1
|
Kwok T, Yeguvapalli S, Chitrala KN. Identification of Genes Crucial for Biological Processes in Breast Cancer Liver Metastasis Relapse. Int J Mol Sci 2024; 25:5439. [PMID: 38791477 PMCID: PMC11122209 DOI: 10.3390/ijms25105439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer, when advancing to a metastatic stage, involves the liver, impacting over 50% of cases and significantly diminishing survival rates. Presently, a lack of tailored therapeutic protocols for breast cancer liver metastasis (BCLM) underscores the need for a deeper understanding of molecular patterns governing this complication. Therefore, by analyzing differentially expressed genes (DEGs) between primary breast tumors and BCLM lesions, we aimed to shed light on the diversities of this process. This research investigated breast cancer liver metastasis relapse by employing a comprehensive approach that integrated data filtering, gene ontology and KEGG pathway analysis, overall survival analysis, identification of the alteration in the DEGs, visualization of the protein-protein interaction network, Signor 2.0, identification of positively correlated genes, immune cell infiltration analysis, genetic alternation analysis, copy number variant analysis, gene-to-mRNA interaction, transcription factor analysis, molecular docking, and identification of potential treatment targets. This study's integrative approach unveiled metabolic reprogramming, suggesting altered PCK1 and LPL expression as key in breast cancer metastasis recurrence.
Collapse
|
2
|
Han W, Shi LZ, Zhang YW, Wang HN. Comment on "Prognostic and Clinical Significance of Human Leukocyte Antigen Class I Expression in Breast Cancer: A Meta‑Analysis". Mol Diagn Ther 2024; 28:237-238. [PMID: 38376655 DOI: 10.1007/s40291-024-00700-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2024] [Indexed: 02/21/2024]
Affiliation(s)
- Wei Han
- Department of General Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, 215300, Jiangsu, People's Republic of China
| | - Li-Zhou Shi
- Department of General Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, 215300, Jiangsu, People's Republic of China
| | - Yu-Wei Zhang
- Nursing Department, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, 215300, Jiangsu, People's Republic of China
| | - Hao-Nan Wang
- Oncology Department, Wuxi Fifth People's Hospital, Wuxi, 214000, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Ka NL, Park MK, Kim SS, Jeon Y, Hwang S, Kim SM, Lim GY, Lee H, Lee MO. NR1D1 Stimulates Antitumor Immune Responses in Breast Cancer by Activating cGAS-STING Signaling. Cancer Res 2023; 83:3045-3058. [PMID: 37395684 PMCID: PMC10538367 DOI: 10.1158/0008-5472.can-23-0329] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/13/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023]
Abstract
Potentiating antitumor immunity is a promising therapeutic approach for treating a variety of cancers, including breast cancer. One potential strategy to promote antitumor immunity is targeting DNA damage response. Given that the nuclear receptor NR1D1 (also known as REV-ERBα) inhibits DNA repair in breast cancer cells, we explored the role of NR1D1 in antitumor CD8+ T-cell responses. First, deletion of Nr1d1 in MMTV-PyMT transgenic mice resulted in increased tumor growth and lung metastasis. Orthotopic allograft experiments suggested that loss of Nr1d1 in tumor cells rather than in stromal cells played a prominent role in increasing tumor progression. Comprehensive transcriptome analyses revealed that biological processes including type I IFN signaling and T cell-mediated immune responses were associated with NR1D1. Indeed, the expression of type I IFNs and infiltration of CD8+ T cells and natural killer cells in tumors were suppressed in Nr1d1-/-;MMTV-PyMT mice. Mechanistically, NR1D1 promoted DNA damage-induced accumulation of cytosolic DNA fragments and activated cGAS-STING signaling, which increased the production of type I IFNs and downstream chemokines CCL5 and CXCL10. Pharmacologic activation of NR1D1 by its ligand, SR9009, enhanced type I IFN-mediated antitumor immunity accompanied by the suppression of tumor progression and lung metastasis. Taken together, these findings reveal the critical role of NR1D1 in enhancing antitumor CD8+ T-cell responses, suggesting that NR1D1 may be a good therapeutic target for breast cancer. SIGNIFICANCE NR1D1 suppresses breast cancer progression and lung metastasis by enhancing antitumor immunity via cGAS-STING pathway activation, which provides potential immunotherapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Na-Lee Ka
