreased in the LPAR1-silenced cells (all P < 0.05). These cellular functions were significantly increased in the LPAR1-overexpressing cells in vitro and in vivo (all P < 0.05). The levels of p-PI3K and p-AKT were significantly decreased in the LPAR1 knockdown cells and significantly increased in the LPAR1-overexpressing cells (all P < 0.05). Conclusions Higher levels of the LPAR1 protein were associated with a poor prognosis. LPAR1 plays essential roles in the invasion, migration, and proliferation of heterogeneous subsets of OSC cell lines and the development of ITH of OSC, possibly by modulating the activity of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ran Cui
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, No.8, North Road of Workers Stadium, Chaoyang District, Beijing, 100020 China
| |
Collapse
|
17
|
Pokhriyal R, Hariprasad R, Kumar L, Hariprasad G. Chemotherapy Resistance in Advanced Ovarian Cancer Patients. BIOMARKERS IN CANCER 2019; 11:1179299X19860815. [PMID: 31308780 PMCID: PMC6613062 DOI: 10.1177/1179299x19860815] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 05/08/2019] [Indexed: 12/26/2022]
Abstract
Ovarian cancer is the seventh most common gynaecologic malignancy seen in women. Majority of the patients with ovarian cancer are diagnosed at the advanced stage making prognosis poor. The standard management of advanced ovarian cancer includes tumour debulking surgery followed by chemotherapy. Various types of chemotherapeutic regimens have been used to treat advanced ovarian cancer, but the most promising and the currently used standard first-line treatment is carboplatin and paclitaxel. Despite improved clinical response and survival to this combination of chemotherapy, numerous patients either undergo relapse or succumb to the disease as a result of chemotherapy resistance. To understand this phenomenon at a cellular level, various macromolecules such as DNA, messenger RNA and proteins have been developed as biomarkers for chemotherapy response. This review comprehensively summarizes the problem that pertains to chemotherapy resistance in advanced ovarian cancer and provides a good overview of the various biomarkers that have been developed in this field.
Collapse
Affiliation(s)
- Ruchika Pokhriyal
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Roopa Hariprasad
- Division of Clinical Oncology, National Institute of Cancer Prevention and Research, Noida, India
| | - Lalit Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
18
|
Guha Majumdar A, Subramanian M. Hydroxychavicol from Piper betle induces apoptosis, cell cycle arrest, and inhibits epithelial-mesenchymal transition in pancreatic cancer cells. Biochem Pharmacol 2019; 166:274-291. [PMID: 31154000 DOI: 10.1016/j.bcp.2019.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer is a major cause of cancer-related mortality around the world. Currently, options for diagnosis and treatment are extremely limited, which culminates in a very high mortality rate. Intensive research spanning more than four decades has met several roadblocks in terms of improvement in overall survival. In this study, we have evaluated the effect of Hydroxychavicol (HC), a naturally occurring and abundantly isolatable allylarene from Piper betle leaves on pancreatic cancer cells. Our investigation reveals that HC inhibits proliferation and epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. HC induces DNA damage, as evidenced by γ-H2AX, 53BP1 induction and comet assay, which further results in mitotic catastrophe and apoptosis. The apoptosis induced by HC is JNK pathway-dependent and caspase-mediated. HC also inhibits migration and invasion of pancreatic cancer cells via a generalized repression of genes involved in EMT. A quantitative real time PCR-based array revealed at least 14 different genes to be differentially expressed upon HC treatment in pancreatic cancer cells. These results show significant potential of HC as an anticancer agent against pancreatic cancer.
Collapse
Affiliation(s)
- Ananda Guha Majumdar
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Mahesh Subramanian
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
19
|
Miksiunas R, Mobasheri A, Bironaite D. Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac Development, Degeneration and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:155-178. [PMID: 30945165 DOI: 10.1007/5584_2019_349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are the most common cause of human death in the developing world. Extensive evidence indicates that various toxic environmental factors and unhealthy lifestyle choices contribute to the risk, incidence and severity of cardiovascular diseases. Alterations in the genetic level of myocardium affects normal heart development and initiates pathological processes leading to various types of cardiac diseases. Homeobox genes are a large and highly specialized family of closely related genes that direct the formation of body structure, including cardiac development. Homeobox genes encode homeodomain proteins that function as transcription factors with characteristic structures that allow them to bind to DNA, regulate gene expression and subsequently control the proper physiological function of cells, tissues and organs. Mutations in homeobox genes are rare and usually lethal with evident alterations in cardiac function at or soon after the birth. Our understanding of homeobox gene family expression and function has expanded significantly during the recent years. However, the involvement of homeobox genes in the development of human and animal cardiac tissue requires further investigation. The phenotype of human congenital heart defects unveils only some aspects of human heart development. Therefore, mouse models are often used to gain a better understanding of human heart function, pathology and regeneration. In this review, we have focused on the role of homeobox genes in the development and pathology of human heart as potential tools for the future development of targeted regenerative strategies for various heart malfunctions.
