1
|
Diskul-Na-Ayudthaya P, Bae SJ, Bae YU, Van NT, Kim W, Ryu S. ANKRD1 Promotes Breast Cancer Metastasis by Activating NF- κB-MAGE-A6 Pathway. Cancers (Basel) 2024; 16:3306. [PMID: 39409926 PMCID: PMC11476229 DOI: 10.3390/cancers16193306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Early detection and surgical excision of tumors have helped improve the survival rate of patients with breast cancer. However, patients with metastatic cancer typically have a poor prognosis. In this study, we propose that ANKRD1 promotes metastasis of breast cancer. ANKRD1 was found to be highly expressed in the MDA-MB-231 and MDA-LM-2 highly metastatic breast cancer cell lines compared to the non-metastatic breast cancer cell lines (MCF-7, ZR-75-30, T47D) and normal breast cancer cells (MCF-10A). Furthermore, high-grade tumors showed increased levels of ANKRD1 compared to low-grade tumors. Both in vitro and in vivo functional studies demonstrated the essential role of ANKRD1 in cancer cell migration and invasion. The previous studies have suggested a significant role of NF-κB and MAGE-A6 in breast cancer metastasis, but the upstream regulators of this axis are not well characterized. Our study suggests that ANKRD1 promotes metastasis of breast cancer by activating NF-κB as well as MAGE-A6 signaling. Our findings show that ANKRD1 is a potential therapeutic target and a diagnostic marker for breast cancer metastasis.
Collapse
Affiliation(s)
- Penchatr Diskul-Na-Ayudthaya
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Seon Joo Bae
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
| | - Yun-Ui Bae
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Konkuk University, Seoul 05030, Republic of Korea;
| | - Ngu Trinh Van
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
| | - Wootae Kim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
- Department of Pathology, College of Medicine, Soonchunhyang University, Asan-si 311151, Republic of Korea
| |
Collapse
|
2
|
Naik A, Lattab B, Qasem H, Decock J. Cancer testis antigens: Emerging therapeutic targets leveraging genomic instability in cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200768. [PMID: 38596293 PMCID: PMC10876628 DOI: 10.1016/j.omton.2024.200768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cancer care has witnessed remarkable progress in recent decades, with a wide array of targeted therapies and immune-based interventions being added to the traditional treatment options such as surgery, chemotherapy, and radiotherapy. However, despite these advancements, the challenge of achieving high tumor specificity while minimizing adverse side effects continues to dictate the benefit-risk balance of cancer therapy, guiding clinical decision making. As such, the targeting of cancer testis antigens (CTAs) offers exciting new opportunities for therapeutic intervention of cancer since they display highly tumor specific expression patterns, natural immunogenicity and play pivotal roles in various biological processes that are critical for tumor cellular fitness. In this review, we delve deeper into how CTAs contribute to the regulation and maintenance of genomic integrity in cancer, and how these mechanisms can be exploited to specifically target and eradicate tumor cells. We review the current clinical trials targeting aforementioned CTAs, highlight promising pre-clinical data and discuss current challenges and future perspectives for future development of CTA-based strategies that exploit tumor genomic instability.
Collapse
Affiliation(s)
- Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Boucif Lattab
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Hanan Qasem
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
3
|
Lin X, Huo J, Su H, Xu Y, Zhang F. The Association of Melanoma-Associated Antigen-C Gene With Clinicopathological Characteristics and Prognosis in Breast Cancer: A Systematic Review and Meta-Analysis. Clin Breast Cancer 2024; 24:7-16. [PMID: 37872029 DOI: 10.1016/j.clbc.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/25/2023]
Abstract
To investigate the correlation of melanoma-associated antigen-C gene expression with clinicopathologic characteristics and prognosis in patients with breast cancer through a meta-analysis. PubMed, Embase, Web of Science, Cochrane, CNKI, Wanfang and VIP databases were searched from the establishment of the databases to December 2022. The New castle-Ottawa Scale (NOS) was used for literature quality evaluation, and meta-analysis of all studies was performed using Rev Man 5.3 and Stata14.0. A total of 11 studies and 1146 samples were included in the meta-analysis. High expression of MAGE-C gene was significantly correlated with tumor grade (OR = 8.06, 95%CI:4.14-15.67, P < .00001), lymph node metastasis (OR = 8.06, 95%CI:4.14-15.67, P < .00001), tumor type (OR = 0.36, 95%CI: 0.23-0.49, P < .00001), tumor stage (OR = 0.14, 95%CI: 0.05-0.38, P = .0001<.05), ER expression (OR = 0.14, 95%CI: 0.05-0.38, P = .0001<.05), HER-2 expression (OR = 0.24, 95%CI:0.11-0.57, P = .001<.05) and tumor embolus (OR = 0.24, 95%CI:0.11-0.57, P = .001<.05). In addition, the MAGE-C expression was correlated with the reduced overall survival (HR = 2.13, 95%CI: 1.52-2.99, P < .0001), recurrence-free survival (HR = 2.59, 95%CI:1.47-4.56, P = .0010) and metastasis-free survival (OR = 2.52, 95%CI: 1.38-4.59, P = .003). The high expression of MAGE-C gene is closely related to some clinicopathological parameters and poor prognosis of breast cancer, which may be used as a potential biomarker to determine the prognosis of breast cancer.
Collapse
Affiliation(s)
- Xunyi Lin
- Department of Thyroid and Breast Surgery, Hebei General Hospital Affiliated to Hebei North University, Shijiazhuang 050051, Hebei Province, China
| | - Jiaxing Huo
- Department of Thyroid and Breast Surgery, Hebei General Hospital Affiliated to Hebei Medicine University, Shijiazhuang 050051, Hebei Province, China
| | - Hang Su
- Department of Thyroid and Breast Surgery, Hebei General Hospital Affiliated to North China University of Science and Technology, Shijiazhuang 050051, Hebei Province, China
| | - Yanbo Xu
- Department of Thyroid and Breast Surgery, Hebei General Hospital Affiliated to North China University of Science and Technology, Shijiazhuang 050051, Hebei Province, China
| | - Fenghua Zhang
- Department of Thyroid and Breast Surgery, Hebei General Hospital, No. 348 Peace West Road, Shijiazhuang 050051, Hebei Province, China.
| |
Collapse
|
4
|
Caramia F, Speed TP, Shen H, Haupt Y, Haupt S. Establishing the Link between X-Chromosome Aberrations and TP53 Status, with Breast Cancer Patient Outcomes. Cells 2023; 12:2245. [PMID: 37759468 PMCID: PMC10526523 DOI: 10.3390/cells12182245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Ubiquitous to normal female human somatic cells, X-chromosome inactivation (XCI) tightly regulates the transcriptional silencing of a single X chromosome from each pair. Some genes escape XCI, including crucial tumour suppressors. Cancer susceptibility can be influenced by the variability in the genes that escape XCI. The mechanisms of XCI dysregulation remain poorly understood in complex diseases, including cancer. Using publicly available breast cancer next-generation sequencing data, we show that the status of the major tumour suppressor TP53 from Chromosome 17 is highly associated with the genomic integrity of the inactive X (Xi) and the active X (Xa) chromosomes. Our quantification of XCI and XCI escape demonstrates that aberrant XCI is linked to poor survival. We derived prognostic gene expression signatures associated with either large deletions of Xi; large amplifications of Xa; or abnormal X-methylation. Our findings expose a novel insight into female cancer risks, beyond those associated with the standard molecular subtypes.
Collapse
Affiliation(s)
- Franco Caramia
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (F.C.); (Y.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Terence P. Speed
- Walter and Eliza Hall Institute for Medical Research, Parkville, VIC 3052, Australia;
| | - Hui Shen
- Van Andel Institute, Grand Rapids, MI 49503, USA;
| | - Ygal Haupt
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (F.C.); (Y.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Sue Haupt
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; (F.C.); (Y.H.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
5
|
Nin DS, Deng LW. Biology of Cancer-Testis Antigens and Their Therapeutic Implications in Cancer. Cells 2023; 12:cells12060926. [PMID: 36980267 PMCID: PMC10047177 DOI: 10.3390/cells12060926] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Tumour-specific antigens have been an area of interest in cancer therapy since their discovery in the middle of the 20th century. In the era of immune-based cancer therapeutics, redirecting our immune cells to target these tumour-specific antigens has become even more relevant. Cancer-testis antigens (CTAs) are a class of antigens with an expression specific to the testis and cancer cells. CTAs have also been demonstrated to be expressed in a wide variety of cancers. Due to their frequency and specificity of expression in a multitude of cancers, CTAs have been particularly attractive as cancer-specific therapeutic targets. There is now a rapid expansion of CTAs being identified and many studies have been conducted to correlate CTA expression with cancer and therapy-resistant phenotypes. Furthermore, there is an increasing number of clinical trials involving using some of these CTAs as molecular targets in pharmacological and immune-targeted therapeutics for various cancers. This review will summarise the current knowledge of the biology of known CTAs in tumorigenesis and the regulation of CTA genes. CTAs as molecular targets and the therapeutic implications of these CTA-targeted anticancer strategies will also be discussed.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD 7, 8 Medical Drive, Singapore 117596, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Lih-Wen Deng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD 7, 8 Medical Drive, Singapore 117596, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- National University Cancer Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
6
|
de Joode K, Veenbergen S, Kransse C, Kortleve D, Debets R, Mathijssen RHJ, Joosse A, Schreurs MWJ, Van der Veldt AAM. Suitability of tumor-associated antibodies as predictive biomarker for response to immune checkpoint inhibitors in patients with melanoma: a short report. J Immunother Cancer 2023; 11:jitc-2022-006467. [PMID: 36750254 PMCID: PMC9906380 DOI: 10.1136/jitc-2022-006467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
In 2019, Fässler et al showed in this journal that the presence of tumor-associated antibodies correlated with response to immune checkpoint inhibitor treatment in patients with metastatic melanoma. The results of this study suggested that tumor-associated antibodies directed against melanocyte-differentiation antigens and the cancer-germline antigen NY-ESO-1 should be further investigated as candidate biomarkers for response to immune checkpoint inhibitors. The aim of the current study was to validate and extend these previous findings. Therefore, we examined the correlation between serum levels of tumor-associated antibodies and tumor response after treatment with immune checkpoint inhibitors in patients with metastatic melanoma.All patients included in this prospective study were diagnosed with advanced stage melanoma and treated with nivolumab or pembrolizumab monotherapy. Blood samples were collected before and during treatment. Serum levels of tumor-associated antibodies against the melanocyte differentiation antigen Melan-A and the cancer germline antigens NY-ESO-1, MAGE-C2, MAGE-A6 and ROPN1B were measured at baseline and during treatment. Differences between responders and non-responders were assessed using the Mann-Whitney U-test, and differences between different overall survival categories with the Kruskal-Wallis test. P values ≤0.05 were considered significant.Serum samples of 58 patients with advanced melanoma with long-term follow-up (>3 years) were collected. In contrast to the findings of Fässler et al, for all antibodies tested, we found no significant differences between serum levels of responders and non-responders before or during treatment with immune checkpoint inhibitors. In addition, no significant differences were found in serum levels of tumor-associated antibodies for different overall survival groups.Although our study included a larger and more mature cohort of patients with longer follow-up, we could not externally validate the findings of Fässler et al In addition, we were not able to identify other cancer germline antigens as predictive biomarkers of response to immune checkpoint inhibitors in patients advanced melanoma. Based on the results of the present study, clinical applicability of tumor-associated antibodies directed against tumor antigens as predictive biomarkers for immune checkpoint inhibitors in patients with advanced melanoma is not feasible.