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Mi Kyung Park
- Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi, Republic of Korea
| | - Seung-Su Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yoon Jeon
- Research Institute, National Cancer Center, Gyeonggi, Republic of Korea
| | - Sewon Hwang
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sun Mi Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi, Republic of Korea
| | - Ga Young Lim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Bio-MAX institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Naik A, Thomas R, Al-Khadairi G, Bacha R, Hendrickx W, Decock J. Cancer testis antigen PRAME: An anti-cancer target with immunomodulatory potential. J Cell Mol Med 2021; 25:10376-10388. [PMID: 34612587 PMCID: PMC8581324 DOI: 10.1111/jcmm.16967] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/29/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022] Open
Abstract
PReferentially expressed Antigen in Melanoma (PRAME) is a cancer testis antigen with restricted expression in somatic tissues and re‐expression in poor prognostic solid tumours. PRAME has been extensively investigated as a target for immunotherapy, however, its role in modulating the anti‐tumour immune response remains largely unknown. Here, we show that PRAME tumour expression is associated with worse survival in the TCGA breast cancer cohort, particularly in immune‐unfavourable tumours. Using direct and indirect co‐culture models, we found that PRAME overexpressing MDA‐MB‐468 breast cancer cells inhibit T cell activation and cytolytic potential, which could be partly restored by silencing of PRAME. Furthermore, silencing of PRAME reduced expression of several immune checkpoints and their ligands, including PD‐1, LAG3, PD‐L1, CD86, Gal‐9 and VISTA. Interestingly, silencing of PRAME induced cancer cell killing to levels similar to anti‐PD‐L1 atezolizumab treatment. Comprehensive analysis of soluble inflammatory mediators and cancer cell expression of immune‐related genes showed that PRAME tumour expression can suppress the expression and secretion of multiple pro‐inflammatory cytokines, and mediators of T cell activation, differentiation and cytolysis. Together, our data indicate that targeting of PRAME offers a potential, novel dual therapeutic approach to specifically target tumour cells and regulate immune activation in the tumour microenvironment.
Collapse
Affiliation(s)
- Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Remy Thomas
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ghaneya Al-Khadairi
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Rim Bacha
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Wouter Hendrickx
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,Cancer Department, Research Branch, Sidra Medicine, Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
5
|
Significant Diagnostic and Prognostic Value of FLAD1 and Related MicroRNAs in Breast Cancer after a Pan-Cancer Analysis. DISEASE MARKERS 2021; 2021:6962526. [PMID: 34336008 PMCID: PMC8321750 DOI: 10.1155/2021/6962526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/30/2021] [Accepted: 06/24/2021] [Indexed: 12/09/2022]
Abstract
The identification of biomarkers plays an important role in the diagnosis and prognosis of cancers. In this study, we explored the diagnostic and prognostic value of the FLAD1 expression across pan-cancer analysis from online databases (Oncomine, cBioPortal, Breast Cancer Gene-Expression Miner, UALCAN, GEO, BCIP, TNMplot, ENCORI, Kaplan-Meier Plotter, and LinkedOmics). We found that FLAD1 was overexpressed in a number of different kinds of cancers, especially in breast cancer, and higher FLAD1 expression level was associated with the HER+, p53 mutant, node-involved, NPI stage 3, basal-like, and triple-negative groups compared with the other subgroups of breast cancer. The FLAD1 expression levels were higher in patients that were 21–40 years old than those in patients of other ages and were higher in the African-American group than in the Caucasian group. We also analyzed the FLAD1-related microRNAs and their prognostic values in breast cancer. This study highlights the significance of FLAD1 in cancers and provides evidence for its potential as a biomarker for the diagnosis and prognosis of cancers.