Collapse
Affiliation(s)
- Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| |
Collapse
|
20
|
Wen X, Wang B, Feng T, Yuan W, Zhou J, Fang T. TNF receptor-associated factor 1 as a biomarker for assessment of non-small cell lung cancer metastasis and overall survival. CLINICAL RESPIRATORY JOURNAL 2018. [PMID: 29528567 DOI: 10.1111/crj.12789] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Non-small cell lung cancer (NSCLC), which comprises 80%-85% of all lung cancer cases, is one of the most common human malignancies. Despite great improvements in diagnostic technology and the introduction of new therapeutic agents in recent years, the 5-year survival rate of NSCLC is still low. Tumor necrosis factor (TNF) receptor-associated factor 1 (TRAF1) plays an important role in the TNF-related apoptosis-inducing ligand (TRAIL) associated signal pathway. METHODS In this study, we aim to illuminate the function of TRAF1 in NSCLC. Toward that end, TRAF1 expression was detected using immunohistochemistry (IHC) in specimens from 200 NSCLC patients. The function of TRAF1 in the A549 and H1299 cell lines was evaluated by colony formation and MTT assays. RESULTS Our data showed that TRAF1 was significantly upregulated in NSCLC tissues. TRAF1 expression was positively associated with NSCLC lymphatic metastasis and clinical stage and was negatively associated with overall patient survival. TRAF1 promoted NSCLC cell proliferation CONCLUSION: TRAF1 expression was positively associated with NSCLC lymphatic metastasis and histological grade and was negatively associated with overall patient survival. TRAF1 may be an important therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Xiaoxing Wen
- Department of Pulmonary Medicine, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| | - Bingping Wang
- Department of Oncology, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| | - Tao Feng
- Department of Pulmonary Medicine, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| | - Wei Yuan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Fang
- Department of Oncology, Shengli Oilfield Central Hospital, Dongying, Shandong Province, China
| |
Collapse
|
21
|
Abstract
The majority of cancer-related deaths result from metastasis, the process by which cancer cells escape the primary tumor site and enter into the blood circulation in order to disseminate to secondary locations throughout the body. Tumor cells found within the circulation are referred to as circulating tumor cells (CTCs), and their detection and enumeration correlate with poor prognosis. The epithelial-to-mesenchymal transition (EMT) is a dynamic process that imparts epithelial cells with mesenchymal-like properties, thus facilitating tumor cell dissemination and contributing to metastasis. However, EMT also results in the downregulation of various epithelial proteins typically utilized by CTC technologies for enrichment and detection of these rare cells, resulting in reduced detection of some CTCs, potentially those with a more metastatic phenotype. In addition to the current clinical role of CTCs as a prognostic biomarker, they also have potential as a predictive biomarker via CTC characterization. However, CTC characterization is complicated by the unknown biological significance of CTCs possessing an EMT-like phenotype, and the ability to capture and understand this CTC subpopulation is an essential step in the utilization of CTCs for patient management. This chapter will review the process of EMT and its contribution to metastasis; discusses current and future clinical applications of CTCs; and describes both traditional and novel methods for CTC enrichment, detection, and characterization with a specific focus on CTCs with an EMT phenotype.