Collapse
Affiliation(s)
- Karlijn de Joode
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Sharon Veenbergen
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Claudia Kransse
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Dian Kortleve
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Marco W J Schreurs
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Astrid A M Van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands .,Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Bhat SS, Mahapatra SD, R S, Sommano SR, Prasad SK. Virtual Screening and Quantitative Structure-Activity Relationship of Moringa oleifera with Melanoma Antigen A (MAGE-A) Genes against the Therapeutics of Non-Small Cell Lung Cancers (NSCLCs). Cancers (Basel) 2022; 14:5052. [PMID: 36291836 PMCID: PMC9600242 DOI: 10.3390/cancers14205052] [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: 08/18/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
In the last decade, there have been significant advancements in the treatment of non-small cell lung cancer, including remarkable gains in detection, diagnosis, and therapy. The emergence of molecular targeted therapies, immunotherapeutic inhibitors, and antiangiogenesis medicines has largely fueled improvements in combination therapy and systemic treatments, all of which have dramatically ameliorated patient outcomes. The Moringa oleifera bioactive compounds have been effective in the suppression of cancers, making them the therapeutic agents of choice for the current investigation to treat MAGE-A presented in NSCLC. The ligand entrants were screened for their pharmacological properties, and 2,2-diphenyl-1,3-benzodioxole was stipulated as the lead candidate. 2,2-Diphenyl-1,3-benzodioxole exhibited better pharmacological properties and superior binding with branched-chain amino acids, making it an ideal candidate to address MAGE-A. The study concluded that addressing MAGE-A to impede their activity and antigenicity can be exploited as immunotarget(s).
Collapse
Affiliation(s)
- Smitha S. Bhat
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru 570 015, Karnataka, India
| | - Shreya Das Mahapatra
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru 570 015, Karnataka, India
| | - Sindhu R
- Department of Microbiology, JSS Academy of Higher Education and Research, Mysuru 570 015, Karnataka, India
| | - Sarana Rose Sommano
- Plant Bioactive Compound Laboratory, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50100, Thailand
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Shashanka K. Prasad
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru 570 015, Karnataka, India
- Plant Bioactive Compound Laboratory, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50100, Thailand
| |
Collapse
|
8
|
Topa J, Grešner P, Żaczek AJ, Markiewicz A. Breast cancer circulating tumor cells with mesenchymal features-an unreachable target? Cell Mol Life Sci 2022; 79:81. [PMID: 35048186 PMCID: PMC8770434 DOI: 10.1007/s00018-021-04064-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/13/2022]
Abstract
Circulating tumor cells (CTCs) mediate dissemination of solid tumors and can be an early sign of disease progression. Moreover, they show a great potential in terms of non-invasive, longitudinal monitoring of cancer patients. CTCs have been extensively studied in breast cancer (BC) and were shown to present a significant phenotypic plasticity connected with initiation of epithelial-mesenchymal transition (EMT). Apart from conferring malignant properties, EMT affects CTCs recovery rate, making a significant portion of CTCs from patients’ samples undetected. Wider application of methods and markers designed to isolate and identify mesenchymal CTCs is required to expand our knowledge about the clinical impact of mesenchymal CTCs. Therefore, here we provide a comprehensive review of clinical significance of mesenchymal CTCs in BC together with statistical analysis of previously published data, in which we assessed the suitability of a number of methods/markers used for isolation of CTCs with different EMT phenotypes, both in in vitro spike-in tests with BC cell lines, as well as clinical samples. Results of spiked-in cell lines indicate that, in general, methods not based on epithelial enrichment only, capture mesenchymal CTCs much more efficiently that CellSearch® (golden standard in CTCs detection), but at the same time are not much inferior to Cell Search®, though large variation in recovery rates of added cells among the methods is observed. In clinical samples, where additional CTCs detection markers are needed, positive epithelial-based CTCs enrichment was the most efficient in isolating CTCs with mesenchymal features from non-metastatic BC patients. From the marker side, PI3K and VIM were contributing the most to detection of CTCs with mesenchymal features (in comparison to SNAIL) in non-metastatic and metastatic BC patients, respectively. However, additional data are needed for more robust identification of markers for efficient detection of CTCs with mesenchymal features.
Collapse
Affiliation(s)
- Justyna Topa
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland
| | - Peter Grešner
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland
| | - Anna J Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
9
|
Beasley HK, Widatalla SE, Whalen DS, Williams SD, Korolkova OY, Namba C, Pratap S, Ochieng J, Sakwe AM. Identification of MAGEC2/CT10 as a High Calcium-Inducible Gene in Triple-Negative Breast Cancer. Front Endocrinol (Lausanne) 2022; 13:816598. [PMID: 35355564 PMCID: PMC8959981 DOI: 10.3389/fendo.2022.816598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 11/29/2022] Open
Abstract
The expression of the melanoma/cancer-testis antigen MAGEC2/CT10 is restricted to germline cells, but like most cancer-testis antigens, it is frequently upregulated in advanced breast tumors and other malignant tumors. However, the physiological cues that trigger the expression of this gene during malignancy remain unknown. Given that malignant breast cancer is often associated with skeletal metastasis and co-morbidities such as cancer-induced hypercalcemia, we evaluated the effect of high Ca2+ on the calcium-sensing receptor (CaSR) and potential mechanisms underlying the survival of triple-negative breast cancer (TNBC) cells at high Ca2+. We show that chronic exposure of TNBC cells to high Ca2+ decreased the sensitivity of CaSR to Ca2+ but stimulated tumor cell growth and migration. Furthermore, high extracellular Ca2+ also stimulated the expression of early response genes such as FOS/FOSB and a unique set of genes associated with malignant tumors, including MAGEC2. We further show that the MAGEC2 proximal promoter is Ca2+ inducible and that FOS/FOSB binds to this promoter in a Ca2+- dependent manner. Finally, downregulation of MAGEC2 strongly inhibited the growth of TNBC cells in vitro. These data suggest for the first time that MAGEC2 is a high Ca2+ inducible gene and that aberrant expression of MAGEC2 in malignant TNBC tissues is at least in part mediated by an increase in circulating Ca2+via the AP-1 transcription factor.
Collapse
Affiliation(s)
- Heather K. Beasley
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Sarrah E. Widatalla
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Diva S. Whalen
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Stephen D. Williams
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Olga Y. Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Clementine Namba
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Siddharth Pratap
- Bioinformatics Core, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Josiah Ochieng
- Bioinformatics Core, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Amos M. Sakwe
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
- *Correspondence: Amos M. Sakwe,
| |
Collapse
|
10
|
Tian Y, Liang P, Zhang L, Zhang X, Wang X, Jin Y, Qi X, Liu Y. High expression of MAGE-C1 gene in colorectal cancer is associated with its poor prognosis. J Gastrointest Oncol 2021; 12:2872-2881. [PMID: 35070414 PMCID: PMC8748057 DOI: 10.21037/jgo-21-739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/06/2021] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND The aim of this study was to explore the relationship between melanoma antigen gene C1 (MAGE-C1) expression and the prognosis for colorectal cancer (CRC), and to establish a mathematical model to comprehensively evaluate the prognosis of patients with CRC. METHODS MAGE-C1 was selected by bioinformatics for its greater expression differences in CRC patients. Immunohistochemistry (IHC) was used to detect the expression level of MAGE-C1 in tissue samples of 156 patients with CRC. Kaplan-Meier analysis was employed to assess the relationship between MAGE-C1 and the prognosis of patients with CRC. Univariate and multivariate Cox regression models analyzed the factors affecting the prognosis of CRC patients. Also, the clinicopathological characteristics of patients and genes with clinical concern were integrated to establish a model to comprehensively predict the prognosis of patients with CRC. RESULTS MAGE-C1 was found to be highly expressed in 28.8% of CRC patients. MAGE-C1 expression was associated with tumor size, number, and metastasis. Survival analysis showed that CRC patients with high expression of MAGE-C1 had a poor prognosis. Regression analysis demonstrated that MAGE-C1 protein status, T stage, differentiation, Kirsten rat sarcoma (KRAS) status, and v-RAF murine sarcoma viral oncogene homolog B1 (BRAF) status were the independent factors influencing the overall survival of patients with CRC. Meanwhile, MAGE-C1 combined with clinicopathological characteristics and hotspot gene mutations could be used to evaluate the prognosis of CRC. CONCLUSIONS Our study shows that MAGE-C1 is differentially expressed in patients with CRC and affects the prognosis of patients. The combination of MAGE-C1, clinicopathological characteristics, and genes with clinical concern can be used to assess the prognosis of CRC.
Collapse
Affiliation(s)
- Yu Tian
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ping Liang
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Lihua Zhang
- Department of Gastrointestinal Surgery, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Xiufen Zhang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaoli Wang
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yufen Jin
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Xiaowei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yankui Liu
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Li S, Shi X, Li J, Zhou X. Pathogenicity of the MAGE family. Oncol Lett 2021; 22:844. [PMID: 34733362 PMCID: PMC8561213 DOI: 10.3892/ol.2021.13105] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
The melanoma antigen gene (MAGE) protein family is a group of highly conserved proteins that share a common homology domain. Under normal circumstances, numerous MAGE proteins are only expressed in reproduction-related tissues; however, abnormal expression levels are observed in a variety of tumor tissues. The MAGE family consists of type I and II proteins, several of which are cancer-testis antigens that are highly expressed in cancer and serve a critical role in tumorigenesis. Therefore, this review will use the relationship between MAGEs and tumors as a starting point, focusing on the latest developments regarding the function of MAGEs as oncogenes, and preliminarily reveal their possible mechanisms.
