1
|
McShane N, Zaborowski A, O'Reilly M, McCartan D, Prichard R. Hormone Receptor Positive Breast Cancer in Young Women: A Review. J Surg Oncol 2025; 131:580-586. [PMID: 39470669 PMCID: PMC12065450 DOI: 10.1002/jso.27963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024]
Abstract
The global incidence of hormone-positive breast cancer (HR+ BC) in young women is rising, though the underlying reasons remain unclear. HR+ disease in younger women appears to represent a distinct clinical entity compared to that in older women, exhibiting distinct clinicopathological characteristics, outcomes and responses to treatment. Despite these differences, there is a paucity of large-volume data focusing on young women with HR+ in contemporary literature. Hormone receptor positive breast cancer in young women is associated with poorer prognoses compared to older women. Additionally, early age onset breast cancer presents unique challenges, including concerns related to fertility, the toxic effects of therapeutic agents, and specific surgical considerations. The purpose of this review is to report the existing literature on HR+ disease in young women.
Collapse
|
2
|
Turova P, Kushnarev V, Baranov O, Butusova A, Menshikova S, Yong ST, Nadiryan A, Antysheva Z, Khorkova S, Guryleva MV, Bagaev A, Lennerz JK, Chernyshov K, Kotlov N. The Breast Cancer Classifier refines molecular breast cancer classification to delineate the HER2-low subtype. NPJ Breast Cancer 2025; 11:19. [PMID: 39979291 PMCID: PMC11842814 DOI: 10.1038/s41523-025-00723-0] [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: 04/19/2024] [Accepted: 01/19/2025] [Indexed: 02/22/2025] Open
Abstract
Current breast cancer classification methods, particularly immunohistochemistry and PAM50, face challenges in accurately characterizing the HER2-low subtype, a therapeutically relevant entity with distinct biological features. This notable gap can lead to misclassification, resulting in inappropriate treatment decisions and suboptimal patient outcomes. Leveraging RNA-seq and machine-learning algorithms, we developed the Breast Cancer Classifier (BCC), a unique transcriptomic classifier for more precise breast cancer subtyping, specifically by delineating and incorporating HER2-low as a distinct subtype. BCC also redefined the PAM50 Normal subtype into other subtypes, disputing its classification as a unique molecular group. Our statistical analysis not only confirmed the reproducibility and accuracy of BCC, but also revealed similarities in prognostic characteristics between the HER2-low and Basal subtypes. Addressing this gap in breast cancer classification is clinically significant because it not only improves treatment stratification, but also uncovers novel molecular and immunohistochemical features associated with the HER2-low and HER2-high subtypes, thereby advancing our understanding of breast cancer heterogeneity and providing guidance in precision oncology.
Collapse
|
3
|
Chen S, Oramas Mogrovejo DM, Huang X, Siegal GP, Wei S. Reduced GATA3 expression during breast cancer progression: A potential anchor for pulmonary metastatic deposition. Pathol Res Pract 2025; 266:155821. [PMID: 39817957 DOI: 10.1016/j.prp.2025.155821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Estrogen receptor (ER) is a direct and reciprocal target gene for GATA3. Previous studies have shown that higher GATA3 expression in primary breast cancer (BC) is associated with a reduced probability of developing lung metastasis when compared to those with metastatic recurrence to other organs. Further, GATA3 downregulates several genes promoting BC lung metastasis and upregulates genes encoding known inhibitors of lung metastasis. In this study, we examined GATA3 expression in 34 consutive cases of paired primary BCs and their pulmonary metastases. Variable levels of GATA3 expression were seen in 94 % primary BCs (mean H-score 239), whereas a significantly reduced GATA3 expression was seen in the paired lung metastases (mean H-score 152; P < 0.0001). However, this trend was not observed for ER (mean H-score 140 vs. 109; P = 0.1). These findings provide further evidence that GATA3 may inhibit pulmonary deposition or sondary growth of BC cells in the lung. The effect of GATA3 in metastatic tumor growth was independent of cell differentiation, and this process is likely mediated by a GATA3-regulated genetic program driven by metastasis-associated genes rather than ER. Further exploring the molecular pathways by which GATA3 regulates downstream targets is pivotal in understanding organ-specific BC dissemination.
Collapse
Affiliation(s)
- Shoujun Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35249, United States
| | - Diana M Oramas Mogrovejo
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35249, United States
| | - Xiao Huang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35249, United States
| | - Gene P Siegal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35249, United States
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35249, United States.
| |
Collapse
|
4
|
Yao TH, Ni Y, Bhadra A, Kang J, Baladandayuthapani V. Robust Bayesian graphical regression models for assessing tumor heterogeneity in proteomic networks. Biometrics 2025; 81:ujae160. [PMID: 39798955 DOI: 10.1093/biomtc/ujae160] [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: 10/24/2023] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 01/15/2025]
Abstract
Graphical models are powerful tools to investigate complex dependency structures in high-throughput datasets. However, most existing graphical models make one of two canonical assumptions: (i) a homogeneous graph with a common network for all subjects or (ii) an assumption of normality, especially in the context of Gaussian graphical models. Both assumptions are restrictive and can fail to hold in certain applications such as proteomic networks in cancer. To this end, we propose an approach termed robust Bayesian graphical regression (rBGR) to estimate heterogeneous graphs for non-normally distributed data. rBGR is a flexible framework that accommodates non-normality through random marginal transformations and constructs covariate-dependent graphs to accommodate heterogeneity through graphical regression techniques. We formulate a new characterization of edge dependencies in such models called conditional sign independence with covariates, along with an efficient posterior sampling algorithm. In simulation studies, we demonstrate that rBGR outperforms existing graphical regression models for data generated under various levels of non-normality in both edge and covariate selection. We use rBGR to assess proteomic networks in lung and ovarian cancers to systematically investigate the effects of immunogenic heterogeneity within tumors. Our analyses reveal several important protein-protein interactions that are differentially associated with the immune cell abundance; some corroborate existing biological knowledge, whereas others are novel findings.
Collapse
Affiliation(s)
- Tsung-Hung Yao
- Department of Biostatistics, University of Michigan at Ann Arbor, Ann Arbor, MI 48109, United States
| | - Yang Ni
- Department of Statistics, Texas A&M University, College Station, TX 77843, United States
| | - Anindya Bhadra
- Department of Statistics, Purdue University, West Lafayette, IN 47907, United States
| | - Jian Kang
- Department of Biostatistics, University of Michigan at Ann Arbor, Ann Arbor, MI 48109, United States
| | | |
Collapse
|
5
|
Bacha R, Alwisi N, Ismail R, Pedersen S, Al-Mansoori L. Unveiling GATA3 Signaling Pathways in Health and Disease: Mechanisms, Implications, and Therapeutic Potential. Cells 2024; 13:2127. [PMID: 39768217 PMCID: PMC11674286 DOI: 10.3390/cells13242127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
GATA binding protein 3 (GATA3), a member of the GATA family transcription factors, is a key player in various physiological and pathological conditions. It is known for its ability to bind to the DNA sequence "GATA", which enables its key role in critical processes in multiple tissues and organs including the immune system, endocrine system, and nervous system. GATA3 also modulates cell differentiation, proliferation, and apoptosis via controlling gene expression. In physiological instances, GATA3 is crucial for maintaining immunological homeostasis by mediating the development of naïve T cells into T helper 2 (Th2). In addition, GATA3 has been demonstrated to play a variety of cellular roles in the growth and maintenance of mammary gland, neuronal, and renal tissues. Conversely, the presence of impaired GATA3 is associated with a variety of diseases, including neurodegenerative diseases, autoimmune diseases, and cancers. Additionally, the altered expression of GATA3 contributes to the worsening of disease progression in hematological malignancies, such as T-cell lymphomas. Therefore, this review explores the multifaceted roles and signaling pathways of GATA3 in health and disease, with a particular emphasis on its potential as a therapeutic and prognostic target for the effective management of diseases.
Collapse
Affiliation(s)
- Rim Bacha
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (R.B.); (N.A.); (R.I.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Nouran Alwisi
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (R.B.); (N.A.); (R.I.)
| | - Rana Ismail
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (R.B.); (N.A.); (R.I.)
| | - Shona Pedersen
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (R.B.); (N.A.); (R.I.)
| | - Layla Al-Mansoori
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
6
|
Sandström J, Bomanson J, Pérez-Tenorio G, Jönsson C, Nordenskjöld B, Fornander T, Lindström LS, Stål O. GATA3 and markers of epithelial-mesenchymal transition predict long-term benefit from tamoxifen in ER-positive breast cancer. NPJ Breast Cancer 2024; 10:78. [PMID: 39242600 PMCID: PMC11379893 DOI: 10.1038/s41523-024-00688-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
GATA binding protein 3 (GATA3) is essential for normal development of the mammary gland and associated with ER-positive breast cancer. Loss of GATA3 has been associated with epithelial-mesenchymal transition (EMT) in experimental studies. We investigated tumoral GATA3 in a cohort of postmenopausal patients with lymph-node negative breast cancer, randomized to adjuvant tamoxifen or control. Nuclear GATA3 expression was assessed with immunohistochemistry and GATA3 gene expression with Agilent microarrays. High GATA3 nuclear expression was associated with a lower rate of distant recurrence in ER-positive breast cancer (HR = 0.60, 95% CI 0.39-0.93). Low gene expression of GATA3 was associated with limited long-term benefit from adjuvant tamoxifen (interaction: p = 0.033). GATA3 gene expression was associated with the epithelial markers CDH1 (E-cadherin) and FOXA1, whereas negatively associated with several mesenchymal markers. Low expression of CDH1 was associated with marginal tamoxifen benefit (HR = 0.80 (0.43-1.49)), whereas patients with higher expression showed a significant benefit (HR = 0.33 (0.20-0.55), interaction: p = 0.029). In ER-positive breast cancer, diminished expression of GATA3 is associated with markers of EMT and poor long-term benefit from tamoxifen.
Collapse
Affiliation(s)
- Josefine Sandström
- Department of Biomedical and Clinical Sciences and Department of Oncology, 581 83 Linköping University, Linköping, Sweden
| | - Jens Bomanson
- Department of Biomedical and Clinical Sciences and Department of Oncology, 581 83 Linköping University, Linköping, Sweden
| | - Gizeh Pérez-Tenorio
- Department of Biomedical and Clinical Sciences and Department of Oncology, 581 83 Linköping University, Linköping, Sweden
| | - Carolin Jönsson
- Department of Biomedical and Clinical Sciences and Department of Oncology, 581 83 Linköping University, Linköping, Sweden
| | - Bo Nordenskjöld
- Department of Biomedical and Clinical Sciences and Department of Oncology, 581 83 Linköping University, Linköping, Sweden
| | - Tommy Fornander
- Department of Oncology and Pathology, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Linda S Lindström
- Department of Oncology and Pathology, Karolinska Institute and University Hospital, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Olle Stål
- Department of Biomedical and Clinical Sciences and Department of Oncology, 581 83 Linköping University, Linköping, Sweden.
| |
Collapse
|
7
|
Wooller SK, Pearl LH, Pearl FMG. Identifying actionable synthetically lethal cancer gene pairs using mutual exclusivity. FEBS Lett 2024; 598:2028-2039. [PMID: 38977941 DOI: 10.1002/1873-3468.14950] [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: 08/03/2023] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 07/10/2024]
Abstract
Mutually exclusive loss-of-function alterations in gene pairs are those that occur together less frequently than may be expected and may denote a synthetically lethal relationship (SSL) between the genes. SSLs can be exploited therapeutically to selectively kill cancer cells. Here, we analysed mutation, copy number variation, and methylation levels in samples from The Cancer Genome Atlas, using the hypergeometric and the Poisson binomial tests to identify mutually exclusive inactivated genes. We focused on gene pairs where one is an inactivated tumour suppressor and the other a gene whose protein product can be inhibited by known drugs. This provided an abundance of potential targeted therapeutics and repositioning opportunities for several cancers. These data are available on the MexDrugs website, https://bioinformaticslab.sussex.ac.uk/mexdrugs.
