1
|
Barnett SN, Cujba AM, Yang L, Maceiras AR, Li S, Kedlian VR, Pett JP, Polanski K, Miranda AMA, Xu C, Cranley J, Kanemaru K, Lee M, Mach L, Perera S, Tudor C, Joseph PD, Pritchard S, Toscano-Rivalta R, Tuong ZK, Bolt L, Petryszak R, Prete M, Cakir B, Huseynov A, Sarropoulos I, Chowdhury RA, Elmentaite R, Madissoon E, Oliver AJ, Campos L, Brazovskaja A, Gomes T, Treutlein B, Kim CN, Nowakowski TJ, Meyer KB, Randi AM, Noseda M, Teichmann SA. An organotypic atlas of human vascular cells. Nat Med 2024; 30:3468-3481. [PMID: 39566559 PMCID: PMC11645277 DOI: 10.1038/s41591-024-03376-x] [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: 11/16/2023] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Abstract
The human vascular system, comprising endothelial cells (ECs) and mural cells, covers a vast surface area in the body, providing a critical interface between blood and tissue environments. Functional differences exist across specific vascular beds, but their molecular determinants across tissues remain largely unknown. In this study, we integrated single-cell transcriptomics data from 19 human organs and tissues and defined 42 vascular cell states from approximately 67,000 cells (62 donors), including angiotypic transitional signatures along the arterial endothelial axis from large to small caliber vessels. We also characterized organotypic populations, including splenic littoral and blood-brain barrier ECs, thus clarifying the molecular profiles of these important cell states. Interrogating endothelial-mural cell molecular crosstalk revealed angiotypic and organotypic communication pathways related to Notch, Wnt, retinoic acid, prostaglandin and cell adhesion signaling. Transcription factor network analysis revealed differential regulation of downstream target genes in tissue-specific modules, such as those of FOXF1 across multiple lung vascular subpopulations. Additionally, we make mechanistic inferences of vascular drug targets within different vascular beds. This open-access resource enhances our understanding of angiodiversity and organotypic molecular signatures in human vascular cells, and has therapeutic implications for vascular diseases across tissues.
Collapse
Affiliation(s)
- Sam N Barnett
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK
| | - Ana-Maria Cujba
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Ana Raquel Maceiras
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Shuang Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Veronika R Kedlian
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - J Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - James Cranley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kazumasa Kanemaru
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lukas Mach
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | | | | | - Zewen K Tuong
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Alik Huseynov
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ioannis Sarropoulos
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rasheda A Chowdhury
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, UK
| | - Elo Madissoon
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Lia Campos
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Tomás Gomes
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Barbara Treutlein
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Chang N Kim
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Anna M Randi
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK.
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
2
|
Wang Y, Wang Z, Yu X, Wang X, Song J, Yu DJ, Ge F. MORE: a multi-omics data-driven hypergraph integration network for biomedical data classification and biomarker identification. Brief Bioinform 2024; 26:bbae658. [PMID: 39692449 DOI: 10.1093/bib/bbae658] [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: 08/19/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
High-throughput sequencing methods have brought about a huge change in omics-based biomedical study. Integrating various omics data is possibly useful for identifying some correlations across data modalities, thus improving our understanding of the underlying biological mechanisms and complexity. Nevertheless, most existing graph-based feature extraction methods overlook the complementary information and correlations across modalities. Moreover, these methods tend to treat the features of each omics modality equally, which contradicts current biological principles. To solve these challenges, we introduce a novel approach for integrating multi-omics data termed Multi-Omics hypeRgraph integration nEtwork (MORE). MORE initially constructs a comprehensive hyperedge group by extensively investigating the informative correlations within and across modalities. Subsequently, the multi-omics hypergraph encoding module is employed to learn the enriched omics-specific information. Afterward, the multi-omics self-attention mechanism is then utilized to adaptatively aggregate valuable correlations across modalities for representation learning and making the final prediction. We assess MORE's performance on datasets characterized by message RNA (mRNA) expression, Deoxyribonucleic Acid (DNA) methylation, and microRNA (miRNA) expression for Alzheimer's disease, invasive breast carcinoma, and glioblastoma. The results from three classification tasks highlight the competitive advantage of MORE in contrast with current state-of-the-art (SOTA) methods. Moreover, the results also show that MORE has the capability to identify a greater variety of disease-related biomarkers compared to existing methods, highlighting its advantages in biomedical data mining and interpretation. Overall, MORE can be investigated as a valuable tool for facilitating multi-omics analysis and novel biomarker discovery. Our code and data can be publicly accessed at https://github.com/Wangyuhanxx/MORE.
Collapse
Affiliation(s)
- Yuhan Wang
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Zhikang Wang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Xuan Yu
- Department of Computer Science, City University of Hong Kong, Kowloon, Hong Kong 999077, China
| | - Xiaoyu Wang
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
- Monash Data Futures Institute, Monash University, Wellington Rd, Clayton, Melbourne, VIC 3800, Australia
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing 210094, China
| | - Fang Ge
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan, Nanjing 210023, China
| |
Collapse
|
3
|
Wang MH, Gao YH, Zhao ZD, Zhang HW. A novel model combining genes associated with disulfidptosis and glycolysis to predict breast cancer prognosis, molecular subtypes, and treatment response. ENVIRONMENTAL TOXICOLOGY 2024; 39:4347-4359. [PMID: 38747344 DOI: 10.1002/tox.24329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 08/09/2024]
Abstract
Breast cancer (BC) is a heterogeneous malignancy with a dismal prognosis. Disulfidptosis is a novel type of regulated cell death that happens in the presence of glucose deficiency and is linked to the metabolic process of glycolysis. However, the mechanism of action of disulfidptosis and glycolysis-related genes (DGRG) in BC, as well as their prognostic value in BC patients, remain unknown. After identifying the differentially expressed DGRG in normal and BC tissues, a number of machine learning algorithms were utilized to select essential prognostic genes to develop a model, including SLC7A11, CACNA1H, SDC1, CHST1, and TFF3. The expression characteristics of these genes were then examined using single-cell RNA sequencing, and BC was classified into three clusters using "ConsensusClusterPlus" based on these genes. The DGRG model's median risk score can categorize BC patients into high-risk and low-risk groups. Furthermore, we investigated variations in clinical landscape, immunoinvasion analysis, tumor immune dysfunction and rejection (TIDE), and medication sensitivity in patients in the DGRG model's high- and low-risk groups. Patients in the low-risk group performed better on immunological and chemotherapeutic therapies and had lower TIDE scores. In conclusion, the DGRG model we developed has significant clinical application potential because it can accurately predict the prognosis of BC, TME, and pharmacological treatment responses.
Collapse
Affiliation(s)
- Mei-Huan Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yue-Hua Gao
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhen-Dan Zhao
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hua-Wei Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
4
|
Said NM, El-Shaer NH. Association of serum trefoil factor 3 and leptin levels with obesity: A case-control study. Cytokine 2024; 181:156690. [PMID: 38996578 DOI: 10.1016/j.cyto.2024.156690] [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: 05/10/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Obesity has a detrimental impact on individuals, communities, and healthcare systems. Trefoil factor 3 is a secretory protein involved in metabolic processes related to weight regulation. However, its relation with obesity is not fully understood. OBJECTIVE We aimed to assess the serum trefoil factor 3 level and to immunohistochemical detect the leptin in obese patients to evaluate their relation to obesity pathogenesis. METHODS As a case-control study, we enrolled 83 non-obese persons as a control group with a BMI (18.5-24.9) and 83 obese persons as a patient group with a BMI > 30. All the study volunteers are subjected to anthropometric measurements, glucose, and lipid profile analysis by colorimetric methods. Serum trefoil factor 3 level was estimated by ELISA and leptin hormone was detected immunohistochemically in the blood using cell block technique. RESULTS ROC curve analysis for TFF3 showed a good relation with obesity with an AUC of 0.891 and a cut-off value of > 96 ng/ml. There was a significant positive correlation between TFF3 and fasting blood sugar, total cholesterol, and triglycerides. The logistic regression analysis showed that TFF3 is a good risk factor for obesity incidence [p = 0.008; OR = 1.117; (95 % CI): 1.029-1.213]. This was confirmed by multiple linear regression that gave an equation for the possibility of predicting BMI using several factors including TFF3 [BMI = 0.821 + 0.051 × TFF3 + 0.044 × FBS + 0.85 × TC]. The more surprising was the ability of the immunohistochemistry cell block technique to detect leptin antigens associated with an obese person blood not only adipose tissue or serum. CONCLUSION Leptin hormone and TFF3 could be good indicators for obesity incidence. Further research with a larger sample size and in different populations could completely approve our results.
Collapse
Affiliation(s)
- Noha Mohamed Said
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt.
| | - Nahla H El-Shaer
- Zoology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
5
|
Guo H, Tan YQ, Huang X, Zhang S, Basappa B, Zhu T, Pandey V, Lobie PE. Small molecule inhibition of TFF3 overcomes tamoxifen resistance and enhances taxane efficacy in ER+ mammary carcinoma. Cancer Lett 2023; 579:216443. [PMID: 37858772 DOI: 10.1016/j.canlet.2023.216443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
Even though tamoxifen has significantly improved the survival of estrogen receptor positive (ER+) mammary carcinoma (MC) patients, the development of drug resistance with consequent disease recurrence has limited its therapeutic efficacy. Trefoil factor-3 (TFF3) has been previously reported to mediate anti-estrogen resistance in ER+MC. Herein, the efficacy of a small molecule inhibitor of TFF3 (AMPC) in enhancing sensitivity and mitigating acquired resistance to tamoxifen in ER+MC cells was investigated. AMPC induced apoptosis of tamoxifen-sensitive and resistant ER+MC cells and significantly reduced cell survival in 2D and 3D culture in vitro. In addition, AMPC reduced cancer stem cell (CSC)-like behavior in ER+MC cells in a BCL2-dependent manner. Synergistic effects of AMPC and tamoxifen were demonstrated in ER+MC cells and AMPC was observed to improve tamoxifen efficacy in tamoxifen-sensitive cells and to re-sensitize cells to tamoxifen in tamoxifen-resistant ER+MC in vitro and in vivo. Additionally, tamoxifen-resistant ER+MC cells were concomitantly resistant to anthracycline, platinum and fluoropyrimidine drugs, but not to Taxanes. Taxane treatment of tamoxifen-sensitive and resistant ER+MC cells increased TFF3 expression indicating a combination vulnerability for tamoxifen-resistant ER+MC cells. Taxanes increased CSC-like behavior of tamoxifen-sensitive and resistant ER+MC cells which was reduced by AMPC treatment. Taxanes synergized with AMPC to promote apoptosis and reduce CSC-like behavior in vitro and in vivo. Hence, AMPC restored the sensitivity of tamoxifen and enhanced the efficacy of Taxanes in tamoxifen-resistant ER+MC. In conclusion, pharmacological inhibition of TFF3 may serve as an effective combinatorial therapeutic strategy for the treatment of tamoxifen-resistant ER+MC.
Collapse
Affiliation(s)
- Hui Guo
- Tsinghua Berkeley Shenzhen Institute and the Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Yan Qin Tan
- Tsinghua Berkeley Shenzhen Institute and the Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Xiaoming Huang
- Tsinghua Berkeley Shenzhen Institute and the Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Shuwei Zhang
- Tsinghua Berkeley Shenzhen Institute and the Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, 570006, India
| | - Tao Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute and the Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China.
| | - Peter E Lobie
- Tsinghua Berkeley Shenzhen Institute and the Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Shenzhen Bay Laboratory, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
6
|
Minegishi K, Dobashi Y, Koyama T, Ishibashi Y, Furuya M, Tsubochi H, Ohmoto Y, Yasuda T, Nomura S. Diagnostic utility of trefoil factor families for the early detection of lung cancer and their correlation with tissue expression. Oncol Lett 2023; 25:139. [PMID: 36909373 PMCID: PMC9996639 DOI: 10.3892/ol.2023.13725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/22/2022] [Indexed: 02/23/2023] Open
Abstract
Trefoil factors (TFFs) are upregulated in numerous types of cancer, including those of the breast, the colon, the lung and the pancreas, suggesting their potential utility as biomarkers for screening. In the present study, the clinical relevance of serum or urinary TFFs as biomarkers were comprehensively evaluated and the correlation with TFF expression levels in lung cancer tissue was examined. Serum and urine were collected from 199 patients with lung cancer and 198 healthy individuals. Concentrations of serum and urinary TFF1, TFF2 and TFF3 were measured using ELISA and the potential of TFF levels to discriminate between cancer and non-cancer samples was evaluated. In 100 of the cancer cases, expression of TFF1-3 was analyzed using immunohistochemical staining of paraffin sections. Furthermore, the relationship between TFF levels and clinicopathological factors among these cancer cases was analyzed using immunohistochemistry of tissue specimens, quantified and statistically analyzed. While serum levels of all TFFs measured using ELISA were significantly higher in patients with lung cancer compared with those in healthy individuals, urinary TFFs were lower. Areas under the curve (AUC) of the receiver operating characteristic curves for serum/urinary TFF1, TFF2 and TFF3 were 0.709/0.594, 0.722/0.501 and 0.663/0.665, respectively. Furthermore, the combination of serum TFF1, TFF2, TFF3 and urinary TFF1 and TFF3 demonstrated the highest AUC (0.826). In the clinicopathological analysis, serum TFF1 was higher in the early pathological T-stage (pTis/1/2) compared with the later stage (pT3/4) and TFF2 was higher in the pN0/1 than the pN2 group. With regards to the histological types, urinary TFF1 was higher in squamous cell carcinoma than adenocarcinoma (AC), but TFF2 tended to be higher in AC. Using immunohistochemical analysis, although TFF1 and TFF3 expression showed positive correlation with serum concentrations, TFF2 was inversely correlated. In conclusion, serum and urinary TFF levels are promising predictive biomarkers, and their measurements provide a useful in vivo and non-invasive diagnostic screening tool. In particular, TFF1 and TFF3 could be surrogate markers of clinicopathological profiles of human lung cancer.