Collapse
Affiliation(s)
- Sanyan Li
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Xiang Shi
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Jingping Li
- Department of Respiratory Medicine, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Xianrong Zhou
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| |
Collapse
|
12
|
Cancer-Testis Antigens in Triple-Negative Breast Cancer: Role and Potential Utility in Clinical Practice. Cancers (Basel) 2021; 13:cancers13153875. [PMID: 34359776 PMCID: PMC8345750 DOI: 10.3390/cancers13153875] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer cells commonly express tumour-associated antigens that can induce immune responses to eradicate the tumour. Triple-negative breast cancer (TNBC) is a form of breast cancer lacking the expression of hormone receptors and cerbB2 (HER2) and tends to be more aggressive and associated with poorer prognoses due to the limited treatment options. Characterisation of biomarkers or treatment targets is thus of great significance in revealing additional therapeutic options. Cancer-testis antigens (CTAs) are tumour-associated antigens that have garnered strong attention as potential clinical biomarkers in targeted immunotherapy due to their cancer-restricted expressions and robust immunogenicity. Previous clinical studies reported that CTAs correlated with negative hormonal status, advanced tumour behaviour and a poor prognosis in a variety of cancers. Various studies also demonstrated the oncogenic potential of CTAs in cell proliferation by inhibiting cell death and inducing metastasis. Multiple clinical trials are in progress to evaluate the role of CTAs as treatment targets in various cancers. CTAs hold great promise as potential treatment targets and biomarkers in cancer, and further research could be conducted on elucidating the mechanism of actions of CTAs in breast cancer or combination therapy with other immune modulators. In the current review, we summarise the current understandings of CTAs in TNBC, addressing the role and utility of CTAs in TNBC, as well as discussing the potential applications and advantage of incorporating CTAs in clinical practise.
Collapse
|
13
|
Park D, Han S, Joo H, Ka HI, Soh S, Park J, Yang Y. Increased Melanoma-Associated Antigen C2 Expression Affords Resistance to Apoptotic Deathin Suspension-Cultured Tumor Cells. J Breast Cancer 2021; 24:138-152. [PMID: 33818016 PMCID: PMC8090803 DOI: 10.4048/jbc.2021.24.e6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/05/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Melanoma-associated antigen C2 (MAGEC2) is an oncogene associated with various types of cancers. However, the biological function of MAGEC2 in circulating tumor cells remains unclear. In this study, we investigated the role of MAGEC2 using adapted suspension cells (ASCs), which were previously developed to study circulating tumor cells (CTCs). METHODS Differential gene expression in adherent cells (ADs) and ASCs was examined using RNA-seq analysis. MAGEC2 expression was assessed using reverse transcription quantitative polymerase chain reaction (RT-qPCR), immunoblotting, and ChIP-seq analysis. Depletion of MAGEC2 expression was performed using siRNA. MAGEC2-depleted ADs and ASCs were used to investigate changes in the proliferation rate and cell cycle. Then, the protein levels of signal transducer and activator of transcription 3 (STAT3), phosphorylated STAT3, and downstream of STAT3 were measured using control and MAGEC2-depleted ADs and ASCs. In ASCs, the direct effect of active STAT3 inhibition with Stattic, a STAT3 inhibitor, was assessed in terms of proliferation and apoptosis. Finally, an Annexin V/7-AAD assay was performed to determine the percentage of apoptotic cells in the Stattic-treated cells. RESULTS MAGEC2 was highly expressed in ASCs when compared with ADs. Depletion of MAGEC2 reduced the proliferation rate and viability of ASCs. To elucidate the underlying mechanism, the level of STAT3 was examined owing to its oncogenic properties. Tyrosine-phosphorylated active STAT3 was highly expressed in ASCs and decreased in MAGEC2-depleted ASCs. Furthermore, on treating ASCs with Stattic, an active STAT3 inhibitor, the cells were markedly sensitive to intrinsic pathway-mediated apoptosis. CONCLUSIONS High MAGEC2 expression may play an important role in the survival of ASCs by maintaining the expression of activated STAT3 to prevent apoptotic cell death.
Collapse
Affiliation(s)
- Doyeon Park
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Sora Han
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Hyunjeong Joo
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Hye In Ka
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | - Sujung Soh
- Department of Biological Sciences, Research Center for Cellular Heterogeneity, Research Institute of Women's Health, Sookmyung Women's University, Seoul, Korea
| | | | - Young Yang
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Korea.
| |
Collapse
|
14
|
Florke Gee RR, Chen H, Lee AK, Daly CA, Wilander BA, Fon Tacer K, Potts PR. Emerging roles of the MAGE protein family in stress response pathways. J Biol Chem 2020; 295:16121-16155. [PMID: 32921631 PMCID: PMC7681028 DOI: 10.1074/jbc.rev120.008029] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
The melanoma antigen (MAGE) proteins all contain a MAGE homology domain. MAGE genes are conserved in all eukaryotes and have expanded from a single gene in lower eukaryotes to ∼40 genes in humans and mice. Whereas some MAGEs are ubiquitously expressed in tissues, others are expressed in only germ cells with aberrant reactivation in multiple cancers. Much of the initial research on MAGEs focused on exploiting their antigenicity and restricted expression pattern to target them with cancer immunotherapy. Beyond their potential clinical application and role in tumorigenesis, recent studies have shown that MAGE proteins regulate diverse cellular and developmental pathways, implicating them in many diseases besides cancer, including lung, renal, and neurodevelopmental disorders. At the molecular level, many MAGEs bind to E3 RING ubiquitin ligases and, thus, regulate their substrate specificity, ligase activity, and subcellular localization. On a broader scale, the MAGE genes likely expanded in eutherian mammals to protect the germline from environmental stress and aid in stress adaptation, and this stress tolerance may explain why many cancers aberrantly express MAGEs Here, we present an updated, comprehensive review on the MAGE family that highlights general characteristics, emphasizes recent comparative studies in mice, and describes the diverse functions exerted by individual MAGEs.
Collapse
Affiliation(s)
- Rebecca R Florke Gee
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Helen Chen
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Anna K Lee
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christina A Daly
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Benjamin A Wilander
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Klementina Fon Tacer
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; School of Veterinary Medicine, Texas Tech University, Amarillo, Texas, USA.
| | - Patrick Ryan Potts
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
15
|
Shuvalov O, Kizenko A, Petukhov A, Aksenov N, Fedorova O, Vorobev M, Daks A, Barlev N. Cancer-testis antigens, semenogelins 1 and 2, exhibit different anti-proliferative effects on human lung adenocarcinoma cells. Cell Death Discov 2020; 6:108. [PMID: 33101710 PMCID: PMC7581521 DOI: 10.1038/s41420-020-00336-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/09/2020] [Accepted: 07/06/2020] [Indexed: 01/21/2023] Open
Abstract
Сancer-testis antigens (CTAs) comprise proteins which are aberrantly expressed in various malignancies, yet under normal situation are restricted to only germ cells. Semenogelins 1 and 2 (SEMG1 and 2, respectively) belong to the family of non-X-linked (autosomal) cancer-testis antigens. They are the major protein ingredients of human semen and share 78% of similarity between them on the gene level. SEMG1/2 gene products regulate the motility and fertility of sperm, as well as provide sperm the antibacterial defense. Besides, SEMG1 and SEMG2 were detected in various malignancies including small cell lung cancer (SCLC). However, the biological role of both SEMG1 and 2 proteins in tumorigenesis has not been fully understood. We demonstrate here that SEMG1 and SEMG2 (SEMGs) exhibit different patterns of expression and sub-cellular localization in non-small cell lung cancer (NSCLC) cell lines. To elucidate the biological properties of SEMGs in NSCLC, we established H1299 cell lines that were stably transduced with either SEMGs-overexpressing or knockdown vectors, respectively. Using fluorescence-based dihydroethidium (DHE) assay we showed that both SEMGs augmented the production of reactive oxygen species (ROS) up to 2 times. Moreover, SEMGs (especially SEMG1) strongly increased the number of Annexin V–positive apoptotic cells manifesting an increased sensitivity to genotoxic drugs including doxorubicin, etoposide, and cisplatin. Taken our results together, SEMGs may arguably play a positive role in tumorigenesis by sensitizing NSCLCs to genotoxic therapy.
Collapse
Affiliation(s)
| | | | - Alexey Petukhov
- Institute of cytology RAS, St-Petersburg, Russia.,Almazov National Medical Research Centre, St-Petersburg, Russia
| | | | | | | | | | - Nickolai Barlev
- Institute of cytology RAS, St-Petersburg, Russia.,MIPT, Doloprudny, Moscow region, Russia
| |
Collapse
|
16
|
Aberrantly enhanced melanoma-associated antigen (MAGE)-A3 expression facilitates cervical cancer cell proliferation and metastasis via actuating Wnt signaling pathway. Biomed Pharmacother 2019; 122:109710. [PMID: 31918280 DOI: 10.1016/j.biopha.2019.109710] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The over-expression of melanoma-associated antigen (MAGE)-A3 in cervical cancer (CC) has been observed in our previous study, suggesting that it possibly take a vital role during the development and metastasis of CC. The present study aimed to investigate the biological function of MAGE-A3 in the progression of CC and explore how it executes its roles. METHODS The mRNA expression of MAGE-A3 in End1/E6E7 and CC cell lines (HeLa, SiHa and C33A) was measured by real-time quantitative reverse transcription PCR (qRT-PCR). Loss- and gain-of-function methods were used to assess the effect of MAGE-A3 on the proliferative, migratory and invasive abilities of HeLa and SiHa cells. Western blot was performed to measure the expression levels of proteins related to epithelial-mesenchymal transition (EMT) and proteins in the Wnt signaling pathway. In vivo tumorigenesis assay was conducted to evaluate the effect of MAGE-A3 on tumor growth. RESULTS MAGE-A3 expression was significantly up-regulated in CC cell lines (HeLa, SiHa and C33A) compared with that in End1/E6E7 cell line. Knockdown of MAGE-A3 could significantly suppress migration, invasion and proliferation in HeLa cells; whereas, overexpression of MAGE-A3 in SiHa cells presented the opposite results. Moreover, knockdown of MAGE-A3 presented a suppressive effect on the activation of EMT and Wnt signaling pathway in HeLa cells, whilst up-regulation of MAGE-A3 exhibited the opponent outcomes in SiHa cells. Through in vivo tumorigenesis assay, we further verified that MAGE-A3 acted as a facilitator in tumor growth. CONCLUSIONS MAGE-A3 is overexpressed in CC cells and possibly facilitates the viability and motility of CC cells via modulating EMT and Wnt signaling. This study implied that MAGE-A3 might be a potential therapeutic target as well as a prognosis predictor for patients with CC.
Collapse
|
17
|
Hendrickson PG, Olson M, Luetkens T, Weston S, Han T, Atanackovic D, Fine GC. The promise of adoptive cellular immunotherapies in hepatocellular carcinoma. Oncoimmunology 2019; 9:1673129. [PMID: 32002284 PMCID: PMC6959455 DOI: 10.1080/2162402x.2019.1673129] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/22/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular Carcinoma (HCC) is one of the leading causes of cancer-related mortality worldwide. Current systemic therapies result only in modest benefits and new therapeutic options are critically needed. Some patients show promising clinical responses to immune checkpoint inhibitors, however, additional immunotherapeutic approaches, such as adoptive cell therapies (ACT), need to be developed. This review summarizes recent ACT studies and discusses the promise and obstacles of this approach. We further discuss ways of improving the efficacy of ACT in HCC including the use of combination therapies and locoregional delivery methods.