Collapse
Affiliation(s)
- Sarah K Wooller
- Bioinformatics Lab, School of Life Sciences, University of Sussex, Brighton, UK
| | - Laurence H Pearl
- Genome Damage Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Frances M G Pearl
- Bioinformatics Lab, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
8
|
Gonzalez T, Nie Q, Chaudhary LN, Basel D, Reddi HV. Methylation signatures as biomarkers for non-invasive early detection of breast cancer: A systematic review of the literature. Cancer Genet 2024; 282-283:1-8. [PMID: 38134587 DOI: 10.1016/j.cancergen.2023.12.003] [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: 10/04/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Early detection of breast cancer would help alleviate the burden of treatment for early-stage breast cancer and help patient prognosis. There is currently no established gene panel that utilizes the potential of DNA methylation as a molecular signature for the early detection of breast cancer. This systematic review aims to identify the optimal methylation biomarkers for a non-invasive liquid biopsy assay and the gaps in knowledge regarding biomarkers for early detection of breast cancer. METHODS Following the PRISMA-ScR method, Pubmed and Google Scholar was searched for publications related to methylation biomarkers in breast cancer over a five-year period. Eligible publications were mined for key data fields such as study aims, cohort demographics, types of breast cancer studied, technologies used, and outcomes. Data was analyzed to address the objectives of the review. RESULTS Literature search identified 112 studies of which based on eligibility criteria, 13 studies were included. 28 potential methylation gene targets were identified, of which 23 were methylated at the promoter region, 1 was methylated in the body of the gene and 4 were methylated at yet to be identified locations. CONCLUSIONS Our evaluation shows that at minimum APC, RASSFI, and FOXA1 genes would be a promising set of genes to start with for the early detection of breast cancer, based on the sensitivity and specificity outlined in the studies. Prospective studies are needed to optimize biomarkers for broader impact in early detection of breast cancer.
Collapse
Affiliation(s)
- Tessa Gonzalez
- Division of Precision Medicine and Cytogenetics, Department of Pathology, Medical College of Wisconsin, CT, USA
| | - Qian Nie
- Division of Precision Medicine and Cytogenetics, Department of Pathology, Medical College of Wisconsin, CT, USA
| | - Lubna N Chaudhary
- Division of Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, CT, USA
| | - Donald Basel
- Division of Genetics, Department of Pediatrics, Medical College of Wisconsin, CT, USA
| | - Honey V Reddi
- Division of Precision Medicine and Cytogenetics, Department of Pathology, Medical College of Wisconsin, CT, USA.
| |
Collapse
|
9
|
Wang E, Henderson M, Yalamanchili P, Cueto J, Islam Z, Dharmani C, Salas M. Potential biomarkers in breast cancer drug development: application of the biomarker qualification evidentiary framework. Biomark Med 2024; 18:265-277. [PMID: 38487948 PMCID: PMC11216506 DOI: 10.2217/bmm-2023-0048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/26/2024] [Indexed: 06/26/2024] Open
Abstract
Breast cancer treatments have evolved rapidly, and clinically meaningful biomarkers have been used to guide therapy. These biomarkers hold utility within the drug development process to increase the efficiency and effectiveness. To this purpose, the US FDA developed an evidentiary framework. Literature searches conducted of literature published between 2016 and 2022 identified biomarkers in breast cancer. These biomarkers were reviewed for drug development utility through the biomarker qualification evidentiary framework. In the breast cancer setting, several promising biomarkers (ctDNA, Ki-67 and PIK3CA) were identified. There is a need for increased transparency regarding the requirements for qualification of specific biomarkers and increased awareness of the processes involved in biomarker qualification.
Collapse
Affiliation(s)
- Eric Wang
- Daiichi-Sankyo, Inc., Basking Ridge, NJ 07920, USA
| | | | - Priyanka Yalamanchili
- Daiichi-Sankyo, Inc., Basking Ridge, NJ 07920, USA
- Rutgers Institute for Pharmaceutical Industry Fellowships, Piscataway, NJ 08854, USA
| | | | | | | | - Maribel Salas
- Daiichi-Sankyo, Inc., Basking Ridge, NJ 07920, USA
- Center for Real-world Effectiveness & Safety of Therapeutics (CREST), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Leyane TS, Jere SW, Houreld NN. Effect of photobiomodulation at 830 nm on gene expression correlated with JAK/STAT signalling in wounded and diabetic wounded fibroblasts in vitro. JOURNAL OF BIOPHOTONICS 2024; 17:e202300230. [PMID: 38010362 DOI: 10.1002/jbio.202300230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023]
Abstract
Treatment of chronic diabetic wounds is an ongoing socio-economic challenge. Dysregulated signalling pathways characterise cells from chronic diabetic wounds. Photobiomodulation (PBM) stimulates healing by eliciting photochemical effects that affect gene regulation. JAK/STAT signalling is a primary signal transduction pathway involved in wound healing. This in vitro study aimed to determine if PBM at 830 nm and a fluence of 5 J/cm2 regulates genes related to JAK/STAT signalling in wounded and diabetic wounded fibroblast cells. A continuous wave diode laser (12.53 mW/cm2 ) was used to irradiate cells. Forty-eight hours post-PBM, RT-qPCR was used to analyse 84 genes related to JAK/STAT signalling. Five genes were upregulated and four downregulated in wounded cell models, while six genes were downregulated in diabetic wounded models. The results show drastic gene expression differences between wounded and diabetic wounded cell models in response to PBM using 830 nm.
Collapse
Affiliation(s)
- Thobekile S Leyane
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Sandy W Jere
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Nicolette N Houreld
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
11
|
Elkhamisy FAA, Eesa AN, Sallam MK, Hussein MF, Abd El-Moeze A. GATA3 Positively Correlates with BCL2 Expression in Indolent and Aggressive Histological Types of Cutaneous Basal Cell Carcinoma. Turk Patoloji Derg 2024; 40:162-169. [PMID: 38668712 PMCID: PMC11402336 DOI: 10.5146/tjpath.2024.13370] [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: 08/26/2023] [Accepted: 03/11/2024] [Indexed: 09/11/2024] Open
Abstract
OBJECTIVE Some histological basal cell carcinoma (BCC) types demonstrate more aggressive behavior than others. They are known as high-risk BCC and are more challenging in therapy, contrary to indolent (low-risk) BCC types. Identifying novel protein markers to predict aggressiveness and potential therapeutic targets in challenging cases is recommended. GATA3 is a transcription factor critical for epithelial and lymphocytic differentiation. This study investigated the immunohistochemical expression of GATA3 in indolent and aggressive BCC and its association with BCL2 expression. MATERIAL AND METHODS Retrospectively collected indolent and aggressive BCC groups (24 cases each) were immunohistochemically stained with anti-GATA3 and BCL2 antibodies. The mean expression score (by area percentage) and TIL counts were determined and compared using ImageJ analysis. Stromal tumor-infiltrating lymphocytes (TIL) were counted per high-power field (HPF) on hematoxylin and eosin (H&E) staining. RESULTS GATA3 and BCL2 expressions were significantly higher in the indolent group than in the aggressive group. GATA3 expression significantly correlated with BCL2 score and TIL counts. Higher GATA3 expression was significantly associated with a more indolent BCC histological type, higher BCL2 expression, and higher TIL count. CONCLUSION GATA3 is a possible target for immunomodulation experiments to improve BCC immunotherapy outcomes.
Collapse
Affiliation(s)
| | | | - Marwa Kamal Sallam
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Kasr Al Ainy Hospitals, Cairo University, Giza, Egypt
| | - Marwa Fathy Hussein
- Department of Dermatology, Cairo University, Faculty of Medicine, Giza, Egypt
| | - Ahmed Abd El-Moeze
- Department of Pathology, Beni-Suef University, Faculty of Medicine, Beni-Suef, Egypt
| |
Collapse
|
12
|
Jacobson DH, Pan S, Fisher J, Secrier M. Multi-scale characterisation of homologous recombination deficiency in breast cancer. Genome Med 2023; 15:90. [PMID: 37919776 PMCID: PMC10621207 DOI: 10.1186/s13073-023-01239-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Homologous recombination is a robust, broadly error-free mechanism of double-strand break repair, and deficiencies lead to PARP inhibitor sensitivity. Patients displaying homologous recombination deficiency can be identified using 'mutational signatures'. However, these patterns are difficult to reliably infer from exome sequencing. Additionally, as mutational signatures are a historical record of mutagenic processes, this limits their utility in describing the current status of a tumour. METHODS We apply two methods for characterising homologous recombination deficiency in breast cancer to explore the features and heterogeneity associated with this phenotype. We develop a likelihood-based method which leverages small insertions and deletions for high-confidence classification of homologous recombination deficiency for exome-sequenced breast cancers. We then use multinomial elastic net regression modelling to develop a transcriptional signature of heterogeneous homologous recombination deficiency. This signature is then applied to single-cell RNA-sequenced breast cancer cohorts enabling analysis of homologous recombination deficiency heterogeneity and differential patterns of tumour microenvironment interactivity. RESULTS We demonstrate that the inclusion of indel events, even at low levels, improves homologous recombination deficiency classification. Whilst BRCA-positive homologous recombination deficient samples display strong similarities to those harbouring BRCA1/2 defects, they appear to deviate in microenvironmental features such as hypoxic signalling. We then present a 228-gene transcriptional signature which simultaneously characterises homologous recombination deficiency and BRCA1/2-defect status, and is associated with PARP inhibitor response. Finally, we show that this signature is applicable to single-cell transcriptomics data and predict that these cells present a distinct milieu of interactions with their microenvironment compared to their homologous recombination proficient counterparts, typified by a decreased cancer cell response to TNFα signalling. CONCLUSIONS We apply multi-scale approaches to characterise homologous recombination deficiency in breast cancer through the development of mutational and transcriptional signatures. We demonstrate how indels can improve homologous recombination deficiency classification in exome-sequenced breast cancers. Additionally, we demonstrate the heterogeneity of homologous recombination deficiency, especially in relation to BRCA1/2-defect status, and show that indications of this feature can be captured at a single-cell level, enabling further investigations into interactions between DNA repair deficient cells and their tumour microenvironment.