Collapse
Affiliation(s)
- Kentaro Minegishi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan
| | - Yoh Dobashi
- Department of Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan.,Department of Pathology, School of Medicine, International University of Health and Welfare Hospital, Nasushiobara, Tochigi 329-2763, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto 602-8566, Japan
| | - Yuko Ishibashi
- Department of Surgery, Breast Surgery, Tokyo Women's Medical University, Adachi Medical Center, Adachi, Tokyo 123-8558, Japan
| | - Miki Furuya
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyoshi Tsubochi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan
| | - Yasukazu Ohmoto
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8505, Japan
| | - Tomohiko Yasuda
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Department of Gastrointestinal Surgery, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Chiba 270-1694, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
7
|
Foo RJK, Tian S, Tan EY, Goh WWB. A novel survival prediction signature outperforms PAM50 and artificial intelligence-based feature-selection methods. Comput Biol Chem 2023; 104:107845. [PMID: 36889140 DOI: 10.1016/j.compbiolchem.2023.107845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/06/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
The robustness of a breast cancer gene signature, the super-proliferation set (SPS), is initially tested and investigated on breast cancer cell lines from the Cancer Cell Line Encyclopaedia (CCLE). Previously, SPS was derived via a meta-analysis of 47 independent breast cancer gene signatures, benchmarked on survival information from clinical data in the NKI dataset. Here, relying on the stability of cell line data and associative prior knowledge, we first demonstrate through Principal Component Analysis (PCA) that SPS prioritizes survival information over secondary subtype information, surpassing both PAM50 and Boruta, an artificial intelligence-based feature-selection algorithm, in this regard. We can also extract higher resolution 'progression' information using SPS, dividing survival outcomes into several clinically relevant stages ('good', 'intermediate', and 'bad) based on different quadrants of the PCA scatterplot. Furthermore, by transferring these 'progression' annotations onto independent clinical datasets, we demonstrate the generalisability of our method on actual patient data. Finally, via the characteristic genetic profiles of each quadrant/stage, we identified efficacious drugs using their gene reversal scores that can shift signatures across quadrants/stages, in a process known as gene signature reversal. This confirms the power of meta-analytical approaches for gene signature inference in breast cancer, as well as the clinical benefit in translating these inferences onto real-world patient data for more targeted therapies.
Collapse
Affiliation(s)
- Reuben Jyong Kiat Foo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Siqi Tian
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ern Yu Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Tan Tock Seng Hospital, Singapore
| | - Wilson Wen Bin Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore; Centre for Biomedical Informatics, Nanyang Technological University, Singapore.
| |
Collapse
|
8
|
An HW, Seok SH, Kwon JW, Choudhury AD, Oh JS, Voon DC, Kim DY, Park JW. The loss of epithelial Smad4 drives immune evasion via CXCL1 while displaying vulnerability to combinatorial immunotherapy in gastric cancer. Cell Rep 2022; 41:111878. [PMID: 36577366 DOI: 10.1016/j.celrep.2022.111878] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/17/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
SMAD4 is frequently mutated and inactivated in human gastric cancer (GC). Although the epithelial cell-autonomous functions of Smad4 have been extensively studied, its contribution to tumor immunity is largely undetermined. Here, we report that the loss of Smad4 expression in GC cells endows them with the ability to evade tumor immunity. Unlike their Smad4-proficient counterparts, Smad4-deficient stomach organoids can evade host immunity to form tumors in immunocompetent mice. Smad4-deficient GC cells show expanded CD133+ cancer stem-like cells while suppressing dendritic cell (DC) differentiation and cytotoxic T cells with granulocytic myeloid-derived suppressor cell (G-MDSC) accumulation through a secretome containing CXCL1. Moreover, Smad4 deficiency increases programmed cell death ligand-1 (PD-L1) and decreases 4-1BBL expressions, indicating a change in immunogenicity. Combinatorial immune checkpoint blockade (ICB) of anti-PD-L1 and anti-CTLA-4 or agonistic anti-4-1BB antibodies effectively treats ICB monotherapy-resistant Smad4-deficient allografts, exposing a specific vulnerability. Collectively, these data provide a rational basis for ICB strategies in treating advanced GC with Smad4 deficiency.
Collapse
Affiliation(s)
- Hyeok-Won An
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do 24341, South Korea
| | - Sang Hyeok Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do 24341, South Korea
| | - Jong-Wan Kwon
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do 24341, South Korea
| | - Anahita Dev Choudhury
- Innovative Cancer Model Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Jeong-Seop Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Dominic C Voon
- Innovative Cancer Model Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan; Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan.
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea.
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do 24341, South Korea.
| |
Collapse
|
9
|
Lin Z, Wan X, Zhang T, Huo H, Zhang X, Li K, Bei W, Guo J, Yang Y. Trefoil factor 3: New highlights in chronic kidney disease research. Cell Signal 2022; 100:110470. [PMID: 36122885 DOI: 10.1016/j.cellsig.2022.110470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
Trefoil factor 3 (TFF3, also known as intestinal trefoil factor) is a small-molecule peptide containing a typical trefoil structure. TFF3 has several biological effects, such as wound healing, immune regulation, neuroprotection, and cell migration and proliferation promotion. Although TFF3 binding sites were identified in rat kidneys more than a decade ago, the specific effects of this small-molecule peptide on kidneys remain unclear. Until recently, much of the research on TFF3 in the kidney field has focused exclusively on its role as a biomarker. Notably, a large prospective randomized study of patients with 29 common clinical diseases revealed that chronic kidney disease (CKD) was associated with the highest serum TFF3 levels, which were 3-fold higher than in acute gastroenteritis, which had the second-highest levels. Examination of each stage of CKD revealed that urine and serum TFF3 levels significantly increased with the progression of CKD. These results suggest that the role of TFF3 in CKD needs further research. The present review summarizes the renal physiological expression, biological functions, and downstream signaling of TFF3, as well as the upstream events that lead to high expression of TFF3 in CKD.
Collapse
Affiliation(s)
- Ziyang Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Xiaofen Wan
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Tao Zhang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Hongyan Huo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Xiaoyu Zhang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Kunping Li
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Weijian Bei
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China.
| |
Collapse
|
10
|
Pandey V, Zhang X, Poh HM, Wang B, Dukanya D, Ma L, Yin Z, Bender A, Periyasamy G, Zhu T, Rangappa KS, Basappa B, Lobie PE. Monomerization of Homodimeric Trefoil Factor 3 (TFF3) by an Aminonitrile Compound Inhibits TFF3-Dependent Cancer Cell Survival. ACS Pharmacol Transl Sci 2022; 5:761-773. [PMID: 36110371 PMCID: PMC9469493 DOI: 10.1021/acsptsci.2c00044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 11/28/2022]
Abstract
Trefoil factor 3 (TFF3) is a secreted protein with an established oncogenic function and a highly significant association with clinical progression of various human malignancies. Herein, a novel small molecule that specifically targets TFF3 homodimeric functions was identified. Utilizing the concept of reversible covalent interaction, 2-amino-4-(4-(6-fluoro-5-methylpyridin-3-yl)phenyl)-5-oxo-4H,5H-pyrano[3,2-c]chromene-3-carbonitrile (AMPC) was identified as a molecule that interacted with TFF3. AMPC monomerized the cellular and secreted TFF3 homodimer at the cysteine (Cys)57-Cys57 residue with subsequent more rapid degradation of the generated TFF3 monomers. Hence, AMPC treatment also resulted in cellular depletion of TFF3 with consequent decreased cell viability in various human carcinoma-derived TFF3 expressing cell lines, including estrogen receptor positive (ER+) mammary carcinoma (MC). AMPC treatment of TFF3 expressing ER+ MC cells significantly suppressed total cell number in a dose-dependent manner. Consistently, exposure of TFF3 expressing ER+ MC cells to AMPC decreased soft agar colony formation, foci formation, and growth in suspension culture and inhibited growth of preformed colonies in 3D Matrigel. AMPC increased apoptosis in TFF3 expressing ER+ MC cells associated with decreased activity of EGFR, p38, STAT3, AKT, and ERK, decreased protein levels of CCND1, CCNE1, BCL2, and BCL-XL, and increased protein levels of TP53, CDKN1A, CASP7, and CASP9. siRNA-mediated depletion of TFF3 expression in ER+ MC cells efficiently abrogated AMPC-stimulated loss of cell viability and CASPASE 3/7 activities. Furthermore, in mice bearing ER+ MC cell-generated xenografts, AMPC treatment significantly impeded xenograft growth. Hence, AMPC exemplifies a novel mechanism by which small molecule drugs may inhibit a dimeric oncogenic protein and provides a strategy to impede TFF3-dependent cancer progression.
Collapse
Affiliation(s)
- Vijay Pandey
- Tsinghua
Berkeley Shenzhen Institute and Institute of Biopharmaceutical and
Health Engineering, Tsinghua Shenzhen International
Graduate School, Shenzhen 518055, PR China
| | - Xi Zhang
- Shenzhen
Bay Laboratory, Shenzhen 518055, PR China
| | - Han-Ming Poh
- Cancer Science
Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599
| | - Baocheng Wang
- Tsinghua
Berkeley Shenzhen Institute and Institute of Biopharmaceutical and
Health Engineering, Tsinghua Shenzhen International
Graduate School, Shenzhen 518055, PR China
| | - Dukanya Dukanya
- Laboratory
of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006 Karnataka, India
| | - Lan Ma
- Tsinghua
Berkeley Shenzhen Institute and Institute of Biopharmaceutical and
Health Engineering, Tsinghua Shenzhen International
Graduate School, Shenzhen 518055, PR China
- Shenzhen
Bay Laboratory, Shenzhen 518055, PR China
| | - Zhinan Yin
- Biomedical
Translational Research Institute, Jinan
University, 601 Huangpu Avenue West, Guangzhou 510632, PR China
- Zhuhai Institute
of Translational Medicine Zhuhai People’s Hospital Affiliated
with Jinan University, Jinan University, Zhuhai, Guangdong 519000, PR China
| | - Andreas Bender
- Centre for
Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Ganga Periyasamy
- DOS in Chemistry, Bangalore University, JB Campus, Bangalore 560001, India
| | - Tao Zhu
- Department
of Oncology of the First Affiliated Hospital, Division of Life Sciences
and Medicine, University of Science and
Technology of China, Hefei, Anhui 230027, China
- Hefei National
Laboratory for Physical Sciences, the CAS Key Laboratory of Innate
Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Kanchugarakoppal S. Rangappa
- Laboratory
of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006 Karnataka, India
| | - Basappa Basappa
- Laboratory
of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006 Karnataka, India
| | - Peter E. Lobie
- Tsinghua
Berkeley Shenzhen Institute and Institute of Biopharmaceutical and
Health Engineering, Tsinghua Shenzhen International
Graduate School, Shenzhen 518055, PR China
- Shenzhen
Bay Laboratory, Shenzhen 518055, PR China
- Cancer Science
Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore 117599
| |
Collapse
|
11
|
Zeng Y, Wei Z, Yu W, Yin R, Yuan Y, Li B, Tang Z, Lu Y, Yang Y. Spatial transcriptomics prediction from histology jointly through Transformer and graph neural networks. Brief Bioinform 2022; 23:6645485. [PMID: 35849101 DOI: 10.1093/bib/bbac297] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/12/2022] [Accepted: 06/29/2022] [Indexed: 12/16/2022] Open
Abstract
The rapid development of spatial transcriptomics allows the measurement of RNA abundance at a high spatial resolution, making it possible to simultaneously profile gene expression, spatial locations of cells or spots, and the corresponding hematoxylin and eosin-stained histology images. It turns promising to predict gene expression from histology images that are relatively easy and cheap to obtain. For this purpose, several methods are devised, but they have not fully captured the internal relations of the 2D vision features or spatial dependency between spots. Here, we developed Hist2ST, a deep learning-based model to predict RNA-seq expression from histology images. Around each sequenced spot, the corresponding histology image is cropped into an image patch and fed into a convolutional module to extract 2D vision features. Meanwhile, the spatial relations with the whole image and neighbored patches are captured through Transformer and graph neural network modules, respectively. These learned features are then used to predict the gene expression by following the zero-inflated negative binomial distribution. To alleviate the impact by the small spatial transcriptomics data, a self-distillation mechanism is employed for efficient learning of the model. By comprehensive tests on cancer and normal datasets, Hist2ST was shown to outperform existing methods in terms of both gene expression prediction and spatial region identification. Further pathway analyses indicated that our model could reserve biological information. Thus, Hist2ST enables generating spatial transcriptomics data from histology images for elucidating molecular signatures of tissues.
Collapse
Affiliation(s)
- Yuansong Zeng
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Zhuoyi Wei
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Weijiang Yu
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Rui Yin
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Yuchen Yuan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Bingling Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhonghui Tang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yutong Lu
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou 510000, China.,Key Laboratory of Machine Intelligence and Advanced Computing (MOE), Guangzhou 510000, China
| |
Collapse
|
12
|
Canet-Jourdan C, Pagès DL, Nguyen-Vigouroux C, Cartry J, Zajac O, Desterke C, Lopez JB, Gutierrez-Mateyron E, Signolle N, Adam J, Raingeaud J, Polrot M, Gonin P, Mathieu JRR, Souquere S, Pierron G, Gelli M, Dartigues P, Ducreux M, Barresi V, Jaulin F. Patient-derived organoids identify an apico-basolateral polarity switch associated with survival in colorectal cancer. J Cell Sci 2022; 135:276070. [PMID: 35703098 DOI: 10.1242/jcs.259256] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
The metastatic progression of cancer remains a major issue in patient treatment. Yet, the molecular and cellular mechanisms underlying this process remains unclear. Here, we use primary explants and organoids from patients harboring mucinous colorectal carcinoma (MUC CRC), a poor prognosis histological form of digestive cancers, to study the architecture, invasive behavior and chemoresistance of tumor cell intermediates. We report that these tumors maintain a robust apico-basolateral polarity as they spread in the peritumoral stroma or organotypic collagen-I gels. We identified two distinct topologies: MUC CRCs either display a conventional "apical-in" polarity or, more frequently, harbor an inverted "apical-out" topology. Transcriptomic analyses combined with interference experiments on organoids showed that TGFb and focal adhesion signaling pathways are the main drivers of polarity orientation. Finally, this apical-out topology is associated with increased resistance to chemotherapeutic treatments in organoids and decreased patient survival in the clinic. Thus, patient-derived organoids have the potential to bridge histological, cellular and molecular analyses to decrypt onco-morphogenic programs and stratify cancer patients.