Collapse
Affiliation(s)
- Peter G. Hendrickson
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Michael Olson
- Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Tim Luetkens
- Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Siani Weston
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Tiffany Han
- Department of Radiology, Norwalk Hospital, Norwalk, CT, USA
| | - Djordje Atanackovic
- Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
18
|
Gu X, Mao Y, Shi C, Ye W, Hou N, Xu L, Chen Y, Zhao W. MAGEC2 Correlates With Unfavorable Prognosis And Promotes Tumor Development In HCC Via Epithelial-Mesenchymal Transition. Onco Targets Ther 2019; 12:7843-7855. [PMID: 31576142 PMCID: PMC6767874 DOI: 10.2147/ott.s213164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
Purpose Although MAGEC2 was first cloned from a human hepatocellular carcinoma (HCC) cDNA library by serum screening, the detailed attributes of MAGEC2 in HCC have rarely been elucidated. Patients and methods In this study, The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases were consulted to analyse the expression of MAGEC2 mRNA in liver cancer. Immunohistochemistry (IHC) analysis was performed to detect MAGEC2 expression in HCC, and the relationship between MAGEC2 expression and the clinicopathological characteristics of HCC patients was evaluated. Then, we employed the short hairpin (sh)RNA-mediated knockdown of MAGEC2 in HCC cell lines to explore the function of MAGEC2 in HCC development. Finally, the expression of epithelial-mesenchymal transition (EMT) markers in HCC xenografts and clinical samples was investigated. Results The results showed a remarkably higher level of MAGEC2 expression in HCC tissues than in noncancerous tissues, and MAGEC2 expression could be used as an independent prognostic factor for overall survival in HCC. Moreover, sh-MAGEC2 inhibited a series of HCC malignant behaviours both in vitro and in vivo. Finally, decreased MAGEC2 expression and low levels of EMT markers were detected in sh-MAGEC2 xenografts, while increased MAGEC2 expression and high levels of EMT markers were observed in invasive and metastatic HCC samples. Conclusion Taken together, our data imply that MAGEC2 is a novel prognostic marker for HCC and that MAGEC2 significantly promotes HCC tumourigenesis by inducing EMT. Targeting MAGEC2 may provide a promising therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Xuefeng Gu
- Medical School, Southeast University, Nanjing, People's Republic of China.,Department of Liver Disease, The Second Hospital of Nanjing, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Yuan Mao
- Department of Hematology and Oncology, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Hospital, Nanjing, People's Republic of China
| | - Chuanbing Shi
- Department of Pathology, Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Wei Ye
- Department of Liver Disease, The Second Hospital of Nanjing, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Ning Hou
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Li Xu
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yan Chen
- Department of Pathology, Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Wei Zhao
- Medical School, Southeast University, Nanjing, People's Republic of China.,Department of Liver Disease, The Second Hospital of Nanjing, Medical School, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
19
|
SPARC correlates with unfavorable outcome and promotes tumor growth in lung squamous cell carcinoma. Exp Mol Pathol 2019; 110:104276. [PMID: 31233732 DOI: 10.1016/j.yexmp.2019.104276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022]
Abstract
Secreted protein acidic and rich in cysteine (SPARC) plays a crucial role in the malignant progression of a number of human cancers. However, the roles of SPARC in lung squamous cell carcinoma (LSCC) remain elusive. In this present study, we first detected SPARC expression and investigated the relationship between SPARC expression and the clinicopathological attributes of LSCC patients. Then we constructed SPARC-overexpression model in LSCC cell line to explore the characteristics of SPARC in LSCC development both in vitro and in vivo. The data demonstrated a remarkably higher level of SPARC in LSCC tissues than in corresponding non-cancerous tissues and elevated SPARC expression was significantly correlated with poor outcome in LSCC patients. Moreover, a serial of phenotypic experiments indicated that SPARC overexpression substantially facilitated the growth and inhibited the apoptosis in LSCC cells and xenografts. Taken together, our results suggest that SPARC is a novel prognostic marker for LSCC prognosis and SPARC significantly promotes LSCC tumorigenesis. Targeting SPARC may provide a novel therapeutic strategy for LSCC management.
Collapse
|
20
|
Trippel A, Halling F, Heymann P, Ayna M, Al-Nawas B, Ziebart T. The expression of melanoma-associated antigen A (MAGE-A) in oral squamous cell carcinoma: an evaluation of the significance for tumor prognosis. Oral Maxillofac Surg 2019; 23:343-352. [PMID: 31093793 DOI: 10.1007/s10006-019-00778-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/02/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Melanoma-associated antigens A had been detected repeatedly in oral squamous cell carcinoma, but not in healthy mucosa. Additionally, patients with MAGE-A expressing cancers are regarded to have a worse survival prognosis, so that MAGE-A are supposed to be part of carcinogenesis. Which role these antigens fulfill within OSCC is still, up today, largely unknown. This study examines the hypothesis that MAGE-A is being produced in OSCC but not in mucosa tissue and if MAGE-A has any correlation to clinical patient's parameters like tumor size, lymph node metastasis, distant metastasis, overall survival, and recurrence. MATERIALS AND METHODS For this purpose, 50 tumor samples and 39 mucosa samples were analyzed by means of PCR and immunohistochemical staining with the antibody 6C1. RESULTS Forty of 41 stained tumor samples showed a positive antibody reaction with a maximum staining rate of 53%. Sixteen mucosa samples showed a mild positive reaction. The PCR revealed a linear expression pattern of MAGE-A in which the genes are proportionally expressed in OSCC. We did not find any relationship between MAGE-A and tumor size, overall survival, or recurrence. There was also no connection between MAGE-A and tumor parameters Hif-1 and LDH. Their expression was detected tendentially in tumors with higher staging, advanced lymph node metastasis, and rising age of the patients. The genes MAGE-A3+6 and MAGE-A4 had a statistically significant correlation with lymph node metastasis (p = 0.007 and p = 0.004). Patients got distant metastasis and influence of MAGE-A on metastatic behavior could not be verified. The genes MAGE-A3 and -A4 are consequently qualified as tumor markers in the field of diagnosis and follow-up of OSCC. CONCLUSIONS AND CLINICAL RELEVANCE Two genes have great potential as target proteins in immunotherapy. The genes MAGE-A3+6 and MAGE-A4 had a statistically significant correlation with lymph node metastasis.
Collapse
Affiliation(s)
- Anna Trippel
- Department of Oral and Maxillofacial Surgery University Medical Center Mainz, Mainz, Germany
| | - Frank Halling
- Department of Oral and Maxillofacial Surgery, Baldingerstrasse, Philipps University of Marburg, University Hospital Giessen and Marburg, Campus Marburg, D-35037, Marburg, Germany
| | - Paul Heymann
- Department of Oral and Maxillofacial Surgery, Baldingerstrasse, Philipps University of Marburg, University Hospital Giessen and Marburg, Campus Marburg, D-35037, Marburg, Germany
| | - Mustafa Ayna
- Center for Dental Implantology, 47051, Duisburg, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery University Medical Center Mainz, Mainz, Germany
| | - Thomas Ziebart
- Department of Oral and Maxillofacial Surgery, Baldingerstrasse, Philipps University of Marburg, University Hospital Giessen and Marburg, Campus Marburg, D-35037, Marburg, Germany.
| |
Collapse
|
21
|
Expression dynamics of Mage family genes during self-renewal and differentiation of mouse pluripotent stem and teratocarcinoma cells. Oncotarget 2019; 10:3248-3266. [PMID: 31143371 PMCID: PMC6524934 DOI: 10.18632/oncotarget.26933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
The biological roles of cancer-testis antigens of the Melanoma antigen (Mage) family in mammalian development, stem cell differentiation and carcinogenesis are largely unknown. In order to understand the involvement of the Mage family genes in maintenance of normal and cancer stem cells, the expression patterns of Mage-a, Mage-b, Mage-d, Mage-e, Mage-h and Mage-l gene subfamilies were analyzed during the self-renewal and differentiation of mouse pluripotent stem and teratocarcinoma cells. Clustering analysis based on the gene expression profiles of undifferentiated and differentiating cell populations revealed strong correlations between Mage expression patterns and differentiation and malignant states. Gene co-expression analysis disclosed the potential contributions of Mage family members in self-renewal and differentiation of pluripotent stem and teratocarcinoma cells. Two gene clusters including Mage-a4 and Mage-a8, Mageb1, Mage-d1, Mage-d2, Mage-e1, Mage-l2 were identified as functional antagonists with opposing roles in the regulation of proliferation and differentiation of mouse pluripotent stem and teratocarcinoma cells. The identified aberrant expression patterns of Mage-a2, Mage-a6, Mage-b4, Mageb-16 and Mage-h1 in teratocarcinoma cells can be considered as specific teratocarcinoma biomarkers promoted the malignant phenotype. Our study first provides a model for the involvement of Mage family members in regulatory networks during the self-renewal and early differentiation of normal and cancerous stem cells for further research of the predicted functional modules and the development of new cancer treatment strategies.
Collapse
|
22
|
Song X, Guo C, Zheng Y, Wang Y, Jin Z, Yin Y. Post-transcriptional regulation of cancer/testis antigen MAGEC2 expression by TRIM28 in tumor cells. BMC Cancer 2018; 18:971. [PMID: 30309319 PMCID: PMC6182782 DOI: 10.1186/s12885-018-4844-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cancer/testis antigen MAGEC2 (also known as HCA587) is highly expressed in a wide variety of tumors and plays an active role in promoting growth and metastasis of tumor cells. However, little is known for the regulation of MAGEC2 expression in cancer cells. METHODS Western blotting and quantitative RT-PCR were performed to analyze MAGEC2 expression. Co-immunoprecipitation assay was applied for detecting the endogenous interaction of MAGEC2 and TRIM28 in tumor cells. Overexpression and knockdown assays were used to examine the effects of TRIM28 on the expression of MAGEC2 protein. Immunohistochemistry (IHC) staining was performed in hepatocellular carcinoma patients to evaluate the association between the expression of MAGEC2 and TRIM28. Proteasome inhibitors MG132 or PS-341 and lysosome inhibitor Chloroquine (CQ) were used to inhibit proteasomal or lysosomal-mediated protein degradation respectively. RESULTS We demonstrate that MAGEC2 interacts with TRIM28 in melanoma cells and MAGEC2 expression in tumor cells depends on the expression of TRIM28. The expression level of MAGEC2 protein was significantly reduced when TRIM28 was depleted in tumor cells, and no changes were observed in MAGEC2 mRNA level. Furthermore, expression levels of MAGEC2 and TRIM28 are positively correlated in MAGEC2-positive human hepatocellular carcinoma tissues (p = 0.0011). Mechanistic studies indicate that the regulatory role of TRIM28 on MAGEC2 protein expression in tumor cells depends on proteasome-mediated pathway. CONCLUSIONS Our findings show that TRIM28 is necessary for MAGEC2 expression in cancer cells, and TRIM28 may serve as a new potential target for immunotherapy of cancer.