Collapse
Affiliation(s)
- Daniel H Jacobson
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Shi Pan
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jasmin Fisher
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
13
|
Thang NX, Han DW, Park C, Lee H, La H, Yoo S, Lee H, Uhm SJ, Song H, Do JT, Park KS, Choi Y, Hong K. INO80 function is required for mouse mammary gland development, but mutation alone may be insufficient for breast cancer. Front Cell Dev Biol 2023; 11:1253274. [PMID: 38020889 PMCID: PMC10646318 DOI: 10.3389/fcell.2023.1253274] [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: 07/05/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
The aberrant function of ATP-dependent chromatin remodeler INO80 has been implicated in multiple types of cancers by altering chromatin architecture and gene expression; however, the underlying mechanism of the functional involvement of INO80 mutation in cancer etiology, especially in breast cancer, remains unclear. In the present study, we have performed a weighted gene co-expression network analysis (WCGNA) to investigate links between INO80 expression and breast cancer sub-classification and progression. Our analysis revealed that INO80 repression is associated with differential responsiveness of estrogen receptors (ERs) depending upon breast cancer subtype, ER networks, and increased risk of breast carcinogenesis. To determine whether INO80 loss induces breast tumors, a conditional INO80-knockout (INO80 cKO) mouse model was generated using the Cre-loxP system. Phenotypic characterization revealed that INO80 cKO led to reduced branching and length of the mammary ducts at all stages. However, the INO80 cKO mouse model had unaltered lumen morphology and failed to spontaneously induce tumorigenesis in mammary gland tissue. Therefore, our study suggests that the aberrant function of INO80 is potentially associated with breast cancer by modulating gene expression. INO80 mutation alone is insufficient for breast tumorigenesis.
Collapse
Affiliation(s)
- Nguyen Xuan Thang
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Dong Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Chanhyeok Park
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Hyeonji Lee
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Hyeonwoo La
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Seonho Yoo
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Heeji Lee
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Sang Jun Uhm
- Department of Animal Science, Sangji University, Wonju, Republic of Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Jeong Tae Do
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Kyoung Sik Park
- Department of Surgery, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Chekhun V, Martynyuk О, Lukianova Y, Mushii O, Zadvornyi T, Lukianova N. FEATURES OF BREAST CANCER IN PATIENTS OF YOUNG AGE: SEARCH FOR DIAGNOSIS OPTIMIZATION AND PERSONALIZED TREATMENT. Exp Oncol 2023; 45:139-150. [PMID: 37824778 DOI: 10.15407/exp-oncology.2023.02.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Indexed: 10/14/2023]
Abstract
The statistical data of the recent decades demonstrate a rapid growth of breast cancer (BCa) incidence and a tendency toward its increase especially in young women. In the structure of morbidity of women in the age group of 18-29 years, BCa ranks first and in the age range of 15-39 years, BCa is one of the leading causes of mortality. According to the data of the epidemiological and clinical studies, the young age is an independent unfavorable prognostic factor of BCa that is associated with an unfavorable prognosis and low survival rates and is considered an important predictor of the disease aggressiveness, a high risk of metastasis and recurrence. The variability of clinicopathological and molecular-biological features of BCa in patients of different age groups as well as the varying course of the disease and different responses to the therapy are mediated by many factors. The analysis of the literature data on the factors and mechanisms of BCa initiation in patients of different age groups demonstrates that the pathogen- esis of BCa depends not only on the molecular-genetic alterations but also on the metabolic disorders caused by the current social and household rhythm of life and nutrition peculiarities. All these factors affect both the general con- dition of the body and the formation of an aggressive microenvironment of the tumor lesion. The identified features of transcriptome and the differential gene expression give evidence of different regulations of the immune response and the metabolic processes in BCa patients of different age groups. Association between the high expression of the components of the stromal microenvironment and the inflammatory immune infiltrate as well as the increased vascu- larization of the tumor lesion has been found in BCa tissue of young patients. Proving the nature of the formation of the landscape comprising molecular-genetic, cytokine, and immune factors of the tumor microenvironment will undoubtedly contribute to our understanding of the mechanisms of tumor growth allowing for the development of algorithms for delineating the groups at high risk of tumor progression, which requires more careful monitoring and personalized treatment approach. Th s will be helpful in the development of innovative technologies for complex BCa treatment.
Collapse
Affiliation(s)
- V Chekhun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine.
| | - О Martynyuk
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - Ye Lukianova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - O Mushii
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - T Zadvornyi
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - N Lukianova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| |
Collapse
|
15
|
Diniz-Gonçalves GS, Hielm-Björkman A, da Silva VB, Ribeiro LGR, da Costa Vieira-Filho CH, Silva LP, Barrouin-Melo SM, Cassali GD, Damasceno KA, Estrela-Lima A. GATA-3 expression and its correlation with prognostic factors and survival in canine mammary tumors. Front Vet Sci 2023; 10:1179808. [PMID: 37483298 PMCID: PMC10356988 DOI: 10.3389/fvets.2023.1179808] [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: 03/04/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction The transcription factor GATA-3 plays a significant role in mammary gland development and differentiation. Recent studies on human oncology have demonstrated its association with favorable pathologic factors in breast cancer. Canine mammary tumours, proposed as comparative and translational study models, have epidemiological, clinical, biological, and genetic characteristics similar to those of human breast cancers. Methods Here, we evaluated the frequency of GATA-3 expression in mammary tumors of dogs and its relationship with prognostic factors and survival. Tumor samples were obtained from 40 female dogs and grouped according to histological type into benign tumors (n = 10), carcinoma in mixed tumors (CMTs) (n = 20), and aggressive tumors (n = 10). CMTs were further separated according to histological grade, and data on clinical staging and diagnosis, histopathological grading, and survival rate were collected. Results GATA-3 and estrogen receptor (ER) expression were higher in benign and well-differentiated carcinomas than in aggressive tumors, which showed greater Ki-67 expression. The expression rate of ER in the studied groups was equivalent to that of GATA-3. We identified a strong positive correlation between GATA-3 and ER expression frequencies and a negative correlation between those of GATA-3 and Ki-67. There were associations between GATA-3 (p < 0.001), Ki-67 (p = 0.003), tumor size (p < 0.001), clinical stage (p = 0.002), lymph node metastasis (p < 0.001), and histological grade (p < 0.001) by univariate survival analysis. The parameters ER (p = 0.015) and GATA-3 (p = 0.005) also influenced survival in a multifactorial manner. Discussion Kaplan-Meier analysis of survival curves validated our previous findings that dogs with GATA-3 expression in ≥79.4% of cells had significantly higher survival rates (p < 0.001). The performance analysis showed that the expression of GATA-3 in ≥79.4% of cells effectively predicted survival or death in dogs with mammary tumors. Collectively, these results suggest that GATA-3 can be a relevant marker in the study of mammary tumor progression and has potential as a prognosis marker for predicting outcomes in canine mammary tumors.
Collapse
Affiliation(s)
| | - Anna Hielm-Björkman
- DogRisk Research Group, Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Vanessa Bonfim da Silva
- Research Center on Mammary Oncology NPqOM/HOSPMEV, Federal University of Bahia, Salvador, Brazil
| | - Lorena Gabriela Rocha Ribeiro
- Laboratory of Animal Pathology, Department of Veterinary Medicine, Federal University of Sergipe, São Cristóvão, Brazil
| | | | - Laís Pereira Silva
- Research Center on Mammary Oncology NPqOM/HOSPMEV, Federal University of Bahia, Salvador, Brazil
| | - Stella Maria Barrouin-Melo
- DogRisk Research Group, Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Geovanni Dantas Cassali
- Laboratory of Comparative Pathology, Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Karine Araújo Damasceno
- Experimental Pathology Laboratory (LAPEX), Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - Alessandra Estrela-Lima
- Research Center on Mammary Oncology NPqOM/HOSPMEV, Federal University of Bahia, Salvador, Brazil
- DogRisk Research Group, Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| |
Collapse
|
16
|
Anstine LJ, Majmudar PR, Aponte A, Singh S, Zhao R, Weber-Bonk KL, Abdul-Karim FW, Valentine M, Seachrist DD, Grennel-Nickelson KE, Cuellar-Vite L, Sizemore GM, Sizemore ST, Webb BM, Thompson CL, Keri RA. TLE3 Sustains Luminal Breast Cancer Lineage Fidelity to Suppress Metastasis. Cancer Res 2023; 83:997-1015. [PMID: 36696357 PMCID: PMC10089698 DOI: 10.1158/0008-5472.can-22-3133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/28/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
Breast cancer subtypes and their phenotypes parallel different stages of the mammary epithelial cell developmental hierarchy. Discovering mechanisms that control lineage identity could provide novel avenues for mitigating disease progression. Here we report that the transcriptional corepressor TLE3 is a guardian of luminal cell fate in breast cancer and operates independently of the estrogen receptor. In luminal breast cancer, TLE3 actively repressed the gene-expression signature associated with highly aggressive basal-like breast cancers (BLBC). Moreover, maintenance of the luminal lineage depended on the appropriate localization of TLE3 to its transcriptional targets, a process mediated by interactions with FOXA1. By repressing genes that drive BLBC phenotypes, including SOX9 and TGFβ2, TLE3 prevented the acquisition of a hybrid epithelial-mesenchymal state and reduced metastatic capacity and aggressive cellular behaviors. These results establish TLE3 as an essential transcriptional repressor that sustains the more differentiated and less metastatic nature of luminal breast cancers. Approaches to induce TLE3 expression could promote the acquisition of less aggressive, more treatable disease states to extend patient survival. SIGNIFICANCE Transcriptional corepressor TLE3 actively suppresses SOX9 and TGFβ transcriptional programs to sustain the luminal lineage identity of breast cancer cells and to inhibit metastatic progression.
Collapse
Affiliation(s)
- Lindsey J. Anstine
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Parth R. Majmudar
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Amy Aponte
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Salendra Singh
- Department of Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Ran Zhao
- Department of Qualitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Kristen L. Weber-Bonk
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Fadi W. Abdul-Karim
- Department of Pathology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mitchell Valentine
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio
| | - Darcie D. Seachrist
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Leslie Cuellar-Vite
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Gina M. Sizemore
- Department of Radiation Oncology and the James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Steven T. Sizemore
- Department of Radiation Oncology and the James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Bryan M. Webb
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
- Department of Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Cheryl L. Thompson
- Department of Public Health Sciences and the Penn State Cancer Institute, Hershey, Pennsylvania
| | - Ruth A. Keri
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
17
|
Geng X, Wang C, Gao X, Chowdhury P, Weiss J, Villegas JA, Saed B, Perera T, Hu Y, Reneau J, Sverdlov M, Wolfe A, Brown N, Harms P, Bailey NG, Inamdar K, Hristov AC, Tejasvi T, Montes J, Barrionuevo C, Taxa L, Casavilca S, de Pádua Covas Lage JLA, Culler HF, Pereira J, Runge JS, Qin T, Tsoi LC, Hong HS, Zhang L, Lyssiotis CA, Ohe R, Toubai T, Zevallos-Morales A, Murga-Zamalloa C, Wilcox RA. GATA-3 is a proto-oncogene in T-cell lymphoproliferative neoplasms. Blood Cancer J 2022; 12:149. [PMID: 36329027 PMCID: PMC9633835 DOI: 10.1038/s41408-022-00745-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Neoplasms originating from thymic T-cell progenitors and post-thymic mature T-cell subsets account for a minority of lymphoproliferative neoplasms. These T-cell derived neoplasms, while molecularly and genetically heterogeneous, exploit transcription factors and signaling pathways that are critically important in normal T-cell biology, including those implicated in antigen-, costimulatory-, and cytokine-receptor signaling. The transcription factor GATA-3 regulates the growth and proliferation of both immature and mature T cells and has recently been implicated in T-cell neoplasms, including the most common mature T-cell lymphoma observed in much of the Western world. Here we show that GATA-3 is a proto-oncogene across the spectrum of T-cell neoplasms, including those derived from T-cell progenitors and their mature progeny, and further define the transcriptional programs that are GATA-3 dependent, which include therapeutically targetable gene products. The discovery that p300-dependent acetylation regulates GATA-3 mediated transcription by attenuating DNA binding has novel therapeutic implications. As most patients afflicted with GATA-3 driven T-cell neoplasms will succumb to their disease within a few years of diagnosis, these findings suggest opportunities to improve outcomes for these patients.