Collapse
Affiliation(s)
| | | | | | - Jérôme Cartry
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| | - Olivier Zajac
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005 Paris, France
| | | | | | | | - Nicolas Signolle
- INSERM Unit U981, Experimental Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Julien Adam
- INSERM Unit U981, Experimental Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Joel Raingeaud
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| | - Mélanie Polrot
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/ US 23, Gustave Roussy, Villejuif, F-94805, France
| | - Patrick Gonin
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/ US 23, Gustave Roussy, Villejuif, F-94805, France
| | | | | | | | - Maximiliano Gelli
- Department of Medical Oncology, Gustave Roussy, Villejuif, F-94805, France
| | - Peggy Dartigues
- Pathology Department, Gustave Roussy, Villejuif, F-94805, France
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Villejuif, F-94805, France.,Paris-Saclay University, Saint-Aubin, F-91190, France
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Verona 37129, Italia
| | - Fanny Jaulin
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| |
Collapse
|
13
|
Abdelrazek MA, Nageb A, Barakat LA, Abouzid A, Elbaz R. BC-DETECT: combined detection of serum HE4 and TFF3 improves breast cancer diagnostic efficacy. Breast Cancer 2022; 29:507-515. [PMID: 34994942 DOI: 10.1007/s12282-021-01328-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/28/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Early accurate breast cancer (BC) diagnosis is critical in disease management. Mammography has been widely used. However, its radiation, and high false-negative and -positive results have always been a concern. We evaluated combined detection of human epididymal protein 4 (HE4) and trefoil factor 3 (TFF3) as substitute method to enhance BC diagnosis. METHODS HE4 and TFF3 blood levels were determined by ELISA in sera of 120 BC patients and 80 women (40 healthy and 40 benign breast disease) as controls. Receiver-operating characteristic curve was applied for evaluation diagnostic power of each biomarker and their combination. RESULTS In BC patients, serum HE4 [5 (2-11.9) vs. 3.1 (1.8-5.4) and 1 (1-3.5); P = 0.022] and TFF3 [5.3 (4.5-6.7) vs. 4.7 (4-4.8) and 3.9 (3-4.4); P = 0.027] were significantly higher than that in benign and healthy groups, respectively. Both HE4 (AUC = 0.783; P < 0.0001) and TFF3 (AUC = 0.759; P < 0.0001) had superior BC diagnostic ability compared to CEA and CA-15.3. Logistic regression analysis revealed simplified index BC-DETECT = HE4 + TFF3, and its values were significantly (P = 0.0132) elevated in BC (10.9 (8.4-17.2) compared to benign (7.2 (5.4-10.1)) and healthy (5.1 (4-6.3)) controls. AUC of BC-DETECT for BC prediction was (AUC = 0.850; P < 0.0001) with sensitivity, specificity, and positive and negative predictive values and accuracy of 84.2%, 70%, 80.8%, 74.7%, and 78.5%, respectively. High BC-DETECT levels were associated with tumor non-luminal subtypes, late stage, high grade, large size, lymph-node invasion, and multiple lesions. CONCLUSIONS BC-DETECT is inexpensive, rapid, and easy to perform and reliably guides BC early detection. Moreover, the association between elevated BC-DETECT values and disease severity may propose its potential role as prognostic marker.
Collapse
Affiliation(s)
- Mohamed A Abdelrazek
- Research and Development Department, Biotechnology Research Center, New Damietta, Egypt.
- Biochemistry Labs, Sherbin Central Hospital, Ministry of Health, Ad Daqahliyah, Egypt.
| | - Ahmed Nageb
- Department of Chemistry, Faculty of Science, Port Said University, Port Fuad, Egypt
| | - Lamiaa A Barakat
- Department of Chemistry, Faculty of Science, Port Said University, Port Fuad, Egypt
| | - Amr Abouzid
- Department of Surgical Oncology, Mansoura Oncology Centre, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rizk Elbaz
- Genetics Unit, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
14
|
Shen M, Yang L, Lei T, Zhang P, Xiao L, Cao S, Chen F, Li L, Ye F, Bu H. Correlation between CA12 and TFF3 and their prediction value of neoadjuvant chemotherapy response in breast cancer. J Clin Pharm Ther 2022; 47:609-618. [PMID: 35229335 DOI: 10.1111/jcpt.13580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/01/2021] [Accepted: 11/16/2021] [Indexed: 02/05/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Compared with other molecular subtypes, hormone receptor-positive breast cancer often shows worse neoadjuvant chemotherapy efficacy. This study aims to explore the relationship between the oestrogen receptor (ER)-related genes carbonic anhydrase 12 (CA12) and trefoil factor 3 (TFF3) and their predictive value of neoadjuvant chemotherapy for breast cancer. METHODS We investigated the relationships between CA12, TFF3 and ER status and their predictive value of anthracycline-taxane neoadjuvant chemotherapy in 115 female breast cancer patients via real-time polymerase chain reaction (RT-PCR) and 4 GEO datasets: GSE41998, GSE25065, GSE20194 and GSE20271. Then, the effects of CA12 and TFF3 on the chemotherapy drugs doxorubicin and docetaxel were verified in vitro in the breast cancer cell lines MCF-7 and BT474. RESULTS AND DISCUSSION The GEO datasets and RT-PCR results showed that the relative expression of both CA12 and TFF3 was higher in oestrogen receptor-positive samples compared with the other samples (p < 0.05). CA12 was significantly correlated with TFF3 (p < 0.05). In MCF-7 cells, inhibition of TFF3 induced downregulation of CA12 and ESR1 (p < 0.05) at both the mRNA and the protein levels, while inhibition of CA12 also downregulated TFF3 and ESR1 (p < 0.05). In BT474 cells, inhibition of TFF3 downregulated CA12 and ESR1 (p < 0.05) at both the mRNA and the protein levels, while inhibition of CA12 led to slight upregulation of TFF3 and ESR1 (p > 0.05). Moreover, GEO datasets and RT-PCR results showed that CA12 and TFF3 were more highly expressed in nonpathological complete response (non-pCR) samples than in pCR samples (p < 0.05). Cell viability assays of MCF-7 and BT474 cells showed that inhibiting CA12 and TFF3 could enhance sensitivity to doxorubicin and docetaxel (p < 0.05). WHAT IS NEW AND CONCLUSION CA12 and TFF3 were correlated with each other, and their high expression might explain the worse efficacy of neoadjuvant chemotherapy in oestrogen receptor-positive breast cancer patients.
Collapse
Affiliation(s)
- Mengjia Shen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Libo Yang
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Lei
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Peichuan Zhang
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Xiao
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyu Cao
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fei Chen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Ye
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Bu
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Hsieh TH, Hsu CY, Yang PJ, Chiu CC, Liang SS, Ou-Yang F, Kan JY, Hou MF, Wang TN, Tsai EM. DEHP mediates drug resistance by directly targeting AhR in human breast cancer. Biomed Pharmacother 2021; 145:112400. [PMID: 34801851 DOI: 10.1016/j.biopha.2021.112400] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 01/25/2023] Open
Abstract
Resistance to chemotherapy and hormonal therapy is a major clinical problem in breast cancer medicine, especially for cancer metastasis and recurrence. Di(2-ethylhexyl)phthalate (DEHP) affects drug resistance by an unknown mechanism of action. Here we analyzed breast cancer patients (N = 457) and found that Σ4MEHP (the sum of MEHP, MEHHP, MECPP and MEOHP concentrations) in urine was significantly higher (P = 0.018) in the recurrent breast cancer group compared with non-recurrent patients. Σ4MEHP-High was positively and significantly correlated with tumor stage (P = 0.005), lymph node status (P = 0.001), estrogen receptor status (P = 0.010), Her2/Neu status (P = 0.004), recurrence (P = 0.000) and tumor size (P = 0.002), as well as an independent prognostic marker (OR = 1.868; 95% CI = 1.424-2.451; P < 0.000) associated with poor survival rates based on a positive Her2/Neu status (P = 0.035). In addition, we found that DEHP inhibited paclitaxel and doxorubicin effects in breast cancer cell lines MCF-7 and MDA-MB-231 and in zebrafish and mouse tumor initiation models. DEHP induced trefoil factor 3 (TFF3) expression through the vinculin/aryl hydrocarbon receptor (AhR)/ERK signaling pathway and induced CYP2D6, CYP2C8 and CYP3A4 expression through the AhR genomic pathway to increase the epithelial-mesenchymal transition (EMT) and doxorubicin metabolism, respectably. DEHP mediated AhR-related alterations in estrogen receptor expression through the ubiquitination system, which decreased tamoxifen effects in AhR knockout mice. These findings suggest a novel therapeutic avenue by targeting AhR in drug-resistant and recurrent breast cancer.
Collapse
Affiliation(s)
- Tsung-Hua Hsieh
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chia-Yi Hsu
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Jing Yang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Shin Liang
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu Ou-Yang
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jung-Yu Kan
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
16
|
Fan J, Feng Y, Cheng Y, Wang Z, Zhao H, Galan EA, Liao Q, Cui S, Zhang W, Ma S. Multiplex gene quantification as digital markers for extremely rapid evaluation of chemo-drug sensitivity. PATTERNS 2021; 2:100360. [PMID: 34693378 PMCID: PMC8515010 DOI: 10.1016/j.patter.2021.100360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/29/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022]
Abstract
Current administrations for precision drug uses are limited in evaluation speed. Here, we propose the use of multiplex gene-based digital markers for the extremely rapid personalized prediction of individual sensitivity to cancer drugs. We first screen the transcriptional profiles by applying two to three gene filters and scoring genes by their impact on drug sensitivity and finalize the gene lists by K-nearest neighbors cross-validation. The digital markers are cancer type dependent, are composed of tens to hundreds of gene expressions, and are rapidly quantified by reverse transcription quantitative real-time PCR (qRT-PCR) within 1–3 h after tumor sampling. The area under the receiver operating characteristic curve reached 0.88 when testing the performance of digital markers on organoids derived from colorectal cancer patient tumors. The algorithm and corresponding graphic user interface were developed to demonstrate the promise of digital markers for extremely rapid drug recommendation. Non-targeted multiplex genes are screened as digital markers for drug sensitivity Transcription level cohort of 10s to 100s genes predicts drug sensitivity Digital markers are quantified using qRT-PCR within 1–3 h Digital markers guide extremely rapid chemo-drug uses after patient hospitalization
In clinical cancer medicine, many patients require immediate chemotherapy after hospitalization. Current administrations for precision drug uses are limited in evaluation speed, including genomic sequencing and tumor organoid evaluation. An extremely rapid evaluation protocol is in high demand to realize drug recommendation within a few hours after tumor sampling. In this work, we have proposed an approach for extremely rapid and personalized drug recommendation.
Collapse
Affiliation(s)
- Jiaqi Fan
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China.,Institute for Brain and Cognitive Sciences (THUIBCS), Tsinghua University, Beijing 100084, China
| | - Yilin Feng
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Yifan Cheng
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Zitian Wang
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Haoran Zhao
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Edgar A Galan
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China
| | - Quanxing Liao
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Shuzhong Cui
- Department of Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Weijie Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shaohua Ma
- Tsinghua University, Shenzhen International Graduate School (SIGS), Shenzhen 518055, China.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Shenzhen 518055, China.,Institute for Brain and Cognitive Sciences (THUIBCS), Tsinghua University, Beijing 100084, China
| |
Collapse
|
17
|
Jurmeister P, Weber K, Villegas S, Karn T, Untch M, Thieme A, Müller V, Taube E, Fasching P, Schmitt WD, Marmé F, Stickeler E, Sinn BV, Jank P, Schem C, Klauschen F, van Mackelenbergh M, Denkert C, Loibl S, Capper D. DNA methylation profiling identifies two distinct subgroups in breast cancers with low hormone receptor expression, mainly associated with HER2 amplification status. Clin Epigenetics 2021; 13:184. [PMID: 34602069 PMCID: PMC8489064 DOI: 10.1186/s13148-021-01176-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/22/2021] [Indexed: 11/25/2022] Open
Abstract
Background Current clinical guidelines suggest that breast cancers with low hormone receptor expression (LowHR) in 1–10% of tumor cells should be regarded as hormone receptor positive. However, clinical data show that these patients have worse outcome compared to patients with hormone receptor expression above 10%. We performed DNA methylation profiling on 23 LowHR breast cancer specimens, including 13 samples with HER2 amplification and compared our results with a reference breast cancer cohort from The Cancer Genome Atlas to clarify the status for this infrequent but important patient subgroup. Results In unsupervised clustering and dimensionality reduction, breast cancers with low hormone receptor expression that lacked HER2 amplification usually clustered with triple negative breast cancer (TNBC) reference samples (8/10; “LowHR TNBC-like”). In contrast, most specimens with low hormone receptor expression and HER2 amplification grouped with hormone receptor positive cancers (11/13; “LowHR HRpos-like”). We observed highly similar DNA methylation patterns of LowHR TNBC-like samples and true TNBCs. Furthermore, the Ki67 proliferation index of LowHR TNBC-like samples and clinical outcome parameters were more similar to TNBCs and differed from LowHR HRpos-like cases.
Conclusions We here demonstrate that LowHR breast cancer comprises two epigenetically distinct groups. Our data strongly suggest that LowHR TNBC-like samples are molecularly, histologically and clinically closely related to TNBC, while LowHR HRpos-like specimens are closely related to hormone receptor positive tumors. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01176-5.