Collapse
Affiliation(s)
- Xiao Song
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing, 100191, China
| | - Chengli Guo
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing, 100191, China
| | - Yutian Zheng
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing, 100191, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing, 100191, China
| | - Zhongtian Jin
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China.
| | - Yanhui Yin
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing, 100191, China.
| |
Collapse
|
23
|
Gordeeva O. Cancer-testis antigens: Unique cancer stem cell biomarkers and targets for cancer therapy. Semin Cancer Biol 2018; 53:75-89. [PMID: 30171980 DOI: 10.1016/j.semcancer.2018.08.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/15/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
Cancer-testis antigens (CTAs) are considered as unique and promising cancer biomarkers and targets for cancer therapy. CTAs are multifunctional protein group with specific expression patterns in normal embryonic and adult cells and various types of cancer cells. CTAs are involved in regulating of the basic cellular processes during development, stem cell differentiation and carcinogenesis though the biological roles and cell functions of CTA families remain largely unclear. Analysis of CTA expression patterns in embryonic germ and somatic cells, pluripotent and multipotent stem cells, cancer stem cells and their cell descendants indicates that rearrangements of characteristic CTA profiles (aberrant expression) could be associated with cancer transformation and failure of the developmental program of cell lineage specification and germ line restriction. Therefore, aberrant CTA profiles can be used as panels of biomarkers for diagnoses and the selection of cancer treatment strategies. Moreover, immunogenic CTAs are prospective targets for cancer immunotherapy. Clinical trials testing broad range of cancer therapeutic vaccines against antigens of MAGEA and NY-ESO-1 families for treating various cancers have shown mixed clinical efficiency, safety and tolerability, suggesting the requirement of in-depth research of CTA expression in normal and cancer stem cells and extensive clinical trials for improving cancer immunotherapy technologies. This review focuses on recent advancement in study of CTAs in normal and cancer cells, particularly in normal and cancer stem cells, and provides a new insight into CTA expression patterns during normal and cancer stem cell lineage development. Additionally, new approaches in development of effective CTA-based therapies exclusively targeting cancer stem cells will be discussed.
Collapse
Affiliation(s)
- Olga Gordeeva
- Laboratory of Cell and Molecular Mechanisms of Histogenesis, Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow, 119334, Russia.
| |
Collapse
|
24
|
STAT3 Interactors as Potential Therapeutic Targets for Cancer Treatment. Int J Mol Sci 2018; 19:ijms19061787. [PMID: 29914167 PMCID: PMC6032216 DOI: 10.3390/ijms19061787] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023] Open
Abstract
Signal transducers and activators of transcription (STATs) mediate essential signaling pathways in different biological processes, including immune responses, hematopoiesis, and neurogenesis. Among the STAT members, STAT3 plays crucial roles in cell proliferation, survival, and differentiation. While STAT3 activation is transient in physiological conditions, STAT3 becomes persistently activated in a high percentage of solid and hematopoietic malignancies (e.g., melanoma, multiple myeloma, breast, prostate, ovarian, and colon cancers), thus contributing to malignant transformation and progression. This makes STAT3 an attractive therapeutic target for cancers. Initial strategies aimed at inhibiting STAT3 functions have focused on blocking the action of its activating kinases or sequestering its DNA binding ability. More recently, the diffusion of proteomic-based techniques, which have allowed for the identification and characterization of novel STAT3-interacting proteins able to modulate STAT3 activity via its subcellular localization, interact with upstream kinases, and recruit transcriptional machinery, has raised the possibility to target such cofactors to specifically restrain STAT3 oncogenic functions. In this article, we summarize the available data about the function of STAT3 interactors in malignant cells and discuss their role as potential therapeutic targets for cancer treatment.
Collapse
|
25
|
Zeng P, Wang Y, Zheng Y, Song X, Yin Y. Cancer‑testis antigen HCA587/MAGEC2 interacts with the general transcription coactivator TAF9 in cancer cells. Mol Med Rep 2017; 17:3226-3231. [PMID: 29257297 DOI: 10.3892/mmr.2017.8260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/20/2017] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma-associated antigen 587/melanoma antigen gene (HCA587/MAGEC2) is a cancer‑testis antigen, which is highly expressed in various types of tumors, but not in normal tissues with the exception of male germ‑line cells. HCA587/MAGEC2 has been previously recognized as a tumor‑specific target for immunotherapy; however, its biological functions have been relatively understudied. To investigate the function of HCA587/MAGEC2, the amino acid sequence of HCA587/MAGEC2 was analyzed by bioinformatics and it was demonstrated that HCA587/MAGEC2 contains a 9‑amino acid transactivation domain which may mediate the interaction of most transcription factors with TATA‑box binding protein associated factor 9 (TAF9), a general transcription coactivator. Co‑immunoprecipitation experiments revealed that HCA587/MAGEC2 interacted with TAF9 in transfected 293T and in A375 melanoma cells endogenously expressing HCA587/MAGEC2, and confirmed the endogenous interaction of HCA587/MAGEC2 and TAF9 within cells. Endogenous HCA587/MAGEC2 and TAF9 were demonstrated to be co‑localized principally in the nucleus of tumor cells using immunofluorescence. Glutathione-S-transferase pull‑down experiments demonstrated that HCA587/MAGEC2 interacts with TAF9 directly and the conserved region in the TAF9 may becrucial for HCA587/MAGEC2 binding. The present study demonstrated that the cancer‑testis antigen HCA587/MAGEC2 directly interacted with TAF9, which may provide novel information for identifying the oncogenic functions of HCA587/MAGEC2 in tumor cells.
Collapse
Affiliation(s)
- Pumei Zeng
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Yutian Zheng
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Xiao Song
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Yanhui Yin
- Department of Immunology, School of Basic Medical Sciences, Key Laboratory of Medical Immunology of Ministry of Health, Peking University Health Science Center, Beijing 100191, P.R. China
| |
Collapse
|
26
|
Jäger D, Lechel A, Tharehalli U, Seeling C, Möller P, Barth TFE, Mellert K. U-CH17P, -M and -S, a new cell culture system for tumor diversity and progression in chordoma. Int J Cancer 2017; 142:1369-1378. [PMID: 29148152 DOI: 10.1002/ijc.31161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/10/2017] [Accepted: 11/06/2017] [Indexed: 01/29/2023]
Abstract
Chordoma is a rare bone tumor with a known intrinsic heterogeneity. Here, we address this tumor heterogeneity in a new cell culture model for tumor diversity and progression in chordoma. The three cell lines U-CH17P, U-CH17M, and U-CH17S were established from a primary sacral chordoma and its derived metastases, a soft tissue and a skin metastasis, respectively. The lesions had divergent differentiation patterns which are conserved in the derived cell lines making them a suitable in vitro model for the analysis of tumorigenesis in chordoma. A common feature of the three cell lines is the expression of typical chordoma markers, such as Brachyury, vimentin, cytokeratins, EMA and S100 protein. A comparison of the genomic aberrations by array comparative genomic hybridization of the cell lines and the corresponding parental tumor tissues revealed that the precursor cells of U-CH17P, U-CH17M and U-CH17S were already present in the primary tumor. Therefore, we show that clonal diversity of this chordoma exists in the primary tumor and that not all of these subclones tend to metastasize. All cell lines had a CDKN2A loss. A comparison of the gene expression profiles of the cell lines revealed significant differences in the expression of several genes like MAGEC2 and SEMA6A known to be associated with the tendency to metastasize or proliferation and migration. Since the underlying mechanisms of tumor progression in chordoma are still largely unclear, the three U-CH17 cell lines are a suitable in vitro model for elucidating chordoma oncobiology.
Collapse
Affiliation(s)
- D Jäger
- Institute of Pathology, University of Ulm, Ulm, Germany
| | - A Lechel
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - U Tharehalli
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | - C Seeling
- Institute of Pathology, University of Ulm, Ulm, Germany
| | - P Möller
- Institute of Pathology, University of Ulm, Ulm, Germany
| | - T F E Barth
- Institute of Pathology, University of Ulm, Ulm, Germany
| | - K Mellert
- Institute of Pathology, University of Ulm, Ulm, Germany
| |
Collapse
|
27
|
Hammerl D, Smid M, Timmermans AM, Sleijfer S, Martens JWM, Debets R. Breast cancer genomics and immuno-oncological markers to guide immune therapies. Semin Cancer Biol 2017; 52:178-188. [PMID: 29104025 DOI: 10.1016/j.semcancer.2017.11.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/30/2017] [Accepted: 11/01/2017] [Indexed: 12/28/2022]
Abstract
There is an increasing awareness of the importance of tumor - immune cell interactions to the evolution and therapy responses of breast cancer (BC). Not surprisingly, numerous studies are currently assessing the clinical value of immune modulation for BC patients. However, till now durable clinical responses are only rarely observed. It is important to realize that BC is a heterogeneous disease comprising several histological and molecular subtypes, which cannot be expected to be equally immunogenic and therefore not equally sensitive to single immune therapies. Here we review the characteristics of infiltrating leukocytes in healthy and malignant breast tissue, the prognostic and predictive values of immune cell subsets across different BC subtypes and the various existing immune evasive mechanisms. Furthermore, we describe the presence of certain groups of antigens as putative targets for treatment, evaluate the outcomes of current clinical immunotherapy trials, and finally, we propose a strategy to better implement immuno-oncological markers to guide future immune therapies in BC.
Collapse
Affiliation(s)
- D Hammerl
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Rotterdam, the Netherlands
| | - M Smid
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Rotterdam, the Netherlands
| | - A M Timmermans
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Rotterdam, the Netherlands
| | - S Sleijfer
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Rotterdam, the Netherlands
| | - J W M Martens
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Rotterdam, the Netherlands
| | - R Debets
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
28
|
McFarlane RJ, Wakeman JA. Meiosis-like Functions in Oncogenesis: A New View of Cancer. Cancer Res 2017; 77:5712-5716. [PMID: 29061671 DOI: 10.1158/0008-5472.can-17-1535] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/03/2017] [Accepted: 08/16/2017] [Indexed: 11/16/2022]
Abstract
Cancer cells have many abnormal characteristics enabling tumors to grow, spread, and avoid immunologic and therapeutic destruction. Central to this is the innate ability of populations of cancer cells to rapidly evolve. One feature of many cancers is that they activate genes that are normally associated with distinct developmental states, including germ cell-specific genes. This has historically led to the proposal that tumors take on embryonal characteristics, the so called embryonal theory of cancer. However, one group of germline genes, not directly associated with embryonic somatic tissue genesis, is the one that encodes the specific factors to drive the unique reductional chromosome segregation of meiosis I, which also results in chromosomal exchanges. Here, we propose that meiosis I-specific modulators of reductional segregation can contribute to oncogenic chromosome dynamics and that the embryonal theory for cancer cell growth/proliferation is overly simplistic, as meiotic factors are not a feature of most embryonic tissue development. We postulate that some meiotic chromosome-regulatory functions contribute to a soma-to-germline model for cancer, in which activation of germline (including meiosis) functions drive oncogenesis, and we extend this to propose that meiotic factors could be powerful sources of targets for therapeutics and biomonitoring in oncology. Cancer Res; 77(21); 5712-6. ©2017 AACR.