Collapse
|
22
|
EMT promoting transcription factors as prognostic markers in human breast cancer. Arch Gynecol Obstet 2017; 295:817-825. [DOI: 10.1007/s00404-017-4304-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
|
23
|
Yamamoto H, Ryu J, Min E, Oi N, Bai R, Zykova TA, Yu DH, Moriyama K, Bode AM, Dong Z. TRAF1 Is Critical for DMBA/Solar UVR-Induced Skin Carcinogenesis. J Invest Dermatol 2017; 137:1322-1332. [PMID: 28131816 DOI: 10.1016/j.jid.2016.12.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/27/2016] [Accepted: 12/30/2016] [Indexed: 12/15/2022]
Abstract
TRAF1 is a member of the TRAF protein family, which regulates the canonical and noncanonical NF-κB signaling cascades. Although aberrant TRAF1 expression in tumors has been reported, the role of TRAF1 remains elusive. Here, we report that TRAF1 is required for solar UV-induced skin carcinogenesis. Immunohistochemical analysis showed that TRAF1 expression is up-regulated in human actinic keratosis and squamous cell carcinoma. In vivo studies indicated that TRAF1 expression levels in mouse skin are induced by short-term solar UV irradiation, and a long-term skin carcinogenesis study showed that deletion of TRAF1 in mice results in a significant inhibition of skin tumor formation. Moreover, we show that TRAF1 is required for solar UV-induced extracellular signal-regulated kinase-5 (ERK5) phosphorylation and the expression of AP-1 family members (c-Fos/c-Jun). Mechanistic studies showed that TRAF1 expression enhances the ubiquitination of ERK5 on lysine 184, which is necessary for its kinase activity and AP-1 activation. Overall, our results suggest that TRAF1 mediates ERK5 activity by regulating the upstream effectors of ERK5 and also by modulating its ubiquitination status. Targeting TRAF1 function might lead to strategies for preventing and treating skin cancer.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Eli Min
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Naomi Oi
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Ruihua Bai
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Tatyana A Zykova
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Dong Hoon Yu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Kenji Moriyama
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA.
| |
Collapse
|
24
|
Sox10 expression in ovarian epithelial tumors is associated with poor overall survival. Virchows Arch 2016; 468:597-605. [PMID: 26951260 DOI: 10.1007/s00428-016-1918-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/08/2016] [Accepted: 02/15/2016] [Indexed: 10/22/2022]
Abstract
Sox10 is a transcription factor regulating the development of several cell lineages and is involved in tumor development. However, the clinicopathological relevance of Sox10 expression in ovarian cancer has not been examined. We assessed expression of Sox10 in ovarian epithelial tumors by immunohistochemistry and assessed its prognostic value by analyzing the correlation between its expression and clinicopathological factors. We used tissue microarrays including 244 ovarian epithelial tumors. Sox10 staining was found in the cytoplasm or nucleus of tumor cells. Malignant serous, mucinous, and endometrioid tumors were significantly more likely to express Sox10 than benign and borderline tumors. Expression patterns in adenocarcinomas were different for histologic subtypes: nuclear Sox10 staining was common in clear-cell adenocarcinomas and serous adenocarcinomas, whereas all cases of mucinous and endometrioid tumors were negative for nuclear staining. Nuclear Sox10 staining was also associated with chemoresistance and shorter overall survival in ovarian adenocarcinomas, notably in high-grade serous adenocarcinoma. Sox10 is expressed in many ovarian carcinomas, suggesting that it might be involved in oncogenesis of ovarian carcinoma. Expression pattern of Sox10 differs between histological subtypes. Nuclear Sox10 expression is an independent indicator of poor prognosis in ovarian adenocarcinomas, notably in high-grade serous adenocarcinomas.
Collapse
|
25
|
E2F1: a promising regulator in ovarian carcinoma. Tumour Biol 2016; 37:2823-31. [PMID: 26749284 DOI: 10.1007/s13277-015-4770-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022] Open
Abstract
E2F is a family of transcription factors that recognized to regulate the expression of genes essential for a wide range of cellular functions, including cell cycle progression, DNA repair, DNA replication, differentiation, proliferation, and apoptosis. E2F1, the most classic member of the E2F family, exhibits a complex role in tumor development regulation. In recent years, a growing body of data suggested an intimate relationship between E2F1 and ovarian carcinoma. And E2F1 was well identified to play dual functions and serve as a useful prognostic indicator in ovarian carcinoma. However, the mechanism underlying E2F1 associated with ovarian carcinoma remains elusive. It is necessary to clarify the fundamental role of E2F1 in ovarian carcinoma. In this review, we tried to sum up the knowledge of E2F1, including its structure and related mechanism. We also attempt to absorb the research achievements and collect the mechanism of E2F1 in ovarian carcinoma.