Collapse
Affiliation(s)
- Philipp Jurmeister
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany. .,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69210, Heidelberg, Germany. .,Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany. .,Institute of Pathology, Ludwig Maximilians University Hospital Munich, Thalkirchner Str. 36, 80337, Munich, Germany.
| | | | - Sonia Villegas
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
| | - Thomas Karn
- Department of Gynecology and Obstetrics, Goethe University, Frankfurt, Germany
| | - Michael Untch
- Department of Gynecology and Obstetrics, Breast Cancer Center, Helios-Klinikum Berlin, Buch, Germany
| | - Anne Thieme
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69210, Heidelberg, Germany
| | - Volkmar Müller
- Department of Obstetrics and Gynecology, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Hamburg, Germany
| | - Eliane Taube
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
| | - Peter Fasching
- Brustzentrum, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wolfgang D Schmitt
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, University Hospital Heidelberg, Heidelberg, Germany
| | - Elmar Stickeler
- Klinik für Gynäkologie und Geburtsmedizin, Universitätsklinikum Aachen, Aachen, Germany
| | - Bruno V Sinn
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
| | - Paul Jank
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg, Marburg, Germany
| | | | - Frederick Klauschen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69210, Heidelberg, Germany.,Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | | | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg, Marburg, Germany
| | | | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69210, Heidelberg, Germany.,Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany.,Department of Neuropathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| |
Collapse
|
18
|
Morgani SM, Su J, Nichols J, Massagué J, Hadjantonakis AK. The transcription factor Rreb1 regulates epithelial architecture, invasiveness, and vasculogenesis in early mouse embryos. eLife 2021; 10:e64811. [PMID: 33929320 PMCID: PMC8131102 DOI: 10.7554/elife.64811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/16/2021] [Indexed: 12/23/2022] Open
Abstract
Ras-responsive element-binding protein 1 (Rreb1) is a zinc-finger transcription factor acting downstream of RAS signaling. Rreb1 has been implicated in cancer and Noonan-like RASopathies. However, little is known about its role in mammalian non-disease states. Here, we show that Rreb1 is essential for mouse embryonic development. Loss of Rreb1 led to a reduction in the expression of vasculogenic factors, cardiovascular defects, and embryonic lethality. During gastrulation, the absence of Rreb1 also resulted in the upregulation of cytoskeleton-associated genes, a change in the organization of F-ACTIN and adherens junctions within the pluripotent epiblast, and perturbed epithelial architecture. Moreover, Rreb1 mutant cells ectopically exited the epiblast epithelium through the underlying basement membrane, paralleling cell behaviors observed during metastasis. Thus, disentangling the function of Rreb1 in development should shed light on its role in cancer and other diseases involving loss of epithelial integrity.
Collapse
Affiliation(s)
- Sophie M Morgani
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Jeffrey Cheah Biomedical Centre Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Jie Su
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Jeffrey Cheah Biomedical Centre Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
19
|
Wu H, Gu J, Zhou D, Cheng W, Wang Y, Wang Q, Wang X. LINC00160 mediated paclitaxel-And doxorubicin-resistance in breast cancer cells by regulating TFF3 via transcription factor C/EBPβ. J Cell Mol Med 2020; 24:8589-8602. [PMID: 32652877 PMCID: PMC7412707 DOI: 10.1111/jcmm.15487] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 05/07/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Chemoresistance represents a major challenge in breast cancer (BC) treatment. This study aimed to probe the roles of LINC00160 in paclitaxel‐ and doxorubicin‐resistant BC cells. Three pairs of BC and adjacent normal tissue were used for lncRNA microarray analysis. Paclitaxel‐resistant MCF‐7 (MCF‐7/Tax) and doxorubicin‐resistant BT474 (BT474/Dox) cells were generated by exposure of parental drug‐sensitive MCF‐7 or BT474 cells to gradient concentrations of drugs. Correlation between LINC00160 expression and clinical response to paclitaxel in BC patients was examined. Short interfering RNAs specifically targeting LINC00160 or TFF3 were designed to construct LINC00160‐ and TFF3‐depleted BC cells to discuss their effects on biological episodes of MCF‐7/Tax and BT474/Dox cells. Interactions among LINC00160, transcription factor C/EBPβ and TFF3 were identified. MCF‐7/Tax and BT474/Dox cells stable silencing of LINC00160 were transplanted into nude mice. Consequently, up‐regulated LINC00160 led to poor clinical response to paclitaxel in BC patients. LINC00160 knockdown reduced drug resistance in MCF‐7/Tax and BT474/Dox cells and reduced cell migration and invasion. LINC00160 recruited C/EBPβ into the promoter region of TFF3 and increased TFF3 expression. LINC00160‐depleted MCF‐7/Tax and BT474/Dox cells showed decreased tumour growth rates in nude mice. Overall, we identified a novel mechanism of LINC00160‐mediated chemoresistance via the C/EBPβ/TFF3 axis, highlighting the potential of LINC00160 for treating BC with chemoresistance.
Collapse
Affiliation(s)
- Huaiguo Wu
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, China
| | - Juan Gu
- Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China.,Department of Pathology, The Fifth People's Hospital of Wuxi, The Medical School of Jiangnan University, Wuxi, China
| | - Daoping Zhou
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China
| | - Wei Cheng
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, China
| | - Yueping Wang
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China.,Department of Biology, College of Arts & Science, Massachusetts University, Boston, MA, USA
| | - Qingping Wang
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China
| | - Xuedong Wang
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, China.,Department of Medical Laboratory Science, The Fifth People's Hospital of Wuxi, Nanjing Medical University, Wuxi, China
| |
Collapse
|
20
|
Kaukonen D, Kaukonen R, Polit L, Hennessy BT, Lund R, Madden SF. Analysis of H3K4me3 and H3K27me3 bivalent promotors in HER2+ breast cancer cell lines reveals variations depending on estrogen receptor status and significantly correlates with gene expression. BMC Med Genomics 2020; 13:92. [PMID: 32620123 PMCID: PMC7333309 DOI: 10.1186/s12920-020-00749-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/25/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The role of histone modifications is poorly characterized in breast cancer, especially within the major subtypes. While epigenetic modifications may enhance the adaptability of a cell to both therapy and the surrounding environment, the mechanisms by which this is accomplished remains unclear. In this study we focus on the HER2 subtype and investigate two histone trimethylations that occur on the histone 3; the trimethylation located at lysine 4 (H3K4me3) found in active promoters and the trimethylation located at lysine 27 (H3K27me3) that correlates with gene repression. A bivalency state is the result of the co-presence of these two marks at the same promoter. METHODS In this study we investigated the relationship between these histone modifications in promoter regions and their proximal gene expression in HER2+ breast cancer cell lines. In addition, we assessed these patterns with respect to the presence or absence of the estrogen receptor (ER). To do this, we utilized ChIP-seq and matching RNA-seq from publicly available data for the AU565, SKBR3, MB361 and UACC812 cell lines. In order to visualize these relationships, we used KEGG pathway enrichment analysis, and Kaplan-Meyer plots. RESULTS We found that the correlation between the three types of promoter trimethylation statuses (H3K4me3, H3K27me3 or both) and the expression of the proximal genes was highly significant overall, while roughly a third of all genes are regulated by this phenomenon. We also show that there are several pathways related to cancer progression and invasion that are associated with the bivalent status of the gene promoters, and that there are specific differences between ER+ and ER- HER2+ breast cancer cell lines. These specific differences that are differentially trimethylated are also shown to be differentially expressed in patient samples. One of these genes, HIF1AN, significantly correlates with patient outcome. CONCLUSIONS This study highlights the importance of looking at epigenetic markings at a subtype specific level by characterizing the relationship between the bivalent promoters and gene expression. This provides a deeper insight into a mechanism that could lead to future targets for treatment and prognosis, along with oncogenesis and response to therapy of HER2+ breast cancer patients.
Collapse
Affiliation(s)
- Damien Kaukonen
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Riina Kaukonen
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Lélia Polit
- Institute Cochin, University Paris Descartes, Paris, France
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Riikka Lund
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Stephen F Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
21
|
Park AY, Han MR, Park KH, Kim JS, Son GS, Lee HY, Chang YW, Park EK, Cha SH, Cho Y, Hong H, Cho KR, Song SE, Woo OH, Lee JH, Cha J, Seo BK. Radiogenomic Analysis of Breast Cancer by Using B-Mode and Vascular US and RNA Sequencing. Radiology 2020; 295:24-34. [PMID: 32013793 DOI: 10.1148/radiol.2020191368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Radiogenomic investigations for breast cancer provide an understanding of tumor heterogeneity and discover image phenotypes of genetic variation. However, there is little research on the correlations between US features of breast cancer and whole-transcriptome profiling. Purpose To explore US phenotypes reflecting genetic alteration relevant to breast cancer treatment and prognosis by comparing US images of tumor with their RNA sequencing results. Materials and Methods From January to October 2016, B-mode and vascular US images in 31 women (mean age, 49 years ± 9 [standard deviation]) with breast cancer were prospectively analyzed. B-mode features included size, shape, echo pattern, orientation, margin, and calcifications. Vascular features were evaluated by using microvascular US and contrast agent-enhanced US: vascular index, vessel morphologic features, distribution, penetrating vessels, enhancement degree, order, margin, internal homogeneity, and perfusion defect. RNA sequencing was conducted with total RNA obtained from a surgical specimen by using next-generation sequencing. US features were compared with gene expression profiles, and ingenuity pathway analysis was used to analyze gene networks, enriched functions, and canonical pathways associated with breast cancer. The P value for differential expression was extracted by using a parametric F test comparing nested linear models. Results Thirteen US features were associated with various patterns of 340 genes (P < .05). Nonparallel orientation at B-mode US was associated with upregulation of TFF1 (log twofold change [log2FC] = 4.0; P < .001), TFF3 (log2FC = 2.5; P < .001), AREG (log2FC = 2.6; P = .005), and AGR3 (log2FC = 2.6; P = .003). Complex vessel morphologic structure was associated with upregulation of FZD8 (log2FC = 2.0; P = .01) and downregulation of IGF1R (log2FC = -2.0; P = .006) and CRIPAK (log2FC = -2.4; P = .01). The top networks with regard to orientation or vessel morphologic structure were associated with cell cycle, death, and proliferation. Conclusion Compared with RNA sequencing, B-mode and vascular US features reflected genomic alterations associated with hormone receptor status, angiogenesis, or prognosis in breast cancer. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ah Young Park
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Mi-Ryung Han
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Kyong Hwa Park
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Jung Sun Kim
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Gil Soo Son
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Hye Yoon Lee
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Young Woo Chang
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Eun Kyung Park
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Sang Hoon Cha
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Yunjung Cho
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Hyosun Hong
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Kyu Ran Cho
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Sung Eun Song
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Ok Hee Woo
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Ju-Han Lee
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Jaehyung Cha
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| | - Bo Kyoung Seo
- From the Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, 123 Jeokgeum-ro, Danwon-gu, Ansan city, Gyeonggi-do, 15355, Republic of Korea (A.Y.P., E.K.P., S.H.C., B.K.S.); Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea (A.Y.P.); Department of Laboratory Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (M.R.H., Y.C., H.H.); Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea (M.R.H.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.H.P.); Division of Hematology/Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.S.K.); Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (G.S.S., H.Y.L., Y.W.C.); Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea (K.R.C., S.E.S.); Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea (O.H.W.); Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.H.L.); and Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Gyeonggi-do, Republic of Korea (J.C.)
| |
Collapse
|
22
|
Kozina N, Mihaljević Z, Lončar MB, Mihalj M, Mišir M, Radmilović MD, Justić H, Gajović S, Šešelja K, Bazina I, Horvatić A, Matić A, Bijelić N, Rođak E, Jukić I, Drenjančević I. Impact of High Salt Diet on Cerebral Vascular Function and Stroke in Tff3-/-/C57BL/6N Knockout and WT (C57BL/6N) Control Mice. Int J Mol Sci 2019; 20:ijms20205188. [PMID: 31635131 PMCID: PMC6829871 DOI: 10.3390/ijms20205188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/20/2019] [Accepted: 10/05/2019] [Indexed: 11/29/2022] Open
Abstract
High salt (HS) dietary intake leads to impaired vascular endothelium-dependent responses to various physiological stimuli, some of which are mediated by arachidonic acid (AA) metabolites. Transgenic Tff3−/− gene knockout mice (Tff3−/−/C57BL/6N) have changes in lipid metabolism which may affect vascular function and outcomes of stroke. We aimed to study the effects of one week of HS diet (4% NaCl) on vascular function and stroke induced by transient occlusion of middle cerebral artery in Tff3−/− and wild type (WT/C57BL/6N) mice. Flow-induced dilation (FID) of carotid artery was reduced in WT-HS mice, but not affected in Tff3−/−-HS mice. Nitric oxide (NO) mediated FID. NO production was decreased with HS diet. On the contrary, acetylcholine-induced dilation was significantly decreased in Tff3−/− mice on both diets and WT-HS mice. HS intake and Tff3 gene depletion affected the structural components of the vessels. Proteomic analysis revealed a significant effect of Tff3 gene deficiency on HS diet-induced changes in neuronal structural proteins and acute innate immune response proteins’ expression and Tff3 depletion, but HS diet did not increase the stroke volume, which is related to proteome modification and upregulation of genes involved mainly in cellular antioxidative defense. In conclusion, Tff3 depletion seems to partially impair vascular function and worsen the outcomes of stroke, which is moderately affected by HS diet.
Collapse
Affiliation(s)
- Nataša Kozina
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Physiology and Immunology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Zrinka Mihaljević
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Physiology and Immunology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Mirela Baus Lončar
- Ruđer Bošković Institute, Department of Molecular Medicine; Bijenička 54, HR-10000 Zagreb, Croatia.
| | - Martina Mihalj
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Physiology and Immunology, J. Huttlera 4, HR-31000 Osijek, Croatia.
- Clinical Hospital Center Osijek, Dept of Dermatology and Venerology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Mihael Mišir
- Clinical Hospital Center Osijek, Neurology Clinic, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Marina Dobrivojević Radmilović
- University of Zagreb, School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia, Šalata 12, HR-10000 Zagreb, Croatia.
| | - Helena Justić
- University of Zagreb, School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia, Šalata 12, HR-10000 Zagreb, Croatia.
| | - Srećko Gajović
- University of Zagreb, School of Medicine, Croatian Institute for Brain Research, Zagreb, Croatia, Šalata 12, HR-10000 Zagreb, Croatia.
| | - Kate Šešelja
- Ruđer Bošković Institute, Department of Molecular Medicine; Bijenička 54, HR-10000 Zagreb, Croatia.
| | - Iva Bazina
- Ruđer Bošković Institute, Department of Molecular Medicine; Bijenička 54, HR-10000 Zagreb, Croatia.
| | - Anita Horvatić
- Proteomics laboratory, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55 HR-10000 Zagreb, Croatia.
| | - Anita Matić
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Physiology and Immunology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Nikola Bijelić
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Histology and Embriology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Edi Rođak
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Histology and Embriology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Ivana Jukić
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Physiology and Immunology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| | - Ines Drenjančević
- Faculty of Medicine Osijek, University Josip Juraj Strossmayer Osijek, Institute and Dept of Physiology and Immunology, J. Huttlera 4, HR-31000 Osijek, Croatia.
| |
Collapse
|
23
|
The Interaction of Helicobacter pylori with TFF1 and Its Role in Mediating the Tropism of the Bacteria Within the Stomach. Int J Mol Sci 2019; 20:ijms20184400. [PMID: 31500233 PMCID: PMC6769565 DOI: 10.3390/ijms20184400] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/27/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori colonises the human stomach and has tropism for the gastric mucin, MUC5AC. The majority of organisms live in the adherent mucus layer within their preferred location, close to the epithelial surface where the pH is near neutral. Trefoil factor 1 (TFF1) is a small trefoil protein co-expressed with the gastric mucin MUC5AC in surface foveolar cells and co-secreted with MUC5AC into gastric mucus. Helicobacter pylori binds with greater avidity to TFF1 dimer, which is present in gastric mucus, than to TFF1 monomer. Binding of H. pylori to TFF1 is mediated by the core oligosaccharide subunit of H. pylori lipopolysaccharide at pH 5.0–6.0. Treatment of H. pylori lipopolysaccharide with mannosidase or glucosidase inhibits its interaction with TFF1. Both TFF1 and H. pylori have a propensity for binding to mucins with terminal non-reducing α- or β-linked N-acetyl-d-glucosamine or α-(2,3) linked sialic acid or Gal-3-SO42−. These findings are strong evidence that TFF1 has carbohydrate-binding properties that may involve a conserved patch of aromatic hydrophobic residues on the surface of its trefoil domain. The pH-dependent lectin properties of TFF1 may serve to locate H. pylori deep in the gastric mucus layer close to the epithelium rather than at the epithelial surface. This restricted localisation could limit the interaction of H. pylori with epithelial cells and the subsequent host signalling events that promote inflammation.