Collapse
Affiliation(s)
- Ramsay J McFarlane
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Bangor, Gwynedd, United Kingdom.
| | - Jane A Wakeman
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Bangor, Gwynedd, United Kingdom
| |
Collapse
|
29
|
Lu C, Meng S, Jin Y, Zhang W, Li Z, Wang F, Wang-Johanning F, Wei Y, Liu H, Tu H, Su D, He A, Cao X, Zhou F. A novel multi-epitope vaccine from MMSA-1 and DKK1 for multiple myeloma immunotherapy. Br J Haematol 2017; 178:413-426. [PMID: 28508448 DOI: 10.1111/bjh.14686] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/02/2017] [Indexed: 01/16/2023]
Abstract
The identification of novel tumour-associated antigens is urgently needed to improve the efficacy of immunotherapy for multiple myeloma (MM). In this study, we identified a membrane protein MMSA-1 (multiple myeloma special antigen-1) that was specifically expressed in MM and exhibited significantly positive correlation with MM. We then identified HLA-A*0201-restricted MMSA-1 epitopes and tested their cytotoxic T lymphocyte (CTL) response. The MMSA-1 epitope SLSLLTIYV vaccine was shown to induce an obvious CTL response in vitro. To improve the immunotherapy, we constructed a multi-epitope peptide vaccine by combining epitopes derived from MMSA-1 and Dickkopf-1 (DKK1). The effector T cells induced by multi-epitope peptide vaccine-loaded dendritic cells lysed U266 cells more effectively than MMSA-1/DKK1 single-epitope vaccine. In myeloma-bearing severe combined immunodeficient mice, the multi-epitope vaccine improved the survival rate significantly compared with single-epitope vaccine. Consistently, multi-epitope vaccine decreased the tumour volume greatly and alleviated bone destruction. The frequencies of CD4+ and CD8+ T cells was significantly increased in mouse blood induced by the multi-epitope vaccine, indicating that it inhibits myeloma growth by changing T cell subsets and alleviating immune paralysis. This study identified a novel peptide from MMSA-1 and the multi-epitope vaccine will be used to establish appropriate individualized therapy for MM.
Collapse
Affiliation(s)
- Chenyang Lu
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shan Meng
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanxia Jin
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Wanggang Zhang
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zongfang Li
- National-local Joint Engineering Research Centre of Biodiagnostics & Biotherapy, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Fang Wang
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Hailing Liu
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Honglei Tu
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dan Su
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Aili He
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xingmei Cao
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fuling Zhou
- Department of Clinical Haematology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
30
|
Planells-Palop V, Hazazi A, Feichtinger J, Jezkova J, Thallinger G, Alsiwiehri NO, Almutairi M, Parry L, Wakeman JA, McFarlane RJ. Human germ/stem cell-specific gene TEX19 influences cancer cell proliferation and cancer prognosis. Mol Cancer 2017; 16:84. [PMID: 28446200 PMCID: PMC5406905 DOI: 10.1186/s12943-017-0653-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 04/18/2017] [Indexed: 12/15/2022] Open
Abstract
Background Cancer/testis (CT) genes have expression normally restricted to the testis, but become activated during oncogenesis, so they have excellent potential as cancer-specific biomarkers. Evidence is starting to emerge to indicate that they also provide function(s) in the oncogenic programme. Human TEX19 is a recently identified CT gene, but a functional role for TEX19 in cancer has not yet been defined. Methods siRNA was used to deplete TEX19 levels in various cancer cell lines. This was extended using shRNA to deplete TEX19 in vivo. Western blotting, fluorescence activated cell sorting and immunofluorescence were used to study the effect of TEX19 depletion in cancer cells and to localize TEX19 in normal testis and cancer cells/tissues. RT-qPCR and RNA sequencing were employed to determine the changes to the transcriptome of cancer cells depleted for TEX19 and Kaplan-Meier plots were generated to explore the relationship between TEX19 expression and prognosis for a range of cancer types. Results Depletion of TEX19 levels in a range of cancer cell lines in vitro and in vivo restricts cellular proliferation/self-renewal/reduces tumour volume, indicating TEX19 is required for cancer cell proliferative/self-renewal potential. Analysis of cells depleted for TEX19 indicates they enter a quiescent-like state and have subtle defects in S-phase progression. TEX19 is present in both the nucleus and cytoplasm in both cancerous cells and normal testis. In cancer cells, localization switches in a context-dependent fashion. Transcriptome analysis of TEX19 depleted cells reveals altered transcript levels of a number of cancer-/proliferation-associated genes, suggesting that TEX19 could control oncogenic proliferation via a transcript/transcription regulation pathway. Finally, overall survival analysis of high verses low TEX19 expressing tumours indicates that TEX19 expression is linked to prognostic outcomes in different tumour types. Conclusions TEX19 is required to drive cell proliferation in a range of cancer cell types, possibly mediated via an oncogenic transcript regulation mechanism. TEX19 expression is linked to a poor prognosis for some cancers and collectively these findings indicate that not only can TEX19 expression serve as a novel cancer biomarker, but may also offer a cancer-specific therapeutic target with broad spectrum potential. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0653-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vicente Planells-Palop
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK
| | - Ali Hazazi
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK
| | - Julia Feichtinger
- Computational Biotechnology and Bioinformatics Group, Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria.,Omics Center Graz, BioTechMed Graz, Graz, Austria
| | - Jana Jezkova
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK
| | - Gerhard Thallinger
- Computational Biotechnology and Bioinformatics Group, Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria.,Omics Center Graz, BioTechMed Graz, Graz, Austria
| | - Naif O Alsiwiehri
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK
| | - Mikhlid Almutairi
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK.,Present address: Department of Zoology, King Saud University, Al-Ryiadh, Saudi Arabia
| | - Lee Parry
- European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Jane A Wakeman
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK
| | - Ramsay J McFarlane
- North West Cancer Research Institute, School of Medical Sciences, Bangor University, Brambell Building, Deiniol Road, Bangor, Gwynedd, LL57 2UW, UK.
| |
Collapse
|
31
|
Lee AK, Potts PR. A Comprehensive Guide to the MAGE Family of Ubiquitin Ligases. J Mol Biol 2017; 429:1114-1142. [PMID: 28300603 DOI: 10.1016/j.jmb.2017.03.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/07/2017] [Accepted: 03/07/2017] [Indexed: 12/28/2022]
Abstract
Melanoma antigen (MAGE) genes are conserved in all eukaryotes and encode for proteins sharing a common MAGE homology domain. Although only a single MAGE gene exists in lower eukaryotes, the MAGE family rapidly expanded in eutherians and consists of more than 50 highly conserved genes in humans. A subset of MAGEs initially garnered interest as cancer biomarkers and immunotherapeutic targets due to their antigenic properties and unique expression pattern that is primary restricted to germ cells and aberrantly reactivated in various cancers. However, further investigation revealed that MAGEs not only drive tumorigenesis but also regulate pathways essential for diverse cellular and developmental processes. Therefore, MAGEs are implicated in a broad range of diseases including neurodevelopmental, renal, and lung disorders, and cancer. Recent biochemical and biophysical studies indicate that MAGEs assemble with E3 RING ubiquitin ligases to form MAGE-RING ligases (MRLs) and act as regulators of ubiquitination by modulating ligase activity, substrate specification, and subcellular localization. Here, we present a comprehensive guide to MAGEs highlighting the molecular mechanisms of MRLs and their physiological roles in germ cell and neural development, oncogenic functions in cancer, and potential as therapeutic targets in disease.
Collapse
Affiliation(s)
- Anna K Lee
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Patrick Ryan Potts
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA.
| |
Collapse
|
32
|
Li Y, Li J, Wang Y, Zhang Y, Chu J, Sun C, Fu Z, Huang Y, Zhang H, Yuan H, Yin Y. Roles of cancer/testis antigens (CTAs) in breast cancer. Cancer Lett 2017; 399:64-73. [PMID: 28274891 DOI: 10.1016/j.canlet.2017.02.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 12/19/2022]
Abstract
Breast cancer is the most common cancer diagnosed and is the second leading cause of cancer death among women in the US. For breast cancer, early diagnosis and efficient therapy remains a significant clinical challenge. Therefore, it is necessary to identify novel tumor associated molecules to target for biomarker development and immunotherapy. In this regard, cancer testis antigens (CTAs) have emerged as a potential clinical biomarker targeting immunotherapy for various malignancies due to the nature of its characteristics. CTAs are a group of tumor associated antigens (TAAs) that display normal expression in immune-privileged organs, but display aberrant expression in several types of cancers, particularly in advanced cancers. Investigation of CTAs for the clinical management of breast malignancies indicates that these TAAs have potential roles as novel biomarkers, with increased specificity and sensitivity compared to those currently used in the clinic. Moreover, TAAs could be therapeutic targets for cancer immunotherapy. This review is an attempt to address the promising CTAs in breast cancer and their possible clinical implications as biomarkers and immunotherapeutic targets with particular focus on challenges and future interventions.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Yifan Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Yanhong Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Jiahui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Chunxiao Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China; Nanjing Maternity and Child Health Medical Institute, Affiliated Obstetrics and Gynecology Hospital, Nanjing Medical University, Nanjing 210004, China
| | - Yi Huang
- Department of Pharmacology and Chemical Biology, Magee Women's Research Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Hansheng Zhang
- School of Public Health, University of Maryland, College Park, MD 20742, USA
| | - Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University. Nanjing 210004, China.
| |
Collapse
|
33
|
Chen X, Wang L, Liu J, Huang L, Yang L, Gao Q, Shi X, Li J, Li F, Zhang Z, Zhao S, Zhang B, Van der Bruggen P, Zhang Y. Expression and prognostic relevance of MAGE-A3 and MAGE-C2 in non-small cell lung cancer. Oncol Lett 2017; 13:1609-1618. [PMID: 28454298 PMCID: PMC5403542 DOI: 10.3892/ol.2017.5665] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/21/2016] [Indexed: 01/06/2023] Open
Abstract
Melanoma-associated antigen (MAGE)-A3 and MAGE-C2 are antigens encoded by cancer-germline genes, and have been recognized as potential prognostic biomarkers and attractive targets for immunotherapy in multiple types of cancer. The present study aimed to analyze the clinicopathological significance of MAGE-A3/C2 expression in non-small cell lung cancer (NSCLC). The association between MAGE-A3/C2 mRNA and protein expression, and the pathological characteristics and overall survival of patients with NSCLC was analyzed. In addition, the functional role of MAGE-A3 in human NSCLC cell line A549 was examined in vitro. MAGE-A3/C2 mRNA expression was identified in 73% (151/206) and 53% (109/206) of patients with NSCLC, respectively. MAGE-A3/C2 protein expression was identified in 58% (44/76) and 53% (40/76) of NSCLC cases, respectively. MAGE-A3 mRNA expression was observed to be associated with smoking history, disease stage and lymph node metastasis. However, no association was identified between MAGE-C2 mRNA expression and the clinicopathological characteristics of patients with NSCLC. MAGE-A3/C2-positive patients had a poorer survival rate compared with MAGE-A3/C2-negative patients. Multivariate analysis identified that MAGE-A3 expression may serve as an independent marker of poor prognosis in patients with NSCLC. Downregulation of MAGE-A3 mRNA expression in A549 cells resulted in lower migration and colony formation rates, and a higher amount of epithelial marker and lower amount of mesenchymal marker expression compared with the control group. These results indicate that MAGE-A3 serves a role in NSCLC cell metastasis through the induction of epithelial-mesenchymal transition. In conclusion, MAGE-A3 may serve as a diagnostic and prognostic biomarker for patients with NSCLC, due to its association with tumor progression and poor clinical outcome.