Collapse
Affiliation(s)
- Xiangrong Geng
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Chenguang Wang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Xin Gao
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Pinki Chowdhury
- Department of Pediatrics, Dayton Children's Hospital, Wright State University Boonshoft School of Medicine, Dayton, OH, USA
| | - Jonathan Weiss
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - José A Villegas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Badeia Saed
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Thilini Perera
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Ying Hu
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - John Reneau
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maria Sverdlov
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Ashley Wolfe
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Noah Brown
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Paul Harms
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Nathanael G Bailey
- Division of Hematopathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kedar Inamdar
- Department of Pathology, Henry Ford Hospital, Detroit, MI, USA
| | - Alexandra C Hristov
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Trilokraj Tejasvi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Jaime Montes
- Department of Pathology, Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru
| | - Carlos Barrionuevo
- Department of Pathology, Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru
| | - Luis Taxa
- Department of Pathology, Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru
| | - Sandro Casavilca
- Department of Pathology, Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru
| | - J Luís Alberto de Pádua Covas Lage
- Department of Hematology, Hemotherapy and Cell Therapy, Faculty of Medicine, Sao Paulo University, Laboratory of Medical Investigation 31 in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology, Sao Paulo, Brazil
| | - Hebert Fabrício Culler
- Department of Hematology, Hemotherapy and Cell Therapy, Faculty of Medicine, Sao Paulo University, Laboratory of Medical Investigation 31 in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology, Sao Paulo, Brazil
| | - Juliana Pereira
- Department of Hematology, Hemotherapy and Cell Therapy, Faculty of Medicine, Sao Paulo University, Non-Hodgkin's Lymphomas and Histiocytic Disorders, Sao Paulo, Brazil
| | - John S Runge
- Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Hanna S Hong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Rintaro Ohe
- Department of Pathology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Tomomi Toubai
- Department of Internal Medicine III, Division of Hematology and Cell Therapy, Yamagata University of Medicine, Yamagata, Japan
| | | | | | - Ryan A Wilcox
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Khalaji A, Haddad S, Yazdani Y, Moslemi M, Alizadeh L, Baradaran B. A bioinformatics-based study on the Cisplatin-resistant lung cancer cells; what are the orchestrators of this phenom? Gene X 2022; 834:146668. [PMID: 35690284 DOI: 10.1016/j.gene.2022.146668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 11/04/2022] Open
Abstract
Lung cancer represents a significant global health issue and is among the central causes of mortality and morbidity around the world. Unfortunately, the majority of lung cancer patients acquire drug resistant to chemotherapy either intrinsically or acquired after Cisplatin treatment. It is indicated that increasing or decreasing the expression of particular genes can affect chemotherapeutic sensitivity or resistance. As a result, gaining a deeper knowledge of the changed expression of genes implicated in lung cancer drug resistance, as well as developing novel therapeutic techniques, are critical targets for continued advancement in lung cancer treatment. In the present study, we aimed to find key regulatory genes in the progression of Cisplatin resistance in A-549 lung cancer cells. In this regard, microarray dataset of Cisplatin-resistant and Cisplatin-sensitive was retrieved from the Gene Expression Omnibus (GEO) with accession number of GSE108214. Then, differentially expressed genes (DEGs) between sensitive and resistant lung cancer cells were obtained by using R software v4.0.2 and related packages. We recognized CEACAM1, DGKA, ARHGEF4, and THSD4 are involved in the drug resistance. Experimentally, Cisplatin-resistant A-549 cells were developed and analyzed by MTT assay. Besides, the expression of candidate genes were analyzed in these cells compared to Cisplatin-sensitive A-549 cells by qRT-PCR. The findings presented that the expression of CEACAM1, DGKA, ARHGEF4, and THSD4 was altered following the induction of Cisplatin resistance in A549 cells.
Collapse
Affiliation(s)
- Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Haddad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Yazdani
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Emam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Moslemi
- Department of Internal Medicine, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Leila Alizadeh
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Identification of Potential Biomarkers for Progression and Prognosis of Bladder Cancer by Comprehensive Bioinformatics Analysis. JOURNAL OF ONCOLOGY 2022; 2022:1802706. [PMID: 35498536 PMCID: PMC9042640 DOI: 10.1155/2022/1802706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/22/2022] [Indexed: 11/23/2022]
Abstract
Background. Bladder cancer (BLCA) is a highly malignant tumor that develops in the urinary system. Identification of biomarkers in progression and prognosis is crucial for the treatment of BLCA. BLCA-related differentially expressed genes (DEGs) were authenticated by screening the DEGs and weighted gene coexpression network analysis (WGCNA). LASSO and SVM-RFE algorithms were utilized to screen the feature genes in BLCA. Survival analysis was performed using the Kaplan–Meier curve provided by the ‘survival' R package. The BLCA samples were clustered by hclust based on the immune score matrix calculated by the single-sample GSEA (ssGSEA) algorithm. The immune, stromal, and ESTIMATE scores of each BLCA patient were calculated by applying the ESTIMATE algorithm. ssGSEA was conducted to explore the function of characteristic genes in BLCA. The expression of characteristic genes in clinical cancer tissue, and the pericancerous tissue of BLCA patients was verified using qRT-PCR assays. A total of 189 BLCA-related DEGs were identified. Fourteen feature genes were defined by LASSO and SVM-RFE algorithms. Five characteristic genes, including SMYD2, GAPDHP1, ATP1A2, CILP, and THSD4, were related to the OS of BLCA. The correlation analysis of five characteristic genes and clinicopathological factors showed that five genes played a role in the progression of BLCA. Additionally, the expression of five characteristic genes in clinical cancer tissues and pericarcinomatous tissues from BLCA patients was verified by qRT-PCR, which was consistent with the result from the public database. Finally, we discovered five prognostic genes linked to BLCA progression, which might serve as a theoretical basis for prognosis and treatment targets for BLCA patients.
Collapse
|
20
|
Bianco G, Coto-Llerena M, Gallon J, Kancherla V, Taha-Mehlitz S, Marinucci M, Konantz M, Srivatsa S, Montazeri H, Panebianco F, Tirunagaru VG, De Menna M, Paradiso V, Ercan C, Dahmani A, Montaudon E, Beerenwinkel N, Kruithof-de Julio M, Terracciano LM, Lengerke C, Jeselsohn RM, Doebele RC, Bidard FC, Marangoni E, Ng CKY, Piscuoglio S. GATA3 and MDM2 are synthetic lethal in estrogen receptor-positive breast cancers. Commun Biol 2022; 5:373. [PMID: 35440675 PMCID: PMC9018745 DOI: 10.1038/s42003-022-03296-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
Synthetic lethal interactions, where the simultaneous but not individual inactivation of two genes is lethal to the cell, have been successfully exploited to treat cancer. GATA3 is frequently mutated in estrogen receptor (ER)-positive breast cancers and its deficiency defines a subset of patients with poor response to hormonal therapy and poor prognosis. However, GATA3 is not yet targetable. Here we show that GATA3 and MDM2 are synthetically lethal in ER-positive breast cancer. Depletion and pharmacological inhibition of MDM2 significantly impaired tumor growth in GATA3-deficient models in vitro, in vivo and in patient-derived organoids/xenograft (PDOs/PDX) harboring GATA3 somatic mutations. The synthetic lethality requires p53 and acts via the PI3K/Akt/mTOR pathway. Our results present MDM2 as a therapeutic target in the substantial cohort of ER-positive, GATA3-mutant breast cancer patients. With MDM2 inhibitors widely available, our findings can be rapidly translated into clinical trials to evaluate in-patient efficacy.
Collapse
Affiliation(s)
- Gaia Bianco
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mairene Coto-Llerena
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - John Gallon
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Venkatesh Kancherla
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Stephanie Taha-Mehlitz
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mattia Marinucci
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martina Konantz
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sumana Srivatsa
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Hesam Montazeri
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Federica Panebianco
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Marta De Menna
- Department of Biomedical Research, Urology Group, University of Bern, Bern, Switzerland
| | - Viola Paradiso
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Caner Ercan
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Ahmed Dahmani
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Luigi M Terracciano
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Pathology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Rinath M Jeselsohn
- Division of Women's Cancers, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Charlotte K Y Ng
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Salvatore Piscuoglio
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland.
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
21
|
Vini R, Rajavelu A, Sreeharshan S. 27-Hydroxycholesterol, The Estrogen Receptor Modulator, Alters DNA Methylation in Breast Cancer. Front Endocrinol (Lausanne) 2022; 13:783823. [PMID: 35360070 PMCID: PMC8961300 DOI: 10.3389/fendo.2022.783823] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/13/2022] [Indexed: 01/01/2023] Open
Abstract
27-hydroxycholesterol (27-HC) is the first known endogenous selective estrogen receptor modulator (SERM), and its elevation from normal levels is closely associated with breast cancer. A plethora of evidence suggests that aberrant epigenetic signatures in breast cancer cells can result in differential responses to various chemotherapeutics and often leads to the development of resistant cancer cells. Such aberrant epigenetic changes are mostly dictated by the microenvironment. The local concentration of oxygen and metabolites in the microenvironment of breast cancer are known to influence the development of breast cancer. Hence, we hypothesized that 27-HC, an oxysterol, which has been shown to induce breast cancer progression via estrogen receptor alpha (ERα) and liver X receptor (LXR) and by modulating immune cells, may also induce epigenetic changes. For deciphering the same, we treated the estrogen receptor-positive cells with 27-HC and identified DNA hypermethylation on a subset of genes by performing DNA bisulfite sequencing. The genes that showed significant DNA hypermethylation were phosphatidylserine synthase 2 (PTDSS2), MIR613, indoleamine 2,3-dioxygenase 1 (IDO1), thyroid hormone receptor alpha (THRA), dystrotelin (DTYN), and mesoderm induction early response 1, family member 3 (MIER). Furthermore, we found that 27-HC weakens the DNMT3B association with the ERα in MCF-7 cells. This study reports that 27-HC induces aberrant DNA methylation changes on the promoters of a subset of genes through modulation of ERα and DNMT3B complexes to induce the local DNA methylation changes, which may dictate drug responses and breast cancer development.