Collapse
|
24
|
Zhang Z. Trefoil factor 3 knock-down prevents autophagy-related gene 12 elevation in colon adenocarcinoma. J Histotechnol 2019; 42:169-176. [PMID: 31362594 DOI: 10.1080/01478885.2019.1633088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Colon cancer, which is considered a common gastrointestinal cancer, has been the third leading cause of cancer mortality in the United States. Colon cancer has various histological sub-types and 90% of them are adenocarcinoma. In recent years, autophagy, the process by which cells are self-cannibalized, has been implicated in pathophysiology of various diseases including colon adenocarcinoma and thus, has become a strong research focus. This has also been true for trefoil factor 3 (TFF3). TFF3 is a small secreted peptide that is present in almost all mucin-secreting tissues, it is most abundant in goblet cells of the gastrointestinal tract and expressed at high protein levels in colon cancer. The present study analyzed the expression of TFF3 and autophagy-related gene ATG12 in cancerous and normal tissue samples collected from patients with colon adenocarcinoma. The expression of both proteins was shown to be increased in cancerous as compared to adjacent non-cancerous tissues. Furthermore, these proteins were shown to be positively correlated using the Pearson's Correlation test in cancerous tissues. Finally, TFF3 was shown to regulate ATG12 in human colon adenocarcinoma cells in vitro. Thus, the data presented here suggest that both TFF3 and ATG12 may be promising potential therapeutic targets to develop novel treatment strategies for patients with colon adenocarcinoma.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Cell Biology, College of Basic Medical Science, China Medical University, Taichung, China
| |
Collapse
|
25
|
Al-Salam S, Sudhadevi M, Awwad A, Al Bashir M. Trefoil factors peptide-3 is associated with residual invasive breast carcinoma following neoadjuvant chemotherapy. BMC Cancer 2019; 19:135. [PMID: 30744593 PMCID: PMC6371459 DOI: 10.1186/s12885-019-5316-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 01/24/2019] [Indexed: 12/12/2022] Open
Abstract
Background Breast carcinoma is the commonest cancer among UAE population and the most common cancer among females. Examination of the 5′ promoter regions of trefoil factor 3 (TFF3) gene has identified putative estrogen and progesterone receptor–DNA binding domains as direct response elements to estrogen and progesterone that are linked to breast functions or steroid regulation. The study was designed to determine the role of TFF3 in breast cancer chemoresistance with the aim of establishing TFF3 expression as a biomarker for drug resistance. Methods In total, 133 cases of breast carcinoma treated with neo-adjuvant therapy were collected. Tissue samples from pre-neoadjuvant therapy as well as tissues from post-neo-adjuvant therapy of those cases were collected and stained with immunohistochemistry for TFF3, Bcl2, BAX, cleaved caspase-3, AKT-1, NF kappa B and Ki-67. Results There was increased expression of TFF3 in residual invasive carcinoma cells. There was a significant correlation between the expression of TFF3 in breast carcinoma cells and response to neoadjuvant chemotherapy (p = 0.0165). There was significant co-expression of TFF3 with AKT1 (p = 0.0365), BCl2 (p = 0.0152), and NF Kappa-B (p = 0.0243) in breast carcinoma cases with residual carcinoma following neoadjuvant therapy which support the role of TFF3 in chemoresistance. Conclusion The expression of TFF3 is significantly associated with residual breast carcinoma following neoadjuvant chemotherapy suggesting its expression is associated with increased resistance to chemotherapy. This is supported by its co-expression with antiapoptotic proteins; BCl2, AKT1 and NF Kappa-B in residual breast carcinoma cells and very low proliferating index and apoptotic bodies in residual tumors. Electronic supplementary material The online version of this article (10.1186/s12885-019-5316-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suhail Al-Salam
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, AlAin, PO Box 17666, United Arab Emirates.
| | - Manjusha Sudhadevi
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, AlAin, PO Box 17666, United Arab Emirates
| | - Aktham Awwad
- Department of Laboratory Medicine, Tawam Hospital, AlAin, United Arab Emirates
| | | |
Collapse
|
26
|
Deng K, Mo S, Liu X, Chen J, Zhang Q, Chen X, Chen J, Dai S. Soy Foods Might Weaken the Sensitivity of Tamoxifen in Premenopausal Patients With Lumina A Subtype of Breast Cancer. Clin Breast Cancer 2019; 19:e337-e342. [PMID: 30733051 DOI: 10.1016/j.clbc.2018.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Based on estrogen active substances, many women consume soy foods in the belief that it could prevent breast cancer (BC). Women with different molecular subtypes would be likely to have diverse reactions to soy foods, especially those with the estrogen-receptor-positive (ER+) subtype. The aim of the current study is to identify the differentially expressed genes (DEGs) on soy foods in premenopausal patients with Lumina A subtype of BC (LABC) after soy food treatment, and to further investigate the critical molecule change. MATERIALS AND METHODS GSE58792 retrieved from Gene Expression Omnibus was analyzed to obtain DEGs using GEO2R. Gene Ontology and pathway enrichment analysis were performed using FunRich and GeneMINIA. Overall survival of critical genes was performed by the Kaplan-Meier plotter online tool. RESULTS A total of 108 DEGs were obtained from the dataset, among which 35 were up-regulated and 73 down-regulated. Soy foods significantly reduced the expression of TFF3, TFF1, GATA3, and ESR1, which were related to the activity of the ER-related pathway and the sensitivity of tamoxifen. Furthermore, the lower expressions of TOX3, FSIP1, ESR1, and CLGN were related to prolonged survival time of patients with BC. The most significant signaling pathways were epithelial-to-mesenchymal transition in up-regulated DEGs, mesenchymal-to-epithelial transition, and mammary gland alveolus development in down-regulated DEGs, which were all related to the development and prognosis of BC. CONCLUSIONS Soy foods could dramatically alter the ER-related gene profile in LABC. Particularly, down-regulated DEGs of TFF3, TFF1, GATA3, and ESR1 might weaken the sensitivity of tamoxifen and increase the efficacy of neoadjuvant chemotherapy in premenopausal patients with LABC.
Collapse
Affiliation(s)
- Kaifeng Deng
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Shanying Mo
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Xuexiang Liu
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Jifei Chen
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Qiaoyun Zhang
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Xiaoli Chen
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Jianming Chen
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China
| | - Shengming Dai
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, People's Republic of China.
| |
Collapse
|
27
|
Pandey V, Zhang M, You M, Zhang W, Chen R, Zhang W, Ma L, Wu ZS, Zhu T, Xu XQ, Lobie PE. Expression of two non-mutated genetic elements is sufficient to stimulate oncogenic transformation of human mammary epithelial cells. Cell Death Dis 2018; 9:1147. [PMID: 30451834 PMCID: PMC6242831 DOI: 10.1038/s41419-018-1177-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/29/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022]
Abstract
Trefoil factor 3 (TFF3) expression is positively associated with advanced clinicopathological features of mammary carcinoma (MC). Herein, we provide evidence for a functional role of TFF3 in oncogenic transformation of immortalized, but otherwise normal human mammary epithelial cells (HMECs), namely, HMEC-hTERT, MCF10A, and MCF12A. Forced expression of TFF3 in immortalized-HMECs enhanced cell proliferation, cell survival, anchorage-independent growth, produced highly disorganised three-dimensional (3D) acinar structures and generated tumours in immunocompromised mice. Forced expression of TFF3 in immortalized-HMECs stimulated STAT3 activity that was required for TFF3-stimulated cell proliferation, survival, and anchorage-independent growth. TFF3 specifically utilised STAT3 activity to govern a transcriptional program, which was required for TFF3-stimulated oncogenic transformation of immortalized-HMECs, including transcriptional upregulation of CCND1 and BCL2. siRNA-mediated depletion or functional inhibition of STAT3 significantly inhibited the TFF3-stimulated transcription of CCND1 and BCL2 and oncogenicity in immortalized-HMECs. Furthermore, DOX-inducible expression of TFF3 in HMEC-hTERT cells also permitted anchorage-independent growth and produced disorganized acinar structures in 3D Matrigel culture. Removal of DOX-induced expression of TFF3 in HMEC-hTERT cells, previously grown with DOX, resulted in efficient normalisation of the disorganized acinar architecture and attenuated cell viability in Matrigel culture. Cumulatively, these findings suggest that TFF3 is a potent oncogene and its increased expression along with hTERT in HMECs is sufficient to produce oncogenic transformation.
Collapse
Affiliation(s)
- Vijay Pandey
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, Guangdong, PR China
| | - Min Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Mingliang You
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Weijie Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Rumei Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wei Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Tianhe District, Guangzhou, Guangdong, PR China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Lan Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, Guangdong, PR China
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiu Qin Xu
- Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, Fujian, PR China.
| | - Peter E Lobie
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, Guangdong, PR China. .,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Lin X, Zhang H, Dai J, Zhang W, Zhang J, Xue G, Wu J. TFF3 Contributes to Epithelial-Mesenchymal Transition (EMT) in Papillary Thyroid Carcinoma Cells via the MAPK/ERK Signaling Pathway. J Cancer 2018; 9:4430-4439. [PMID: 30519349 PMCID: PMC6277656 DOI: 10.7150/jca.24361] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 08/24/2018] [Indexed: 12/20/2022] Open
Abstract
Trefoil factor 3 (TFF3) was found to be overexpressed in many types of tumours. Evidence has shown that TFF3 plays an important role in tumour proliferation, migration and invasion metastasis. However, the impact of TFF3 on patients' clinicopathological characteristics and underlying mechanisms remain unknown in papillary thyroid carcinoma (PTC). In this study, the expression of TFF3 and the epithelial-mesenchymal transition (EMT) transcriptional factor Snail in PTC and para-carcinoma specimens was evaluated by immunohistochemistry (IHC) and Western blot, and the possible associations with lymph node (LN) metastasis and other clinicopathological parameters were analysed. In vitro, the effect of TFF3 on the malignant behaviour of TPC-1 cells was evaluated by cell proliferation assays, cell adhesion assays, colony formation assays, wound-healing assays and transwell chamber invasion assays. EMT markers and regulatory molecules were detected by quantitative RT-PCR (qRT-PCR) and Western blot analysis in the TFF3-knockdown groups and shRNA control group. The results showed that TFF3 was upregulated in PTC tissue and was associated with lymph node metastasis (P=0.0001), pathological grade (P=0.0002) and Snail expression (P=0.0001). The knockdown of TFF3 markedly inhibited the abilities of TPC-1 cell proliferation, adhesion, colony formation, migration and invasion. Mechanically, the results demonstrated that TFF3 might activate the MAPK/ERK signalling pathways, affect the expression of the transcription factors snail and slug in addition to affecting EMT associated markers E-cadherin and N-cadherin, and accelerate the progression of EMT in TPC-1 cells. These findings indicate that TFF3 might promote the metastatic potential of PTC by promoting the EMT process through cascades of MAPK/ERK pathways.
Collapse
Affiliation(s)
- Xu Lin
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, 075000, China
| | - Huiqin Zhang
- Department of Ophthalmology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Jin Dai
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, 075000, China
| | - Wenjing Zhang
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, 075000, China
| | - Jing Zhang
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, 075000, China
| | - Gang Xue
- Department of Otorhinolaryngology Head and Neck Surgery, Hebei North University, Zhangjiakou, 075000, China
| | - Jingfang Wu
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, 075000, China
| |
Collapse
|
29
|
TFF1 and TFF3 mRNAs Are Higher in Blood from Breast Cancer Patients with Metastatic Disease than Those without. JOURNAL OF ONCOLOGY 2018; 2018:4793498. [PMID: 29977293 PMCID: PMC6011051 DOI: 10.1155/2018/4793498] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 03/03/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Introduction Breast cancer metastasis occurs when tumor cells dissociate from the primary tumor and migrate to distant organs through the peripheral bloodstream or lymphatic drainage. Circulating tumor cells (CTCs) originate from primary sites or metastases and circulate in the patients' bloodstream. Molecular assays for the detection and molecular characterization of CTCs can serve as a liquid biopsy and can represent an alternative to invasive biopsies as a source of tumor tissue in the metastatic patients. Patients and Methods We analyzed the presence of CTCs in the peripheral blood of 50 breast cancer patients by quantitative real-time reverse transcriptase polymerase chain reaction (RT-qPCR) to detect trefoil factor family (TFF) 1 and 3 genes. Results We found significant difference in the level of both TFF1 and TFF3 mRNA in the blood of nonmetastatic versus metastatic breast cancer patients (p= 0.001 and p= 0.038, respectively). TFF1 mRNA was detected at higher levels in 34.6% of metastatic breast cancer patients as compared to 0% of nonmetastatic (p= 0.002). As regards TFF3 mRNA, it was detected at higher levels in 46.2% of metastatic breast cancer patients as compared to 4% of nonmetastatic (p= 0.026). Moreover, we found that the high level of both TFF1 and TFF3 mRNA was related to estrogen status of the patients. The detection of high level of TFF1 mRNA in CTCs was associated with bone metastases (77.8%), while that of TFF3 was related to lymph node involvement (75%) and lung metastases (68.8%). Conclusion The combined measurement of both TFF1 and TFF3 mRNA level for differentiation of metastatic from nonmetastatic breast cancer gave 57.69% sensitivity and 83.3% specificity.