Collapse
Affiliation(s)
- Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Liping Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jinyan Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lan Huang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qun Gao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaojuan Shi
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jieyao Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Song Zhao
- Department of Cerebral Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Bin Zhang
- Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pierre Van der Bruggen
- Ludwig Institute for Cancer Research Brussels Branch, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Engineering Key Laboratory for Cell Therapy of Henan, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
34
|
Wang J, Song X, Guo C, Wang Y, Yin Y. Establishment of MAGEC2-knockout cells and functional investigation of MAGEC2 in tumor cells. Cancer Sci 2016; 107:1888-1897. [PMID: 27636589 PMCID: PMC5198962 DOI: 10.1111/cas.13082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/10/2016] [Accepted: 09/13/2016] [Indexed: 01/23/2023] Open
Abstract
Cancer/testis antigen MAGEC2, a member of the type I melanoma‐associated antigen family, is expressed in a wide variety of cancer types but not in normal somatic cells. MAGEC2 has long been recognized as a tumor‐specific target, however, its functions remain largely unknown. In this study, we established MAGEC2‐knockout A375 melanoma cell lines using the CRISPR/Cas9 system. Seven clonal cell lines were generated by using four single guide RNAs targeting the coding region of the MAGEC2 gene, which produced indels that abolished MAGEC2 protein expression. To identify the differentially expressed protein profiles associated with MAGEC2 loss, isobaric tag for relative quantitation‐based comparative proteomics experiments were carried out on the MAGEC2‐knockcout and control A375 cells. Mining of the proteomics data identified a total 224 (61.6% upregulated and 38.4% downregulated) proteins to be significantly altered in expression level in MAGEC2‐knockcout cells. Ingenuity Pathway Analysis indicated that the significantly altered proteins were involved in critical neoplasia‐related biological functions such as cell death, proliferation, and movement. Gene ontology analysis identified “apoptosis signaling” as the top‐most upregulated pathway associated with MAGEC2 loss. We showed that knockout or knockdown of the MAGEC2 gene sensitized melanoma cells to tumor necrosis factor‐α‐induced apoptosis. Interestingly, actin‐based motility by Rho and RhoA signaling, known to promote cell migration, were also identified as the top downregulated pathways in MAGEC2‐knockout A375 cells. In short, our study provides a suitable cell model for exploring the biological functions of MAGEC2 in malignant cells, and sheds light on the molecular pathway by which MAGEC2 promotes tumor development.
Collapse
Affiliation(s)
- Jingjing Wang
- Key Laboratory of Medical Immunology, Ministry of Health, Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiao Song
- Key Laboratory of Medical Immunology, Ministry of Health, Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chengli Guo
- Key Laboratory of Medical Immunology, Ministry of Health, Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ying Wang
- Key Laboratory of Medical Immunology, Ministry of Health, Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yanhui Yin
- Key Laboratory of Medical Immunology, Ministry of Health, Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
35
|
The cancer/testis antigen MAGEC2 promotes amoeboid invasion of tumor cells by enhancing STAT3 signaling. Oncogene 2016; 36:1476-1486. [PMID: 27775077 DOI: 10.1038/onc.2016.314] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/18/2016] [Accepted: 07/22/2016] [Indexed: 12/11/2022]
Abstract
The biological function of MAGEC2, a cancer/testis antigen highly expressed in various cancers, remains largely unknown. Here we demonstrate that expression of MAGEC2 induces rounded morphology and amoeboid-like movement of tumor cells in vitro and promotes tumor metastasis in vivo. The pro-metastasis effect of MAGEC2 was mediated by signal transducer and activator of transcription 3 (STAT3) activation. Mechanistically, MAGEC2 interacts with STAT3 and inhibits the polyubiquitination and proteasomal degradation of STAT3 in the nucleus of tumor cells, resulting in accumulation of phosphorylated STAT3 and enhanced transcriptional activity. Notably, expression levels of MAGEC2 and phosphorylated STAT3 are positively correlated and both are associated with incidence of metastasis in human hepatocellular carcinoma. This study not only reveals a previously unappreciated role of MAGEC2 in promoting tumor metastasis, but also identifies a new molecular mechanism by which MAGEC2 sustains hyperactivation of STAT3 in the nucleus of tumor cells. Thus, MAGEC2 may represent a new antitumor metastasis target for treatment of cancer.
Collapse
|
36
|
Kunert A, van Brakel M, van Steenbergen-Langeveld S, da Silva M, Coulie PG, Lamers C, Sleijfer S, Debets R. MAGE-C2-Specific TCRs Combined with Epigenetic Drug-Enhanced Antigenicity Yield Robust and Tumor-Selective T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2016; 197:2541-52. [PMID: 27489285 DOI: 10.4049/jimmunol.1502024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 07/03/2016] [Indexed: 01/22/2023]
Abstract
Adoptive T cell therapy has shown significant clinical success for patients with advanced melanoma and other tumors. Further development of T cell therapy requires improved strategies to select effective, yet nonself-reactive, TCRs. In this study, we isolated 10 TCR sequences against four MAGE-C2 (MC2) epitopes from melanoma patients who showed clinical responses following vaccination that were accompanied by significant frequencies of anti-MC2 CD8 T cells in blood and tumor without apparent side effects. We introduced these TCRs into T cells, pretreated tumor cells of different histological origins with the epigenetic drugs azacytidine and valproate, and tested tumor and self-reactivities of these TCRs. Pretreatment of tumor cells upregulated MC2 gene expression and enhanced recognition by T cells. In contrast, a panel of normal cell types did not express MC2 mRNA, and similar pretreatment did not result in recognition by MC2-directed T cells. Interestingly, the expression levels of MC2, but not those of CD80, CD86, or programmed death-ligand 1 or 2, correlated with T cell responsiveness. One of the tested TCRs consistently recognized pretreated MC2(+) cell lines from melanoma, head and neck, bladder, and triple-negative breast cancers but showed no response to MHC-eluted peptides or peptides highly similar to MC2. We conclude that targeting MC2 Ag, combined with epigenetic drug-enhanced antigenicity, allows for significant and tumor-selective T cell responses.
Collapse
Affiliation(s)
- Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| | - Mandy van Brakel
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| | - Sabine van Steenbergen-Langeveld
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| | - Marvin da Silva
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| | - Pierre G Coulie
- de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Cor Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| | - Stefan Sleijfer
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, the Netherlands; and
| |
Collapse
|
37
|
Park HJ, Kim MK, Choi KS, Jeong JW, Bae SK, Kim HJ, Bae MK. Neuromedin B receptor antagonism inhibits migration, invasion, and epithelial-mesenchymal transition of breast cancer cells. Int J Oncol 2016; 49:934-42. [PMID: 27571778 DOI: 10.3892/ijo.2016.3590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/02/2016] [Indexed: 11/05/2022] Open
Abstract
Neuromedin B (NMB) acts as an autocrine growth factor and a pro-angiogenic factor. Its receptor, NMB receptor (NMB-R), is overexpressed in solid tumors. In the present study, we showed that an NMB-R antagonist, PD168368, suppresses migration and invasion of the human breast cancer cell line MDA-MB-231. In addition, PD168368 reduced epithelial-mesenchymal transition (EMT) of breast cancer cells by E-cadherin upregulation and vimentin downregulation. Moreover, we found that PD168368 potently inhibits in vivo metastasis of breast cancer. Taken together, these findings suggest that NMB-R antagonism may be an alternative approach to prevent breast cancer metastasis, and targeting NMB-R may provide a novel therapeutic strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Hyun-Joo Park
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Mi-Kyoung Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Kyu-Sil Choi
- Molecular and Cellular Imaging Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul 136‑710, Republic of Korea
| | - Joo-Won Jeong
- School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Soo-Kyung Bae
- Department of Dental Pharmacology, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Hyung Joon Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Moon-Kyoung Bae
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, Republic of Korea
| |
Collapse
|
38
|
Oncogenic cancer/testis antigens: prime candidates for immunotherapy. Oncotarget 2016; 6:15772-87. [PMID: 26158218 PMCID: PMC4599236 DOI: 10.18632/oncotarget.4694] [Citation(s) in RCA: 247] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/21/2015] [Indexed: 12/15/2022] Open
Abstract
Recent developments have set the stage for immunotherapy as a supplement to conventional cancer treatment. Consequently, a significant effort is required to further improve efficacy and specificity, particularly the identification of optimal therapeutic targets for clinical testing. Cancer/testis antigens are immunogenic, highly cancer-specific, and frequently expressed in various types of cancer, which make them promising candidate targets for cancer immunotherapy, including cancer vaccination and adoptive T-cell transfer with chimeric T-cell receptors. Our current understanding of tumor immunology and immune escape suggests that targeting oncogenic antigens may be beneficial, meaning that identification of cancer/testis antigens with oncogenic properties is of high priority. Recent work from our lab and others provide evidence that many cancer/testis antigens, in fact, have oncogenic functions, including support of growth, survival and metastasis. This novel insight into the function of cancer/testis antigens has the potential to deliver more effective cancer vaccines. Moreover, immune targeting of oncogenic cancer/testis antigens in combination with conventional cytotoxic therapies or novel immunotherapies such as checkpoint blockade or adoptive transfer, represents a highly synergistic approach with the potential to improve patient survival.