Collapse
Affiliation(s)
- Ravindran Vini
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Arumugam Rajavelu
- Pathogen Biology, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology, Chennai, India
- *Correspondence: Arumugam Rajavelu, ; Sreeja Sreeharshan,
| | - Sreeja Sreeharshan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- *Correspondence: Arumugam Rajavelu, ; Sreeja Sreeharshan,
| |
Collapse
|
22
|
Kim HJ, Kim S, Freedman RA, Partridge AH. The impact of young age at diagnosis (age <40 years) on prognosis varies by breast cancer subtype: A U.S. SEER database analysis. Breast 2021; 61:77-83. [PMID: 34923225 PMCID: PMC8693310 DOI: 10.1016/j.breast.2021.12.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 12/20/2022] Open
Abstract
Purpose Women under 40 years old are at increased risk for developing human epidermal growth factor receptor 2 (HER2) positive or triple negative subtype and more advanced breast cancer, yet young age itself has also historically been an independent prognostic factor. Methods Using the Surveillance, Epidemiology, and End Results (SEER) Program, we examined data for 271,173 women with stage I-III breast cancer between 2010 and 2015. Using Fine and Gray regression models to account for competing risks, we examined the risk of breast cancer-specific death by age and clinical subtypes, considering grade, hormone receptor (HR) and HER2 status, adjusting for demographic, clinical and treatment variables. Results Of 271,173 women eligible for analysis, 14,109 were <40 years of age. Women under 40 years old were more likely to be non-white, uninsured, and to have higher stage, higher grade, HER2-positive and triple-negative subtype disease (all, p < 0.001). Compared to women ages 40–60, women ages <40 had higher breast cancer mortality (hazard ratio, 1.8; 95% confidence interval (CI) 1.6–1.9) in unadjusted analysis. In models controlling for demographic, clinical and treatment factors, young age was significantly associated with an increased risk of breast cancer mortality among women with HR-positive, lower grade disease (hazard ratio 1.7; 95% CI 1.4–2.1) but not for women with high grade/HR-positive, HER2-positive, or triple-negative disease. Women age >75 had increased breast cancer mortality in all subtypes. Conclusion With modern clinical subtyping, age under 40 remains independently associated with worse outcomes in 30 months follow-up only in HR-positive, lower grade disease. Young women present with more advanced and aggressive types of breast cancer. Young age is not an independent prognostic factor in HER2+ breast cancer or TNBC. Young age is independently associated with poor outcomes in HR+/lower grade disease.
Collapse
Affiliation(s)
- Hee Jeong Kim
- Department of Surgery, College of Medicine, University of Ulsan, Asan Medical Center, Seoul, South Korea
| | - Seonok Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, Seoul, Republic of Korea
| | - Rachel A Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
23
|
Aman S, Li Y, Cheng Y, Yang Y, Lv L, Li B, Xia K, Li S, Wu H. DACH1 inhibits breast cancer cell invasion and metastasis by down-regulating the transcription of matrix metalloproteinase 9. Cell Death Discov 2021; 7:351. [PMID: 34772908 PMCID: PMC8590022 DOI: 10.1038/s41420-021-00733-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Human Dachshund homolog 1 (DACH1) is usually defined as a tumor suppressor, which plays an influential role in tumor growth and metastasis in a variety of cancer cells. However, the underlying mechanisms in these process are not yet fully clarified. In this study, DACH1 inhibited the invasion and metastasis of breast cancer cells by decreasing MMP9 expression. Mechanistically, DACH1 represses the transcriptional level of MMP9 by interacting with p65 and c-Jun at the NF-κB and AP-1 binding sites in MMP9 promoter respectively, and the association of DACH1 and p65 promote the recruitment of HDAC1 to the NF-κB binding site in MMP9 promoter, resulting in the reduction of the acetylation level and the transcriptional activity of p65. Accordingly, the level of MMP9 was decreased. In conclusion, we found a new mechanism that DACH1 could inhibit the metastasis of breast cancer cells by inhibiting the expression of MMP9.
Collapse
Affiliation(s)
- Sattout Aman
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Yanan Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Yunmeng Cheng
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Yuxi Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Linlin Lv
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Bowen Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Kangkai Xia
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China.
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China.
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, Dalian, China.
- 2 Ling Gong Road, Dalian, 116024, Liaoning, China.
| |
Collapse
|
24
|
Liu J, Huang Z, Chen HN, Qin S, Chen Y, Jiang J, Zhang Z, Luo M, Ye Q, Xie N, Zhou ZG, Wei Y, Xie K, Huang C. ZNF37A promotes tumor metastasis through transcriptional control of THSD4/TGF-β axis in colorectal cancer. Oncogene 2021; 40:3394-3407. [PMID: 33875786 DOI: 10.1038/s41388-021-01713-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 02/05/2023]
Abstract
Poorly differentiated colorectal cancer (CRC) is characterized by aggressive invasion and stromal fibroblast activation, which results in rapid progression and poor therapeutic consequences. However, the regulatory mechanism involved remains unclear. Here, we showed that ZNF37A, a member of KRAB-ZFP family, was upregulated in poorly differentiated CRCs and associated with tumor metastasis. ZNF37A enhanced the metastatic potential of multiple CRC cell lines and promoted distant metastasis in an orthotopic CRC model. Further investigation attributed the ZNF37A-exacerbated metastasis to increased extracellular TGF-β and the consequent activation of cancer-associated fibroblasts (CAFs) in tumor microenvironment (TME). Mechanistically, ZNF37A formed a complex with KAP1 and bound to the promoter of THSD4, a TME modulator, to suppress its transcription, which is required for ZNF37A-mediated TGF-β activation and CRC metastasis. Collectively, our study indicates that ZNF37A promotes TGF-β signaling in CRC cells and activates CAFs by transcriptionally repressing THSD4 to drive CRC metastasis, implicating ZNF37A as a potential biomarker for CRC differentiation and progression.
Collapse
Affiliation(s)
- Jiayang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Hai-Ning Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Yan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Qin Ye
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Zong-Guang Zhou
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China. .,School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
25
|
Majed SO, Mustafa SA. MACE-Seq-based coding RNA and TrueQuant-based small RNA profile in breast cancer: tumor-suppressive miRNA-1275 identified as a novel marker. BMC Cancer 2021; 21:473. [PMID: 33910530 PMCID: PMC8082896 DOI: 10.1186/s12885-021-08218-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
Introduction Disruption of cellular processes in the breast by abnormally expressed miRNA is characterized to develop cancer. We aimed to identify the differential expression of small RNAs (sRNAs) and mRNAs in formalin-fixed paraffin-embedded (FFPE) tissue of the breast cancer (BC) and normal adjacent tissue (NAT). Another aim is to determine the differential expression of miR-1275 as a novel biomarker for BC and also identify its target genes. Methods TrueQuant method for analysis of sRNA expression and MACE-sequencing method for analysis of gene expression were used analyzing. The RT-qPCR technique was used to confirm miR-1275 down expression. Target genes of miR-1275 were computationally identified using target prediction sites and also the expression level of them was experimentally determined among the expressed genes. Results TrueQuant findings showed that 1400 sRNAs were differentially expressed in the FFPE tissue of two Kurdish cases with BC, as compared to NAT. Among the sRNAs, 29 small RNAs were shown to be significantly downregulated in BC cells. The RT-qPCR results confirmed that miR-1275 was significantly down-expressed in 20 Kurdish cases with BC compared to NAT. However, Overall survival (OS) analysis revealed that the correlation between the expression level of miR-1275 and clinical significance was highly corrected in cases with BC (OS rate: P = 0.0401). The MACE-seq results revealed that 26,843 genes were differentially expressed in the BC tissue compared to NAT, but 7041 genes were displayed in a scatter plot. Furthermore, putative target genes (DVL3, PPP2R2D, THSD4, CREB1, SYT7, and PRKACA) were computationally identified as direct targets of miR-1275 in several target predicted sites. The MACE-seq results revealed that the expression level of these targets was increased in BC tissue compared to NAT. The level of these targets was negatively associated with miR-1275 expression. Finally, the role of down-regulated miR-1275 on its targets in biological mechanisms of BC cells was identified; including cell growth, proliferation, movement, invasion, metastasis, and apoptosis. Conclusion Down-expressed miR-1275, a tumor suppressor, is a novel biomarker for early detection of BC. DVL3, PPP2R2D, THSD4, CREB1, SYT7, and PRKACA are newly identified to be targeted by miR-1275.
Collapse
Affiliation(s)
- Sevan Omer Majed
- Biology Department, College of Education, Salahaddin University-Erbil, Erbil, Iraq.
| | - Suhad Asad Mustafa
- Research Center, Molecular Genetics lab, Salahaddin University-Erbil, Erbil, Iraq
| |
Collapse
|
26
|
LncRNA GNAS-AS1 facilitates ER+ breast cancer cells progression by promoting M2 macrophage polarization via regulating miR-433-3p/GATA3 axis. Biosci Rep 2021; 40:225295. [PMID: 32538432 PMCID: PMC7327181 DOI: 10.1042/bsr20200626] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Objective: ER+ breast cancer is the most common type of breast cancer, which seriously affects the physical and mental health of women. Recently, lncRNAs mediated tumor-associated macrophages (TAM) were identified to involve in tumorigenesis. Therefore, the present study aimed at demonstrating the regulatory network of GNAS-AS1 in TAM-mediated ER+ breast cancer progress. Methods: The expression levels of genes were evaluated using qRT-PCR. The proportions of polarized macrophages (M1, M2) were assessed by flow cytometry. Cell proliferation, migration and invasion were evaluated by CCK-8, wound healing and transwell assay, respectively. Double-luciferase reporter system was used to detect the interaction between molecules. Western blot was applied to test protein levels. Results: The expression of GNAS-AS1 was obviously increased in ER+ breast cancer tissues and cell lines, as well as M2 macrophages. GNAS-AS1 facilitated the capabilities of proliferation, migration and invasion of ER+ breast cancer cells by accelerating M2 macrophage polarization via directly sponging miR-433-3p. GATA3, as a target of miR-433-3p, could positively regulate by GNAS-AS1. Furthermore, either miR-433-3p overexpression or GATA3 knockdown impaired the effects of GNAS-AS1 on M2 macrophage polarization and ER+ breast cancer cells progression. Conclusion: GNAS-AS1/miR-433-3p/GATA3 axis promoted proliferation, metastasis of ER+ breast cancer cells by accelerating M2 macrophage polarization. The mechanism may provide a new strategy and target for ER+ breast cancer treatment.
Collapse
|
27
|
Shin EM, Huynh VT, Neja SA, Liu CY, Raju A, Tan K, Tan NS, Gunaratne J, Bi X, Iyer LM, Aravind L, Tergaonkar V. GREB1: An evolutionarily conserved protein with a glycosyltransferase domain links ERα glycosylation and stability to cancer. SCIENCE ADVANCES 2021; 7:7/12/eabe2470. [PMID: 33731348 PMCID: PMC7968844 DOI: 10.1126/sciadv.abe2470] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 01/29/2021] [Indexed: 05/03/2023]
Abstract
What covalent modifications control the temporal ubiquitination of ERα and hence the duration of its transcriptional activity remain poorly understood. We show that GREB1, an ERα-inducible enzyme, catalyzes O-GlcNAcylation of ERα at residues T553/S554, which stabilizes ERα protein by inhibiting association with the ubiquitin ligase ZNF598. Loss of GREB1-mediated glycosylation of ERα results in reduced cellular ERα levels and insensitivity to estrogen. Higher GREB1 expression in ERα+ve breast cancer is associated with greater survival in response to tamoxifen, an ERα agonist. Mice lacking Greb1 exhibit growth and fertility defects reminiscent of phenotypes in ERα-null mice. In summary, this study identifies GREB1, a protein with an evolutionarily conserved domain related to DNA-modifying glycosyltransferases of bacteriophages and kinetoplastids, as the first inducible and the only other (apart from OGT) O-GlcNAc glycosyltransferase in mammalian cytoplasm and ERα as its first substrate.
Collapse
Affiliation(s)
- Eun Myoung Shin
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Vinh Thang Huynh
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sultan Abda Neja
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Chia Yi Liu
- Bioprocessing Technology Institute (BTI), A*STAR, Singapore, Singapore
| | - Anandhkumar Raju
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Kelly Tan
- Bioprocessing Technology Institute (BTI), A*STAR, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive,, Singapore 637551, Singapore
| | - Jayantha Gunaratne
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117594, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute (BTI), A*STAR, Singapore, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Lakshminarayan M Iyer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Vinay Tergaonkar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research, Singapore 138673, Singapore.