Collapse
|
30
|
Tolušić Levak M, Mihalj M, Koprivčić I, Lovrić I, Novak S, Bijelić N, Baus-Lončar M, Belovari T, Kralik K, Pauzar B. Differential Expression of TFF Genes and Proteins in Breast Tumors. Acta Clin Croat 2018; 57:264-277. [PMID: 30431719 PMCID: PMC6532012 DOI: 10.20471/acc.2018.57.02.06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
SUMMARY – The objective of this study was to determine differential expression of TFF1, TFF2 and TFF3 genes and proteins in breast tumor subtypes. In addition, we investigated the correlation between TFF genes within tumor subgroups, and TFF genes with clinical and pathologic characteristics of the tumor. Study group included 122 patients with surgically removed breast tumors. Samples were investigated using qRT-PCR and immunohistochemistry. TFF1 and TFF3 genes and proteins were expressed in breast tumors, while the levels of TFF2 gene and protein expression were very low or undetectable. TFF1 was significantly more expressed in benign tumors, while TFF3 was more expressed in malignant tumors. Gene and protein expression of both TFF1 and TFF3 was greater in lymph node-negative tumors, hormone positive tumors, tumors with moderate levels of Ki67 expression, and in grade II tumors. A strong positive correlation was found between TFF1 and TFF3 genes, and the expression of both negatively correlated with Ki67 and the level of tumor histologic differentiation. Our results suggest that TFF1 and TFF3, but not TFF2, may have a role in breast tumor pathogenesis and could be used in the assessment of tumor differentiation and malignancy.
Collapse
Affiliation(s)
| | - Martina Mihalj
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Ivan Koprivčić
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Department of Surgery, Osijek University Hospital Centre, Osijek, Croatia
| | - Ivana Lovrić
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Department of Anatomy, Histology and Embryology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Sanja Novak
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Nikola Bijelić
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Mirela Baus-Lončar
- Department of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Tatjana Belovari
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Kristina Kralik
- Department of Medical Statistics and Informatics, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Biljana Pauzar
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Department of Clinical Cytology, Osijek University Hospital Centre, Osijek, Croatia
| |
Collapse
|
31
|
Glycolytic inhibitor 2-Deoxy-d-Glucose activates migration and invasion in glioblastoma cells through modulation of the miR-7-5p/TFF3 signaling pathway. Biochem Biophys Res Commun 2018; 499:829-835. [DOI: 10.1016/j.bbrc.2018.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/01/2018] [Indexed: 02/06/2023]
|
32
|
Comprehensive Evaluation of TFF3 Promoter Hypomethylation and Molecular Biomarker Potential for Prostate Cancer Diagnosis and Prognosis. Int J Mol Sci 2017; 18:ijms18092017. [PMID: 28930171 PMCID: PMC5618665 DOI: 10.3390/ijms18092017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/08/2017] [Accepted: 09/13/2017] [Indexed: 01/03/2023] Open
Abstract
Overdiagnosis and overtreatment of clinically insignificant tumors remains a major problem in prostate cancer (PC) due to suboptimal diagnostic and prognostic tools. Thus, novel biomarkers are urgently needed. In this study, we investigated the biomarker potential of Trefoil factor 3 (TFF3) promoter methylation and RNA expression levels for PC. Initially, by quantitative methylation specific PCR (qMSP) analysis of a large radical prostatectomy (RP) cohort (n = 292), we found that the TFF3 promoter was significantly hypomethylated in PC compared to non-malignant (NM) prostate tissue samples (p < 0.001) with an AUC (area under the curve) of 0.908 by receiver operating characteristics (ROC) curve analysis. Moreover, significant TFF3 promoter hypomethylation (p ≤ 0.010) as well as overexpression (p < 0.001) was found in PC samples from another large independent patient sample set (498 PC vs. 67 NM) analyzed by Illumina 450K DNA methylation arrays and/or RNA sequencing. TFF3 promoter methylation and transcriptional expression levels were inversely correlated, suggesting that epigenetic mechanisms contribute to the regulation of gene activity. Furthermore, low TFF3 expression was significantly associated with high ERG, ETS transcription factor (ERG) expression (p < 0.001), as well as with high Gleason score (p < 0.001), advanced pathological T-stage (p < 0.001), and prostate-specific antigen (PSA) recurrence after RP (p = 0.013; univariate Cox regression analysis). There were no significant associations between TFF3 promoter methylation levels, ERG status, or PSA recurrence in these RP cohorts. In conclusion, our results demonstrated diagnostic biomarker potential of TFF3 promoter hypomethylation for PC as well as prognostic biomarker potential of TFF3 RNA expression. To the best of our knowledge, this is the most comprehensive study of TFF3 promoter methylation and transcriptional expression in PC to date.
Collapse
|
33
|
Pandey V, Zhang M, Chong QY, You M, Raquib AR, Pandey AK, Liu DX, Liu L, Ma L, Jha S, Wu ZS, Zhu T, Lobie PE. Hypomethylation associated enhanced transcription of trefoil factor-3 mediates tamoxifen-stimulated oncogenicity of ER+ endometrial carcinoma cells. Oncotarget 2017; 8:77268-77291. [PMID: 29100386 PMCID: PMC5652779 DOI: 10.18632/oncotarget.20461] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/16/2017] [Indexed: 12/20/2022] Open
Abstract
Tamoxifen (TAM) is widely used as an adjuvant therapy for women with breast cancer (BC). However, TAM possesses partial oestrogenic activity in the uterus and its use has been associated with an increased incidence of endometrial carcinoma (EC). The molecular mechanism for these observations is not well understood. Herein, we demonstrated that forced expression of Trefoil factor 3 (TFF3), in oestrogen receptor-positive (ER+) EC cells significantly increased cell cycle progression, cell survival, anchorage-independent growth, invasiveness and tumour growth in xenograft models. Clinically, elevated TFF3 protein expression was observed in EC compared with normal endometrial tissue, and its increased expression in EC was significantly associated with myometrial invasion. TAM exposure increased expression of TFF3 in ER+ EC cells and its elevated expression resulted in increased oncogenicity and invasiveness. TAM-stimulated expression of TFF3 in EC cells was associated with hypomethylation of the TFF3 promoter sequence and c-JUN/SP1-dependent transcriptional activation. In addition, small interfering (si) RNA-mediated depletion or polyclonal antibody inhibition of TFF3 significantly abrogated oncogenicity and invasiveness in EC cells consequent to TAM induction or forced expression of TFF3. Hence, TAM-stimulated upregulation of TFF3 in EC cells was critical in promoting EC progression associated with TAM treatment. Importantly, inhibition of TFF3 function might be an attractive molecular modality to abrogate the stimulatory effects of TAM on endometrial tissue and to limit the progression of EC.
Collapse
Affiliation(s)
- Vijay Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Min Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, P.R. China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Mingliang You
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Amit K Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Dong-Xu Liu
- School of Science, Auckland University of Technology, Auckland, New Zealand
| | - Liang Liu
- Department of Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, P.R China.,Department of Radiology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, P.R China
| | - Lan Ma
- Tsinghua Berkeley Shenzhen Institute, Division of Life Sciences & Health, Tsinghua University Graduate School, Shenzhen, P.R China
| | - Sudhakar Jha
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, P.R China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, P.R. China
| | - Peter E Lobie
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, National University of Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Division of Life Sciences & Health, Tsinghua University Graduate School, Shenzhen, P.R China
| |
Collapse
|
34
|
Diao S, Zheng Q, Gao J, Yao Y, Ren S, Liu Y, Xu Y. Trefoil factor 3 contributes to the malignancy of glioma via regulating HIF-1α. Oncotarget 2017; 8:76770-76782. [PMID: 29100347 PMCID: PMC5652741 DOI: 10.18632/oncotarget.20010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
Trefoil factor 3 (TFF3) plays significant roles in several solid tumors. However, the expression pattern and function of TFF3 in glioblastoma (GBM) have not been reported. Here, we report that expression level of TFF3 significantly elevated in glioma and correlated with the prognosis of glioma patients. Then we found TFF3 promotes proliferation, invasion, and migration and inhibits apoptosis of glioma cells in vitro, and delayed tumor progression in subcutaneous xenograft nude mice, and prolonged the median survival time in orthotopic xenograft mice. Moreover, knockdown of TFF3 reduced the expression of HIF-1α through a hypoxia-independent manner. These findings suggest that targeting TFF3 may offer a novel strategy for therapeutic intervention of malignant gliomas.
Collapse
Affiliation(s)
- Shuo Diao
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medical College, China Medical University, Shenyang, People's Republic of China
| | - Jian Gao
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, People's Republic of China
| | - Yiqun Yao
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Siyang Ren
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Yongjian Liu
- Department of Interventional Therapy, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Yinghui Xu
- Department of Neurosurgery, First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| |
Collapse
|
35
|
Ishibashi Y, Ohtsu H, Ikemura M, Kikuchi Y, Niwa T, Nishioka K, Uchida Y, Miura H, Aikou S, Gunji T, Matsuhashi N, Ohmoto Y, Sasaki T, Seto Y, Ogawa T, Tada K, Nomura S. Serum TFF1 and TFF3 but not TFF2 are higher in women with breast cancer than in women without breast cancer. Sci Rep 2017; 7:4846. [PMID: 28687783 PMCID: PMC5501858 DOI: 10.1038/s41598-017-05129-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/24/2017] [Indexed: 12/31/2022] Open
Abstract
Breast cancer remains a common malignancy in women, but the take-up for breast cancer screening programs in Japan is still low, possibly due to its perceived inconvenience. TFF1 and TFF3 are expressed in both breast cancer tissue and normal breast. Serum trefoil proteins were reported as cancer screening markers for gastric, prostate, lung, pancreatic cancer and cholangio carcinoma. The purpose of this study was to examine whether serum trefoil proteins could be screening biomarkers for breast cancer. Serum trefoil proteins in 94 breast cancer patients and 84 health check females were measured by ELISA. Serum TFF1 and TFF3 were significantly higher and serum TFF2 was significantly lower in breast cancer patients. Area under the curve of receiver operating characteristic of TFF1, TFF2, and TFF3 was 0.69, 0.83, and. 0.72, respectively. AUC of the combination of TFF1, TFF2, and TFF3 was 0.96. Immunohistochemically, TFF1 expression was positive in 56.5% and TFF3 was positive in 73.9% of breast cancers, while TFF2 was negative in all tumors. Serum TFF1 had positive correlation with expression of TFF1 in breast cancer tissue. Serum concentrations of TFF1 and TFF3 but not TFF2 are higher in women with breast cancer than in women without breast cancer.
Collapse
Affiliation(s)
- Yuko Ishibashi
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ohtsu
- Center of Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masako Ikemura
- Department of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Yasuko Kikuchi
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takayoshi Niwa
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kotoe Nishioka
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Uchida
- Breast Center, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Hirona Miura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Susumu Aikou
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | - Yasukazu Ohmoto
- Otsuka Pharmaceutical Tokusima Research Institute, Tokyo, Japan
| | - Takeshi Sasaki
- Department of Pathology, The University of Tokyo Hospital, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihisa Ogawa
- Breast Center, Dokkyo Medical University Koshigaya Hospital, Tokyo, Japan
| | - Keiichiro Tada
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
36
|
Release of HER2 repression of trefoil factor 3 (TFF3) expression mediates trastuzumab resistance in HER2+/ER+ mammary carcinoma. Oncotarget 2017; 8:74188-74208. [PMID: 29088778 PMCID: PMC5650333 DOI: 10.18632/oncotarget.18431] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/10/2017] [Indexed: 12/15/2022] Open
Abstract
HER2+/ER+ breast cancer, a subset of the luminal B subtype, makes up approximately 10% of all breast cancers. The bidirectional crosstalk between HER2 and estrogen receptor (ER) in HER2+/ER+ breast cancer contributes to resistance towards both anti-estrogens and HER2-targeted therapies. TFF3 promotes breast cancer progression and has been implicated in anti-estrogen resistance in breast cancer. Herein, we investigated the cross-regulation between HER2 and estrogen-responsive TFF3, and the role of TFF3 in mediating trastuzumab resistance in HER2+/ER+ breast cancer. TFF3 expression was decreased by HER2 activation, and increased by inhibition of HER2 with trastuzumab in HER2+/ER+ breast cancer cells, partially in an ERα-independent manner. In contrast, the forced expression of TFF3 activated the entire HER family of receptor tyrosine kinases (HER1-4). Hence, HER2 negatively regulates its own signalling through the transcriptional repression of TFF3, while trastuzumab inhibition of HER2 results in increased TFF3 expression to compensate for the loss of HER2 signalling. In HER2+/ER+ breast cancer cells with acquired trastuzumab resistance, TFF3 expression was markedly upregulated and associated with a corresponding decrease in HER signalling. siRNA mediated depletion or small molecule inhibition of TFF3 decreased the survival and growth advantage of the trastuzumab resistant cells without re-sensitization to trastuzumab. Furthermore, TFF3 inhibition abrogated the enhanced cancer stem cell-like behaviour in trastuzumab resistant HER2+/ER+ breast cancer cells. Collectively, TFF3 may function as a potential biomarker and therapeutic target in trastuzumab resistant HER2+/ER+ breast cancer.