Collapse
|
26
|
Hu L, Cong L. Fibroblast growth factor 19 is correlated with an unfavorable prognosis and promotes progression by activating fibroblast growth factor receptor 4 in advanced-stage serous ovarian cancer. Oncol Rep 2015; 34:2683-91. [PMID: 26323668 DOI: 10.3892/or.2015.4212] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/22/2015] [Indexed: 11/05/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) has been confirmed to be associated with the progression and prognosis of ovarian cancer, while the underlying mechanism has not been well elucidated and the clinical significance of its ligand, fibroblast growth factor 19 (FGF19), has not been explored. To study the clinical significance of FGF19 in advanced‑stage serous ovarian cancer, we detected the expression of FGF19 and FGFR4 by immunohistochemistry (IHC), evaluated the correlation between FGF19 and clinicopathological factors by Chi-square (χ2) test, and analyzed the association between FGF19, FGFR4 and the overall survival rate using the Kaplan‑Meier method. As a result, we demonstrated that high expression of FGF19 and FGFR4 both predicted unfavorable prognosis (P=0.033 and 0.018, respectively), whereas FGF19-FGFR4 double high expression was a more sensitive prognostic factor of advanced-stage serous ovarian cancer (P<0.001). With experiments in vitro, we demonstrated that both recombinant FGF19 and secreted FGF19 promoted ovarian cancer proliferation and invasion by activating FGFR4 and the subsequent AKT-MAPK signaling pathway, suggesting that FGF19-FGFR4 signaling may auto-activate in a paracrine or autocrine manner. In conclusion, FGF19-FGFR4 double high expression was a more sensitive prognostic factor than FGF19 or FGFR4 alone in advanced-stage serous ovarian cancer. The FGF19-FGFR4 signaling pathway can promote ovarian cancer proliferation and invasion by the AKT-MAPK signaling pathway, indicating that FGF19 could be a potential therapeutic drug target of advanced-stage serous ovarian cancer.
Collapse
Affiliation(s)
- Lingling Hu
- Department of Gynecology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Lanxiang Cong
- Department of Gynecology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
27
|
Chen F, Liu N. A 10-gene expression signature of Notch pathway predicts recurrence in ovarian carcinoma. Oncol Lett 2015; 10:1704-1708. [PMID: 26622736 DOI: 10.3892/ol.2015.3382] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 06/08/2015] [Indexed: 12/31/2022] Open
Abstract
Patients with ovarian carcinoma are at high risk of tumor recurrence. In the present study, 81 Notch pathway genes were selected to find recurrence-related genes in The Cancer Genome Atlas dataset. A 10-gene signature (FZD4, HES1, PSEN2, JAG2, PPARG, FOS, HEY1, CDC16, MFNG, and EP300) was identified and validated that is associated with recurrence-free survival time, but not with overall survival time, in the TCGA dataset and in other two independent datasets, GSE9891 and GSE30161. This gene signature gave a significant performance in discriminating patients at high risk of recurrence from those at low risk, as measured by the area under the receiver operating characteristic curve. Cox proportional hazards regression analyses demonstrated that the prognostic value of this 10-gene set is independent of other clinical variables in all three datasets. The potential as a biomarker for predicting high- and low-risk subgroups for recurrence in ovarian cancer patients deserves further investigation in prospective patient cohorts in the future.
Collapse
Affiliation(s)
- Fang Chen
- Department of Obstetrics and Gynecology, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Naifu Liu
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
28
|
Wang A, Guo C, Sun Y, Lu L, Wang Y, Wang Q, Zhang Y, Zhang H, Wang L, Gu Y, Liu A. Overexpression of CUEDC2 Predicts Poor Prognosis in Ovarian Serous Carcinomas. J Cancer 2015; 6:542-7. [PMID: 26000046 PMCID: PMC4439940 DOI: 10.7150/jca.11420] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/22/2015] [Indexed: 12/22/2022] Open
Abstract
CUEDC2, a newly reported protein, plays critical roles in many biological processes, such as cell cycle, inflammation and tumorigenesis, however, its expression in ovarian serious carcinoma is still poorly understood. In this study, we performed an immunohistochemical study on 101 cases of ovarian serous carcinoma tissues to investigate whether CUEDC2 is a useful biomarker to evaluate the progression of ovarian serous carcinomas. The data showed that the overexpression of CUEDC2 was observed in 59.4% of ovarian serous carcinoma tissue samples and correlated with histopathological grade, patient age at diagnosis, FIGO stage and recurrence. To assess the clinical relevance of CUEDC2, we analyzed the survival follow-up information, the results showed that CUEDC2-positive expression was associated with a shorter disease-free survival time, the median disease-free survival time of CUEDC2-positive patients was 36.0 months compared with 53.9 months of CUEDC2-negative ones (Log-rank χ2=6.149, P=0.013). Collectively, our results suggested that CUEDC2 may be a promising biomarker to evaluate the progression of serous ovarian carcinoma and to predict likely relapse of ovarian serous carcinoma.