Collapse
|
39
|
TCR-engineered T cells to treat tumors: Seeing but not touching? Semin Immunol 2016; 28:10-21. [PMID: 26997556 DOI: 10.1016/j.smim.2016.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Adoptive transfer of T cells gene-engineered with T cell receptors (TCRs) has proven its feasibility and therapeutic potential in the treatment of malignant tumors. To ensure further clinical development of TCR gene therapy, it is necessary to accurately select TCRs that demonstrate antigen-selective responses that are restricted to tumor cells and, at the same time, include strategies that restore or enhance the entry, migration and local accumulation of T cells in tumor tissues. Here, we present the current standing of TCR-engineered T cell therapy, discuss and propose procedures to select TCRs as well as strategies to sensitize the tumor to T cell trafficking, and provide a rationale for combination therapies with TCR-engineered T cells.
Collapse
|
40
|
Nabavi S, Schmolze D, Maitituoheti M, Malladi S, Beck AH. EMDomics: a robust and powerful method for the identification of genes differentially expressed between heterogeneous classes. Bioinformatics 2016; 32:533-41. [PMID: 26515818 PMCID: PMC4743632 DOI: 10.1093/bioinformatics/btv634] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/16/2015] [Accepted: 10/24/2015] [Indexed: 01/05/2023] Open
Abstract
MOTIVATION A major goal of biomedical research is to identify molecular features associated with a biological or clinical class of interest. Differential expression analysis has long been used for this purpose; however, conventional methods perform poorly when applied to data with high within class heterogeneity. RESULTS To address this challenge, we developed EMDomics, a new method that uses the Earth mover's distance to measure the overall difference between the distributions of a gene's expression in two classes of samples and uses permutations to obtain q-values for each gene. We applied EMDomics to the challenging problem of identifying genes associated with drug resistance in ovarian cancer. We also used simulated data to evaluate the performance of EMDomics, in terms of sensitivity and specificity for identifying differentially expressed gene in classes with high within class heterogeneity. In both the simulated and real biological data, EMDomics outperformed competing approaches for the identification of differentially expressed genes, and EMDomics was significantly more powerful than conventional methods for the identification of drug resistance-associated gene sets. EMDomics represents a new approach for the identification of genes differentially expressed between heterogeneous classes and has utility in a wide range of complex biomedical conditions in which sample classes show within class heterogeneity. AVAILABILITY AND IMPLEMENTATION The R package is available at http://www.bioconductor.org/packages/release/bioc/html/EMDomics.html.
Collapse
Affiliation(s)
- Sheida Nabavi
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Daniel Schmolze
- Department of Pathology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Mayinuer Maitituoheti
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA and
| | - Sadhika Malladi
- Department of Pathology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA, The Harker School, San Jose, CA, USA
| | - Andrew H Beck
- Department of Pathology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Moarii M, Reyal F, Vert JP. Integrative DNA methylation and gene expression analysis to assess the universality of the CpG island methylator phenotype. Hum Genomics 2015; 9:26. [PMID: 26463173 PMCID: PMC4603341 DOI: 10.1186/s40246-015-0048-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/01/2015] [Indexed: 01/26/2023] Open
Abstract
Background The CpG island methylator phenotype (CIMP) was first characterized in colorectal cancer but since has been extensively studied in several other tumor types such as breast, bladder, lung, and gastric. CIMP is of clinical importance as it has been reported to be associated with prognosis or response to treatment. However, the identification of a universal molecular basis to define CIMP across tumors has remained elusive. Results We perform a genome-wide methylation analysis of over 2000 tumor samples from 5 cancer sites to assess the existence of a CIMP with common molecular basis across cancers. We then show that the CIMP phenotype is associated with specific gene expression variations. However, we do not find a common genetic signature in all tissues associated with CIMP. Conclusion Our results suggest the existence of a universal epigenetic and transcriptomic signature that defines the CIMP across several tumor types but does not indicate the existence of a common genetic signature of CIMP. Electronic supplementary material The online version of this article (doi:10.1186/s40246-015-0048-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matahi Moarii
- CBIO-Centre for Computational Biology, Mines Paristech, PSL-Research University, 35 Rue Saint-Honore, Fontainebleau, F-77300, France. .,Department of Bioinformatics, Biostatistics and System Biology, Institut Curie, 11-13 Rue Pierre et Marie Curie, Paris, F-75248, France. .,U900, INSERM, 11-13 Rue Pierre et Marie Curie, Paris, F-75248, France.
| | - Fabien Reyal
- UMR932, Immunity and Cancer Team, Institut Curie, 26 Rue d'Ulm, Paris, 75006, France. .,Department of Translational Research, Residual Tumor and Response to Treatment Team, Institut Curie, 26 Rue d'Ulm, Paris, 75006, France. .,Department of Surgery, Institut Curie, 26 Rue d'Ulm, Paris, 75006, France.
| | - Jean-Philippe Vert
- CBIO-Centre for Computational Biology, Mines Paristech, PSL-Research University, 35 Rue Saint-Honore, Fontainebleau, F-77300, France. .,Department of Bioinformatics, Biostatistics and System Biology, Institut Curie, 11-13 Rue Pierre et Marie Curie, Paris, F-75248, France. .,U900, INSERM, 11-13 Rue Pierre et Marie Curie, Paris, F-75248, France.
| |
Collapse
|
42
|
SINKOVICS JOSEPHG. The cnidarian origin of the proto-oncogenes NF-κB/STAT and WNT-like oncogenic pathway drives the ctenophores (Review). Int J Oncol 2015; 47:1211-29. [PMID: 26239915 PMCID: PMC4583530 DOI: 10.3892/ijo.2015.3102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 01/09/2023] Open
Abstract
The cell survival pathways of the diploblastic early multicellular eukaryotic hosts contain and operate the molecular machinery resembling those of malignantly transformed individual cells of highly advanced multicellular hosts (including Homo). In the present review, the STAT/NF-κB pathway of the cnidarian Nematostella vectensis is compared with that of human tumors (malignant lymphomas, including Reed-Sternberg cells) pointing out similarities, including possible viral initiation in both cases. In the ctenophore genome and proteome, β-catenin gains intranuclear advantages due to a physiologically weak destructive complex in the cytoplasm, and lack of natural inhibitors (the dickkopfs). Thus, a scenario similar to what tumor cells initiate and achieve is presented through several constitutive loss-of-function type mutations in the destructive complex and in the elimination of inhibitors. Vice versa, malignantly transformed individual cells of advanced multicellular hosts assume pheno-genotypic resemblance to cells of unicellular or early multicellular hosts, and presumably to their ancient predecessors, by returning to the semblance of immortality and to the resumption of the state of high degree of resistance to physicochemical insults. Human leukemogenic and oncogenic pathways are presented for comparisons. The supreme bioengineers RNA/DNA complex encoded both the malignantly transformed immortal cell and the human cerebral cortex. The former generates molecules for the immortality of cellular life in the Universe. The latter invents the inhibitors of the process in order to gain control over it.
Collapse
Affiliation(s)
- JOSEPH G. SINKOVICS
- St. Joseph Hospital's Cancer Institute Affiliated with the H.L. Moffitt Comprehensive Cancer Center; Department of Molecular Medicine, The University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
43
|
Weon JL, Potts PR. The MAGE protein family and cancer. Curr Opin Cell Biol 2015; 37:1-8. [PMID: 26342994 DOI: 10.1016/j.ceb.2015.08.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022]
Abstract
The Melanoma Antigen Gene (MAGE) protein family is a large, highly conserved group of proteins that share a common MAGE homology domain. Intriguingly, many MAGE proteins are restricted in expression to reproductive tissues, but are aberrantly expressed in a wide variety of cancer types. Originally discovered as antigens on tumor cells and developed as cancer immunotherapy targets, recent literature suggests a more prominent role for MAGEs in driving tumorigenesis. This review will highlight recent developments into the function of MAGEs as oncogenes, their mechanisms of action in regulation of ubiquitin ligases, and outstanding questions in the field.
Collapse
Affiliation(s)
- Jenny L Weon
- Departments of Physiology, Pharmacology, and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, United States
| | - Patrick Ryan Potts
- Departments of Physiology, Pharmacology, and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
44
|
Lu Z, Jiao D, Qiao J, Yang S, Yan M, Cui S, Liu Z. Restin suppressed epithelial-mesenchymal transition and tumor metastasis in breast cancer cells through upregulating mir-200a/b expression via association with p73. Mol Cancer 2015; 14:102. [PMID: 25972084 PMCID: PMC4429374 DOI: 10.1186/s12943-015-0370-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 04/21/2015] [Indexed: 12/31/2022] Open
Abstract
Background Restin belongs to MAGE superfamily and is known as MAGE H1. Restin was firstly cloned from HL-60 cells treated with all-trans retinoic acid (ATRA). Previous studies showed a pro-apoptotic role of Restin in several cell lines. However, little information is available on its expression patterns and functions in vivo. Our study was performed to detect if Restin plays a role in breast cancer cells in vitro and in vivo. Methods and results Real-time PCR and western blot were conducted to detect Restin expression in multiple breast cancer cell lines and Restin level was negatively related with cell motility. Restin overexpression and knockdown stable cell lines were established by transducing lentivirus into MCF-7 and MDA-MB-231 cells. Cell morphology, wound closure assay, transwell migration and invasion assays were performed to detect if Restin inhibited EMT. Our data showed that Restin overexpressed cells exhibited classical epithelial cell morphology, and Restin overexpression resulted in activation of epithelial markers and suppression of mesenchymal markers, and inhibition of cell migration and invasion. Tumor xenograft model was used to characterize the biological functions of Restin in vivo. We found that Restin overexpression led to reduced lung metastasis. Real-time PCR, western blot, luciferase assay and ChIP assay were performed to identify the potential targets of Restin and the underlying molecular mechanisms. Among several master regulators of EMT, only ZEB1/2 levels were dramatically inhibited by Restin. Unexpectedly, Restin indirectly regulated ZEB1/2 expression at post-transcriptional level. We further identified mir-200a/b, well-characterized mediators controlling ZEB1/2 expression, were transcriptionally activated by Restin and the regulation was dependent on the p53 binding site in mir-200b/a/429 promoter. Further mechanical studies demonstrated Restin interacted with p73, one of p53 family members, which contributed to Restin-mediated activation of mir-200a/b and suppression of ZEB1/2. Conclusions Taken together, our results suggest that Restin inhibits EMT and tumor metastasis by controlling the expression of the tumor metastasis suppressor mir-200a/b via association with p73. Our findings not only establish a mechanistic link between Restin, EMT and tumor metastasis, but also provide strong evidence supporting the notion that MAGE Group II proteins may exert a tumor suppressive effect in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0370-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenduo Lu
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Dechuang Jiao
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Jianghua Qiao
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Sen Yang
- Department of Pathogen Biology, Basic Medical College of Zhengzhou University, #100 Science Road, Zhengzhou, 450001, People's Republic of China.
| | - Min Yan
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Shude Cui
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Zhenzhen Liu
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| |
Collapse
|