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| |
Collapse
|
28
|
Sun H, Cui Y, Wang H, Liu H, Wang T. Comparison of methods for the detection of outliers and associated biomarkers in mislabeled omics data. BMC Bioinformatics 2020; 21:357. [PMID: 32795265 PMCID: PMC7646480 DOI: 10.1186/s12859-020-03653-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/10/2020] [Indexed: 02/08/2023] Open
Abstract
Background Previous studies have reported that labeling errors are not uncommon in omics data. Potential outliers may severely undermine the correct classification of patients and the identification of reliable biomarkers for a particular disease. Three methods have been proposed to address the problem: sparse label-noise-robust logistic regression (Rlogreg), robust elastic net based on the least trimmed square (enetLTS), and Ensemble. Ensemble is an ensembled classification based on distinct feature selection and modeling strategies. The accuracy of biomarker selection and outlier detection of these methods needs to be evaluated and compared so that the appropriate method can be chosen. Results The accuracy of variable selection, outlier identification, and prediction of three methods (Ensemble, enetLTS, Rlogreg) were compared for simulated and an RNA-seq dataset. On simulated datasets, Ensemble had the highest variable selection accuracy, as measured by a comprehensive index, and lowest false discovery rate among the three methods. When the sample size was large and the proportion of outliers was ≤5%, the positive selection rate of Ensemble was similar to that of enetLTS. However, when the proportion of outliers was 10% or 15%, Ensemble missed some variables that affected the response variables. Overall, enetLTS had the best outlier detection accuracy with false positive rates < 0.05 and high sensitivity, and enetLTS still performed well when the proportion of outliers was relatively large. With 1% or 2% outliers, Ensemble showed high outlier detection accuracy, but with higher proportions of outliers Ensemble missed many mislabeled samples. Rlogreg and Ensemble were less accurate in identifying outliers than enetLTS. The prediction accuracy of enetLTS was better than that of Rlogreg. Running Ensemble on a subset of data after removing the outliers identified by enetLTS improved the variable selection accuracy of Ensemble. Conclusions When the proportion of outliers is ≤5%, Ensemble can be used for variable selection. When the proportion of outliers is > 5%, Ensemble can be used for variable selection on a subset after removing outliers identified by enetLTS. For outlier identification, enetLTS is the recommended method. In practice, the proportion of outliers can be estimated according to the inaccuracy of the diagnostic methods used.
Collapse
Affiliation(s)
- Hongwei Sun
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan City, 030001, Shanxi, China.,Department of Health Statistics, School of Public Health and Management, Binzhou Medical University, City, Yantai, 264003, Shandong, China
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, 48824, USA
| | - Hui Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan City, 030001, Shanxi, China
| | - Haixia Liu
- Department of Health Statistics, School of Public Health and Management, Binzhou Medical University, City, Yantai, 264003, Shandong, China
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan City, 030001, Shanxi, China.
| |
Collapse
|
29
|
Hruschka N, Kalisz M, Subijana M, Graña-Castro O, Del Cano-Ochoa F, Brunet LP, Chernukhin I, Sagrera A, De Reynies A, Kloesch B, Chin SF, Burgués O, Andreu D, Bermejo B, Cejalvo JM, Sutton J, Caldas C, Ramón-Maiques S, Carroll JS, Prat A, Real FX, Martinelli P. The GATA3 X308_Splice breast cancer mutation is a hormone context-dependent oncogenic driver. Oncogene 2020; 39:5455-5467. [PMID: 32587399 PMCID: PMC7410826 DOI: 10.1038/s41388-020-1376-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022]
Abstract
As the catalog of oncogenic driver mutations is expanding, it becomes clear that alterations in a given gene might have different functions and should not be lumped into one class. The transcription factor GATA3 is a paradigm of this. We investigated the functions of the most common GATA3 mutation (X308_Splice) and five additional mutations, which converge into a neoprotein that we called "neoGATA3," associated with excellent prognosis in patients. Analysis of available molecular data from >3000 breast cancer patients revealed a dysregulation of the ER-dependent transcriptional response in tumors carrying neoGATA3-generating mutations. Mechanistic studies in vitro showed that neoGATA3 interferes with the transcriptional programs controlled by estrogen and progesterone receptors, without fully abrogating them. ChIP-Seq analysis indicated that ER binding is reduced in neoGATA3-expressing cells, especially at distal regions, suggesting that neoGATA3 interferes with the fine tuning of ER-dependent gene expression. This has opposite outputs in distinct hormonal context, having pro- or anti-proliferative effects, depending on the estrogen/progesterone ratio. Our data call for functional analyses of putative cancer drivers to guide clinical application.
Collapse
Affiliation(s)
- Natascha Hruschka
- Institute of Cancer Research, Medical University Vienna, Comprehensive Cancer Center, Vienna, Austria
| | - Mark Kalisz
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, CIBERONC, Madrid, Spain
| | - Maria Subijana
- Institute of Cancer Research, Medical University Vienna, Comprehensive Cancer Center, Vienna, Austria
| | - Osvaldo Graña-Castro
- Bioinformatics Unit, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Francisco Del Cano-Ochoa
- Department of Genome Dynamics and Function, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Laia Paré Brunet
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Igor Chernukhin
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Ana Sagrera
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, CIBERONC, Madrid, Spain
| | - Aurelien De Reynies
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013, Paris, France
| | - Bernhard Kloesch
- Institute of Cancer Research, Medical University Vienna, Comprehensive Cancer Center, Vienna, Austria
| | - Suet-Feung Chin
- Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Octavio Burgués
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Pathology Department, Hospital Clínico Universitario-CIBERONC, Valencia, Spain
| | - David Andreu
- Laboratory of Proteomics and Protein Chemistry, Universitat Pompeu Fabra, Barcelona, Spain
| | - Begoña Bermejo
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Oncology and Hematology Department, Hospital Clínico Universitario-CIBERONC, Valencia, Spain
| | - Juan Miguel Cejalvo
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Oncology and Hematology Department, Hospital Clínico Universitario-CIBERONC, Valencia, Spain
| | - Joe Sutton
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Carlos Caldas
- Department of Oncology, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Santiago Ramón-Maiques
- Department of Genome Dynamics and Function, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - Aleix Prat
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, CIBERONC, Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Paola Martinelli
- Institute of Cancer Research, Medical University Vienna, Comprehensive Cancer Center, Vienna, Austria.
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, CIBERONC, Madrid, Spain.
- Cancer Cell Signaling Department, Boehringer-Ingelheim RCV, Vienna, Austria.
| |
Collapse
|
30
|
Entry and exit of chemotherapeutically-promoted cellular dormancy in glioblastoma cells is differentially affected by the chemokines CXCL12, CXCL16, and CX3CL1. Oncogene 2020; 39:4421-4435. [PMID: 32346064 PMCID: PMC7253351 DOI: 10.1038/s41388-020-1302-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a malignant brain tumor that evades therapy regimens. Since cellular dormancy is one strategy for surviving, and since chemokines determine the environmental conditions in which dormancy occurs, we investigated how chemokines affect temozolomide (TMZ)-promoted cellular dormancy entry and exit in GBM cells. TMZ administration over ten days promoted cellular dormancy entry, whereas discontinuing TMZ for a further 15 days resulted in resumption of proliferation. Co-administration of a chemokine cocktail containing CXCL12, CXCL16, and CX3CL1 resulted in both delayed entry and exit from cellular dormancy. A microarray-based transcriptome analysis in LN229 GBM cells revealed that cellular dormancy entry was characterized by an increased expression of CCL2 and SAA2, while THSD4, FSTL3, and VEGFC were upregulated during dormancy exit. Co-stimulation with the chemokine cocktail reduced upregulation of identified genes. After verifying the appearance of identified genes in human GBM primary cultures and ex vivo samples, we clarified whether each chemokine alone impacts cellular dormancy mechanisms using specific antagonists and selective CRISPR/Cas9 clones. While expression of CCL2 and SAA2 in LN229 cells was altered by the CXCL12-CXCR4-CXCR7 axis, CXCL16 and CX3CL1 contributed to reduced upregulation of THSD4 and, to a weaker extent, of VEGFC. The influence on FSTL3 expression depended on the entire chemokine cocktail. Effects of chemokines on dormancy entry and exit-associated genes were detectable in human GBM primary cells, too, even if in a more complex, cell-specific manner. Thus, chemokines play a significant role in the regulation of TMZ-promoted cellular dormancy in GBMs.
Collapse
|
31
|
Wu J, Li X, Luo F, Yan J, Yang K. Screening key miRNAs and genes in prostate cancer by microarray analysis. Transl Cancer Res 2020; 9:856-868. [PMID: 35117431 PMCID: PMC8799076 DOI: 10.21037/tcr.2019.12.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/29/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is the second most frequent cancer and the fifth leading cause of cancer-related death in men while the mechanisms remain unclear. METHODS Differentially expressed mRNAs (DEmRNAs) and miRNAs (DEmiRNAs) between PCa and non-tumor controls were identified by using microarray analysis. Functional annotation of DEmRNAs, construction of protein-protein interaction (PPI) network and prediction of upstream transcription factors and downstream target DEmRNAs of DEmiRNAs were conducted to further research functions of key DEmRNAs and DEmiRNAs. Validation of selected DEmRNAs and survival analysis were conducted by using The Cancer Genome Atlas (TCGA). RESULTS Total of 91 DEmRNAs and 62 DEmiRNAs were obtained. Thrombospondin-4 precursor (THBS4) was the most significantly up-regulated DEmRNA whose product was predicted to interact with the hub protein of the PCa-specific PPI network, collagen type I alpha 1 chain (COL1A1). Both ATP binding cassette subfamily C member 4 (ABCC4) and endothelin receptor type B (EDNRB) have great prognostic value for PCa. Thrombospondin type 1 domain containing 4 (THSD4) was a down-regulated DEmRNA regulated by several cancer-related miRNAs including has-miR-107, hsa-miR-3175 and hsa-miR-484. Two miRNAs (hsa-miR-428 and hsa-miR-4284) involve in PCa by regulating BMP5-BAMBI interaction and TGF-beta signaling pathway. The expression of selected DEmRNAs between PCa and non-tumor controls in TCGA was consistent with that in our microarray analysis, generally. CONCLUSIONS Key DEmRNAs and DEmiRNAs between PCa and non-tumor controls were identified in this study which provided clues for exploring the molecular mechanism and developing potential biomarkers and therapeutic target sites for PCa.
Collapse
Affiliation(s)
- Jianhui Wu
- Department of Urology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Xuemei Li
- Department of Endocrinology, Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Fei Luo
- Department of Urology, Tianjin Union Medical Center, Tianjin 300121, China
| | - Jun Yan
- Department of Pathology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Kuo Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
32
|
Romano O, Miccio A. GATA factor transcriptional activity: Insights from genome-wide binding profiles. IUBMB Life 2019; 72:10-26. [PMID: 31574210 DOI: 10.1002/iub.2169] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/05/2019] [Indexed: 01/07/2023]
Abstract
The members of the GATA family of transcription factors have homologous zinc fingers and bind to similar sequence motifs. Recent advances in genome-wide technologies and the integration of bioinformatics data have led to a better understanding of how GATA factors regulate gene expression; GATA-factor-induced transcriptional and epigenetic changes have now been analyzed at unprecedented levels of detail. Here, we review the results of genome-wide studies of GATA factor occupancy in human and murine cell lines and primary cells (as determined by chromatin immunoprecipitation sequencing), and then discuss the molecular mechanisms underlying the mediation of transcriptional and epigenetic regulation by GATA factors.