Collapse
Affiliation(s)
- Aichun Wang
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China ; 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Chao Guo
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yunfei Sun
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Lijuan Lu
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Yun Wang
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Qiong Wang
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yan Zhang
- 3. Department of Obsteristics and Gynecology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Hui Zhang
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Li Wang
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Yiqun Gu
- 2. Department of Pathology, Haidian Maternal & Children Health Hospital, Beijing, 100080, China
| | - Aijun Liu
- 1. Department of Pathology, People's Liberation Army General Hospital, Beijing, 100853, China
| |
Collapse
|
29
|
Discover the molecular biomarker associated with cell death and extracellular matrix module in ovarian cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:735689. [PMID: 25861644 PMCID: PMC4378326 DOI: 10.1155/2015/735689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/10/2015] [Accepted: 02/15/2015] [Indexed: 02/01/2023]
Abstract
High throughput technologies have provided many new research methods for ovarian cancer investigation. In tradition, in order to find the underlying functional mechanisms of the survival-associated genes, gene sets enrichment analysis (GSEA) is always regarded as the important choice. However, GSEA produces too many candidate genes and cannot discover the signaling transduction cascades. In this work, we have used a network-based strategy to optimize the discovery of biomarkers using multifactorial data, including patient expression, clinical survival, and protein-protein interaction (PPI) data. The biomarkers discovered by this strategy belong to the network-based biomarker, which is apt to reveal the underlying functional mechanisms of the biomarker. In this work, over 400 expression arrays in ovarian cancer have been analyzed: the results showed that cell death and extracellular module are the main themes related to ovarian cancer progression.
Collapse
|
30
|
Kwon AY, Kim GI, Jeong JY, Song JY, Kwack KB, Lee C, Kang HY, Kim TH, Heo JH, An HJ. VAV3 Overexpressed in Cancer Stem Cells Is a Poor Prognostic Indicator in Ovarian Cancer Patients. Stem Cells Dev 2015; 24:1521-35. [PMID: 25715123 DOI: 10.1089/scd.2014.0588] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ovarian carcinoma is a highly lethal malignancy due to frequent relapse and drug resistance. Cancer stem cells (CSCs) are thought to contribute significantly to disease relapse and drug resistance. In this study, a subpopulation of CSCs of ovarian carcinoma was isolated and the genes differentially expressed in these cells were identified to characterize CSCs and to find candidate biomarkers. Ovarian carcinoma cells from patients were primarily cultured, and spheroid-forming cells (SFCs) were isolated. The characteristic genes of SFCs were identified through cDNA microarray and validation by quantitative real-time polymerase chain reaction and immunohistochemistry, and the association of their expression with clinicopathologic parameters was analyzed. GSC (4.26-fold), VAV3 (7.05-fold), FOXA2 (12.06-fold), LEF1 (17.26-fold), COMP (21.33-fold), GRIN2A (9.36-fold), CD86 (23.14-fold), PYY (4.18-fold), NKX3-2 (10.35-fold), and PDK4 (74.26-fold) were significantly upregulated in SFCs compared with parental cancer cells. With validation for human ovarian carcinomas, LEF1, PYY, NKX3-2, and WNT3A were significantly upregulated in chemoresistant cancers compared with chemosensitive cancers. Overexpression of LEF1, VAV3, and NKX3-2 was significantly associated with distant metastasis by immunohistochemistry. VAV3 overexpression was an independent poor survival indicator (hazard ratio=15.27, P<0.05) by multivariate Cox analysis. The further functional assay revealed that VAV3 knockdown regulated CSC activation and ovarian cancer cell proliferation and sensitized paclitaxel (PTX)-resistant cancer cells to PTX treatment. Taken together, we identified by high-throughput analysis of CSCs that VAV3 overexpression is a novel biomarker for poor prognosis and survival in ovarian carcinoma.
Collapse
Affiliation(s)
- Ah-Young Kwon
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Gwang-Il Kim
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Ju-Yeon Jeong
- 2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Ji-Ye Song
- 2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Kyu-Beom Kwack
- 3 Department of Biomedical Science, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Chan Lee
- 4 Department of Gynecologic Oncology, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Hae-Youn Kang
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Tae-Heon Kim
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Jin-Hyung Heo
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| | - Hee Jung An
- 1 Department of Pathology, College of Medicine, CHA University , Sungnam, Republic of Korea.,2 Institute for Clinical Research, College of Medicine, CHA University , Sungnam, Republic of Korea
| |
Collapse
|