Collapse
|
37
|
El-Balat A, Schmeil I, Karn T, Becker S, Sänger N, Holtrich U, Arsenic R. TFF3 Expression as Stratification Marker in Borderline Epithelial Tumors of the Ovary. Pathol Oncol Res 2017; 24:277-282. [PMID: 28470574 DOI: 10.1007/s12253-017-0240-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/25/2017] [Indexed: 12/17/2022]
Abstract
Borderline tumors (BOT) of the ovary account for 10% to 20% of ovarian neoplasms. Like ovarian cancer, BOT encompass several different histological subtypes (serous, mucinous, endometrioid, clear cell, transitional cell and mixed) with serous (SBOT) and mucinous (MBOT) the most common. Current hypotheses suggest low-grade serous carcinoma may develop in a stepwise fashion from SBOT whereas the majority of high grade serous carcinomas develop rapidly presumably from inclusion cysts or ovarian surface epithelium. The pathogenesis of mucinous ovarian tumors is still puzzling. Molecular markers could help to better define relationships between such entities. Trefoil factor-3 (TFF3) is an estrogen-regulated gene associated with prognosis in different types of cancer. It has also been included in a recent marker panel predicting subtypes of ovarian carcinoma. We analyzed the expression of TFF3 by immunohistochemistry in a cohort of 137 BOT and its association with histopathological features. Overall expression rate of TFF3 was 21.9%. None of the BOT with serous and endometrioid histology displayed strong TFF3 expression. On the other hand, TFF3 was highly expressed in 61.4% of MBOT cases and 33.3% of BOT with mixed histology (P < 0.001) suggesting a potential function of the protein in that subtypes. Associations of TFF3 expression with FIGO stage and micropapillary pattern were significant in the overall cohort but confounded by their correlation with histological subtypes. The highly specific expression of TFF3 in MBOT may help to further clarify potential relationships of tumors with mucinous histology and warrants further studies.
Collapse
Affiliation(s)
- Ahmed El-Balat
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Iryna Schmeil
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Karn
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Sven Becker
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Nicole Sänger
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Uwe Holtrich
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ruza Arsenic
- Institute of Pathology, Charite University Hospital, Chariteplatz 1, 10117, Berlin, Germany
| |
Collapse
|
38
|
Epigenetic basis of cancer health disparities: Looking beyond genetic differences. Biochim Biophys Acta Rev Cancer 2017; 1868:16-28. [PMID: 28108348 DOI: 10.1016/j.bbcan.2017.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/07/2017] [Accepted: 01/16/2017] [Indexed: 12/18/2022]
Abstract
Despite efforts at various levels, racial health disparities still exist in cancer patients. These inequalities in incidence and/or clinical outcome can only be explained by a multitude of factors, with genetic basis being one of them. Several investigations have provided convincing evidence to support epigenetic regulation of cancer-associated genes, which results in the differential transcriptome and proteome, and may be linked to a pre-disposition of individuals of certain race/ethnicity to early or more aggressive cancers. Recent technological advancements and the ability to quickly analyze whole genome have aided in these efforts, and owing to their relatively easy detection, methylation events are much well-characterized, than the acetylation events, across human populations. The early trend of investigating a pre-determined set of genes for differential epigenetic regulation is paving way for more unbiased screening. This review summarizes our current understanding of the epigenetic events that have been tied to the racial differences in cancer incidence and mortality. A better understanding of the epigenetics of racial diversity holds promise for the design and execution of novel strategies targeting the human epigenome for reducing the disparity gaps.
Collapse
|
39
|
Stiegel MA, Pleil JD, Sobus JR, Angrish MM, Morgan MK. Kidney injury biomarkers and urinary creatinine variability in nominally healthy adults. Biomarkers 2015; 20:436-52. [DOI: 10.3109/1354750x.2015.1094136] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- M. A. Stiegel
- Department of Environmental Sciences and Engineering, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA,
- ORISE, US EPA, Research Triangle Park, NC, USA, and
| | - J. D. Pleil
- National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - J. R. Sobus
- National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | | | - M. K. Morgan
- National Exposure Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
40
|
Thomsen KG, Lyng MB, Elias D, Vever H, Knoop AS, Lykkesfeldt AE, Lænkholm AV, Ditzel HJ. Gene expression alterations associated with outcome in aromatase inhibitor-treated ER+ early-stage breast cancer patients. Breast Cancer Res Treat 2015; 154:483-94. [PMID: 26585578 DOI: 10.1007/s10549-015-3644-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 11/13/2015] [Indexed: 01/05/2023]
Abstract
Aromatase inhibitors (AI), either alone or together with chemotherapy, have become the standard adjuvant treatment for postmenopausal, estrogen receptor-positive (ER+) breast cancer. Although AIs improve overall survival, resistance is still a major clinical problem, thus additional biomarkers predictive of outcome of ER+ breast cancer patients treated with AIs are needed. Global gene expression analysis was performed on ER+ primary breast cancers from patients treated with adjuvant AI monotherapy; half experienced recurrence (median follow-up 6.7 years). Gene expression alterations were validated by qRT-PCR, and functional studies evaluating the effect of siRNA-mediated gene knockdown on cell growth were performed. Twenty-six genes, including TFF3, DACH1, RGS5, and GHR, were shown to exhibit altered expression in tumors from patients with recurrence versus non-recurrent (fold change ≥1.5, p < 0.05), and the gene expression alterations were confirmed using qRT-PCR. Ten of these 26 genes could be linked in a network associated with cellular proliferation, growth, and development. TFF3, which encodes for trefoil factor 3 and is an estrogen-responsive oncogene shown to play a functional role in tamoxifen resistance and metastasis of ER+ breast cancer, was also shown to be upregulated in an AI-resistant cell line model, and reduction of TFF3 levels using TFF3-specific siRNAs decreased the growth of both the AI-resistant and -sensitive parental cell lines. Moreover, overexpression of TFF3 in parental AI-sensitive MCF-7/S0.5 cells resulted in reduced sensitivity to the AI exemestane, whereas TFF3 overexpression had no effect on growth in the absence of exemestane, indicating that TFF3 mediates growth and survival signals that abrogate the growth inhibitory effect of exemestane. We identified a panel of 26 genes exhibiting altered expression associated with disease recurrence in patients treated with adjuvant AI monotherapy, including TFF3, which was shown to exhibit a growth- and survival-promoting effect in the context of AI treatment.
Collapse
Affiliation(s)
- Karina G Thomsen
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Maria B Lyng
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Daniel Elias
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Henriette Vever
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Ann S Knoop
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Anne E Lykkesfeldt
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | | | - Henrik J Ditzel
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark.
- Department of Oncology, Odense University Hospital, 5000, Odense, Denmark.
| |
Collapse
|
41
|
Lau WH, Pandey V, Kong X, Wang XN, Wu Z, Zhu T, Lobie PE. Trefoil Factor-3 (TFF3) Stimulates De Novo Angiogenesis in Mammary Carcinoma both Directly and Indirectly via IL-8/CXCR2. PLoS One 2015; 10:e0141947. [PMID: 26559818 PMCID: PMC4641663 DOI: 10.1371/journal.pone.0141947] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/13/2015] [Indexed: 12/19/2022] Open
Abstract
Mammary carcinoma cells produce pro-angiogenic factors to stimulate angiogenesis and tumor growth. Trefoil factor-3 (TFF3) is an oncogene secreted from mammary carcinoma cells and associated with poor prognosis. Herein, we demonstrate that TFF3 produced in mammary carcinoma cells functions as a promoter of tumor angiogenesis. Forced expression of TFF3 in mammary carcinoma cells promoted proliferation, survival, invasion and in vitro tubule formation of human umbilical vein endothelial cells (HUVEC). MCF7-TFF3 cells with forced expression of TFF3 generated tumors with enhanced microvessel density as compared to tumors formed by vector control cells. Depletion of TFF3 in mammary carcinoma cells by siRNA concordantly decreased the angiogenic behavior of HUVEC. Forced expression of TFF3 in mammary carcinoma cells stimulated IL-8 transcription and subsequently enhanced IL-8 expression in both mammary carcinoma cells and HUVEC. Depletion of IL-8 in mammary carcinoma cells with forced expression of TFF3, or antibody inhibition of IL-8, partially abrogated mammary carcinoma cell TFF3-stimulated HUVEC angiogenic behavior in vitro, as did inhibition of the IL-8 receptor, CXCR2. Depletion of STAT3 by siRNA in MCF-7 cells with forced expression of TFF3 partially diminished the angiogenic capability of TFF3 on stimulation of cellular processes of HUVEC. Exogenous recombinant hTFF3 also directly promoted the angiogenic behavior of HUVEC. Hence, TFF3 is a potent angiogenic factor and functions as a promoter of de novo angiogenesis in mammary carcinoma, which may co-coordinate with the growth promoting and metastatic actions of TFF3 in mammary carcinoma to enhance tumor progression.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blotting, Western
- Breast Neoplasms/blood supply
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cells, Cultured
- Coculture Techniques
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Interleukin-8/genetics
- Interleukin-8/metabolism
- MCF-7 Cells
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Fluorescence
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Peptides/genetics
- Peptides/metabolism
- RNA Interference
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Transplantation, Heterologous
- Trefoil Factor-3
Collapse
Affiliation(s)
- Wai-Hoe Lau
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vijay Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xiangjun Kong
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiao-Nan Wang
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
- Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - ZhengSheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
- * E-mail: (PEL); (TZ)
| | - Peter E Lobie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- National University Cancer Institute of Singapore, National Health System, National University of Singapore, Singapore, Singapore
- * E-mail: (PEL); (TZ)
| |
Collapse
|
42
|
Xiao P, Ling H, Lan G, Liu J, Hu H, Yang R. Trefoil factors: Gastrointestinal-specific proteins associated with gastric cancer. Clin Chim Acta 2015; 450:127-34. [PMID: 26265233 DOI: 10.1016/j.cca.2015.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/11/2022]
Abstract
Trefoil factor family (TFF), composed of TFF1, TFF2, and TFF3, is a cluster of secreted peptides characterized by trefoil domain (s) and C-terminal dimerization domain. TFF1, a gastric tumor suppressor, is a single trefoil peptide originally detected in breast cancer cell lines but expressed mainly in the stomach; TFF2, a candidate of gastric cancer suppressor with two trefoil domains, is abundant in the stomach and duodenal Brunner's glands; and TFF3 is another single trefoil peptide expressed throughout the intestine which can promote the development of gastric carcinoma. According to multiple studies, TFFs play a regulatory function in the mammals' digestive system, namely in mucosal protection and epithelial cell reconstruction, tumor suppression or promotion, signal transduction and the regulation of proliferation and apoptosis. Action mechanisms of TFFs remain unresolved, but the recent demonstration of a GKN (gastrokine) 2-TFF1 heterodimer implicates structural and functional interplay with gastrokines. This review aims to encapsulate the structural and biological characteristics of TFF.
Collapse
Affiliation(s)
- Ping Xiao
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui Ling
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China.
| | - Gang Lan
- Key Laboratory for Atherosclerology of Hunan Province, Cardiovascular Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Jiao Liu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Haobin Hu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Ruirui Yang
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| |
Collapse
|
43
|
Milioli HH, Vimieiro R, Riveros C, Tishchenko I, Berretta R, Moscato P. The Discovery of Novel Biomarkers Improves Breast Cancer Intrinsic Subtype Prediction and Reconciles the Labels in the METABRIC Data Set. PLoS One 2015; 10:e0129711. [PMID: 26132585 PMCID: PMC4488510 DOI: 10.1371/journal.pone.0129711] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/12/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The prediction of breast cancer intrinsic subtypes has been introduced as a valuable strategy to determine patient diagnosis and prognosis, and therapy response. The PAM50 method, based on the expression levels of 50 genes, uses a single sample predictor model to assign subtype labels to samples. Intrinsic errors reported within this assay demonstrate the challenge of identifying and understanding the breast cancer groups. In this study, we aim to: a) identify novel biomarkers for subtype individuation by exploring the competence of a newly proposed method named CM1 score, and b) apply an ensemble learning, as opposed to the use of a single classifier, for sample subtype assignment. The overarching objective is to improve class prediction. METHODS AND FINDINGS The microarray transcriptome data sets used in this study are: the METABRIC breast cancer data recorded for over 2000 patients, and the public integrated source from ROCK database with 1570 samples. We first computed the CM1 score to identify the probes with highly discriminative patterns of expression across samples of each intrinsic subtype. We further assessed the ability of 42 selected probes on assigning correct subtype labels using 24 different classifiers from the Weka software suite. For comparison, the same method was applied on the list of 50 genes from the PAM50 method. CONCLUSIONS The CM1 score portrayed 30 novel biomarkers for predicting breast cancer subtypes, with the confirmation of the role of 12 well-established genes. Intrinsic subtypes assigned using the CM1 list and the ensemble of classifiers are more consistent and homogeneous than the original PAM50 labels. The new subtypes show accurate distributions of current clinical markers ER, PR and HER2, and survival curves in the METABRIC and ROCK data sets. Remarkably, the paradoxical attribution of the original labels reinforces the limitations of employing a single sample classifiers to predict breast cancer intrinsic subtypes.
Collapse
Affiliation(s)
- Heloisa Helena Milioli
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Environmental and Life Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Renato Vimieiro
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Centro de Informática, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Carlos Riveros
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Inna Tishchenko
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Regina Berretta
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Pablo Moscato
- Priority Research Centre for Bioinformatics, Biomarker Discovery and Information-Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Electrical Engineering and Computer Science, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
44
|
May FEB, Westley BR. TFF3 is a valuable predictive biomarker of endocrine response in metastatic breast cancer. Endocr Relat Cancer 2015; 22:465-79. [PMID: 25900183 PMCID: PMC4455223 DOI: 10.1530/erc-15-0129] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2015] [Indexed: 12/13/2022]
Abstract
The stratification of breast cancer patients for endocrine therapies by oestrogen or progesterone receptor expression is effective but imperfect. The present study aims were to validate microarray studies that demonstrate TFF3 regulation by oestrogen and its association with oestrogen receptors in breast cancer, to evaluate TFF3 as a biomarker of endocrine response, and to investigate TFF3 function. Microarray data were validated by quantitative RT-PCR and northern and western transfer analyses. TFF3 was induced by oestrogen, and its induction was inhibited by antioestrogens, tamoxifen, 4-hydroxytamoxifen and fulvestrant in oestrogen-responsive breast cancer cells. The expression of TFF3 mRNA was associated with oestrogen receptor mRNA in breast tumours (Pearson's coefficient=0.762, P=0.000). Monoclonal antibodies raised against the TFF3 protein detected TFF3 by immunohistochemistry in oesophageal submucosal glands, intestinal goblet and neuroendocrine cells, Barrett's metaplasia and intestinal metaplasia. TFF3 protein expression was associated with oestrogen receptor, progesterone receptor and TFF1 expression in malignant breast cells. TFF3 is a specific and sensitive predictive biomarker of response to endocrine therapy, degree of response and duration of response in unstratified metastatic breast cancer patients (P=0.000, P=0.002 and P=0.002 respectively). Multivariate binary logistic regression analysis demonstrated that TFF3 is an independent biomarker of endocrine response and degree of response, and this was confirmed in a validation cohort. TFF3 stimulated migration and invasion of breast cancer cells. In conclusion, TFF3 expression is associated with response to endocrine therapy, and outperforms oestrogen receptor, progesterone receptor and TFF1 as an independent biomarker, possibly because it mediates the malign effects of oestrogen on invasion and metastasis.