Collapse
Affiliation(s)
- Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Annarita Miccio
- Laboratory of chromatin and gene regulation during development, Imagine Institute, INSERM UMR, Paris, France.,Paris Descartes, Sorbonne Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
33
|
Fararjeh AFS, Tu SH, Chen LC, Liu YR, Lin YK, Chang HL, Chang HW, Wu CH, Hwang-Verslues WW, Ho YS. The impact of the effectiveness of GATA3 as a prognostic factor in breast cancer. Hum Pathol 2018; 80:219-230. [PMID: 29902578 DOI: 10.1016/j.humpath.2018.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/24/2018] [Accepted: 06/01/2018] [Indexed: 01/21/2023]
Abstract
The transcription factor GATA3 plays a significant role in mammary gland development and differentiation. We analyzed expression of GATA3 in breast cancer (BC) cell lines and clinical specimens from BC patients in Taiwan. Semiquantitative reverse-transcription polymerase chain reaction (RT-PCR), quantitative real-time PCR were carried out to determine the mRNA level of GATA3 from 241 pairs of matched tumor and adjacent normal tissues from anonymous female donors. GATA3 immunohistochemistry (IHC) staining and H-score were performed (n = 25). Inducing and silencing of GATA3 were done by exposure MCF-7 cell line to nicotine or curcumin, respectively. GATA3 expression was detected in most of the estrogen receptor-positive (ER+) tumor specimens (176/241, 73%) compared with paired normal tissues (65/241, 27%) (P < .001). The GATA3 level was highest in Luminal A, and independent t-tests revealed higher GATA3 was associated with ER+ (P = .018) and BC stages (stage II, and stage IV). Nuclear protein expression of GATA3 was detected in tumor tissues (P < .001) with higher H-score in Luminal A patients (P = .012). Kaplan-Meier survival analyses showed that ER+/progesterone receptor (PgR)+ and lower grade BC patients with relatively high GATA3 had better clinical overall survival (OS). GATA3 regulates ERα and BCL-2 as BC luminal subtype markers. Cox univariate and multivariate analyses demonstrated that the expression of GATA3 was an effective predictor of the risk of death. We demonstrated a correlation between GATA3 expression and only ER+ and suggest that a higher GATA3 expression is a good prognostic factor for OS for ER+ BC patients.
Collapse
Affiliation(s)
- Abdul-Fattah Salah Fararjeh
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, 110 Taipei, Taiwan
| | - Shih-Hsin Tu
- Breast Medical Center, Taipei Medical University Hospital, Taipei, Taiwan; Taipei Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Ching Chen
- Breast Medical Center, Taipei Medical University Hospital, Taipei, Taiwan; Taipei Cancer Center, Taipei Medical University Hospital, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Yen-Kuang Lin
- Research Center of Biostatistics, Taipei Medical University, Taipei, Taiwan
| | - Hang-Lung Chang
- Department of General Surgery, EnChun Kong Hospital, New Taipei City, Taiwan
| | - Hui-Wen Chang
- Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Hsiung Wu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of General Surgery, EnChun Kong Hospital, New Taipei City, Taiwan
| | | | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Department of Laboratory Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
34
|
Huang L, Guo Y, Cao D, Liu X, Zhang L, Cao K, Hu T, Qi Y, Xu C. Effects of Helicobacter pylori on the expression levels of GATA-3 and connexin 32 and the GJIC function in gastric epithelial cells and their association by promoter analysis. Oncol Lett 2018; 16:1650-1658. [PMID: 30008849 PMCID: PMC6036278 DOI: 10.3892/ol.2018.8796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
The present study aimed to explore the effects of Helicobacter pylori (H. pylori) infection on the expression of transcription factor GATA binding protein 3 (GATA-3) and connexin 32 (Cx32) in cultured gastric mucosa cells, and their association with each other. GES-1 cells were co-cultured with East Asian type cytotoxin-associated gene A+ H. pylori in the H. pylori group, and without H. pylori culture in the control group. Additionally, Mongolian gerbils were gavaged with H. pylori, and later the gastric antrum tissues were collected. The GATA-3 and Cx32 mRNA and protein expression levels were detected by a reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. The scratch labeling fluorescent dye tracer (SLDT) technique was used to detect the gap junctional intercellular communication (GJIC) function. GATA-3 small interfering RNA (siRNA) was transfected into BGC823 cells and its effect on Cx32 expression levels was detected. The impact of GATA-3 on Cx32 promoter transcriptional activity was detected using a dual luciferase reporter assay. The results revealed that H. pylori infection increased GATA-3 expression and decreased Cx32 expression in GES-1 cells and in animal gastric tissues compared with their respective controls, whilst in BGC823 cells, GATA-3 siRNA increased Cx32 expression compared with the control. In the SLDT experiment of GES-1 cells with H. pylori infection, the fluorescent dye was primarily limited to a single cell row close to the scratch, and only a limited amount of dye passing to the second cell row, indicating that the GJIC function was substantially reduced or absent compared with the control group, where the fluorescence dye transferred to the neighboring cells of 3–4 rows, indicating a stronger GJIC function comparatively. GATA-3 inhibited the expression of the luciferase reporter gene, compared with the controls, suggesting that GATA-3 inhibited the expression of Cx32 by binding to Cx32 promoter sites. These results indicated that H. pylori-increased GATA-3 expression, which downregulated Cx32 expression, may serve an important function in gastric carcinogenesis, and GATA-3 siRNA may serve a function in the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Lihua Huang
- Center for Medical Experiments, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yinjie Guo
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Dan Cao
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaoming Liu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Linfang Zhang
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Tingzi Hu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yong Qi
- Clinical Laboratory, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Canxia Xu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
35
|
Wang MX, Ren JT, Tang LY, Ren ZF. Molecular features in young vs elderly breast cancer patients and the impacts on survival disparities by age at diagnosis. Cancer Med 2018; 7:3269-3277. [PMID: 29761914 PMCID: PMC6051220 DOI: 10.1002/cam4.1544] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/03/2018] [Accepted: 04/15/2018] [Indexed: 01/02/2023] Open
Abstract
Young and elderly breast cancer patients are more likely to have a poorer outcome than middle‐aged patients. The intrinsic molecular features for this disparity are unclear. We obtained data from the Cancer Genome Atlas (TCGA) on May 15, 2017 to test the potential mediation effects of the molecular features on the association between age and prognosis with a four‐step approach. The relative contributions of the molecular features (PAM50 subtype, risk stratification, DNAm age, and mutations in TP53,PIK3CA,MLL3,CDH1,GATA3, and MAP3K1) to age disparities in survival were estimated by Cox proportional hazard models with or without the features. Young patients were significantly more likely to have basal‐like subtype, GATA3 mutations, and younger DNA methylation (DNAm) age than middle‐aged patients (P < .05). Both the young and elderly patients had a significantly increased risk of breast cancer recurrence after adjusted by race, tumor size, and node status (Hazard ratio [HR] (95% confidence interval [CI]): 2.81 [1.44, 5.45], 2.37 [1.45, 3.89], respectively). This increased risk was weakened in the young patients after further adjustments in the molecular features, particularly basal‐like subtype, GATA3 mutations, and DNAm age (HR [95%CI]: 1.87 [0.81, 4.32]), resulting in 33.5% decreased risk of recurrence. Meanwhile, the adjustments of the molecular features did not alter the recurrence risk for the elderly patients. Compared with middle‐aged patients of breast cancer, poorer prognosis of elderly patients may be caused by aging, while poorer prognosis of young patients was probably mediated through intrinsic characteristics, such as basal‐like subtype, GATA3 mutations, and DNAm age of the cancerous tissues.
Collapse
Affiliation(s)
- Mei-Xia Wang
- The School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jun-Ting Ren
- The School of Public Health, Sun Yat-sen University, Guangzhou, China.,Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ze-Fang Ren
- The School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
Takaku M, Grimm SA, Roberts JD, Chrysovergis K, Bennett BD, Myers P, Perera L, Tucker CJ, Perou CM, Wade PA. GATA3 zinc finger 2 mutations reprogram the breast cancer transcriptional network. Nat Commun 2018. [PMID: 29535312 PMCID: PMC5849768 DOI: 10.1038/s41467-018-03478-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
GATA3 is frequently mutated in breast cancer; these mutations are widely presumed to be loss-of function despite a dearth of information regarding their effect on disease course or their mechanistic impact on the breast cancer transcriptional network. Here, we address molecular and clinical features associated with GATA3 mutations. A novel classification scheme defines distinct clinical features for patients bearing breast tumors with mutations in the second GATA3 zinc-finger (ZnFn2). An engineered ZnFn2 mutant cell line by CRISPR–Cas9 reveals that mutation of one allele of the GATA3 second zinc finger (ZnFn2) leads to loss of binding and decreased expression at a subset of genes, including Progesterone Receptor. At other loci, associated with epithelial to mesenchymal transition, gain of binding correlates with increased gene expression. These results demonstrate that not all GATA3 mutations are equivalent and that ZnFn2 mutations impact breast cancer through gain and loss-of function. In breast cancer GATA3 is known to be frequently mutated, but the function of these mutations is unclear. Here, the authors utilise CRISPR-Cas9 to model frame-shift mutations in zinc finger 2 of GATA3, highlighting that GATA3 mutation can have gain- or loss-of function effects in breast cancer.
Collapse
Affiliation(s)
- Motoki Takaku
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Sara A Grimm
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - John D Roberts
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Kaliopi Chrysovergis
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Brian D Bennett
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Page Myers
- Comparative Medicine Branch, National Institute of Environmental Health Sciences, Research Triangle Park, 27709, Durham, NC, USA
| | - Lalith Perera
- Laboratory of Genome Integrity and Structural Biology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Charles J Tucker
- Fluorescence Microscopy and Imaging Center, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center and Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Paul A Wade
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA.
| |
Collapse
|
37
|
Bianco TM, Abdalla DR, Desidério CS, Thys S, Simoens C, Bogers JP, Murta EFC, Michelin MA. The influence of physical activity in the anti-tumor immune response in experimental breast tumor. Immunol Lett 2017; 190:148-158. [PMID: 28818640 DOI: 10.1016/j.imlet.2017.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/03/2017] [Accepted: 08/09/2017] [Indexed: 12/31/2022]
Abstract
This study aimed to investigate the influence of physical activity in innate immunity to conduce to an effective antitumoral immune response analyzing the phenotype and activation status of infiltrating cells. We analysed the intracellular cytokines and the transcription factors of tumor infiltrating lymphocytes (TILS) and spleen leukocytes. The Nos2 gene expression was evaluated in spleen cells and futhermore the ROS production was measured and spleen cells; another cell evaluated was dendritic cells (TIDCs), their cytokines expression and membrane molecules; finally to understood the results obtained, we analysed the dendritic cells obtained from bone marrow. Were used female Balb/c mice divided into 4 groups: two controls without tumor, sedentary (GI) and trained (GII) and two groups with tumor, sedentary (GIII) or trained (GIV). The physical activity (PA) was realized acoording swimming protocol. Tumor was induced by injection of 4T1 cells. All experiments were performed in biological triplicate. After the experimental period, the tumor was removed and the cells were identified by flow cytometry with labeling to CD4, CD8, CD11c, CD11b, CD80, CD86 and Ia, and intracelular staining IL-10, IL-12, TNF-α, IFN-γ, IL-17, Tbet, GATA3, RORγt and FoxP3. The bone marrow of the animals was obtained to analyse the derivated DCs by flow cytometry and culture cells to obtain the supernatant to measure the cytokines. Our results demonstrated that the PA inhibit the tumoral growth although not to change the number of TILS, but reduced expression of GATA-3, ROR-γT, related with poor prognosis, and TNF-α intracellular; however occur one significantly reduction in TIDCS, but these cells expressed more co-stimulatory and presentation molecules. Furthermore, we observed that the induced PA stimulated the gene expression of Tbet and the production of inflammatory cytokines suggesting an increase of Th1 systemic response. The results evaluating the systemic influence in DCs showed that the PA improve significantly the number of those cells in bone marrow as well the number of co-stimulatory molecules. Therefore, we could conclude that PA influence the innate immunity by interfering to promote in process of maturation of DCs both in tumor and systemically, that by its turn promote a modification in acquired immune cells, representing by T helper to induce an important alteration transcription factors that are responsible to maintain a suppressive microenviroment, and thereby, allowing the latter cells can thus activate antitumor immune response. The PA was able improve the Th1 systemic response by enhance to Tbet gene expression, promote a slightly increased of Th1-type cytokines and decrease Gata3 and Foxp3 gene expression in which can inhibit the Th1 immune response.
Collapse
Affiliation(s)
- Thiago M Bianco
- Oncology Research Institute (Instituto de Pesquisa em Oncologia-IPON), Federal University of the Triângulo Mineiro (UFTM), Brazil
| | - Douglas R Abdalla
- Oncology Research Institute (Instituto de Pesquisa em Oncologia-IPON), Federal University of the Triângulo Mineiro (UFTM), Brazil
| | - Chamberttan S Desidério
- Oncology Research Institute (Instituto de Pesquisa em Oncologia-IPON), Federal University of the Triângulo Mineiro (UFTM), Brazil
| | - Sofie Thys
- Laboratory of Cell Biology and Histology, University of Antwerp, Belgium
| | - Cindy Simoens
- Laboratory of Cell Biology and Histology, University of Antwerp, Belgium
| | - John-Paul Bogers
- Laboratory of Cell Biology and Histology, University of Antwerp, Belgium
| | - Eddie F C Murta
- Oncology Research Institute (IPON)/Discipline of Gynecology and Obstetrics, UFTM, Brazil
| | - Márcia A Michelin
- Oncology Research Institute (IPON)/Discipline of Immunology, UFTM, Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
38
|
GATA3 interacts with and stabilizes HIF-1α to enhance cancer cell invasiveness. Oncogene 2017; 36:4243-4252. [PMID: 28263977 PMCID: PMC5537608 DOI: 10.1038/onc.2017.8] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/23/2016] [Accepted: 01/11/2017] [Indexed: 12/11/2022]
Abstract
GATA binding protein 3 (GATA3) is indispensable in development of human organs. However, the role of GATA3 in cancers remains elusive. Hypoxia inducible factor (HIF)-1 plays an important role in pathogenesis of human cancers. Regulation of HIF-1α degradation is orchestrated through collaboration of its interacting proteins. In this study, we discover that GATA3 is upregulated in head and neck squamous cell carcinoma (HNSCC) and is an independent predictor for poor disease-free survival. GATA3 promotes invasive behaviours of HNSCC and melanoma cells in vitro and in immunodeficient mice. Mechanistically, GATA3 physically associates with HIF-1α under hypoxia to inhibit ubiquitination and proteasomal degradation of HIF-1α, which is independent of HIF-1α prolyl hydroxylation. Chromatin immunoprecipitation assays show that the GATA3/HIF-1α complex binds to and regulates HIF-1 target genes, which is also supported by the microarray analysis. Notably, the GATA3-mediated invasiveness can be significantly reversed by HIF-1α knockdown, suggesting a critical role of HIF-1α in the underlying mechanism of GATA3-mediated effects. Our findings suggest that GATA3 stabilizes HIF-1α to enhance cancer invasiveness under hypoxia and support the GATA3/HIF-1α axis as a potential therapeutic target for cancer treatment.
Collapse
|
39
|
Identification of the Key Genes and Pathways in Esophageal Carcinoma. Gastroenterol Res Pract 2016; 2016:2968106. [PMID: 27818681 PMCID: PMC5080515 DOI: 10.1155/2016/2968106] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/01/2016] [Accepted: 07/11/2016] [Indexed: 12/17/2022] Open
Abstract
Objective. Esophageal carcinoma (EC) is a frequently common malignancy of gastrointestinal cancer in the world. This study aims to screen key genes and pathways in EC and elucidate the mechanism of it. Methods. 5 microarray datasets of EC were downloaded from Gene Expression Omnibus. Differentially expressed genes (DEGs) were screened by bioinformatics analysis. Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and protein-protein interaction (PPI) network construction were performed to obtain the biological roles of DEGs in EC. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to verify the expression level of DEGs in EC. Results. A total of 1955 genes were filtered as DEGs in EC. The upregulated genes were significantly enriched in cell cycle and the downregulated genes significantly enriched in Endocytosis. PPI network displayed CDK4 and CCT3 were hub proteins in the network. The expression level of 8 dysregulated DEGs including CDK4, CCT3, THSD4, SIM2, MYBL2, CENPF, CDCA3, and CDKN3 was validated in EC compared to adjacent nontumor tissues and the results were matched with the microarray analysis. Conclusion. The significantly DEGs including CDK4, CCT3, THSD4, and SIM2 may play key roles in tumorigenesis and development of EC involved in cell cycle and Endocytosis.
Collapse
|
40
|
Gain- and Loss-of-Function Mutations in the Breast Cancer Gene GATA3 Result in Differential Drug Sensitivity. PLoS Genet 2016; 12:e1006279. [PMID: 27588951 PMCID: PMC5010247 DOI: 10.1371/journal.pgen.1006279] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/04/2016] [Indexed: 01/02/2023] Open
Abstract
Patterns of somatic mutations in cancer genes provide information about their functional role in tumourigenesis, and thus indicate their potential for therapeutic exploitation. Yet, the classical distinction between oncogene and tumour suppressor may not always apply. For instance, TP53 has been simultaneously associated with tumour suppressing and promoting activities. Here, we uncover a similar phenomenon for GATA3, a frequently mutated, yet poorly understood, breast cancer gene. We identify two functional classes of frameshift mutations that are associated with distinct expression profiles in tumours, differential disease-free patient survival and gain- and loss-of-function activities in a cell line model. Furthermore, we find an estrogen receptor-independent synthetic lethal interaction between a GATA3 frameshift mutant with an extended C-terminus and the histone methyltransferases G9A and GLP, indicating perturbed epigenetic regulation. Our findings reveal important insights into mutant GATA3 function and breast cancer, provide the first potential therapeutic strategy and suggest that dual tumour suppressive and oncogenic activities are more widespread than previously appreciated. Cancer is a disease caused by genetic mutations. Mutation patterns are often indicative of a gene’s function as either tumour promoting or tumour suppressive. Here we describe the frequently mutated, but poorly studied, breast cancer gene GATA3 as a rare exception: We discover that two different functional classes of mutations in this gene can lead to either gain- or loss-of-function activities. The most common type of mutations, resulting in an unusually extended protein, is associated with differential gene expression and decreased disease-free survival. This mutant, in contrast to other mutations or the non-mutated protein, renders cells specifically vulnerable to inhibitors of two chromatin-modifying enzymes, the histone methyltransferases G9A and GLP. Our findings shed light on the functional consequences of frequent GATA3 mutations in breast cancer and represent a first lead toward personalised therapy for a large subgroup of breast cancer patients.
Collapse
|
41
|
Azim HA, Nguyen B, Brohée S, Zoppoli G, Sotiriou C. Genomic aberrations in young and elderly breast cancer patients. BMC Med 2015; 13:266. [PMID: 26467651 PMCID: PMC4606505 DOI: 10.1186/s12916-015-0504-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/01/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Age at breast cancer diagnosis is a known prognostic factor. Previously, several groups including ours have shown that young age at diagnosis is associated with higher prevalence of basal-like tumors and aggressive tumor phenotypes. Yet the impact of age at diagnosis on the genomic landscape of breast cancer remains unclear. In this study, we examined the pattern of somatic mutations, chromosomal copy number variations (CNVs) and transcriptomic profiles in young and elderly breast cancer patients. METHODS Analyses were performed on The Cancer Genome Atlas (TCGA) dataset. Patients with metastatic disease at diagnosis, classified as normal-like by PAM50 or had missing clinical information were excluded. Young patients were defined as ≤45 years of age, while elderly patients were those ≥70 years of age at breast cancer diagnosis. The remaining patients were classified as "intermediate". We evaluated the association between age at diagnosis and somatic mutations, CNV and gene expression in a logistic regression model adjusting for tumor size, nodal status, histology and breast cancer subtype. All analyses were corrected for multiple testing using the Benjamini-Hochberg approach. RESULTS In this study, 125, 486 and 169 patients were ≤45, 46-69 and ≥70 years of age, respectively. Older patients had more somatic mutations (n = 44 versus 35 versus 31; P = 0.0009) and more CNVs, especially in ductal tumors (P = 0.02). Eleven mutations were independently associated with age at diagnosis, of which only GATA3 was associated with young age (15.2% versus 8.2% versus 9%; P = 0.003). Only two CNV events were independently associated with age, with more chr18p losses in older patients and more chr6q27 deletions in younger ones. Younger age at diagnosis was associated with higher expression of gene signatures related to proliferation, stem cell features and endocrine resistance. CONCLUSIONS Age adds a layer of biological complexity beyond breast cancer molecular subtypes, classic pathological and clinical variables, worthy of further consideration in future drug development as we seek to refine therapeutic strategies in the era of personalized medicine.
Collapse
Affiliation(s)
- Hatem A Azim
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, 121, Brussels, Belgium.
| | - Bastien Nguyen
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, 121, Brussels, Belgium.
| | - Sylvain Brohée
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, 121, Brussels, Belgium.
| | - Gabriele Zoppoli
- Department of Internal Medicine, University of Genova and IRCCS AOU San Martino - IST, Genoa, Italy.
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo, 121, Brussels, Belgium.
| |
Collapse
|
42
|
Du F, Yuan P, Wang T, Zhao J, Zhao Z, Luo Y, Xu B. The Significance and Therapeutic Potential of GATA3 Expression and Mutation in Breast Cancer: A Systematic Review. Med Res Rev 2015; 35:1300-15. [PMID: 26313026 DOI: 10.1002/med.21362] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/12/2015] [Accepted: 07/23/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Feng Du
- Department of Medical Oncology, Cancer Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100021 China
| | - Peng Yuan
- Department of Medical Oncology, Cancer Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100021 China
| | - Teng Wang
- Tumor Marker Research Center, Cancer Institute and Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100191 China
| | - Jiuda Zhao
- Department of Medical Oncology, Cancer Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100021 China
| | - Zitong Zhao
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100191 China
| | - Yang Luo
- Department of Medical Oncology, Cancer Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100021 China
| | - Binghe Xu
- Department of Medical Oncology, Cancer Hospital; Chinese Academy of Medical Sciences, Peking Union Medical College; Beijing 100021 China
| |
Collapse
|