Collapse
Affiliation(s)
- Felicity E B May
- Department of Pathology Faculty of Medical Sciences, Northern Institute for Cancer Research and Newcastle University Institute for Ageing, University of Newcastle-upon-Tyne, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Bruce R Westley
- Department of Pathology Faculty of Medical Sciences, Northern Institute for Cancer Research and Newcastle University Institute for Ageing, University of Newcastle-upon-Tyne, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| |
Collapse
|
45
|
Wang XN, Wang SJ, Pandey V, Chen P, Li Q, Wu ZS, Wu Q, Lobie PE. Trefoil factor 3 as a novel biomarker to distinguish between adenocarcinoma and squamous cell carcinoma. Medicine (Baltimore) 2015; 94:e860. [PMID: 25997063 PMCID: PMC4602872 DOI: 10.1097/md.0000000000000860] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In carcinoma, such as of the lung, the histological subtype is important to select an appropriate therapeutic strategy for patients. However, carcinomas with poor differentiation cannot always be distinguished on the basis of morphology alone nor on clinical findings. Hence, delineation of poorly differentiated adenocarcinoma and squamous cell carcinoma, the 2 most common epithelial-origin carcinomas, is pivotal for selection of optimum therapy. Herein, we explored the potential utility of trefoil factor 3 (TFF3) as a biomarker for primary lung adenocarcinoma and extrapulmonary adenocarcinomas derived from different organs. We observed that 90.9% of lung adenocarcinomas were TFF3-positive, whereas no expression of TFF3 was observed in squamous cell carcinomas. The subtype of lung carcinoma was confirmed by four established biomarkers, cytokeratin 7 and thyroid transcription factor 1 for adenocarcinoma and P63 and cytokeratin 5/6 for squamous cell carcinoma. Furthermore, expression of TFF3 mRNA was observed by quantitative PCR in all of 11 human lung adenocarcinoma cell lines and highly correlated with markers of the adenocarcinomatous lineage. In contrast, little or no expression of TFF3 was observed in 4 lung squamous cell carcinoma cell lines. By use of forced expression, or siRNA-mediated depletion of TFF3, we determined that TFF3 appeared to maintain rather than promote glandular differentiation of lung carcinoma cells. In addition, TFF3 expression was also determined in adenocarcinomas from colorectum, stomach, cervix, esophagus, and larynx. Among all these extrapulmonary carcinomas, 93.7% of adenocarcinomas exhibited TFF3 positivity, whereas only 2.9% of squamous cell carcinomas were TFF3-positive. Totally, 92.9% of both pulmonary and extrapulmonary adenocarcinomas exhibited TFF3 positivity, whereas only 1.5% of squamous cell carcinomas were TFF3-positive. In conclusion, TFF3 is preferentially expressed in adenocarcinoma and may function as an additional biomarker for distinguishing adenocarcinoma from squamous cell carcinoma.
Collapse
Affiliation(s)
- Xiao-Nan Wang
- From the Department of Pathology (X-NW, S-JW, PC, QL, Z-SW, QW); Laboratory of Pathogenic Microbiology and Immunology, Anhui Medical University, Hefei, Anhui, People's Republic of China (X-NW); Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore (VP, PEL); and National Cancer Institute of Singapore, National University Health System, Singapore (PEL). These authors contributed equally to this work
| | | | | | | | | | | | | | | |
Collapse
|
46
|
mRNA profiling reveals determinants of trastuzumab efficiency in HER2-positive breast cancer. PLoS One 2015; 10:e0117818. [PMID: 25710561 PMCID: PMC4339844 DOI: 10.1371/journal.pone.0117818] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 12/30/2014] [Indexed: 12/30/2022] Open
Abstract
Intrinsic and acquired resistance to the monoclonal antibody drug trastuzumab is a major problem in the treatment of HER2-positive breast cancer. A deeper understanding of the underlying mechanisms could help to develop new agents. Our intention was to detect genes and single nucleotide polymorphisms (SNPs) affecting trastuzumab efficiency in cell culture. Three HER2-positive breast cancer cell lines with different resistance phenotypes were analyzed. We chose BT474 as model of trastuzumab sensitivity, HCC1954 as model of intrinsic resistance, and BTR50, derived from BT474, as model of acquired resistance. Based on RNA-Seq data, we performed differential expression analyses on these cell lines with and without trastuzumab treatment. Differentially expressed genes between the resistant cell lines and BT474 are expected to contribute to resistance. Differentially expressed genes between untreated and trastuzumab treated BT474 are expected to contribute to drug efficacy. To exclude false positives from the candidate gene set, we removed genes that were also differentially expressed between untreated and trastuzumab treated BTR50. We further searched for SNPs in the untreated cell lines which could contribute to trastuzumab resistance. The analysis resulted in 54 differentially expressed candidate genes that might be connected to trastuzumab efficiency. 90% of 40 selected candidates were validated by RT-qPCR. ALPP, CALCOCO1, CAV1, CYP1A2 and IGFBP3 were significantly higher expressed in the trastuzumab treated than in the untreated BT474 cell line. GDF15, IL8, LCN2, PTGS2 and 20 other genes were significantly higher expressed in HCC1954 than in BT474, while NCAM2, COLEC12, AFF3, TFF3, NRCAM, GREB1 and TFF1 were significantly lower expressed. Additionally, we inferred SNPs in HCC1954 for CAV1, PTGS2, IL8 and IGFBP3. The latter also had a variation in BTR50. 20% of the validated subset have already been mentioned in literature. For half of them we called and analyzed SNPs. These results contribute to a better understanding of trastuzumab action and resistance mechanisms.
Collapse
|
47
|
Belovari T, Bijelić N, Tolušić Levak M, Baus Lončar M. Trefoil factor family peptides TFF1 and TFF3 in the nervous tissues of developing mouse embryo. Bosn J Basic Med Sci 2015; 15:33-7. [PMID: 25725142 DOI: 10.17305/bjbms.2015.251] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 01/28/2023] Open
Abstract
Trefoil factor family peptides (TFF1, TFF2, and TFF3) are predominantly found in mucous epithelia of various organs. However, they have also been reported in the nervous tissue, particularly mouse, rat, porcine, and human brain. The aim of this research was to determine the presence of TFF1 and TFF3 in the nervous system of developing mouse embryo. Mouse embryos, at the stages E15 to E17 were isolated, fixed in 4% paraformaldehyde and embedded in paraffin blocks. Sagittal 6µm sections were made, processed for immunohistochemistry, and incubated with anti-TFF1 or anti-TFF3 primary polyclonal rabbit antibodies. Labeled streptavidin-biotin method was used for TFF detection. TFF1 and 3 were found in the cytoplasm of ganglion cell somata, while TFF3 staining was also visible in the cytoplasm of neurons in different areas and nuclei of brain and medulla oblongata. Neurons in the gray matter of spinal cord were also TFF1 and TFF3 positive, and signal for both peptides was found in the choroid plexus. TFF peptides might be involved in the complex processes of nervous system development and differentiation and brain plasticity.
Collapse
Affiliation(s)
- Tatjana Belovari
- Department of Histology and Embryology, Faculty of Medicine, University of Osijek, Osijek.
| | | | | | | |
Collapse
|
48
|
Gu J, Zheng L, Zhang L, Chen S, Zhu M, Li X, Wang Y. TFF3 and HER2 expression and their correlation with survival in gastric cancer. Tumour Biol 2014; 36:3001-7. [PMID: 25514872 DOI: 10.1007/s13277-014-2933-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 12/03/2014] [Indexed: 12/17/2022] Open
Abstract
The molecular biomarkers human epidermal growth factor receptor-2 (HER2) and trefoil factor 3 (TFF3) are reported to play important roles in the pathogenesis of gastric cancer (GC). In this study, we investigated the clinicopathological and prognostic significance of TFF3 and HER2 expression in GC and explored the correlation between these two biomarkers. Ninety-two patients who were diagnosed with GC were enrolled. TFF3 and HER2 expression was determined on tumor tissues. The results showed that TFF3 and HER2 were positively expressed in 42.7 and 10.9% of the cases, respectively. There were significantly higher rates of TFF3 positivity in patients with deep invasive tumors and advanced stage ones. Patients with negative TFF3 staining survived longer than those with the presence of TFF3, with 5-year overall survival (OS) rates of 57.1 ± 7.1 and 39.5 ± 7.5%, respectively (P = 0.033). However, HER2 positivity was not significantly associated with OS (P = 0.262). Multivariate analysis demonstrated TFF3 expression to be an independent indicator for short-term survival, with a hazard ratio of 2.327 (95% confidence interval (CI), 1.202-4.507, P = 0.012). There was a trend that the expression of TFF3 was more frequent in HER2 negative tumors than in HER2 positive ones (positive rates: 16.3 vs. 4.7%, P = 0.098). Patients with HER2-negative/TFF3-negative GC presented higher OS than those with other phenotypes (P = 0.009). This study suggests that TFF3 is an independent indicator for survival in GC, while HER2 is not associated with the outcome. Patients with HER2-negative/TFF3-negative GC have the best outcome.
Collapse
Affiliation(s)
- Jianchun Gu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Ahmed ARH, Muhammad EMS. E-cadherin and CD10 expression in atypical hyperplastic and malignant endometrial lesions. J Egypt Natl Canc Inst 2014; 26:211-7. [PMID: 25282623 DOI: 10.1016/j.jnci.2014.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/27/2014] [Accepted: 08/29/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Loss of E-cadherin is a critical step for development and progression of malignant tumors. CD10; a marker of non-neoplastic and neoplastic endometrial stroma, is associated with aggressiveness of many epithelial malignancies. AIMS To evaluate expression and correlation of E-cadherin and CD10 in endometrial lesions and their possible role in differentiating atypical endometrial hyperplasia from endometrial carcinoma. The association of E-cadherin and CD10 expression with clinico-pathological parameters of endometrial carcinoma was also investigated. MATERIALS AND METHODS Fifty four cases including 28 endometrial carcinomas; 19 endometrial hyperplasia and 7 cases of normal endometrial changes were enrolled for this study. The expression of E-cadherin and CD10 was evaluated by immunohistochemistry using the streptavidin-biotin technique. RESULTS There was a strong association between malignant change of endometrial glands and membrano-cytoplasmic localization of E-cadherin (p<0.001). Expression of E-cadherin but not CD10 was significantly higher in endometrial carcinomas compared to atypical endometrial hyperplasia (p<0.01). Expression of E-cadherin was not associated with CD10 expression in different endometrial lesions. High grade tumors expressed low levels of both E-cadherin (p<0.01) and CD10 (p<0.05) and serous endometrial carcinoma had low E-cadherin and CD10 expression compared to endometrioid carcinoma (p<0.01 and <0.05, respectively). Expression of both molecules showed no association with depth of tumor invasion or FIGO stage. Tumors with lower E-cadherin or CD10 expression had higher rates of vascular tumor emboli (p<0.01 and <0.07, respectively). CONCLUSIONS Although expression of E-cadherin and CD10 in endometrial lesions was not correlated, reduced expression of both molecules could be critical for progression of endometrial carcinoma.
Collapse
Affiliation(s)
- Ahmed R H Ahmed
- Department of Pathology, Faculty of Medicine, Sohag University, Egypt.
| | - Eman M S Muhammad
- Department of Pathology, Faculty of Medicine, Sohag University, Egypt.
| |
Collapse
|
50
|
Pandey V, Wu ZS, Zhang M, Li R, Zhang J, Zhu T, Lobie PE. Trefoil factor 3 promotes metastatic seeding and predicts poor survival outcome of patients with mammary carcinoma. Breast Cancer Res 2014; 16:429. [PMID: 25266665 PMCID: PMC4303111 DOI: 10.1186/s13058-014-0429-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 08/15/2014] [Indexed: 12/14/2022] Open
Abstract
Introduction Recurrence or early metastasis remains the predominant cause of mortality in patients with estrogen receptor positive (ER+) mammary carcinoma (MC). However, the molecular mechanisms underlying the initial progression of ER+ MC to metastasis remains poorly understood. Trefoil factor 3 (TFF3) is an estrogen-responsive oncogene in MC. Herein, we provide evidence for a functional role of TFF3 in metastatic progression of ER+ MC. Methods The association of TFF3 expression with clinicopathological parameters and survival outcome in a cohort of MC patients was assessed by immunohistochemistry. The expression of TFF3 in MCF7 and T47D cells was modulated by forced expression or siRNA-mediated depletion of TFF3. mRNA and protein levels were determined using qPCR and western blot. The functional effect of modulation of TFF3 expression in MC cells was determined in vitro and in vivo. Mechanistic analyses were performed using reporter constructs, modulation of signal transducer and activator of transcription 3 (STAT3) expression, and pharmacological inhibitors against c-SRC and STAT3 activity. Results TFF3 protein expression was positively associated with larger tumour size, lymph node metastasis, higher stage, and poor survival outcome. Forced expression of TFF3 in ER+ MC cells stimulated colony scattering, cell adhesion to a Collagen I-coated matrix, colony formation on a Collagen I- or Matrigel-coated matrix, endothelial cell adhesion, and transmigration through an endothelial cell barrier. In vivo, forced expression of TFF3 in MCF7 cells stimulated the formation of metastatic nodules in animal lungs. TFF3 regulation of the mRNA levels of epithelial, mesenchymal, and metastatic-related genes in ER+ MC cells were consistent with the altered cell behaviour. Forced expression of TFF3 in ER+ MC cells stimulated phosphorylation of c-SRC that subsequently increased STAT3 activity, which lead to the downregulation of E-cadherin. siRNA-mediated depletion of TFF3 reduced the invasiveness of ER+ MC cells. Conclusions TFF3 expression predicts metastasis and poor survival outcome of patients with MC and functionally stimulates cellular invasion and metastasis of ER+ MC cells. Adjuvant functional inhibition of TFF3 may therefore be considered to ameliorate outcome of ER+ MC patients. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0429-3) contains supplementary material, which is available to authorized users.
Collapse
|