Collapse
|
6
|
Naik A, Decock J. Targeting of lactate dehydrogenase C dysregulates the cell cycle and sensitizes breast cancer cells to DNA damage response targeted therapy. Mol Oncol 2021; 16:885-903. [PMID: 34050611 PMCID: PMC8847988 DOI: 10.1002/1878-0261.13024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/21/2021] [Accepted: 05/27/2021] [Indexed: 12/31/2022] Open
Abstract
The cancer testis antigen (CTA) lactate dehydrogenase C (LDHC) is a promising anticancer target with tumor-specific expression and immunogenicity. Interrogation of breast cancer patient cohorts from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) indicate that upregulation of LDHC expression correlates with unfavorable prognosis. Although the role of LDHC is well characterized in spermatocytes, its role in tumors remains largely unknown. We investigated whether LDHC is involved in regulating genomic stability and whether it could be targeted to affect tumor cellular fitness. Silencing LDHC in four breast cancer cell lines significantly increased the presence of giant cells, nuclear aberrations, DNA damage, and apoptosis. LDHC-silenced cells demonstrated aberrant cell cycle progression with differential expression of cell cycle checkpoint and DNA damage response regulators. In addition, LDHC silencing-induced microtubule destabilization, culminating in increased mitotic catastrophe and reduced long-term survival. Notably, the clonogenicity of LDHC-silenced cells was further reduced by treatment with the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib and with the DNA-damaging drug cisplatin. This study supports the therapeutic potential of targeting LDHC to mitigate cancer cell survival and improve sensitivity to agents that cause DNA damage or inhibit its repair.
Collapse
Affiliation(s)
- Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
7
|
Davison C, Morelli R, Knowlson C, McKechnie M, Carson R, Stachtea X, McLaughlin KA, Prise VE, Savage K, Wilson RH, Mulligan KA, Wilson PM, Ladner RD, LaBonte MJ. Targeting nucleotide metabolism enhances the efficacy of anthracyclines and anti-metabolites in triple-negative breast cancer. NPJ Breast Cancer 2021; 7:38. [PMID: 33824328 PMCID: PMC8024381 DOI: 10.1038/s41523-021-00245-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) remains the most lethal breast cancer subtype with poor response rates to the current chemotherapies and a lack of additional effective treatment options. We have identified deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase) as a critical gatekeeper that protects tumour DNA from the genotoxic misincorporation of uracil during treatment with standard chemotherapeutic agents commonly used in the FEC regimen. dUTPase catalyses the hydrolytic dephosphorylation of deoxyuridine triphosphate (dUTP) to deoxyuridine monophosphate (dUMP), providing dUMP for thymidylate synthase as part of the thymidylate biosynthesis pathway and maintaining low intracellular dUTP concentrations. This is crucial as DNA polymerase cannot distinguish between dUTP and deoxythymidylate triphosphate (dTTP), leading to dUTP misincorporation into DNA. Targeting dUTPase and inducing uracil misincorporation during the repair of DNA damage induced by fluoropyrimidines or anthracyclines represents an effective strategy to induce cell lethality. dUTPase inhibition significantly sensitised TNBC cell lines to fluoropyrimidines and anthracyclines through imbalanced nucleotide pools and increased DNA damage leading to decreased proliferation and increased cell death. These results suggest that repair of treatment-mediated DNA damage requires dUTPase to prevent uracil misincorporation and that inhibition of dUTPase is a promising strategy to enhance the efficacy of TNBC chemotherapy.
Collapse
Affiliation(s)
- Craig Davison
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Roisin Morelli
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Catherine Knowlson
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Melanie McKechnie
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Robbie Carson
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Xanthi Stachtea
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | | | | | - Kienan Savage
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Richard H Wilson
- Translational Research Centre, University of Glasgow, Glasgow, UK
| | | | | | - Robert D Ladner
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Melissa J LaBonte
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
- Medicine, Dentistry and Biomedical Sciences: Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
8
|
Song S, Tian B, Zhang M, Gao X, Jie L, Liu P, Li J. Diagnostic and prognostic value of thymidylate synthase expression in breast cancer. Clin Exp Pharmacol Physiol 2021; 48:279-287. [PMID: 33030246 PMCID: PMC7820961 DOI: 10.1111/1440-1681.13415] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 09/06/2020] [Accepted: 10/01/2020] [Indexed: 01/27/2023]
Abstract
Nucleotide metabolism is the driving force of cell proliferation, and thymidylate synthase (TYMS) catalyzes a rate-limiting step in the initial synthesis of nucleotides. Previous studies reported that TYMS activity significantly affected the proliferation of tumour cells. However, the diagnostic and prognostic significance of TYMS expression in breast cancer remains unclear. Here, we used the Breast Cancer Integrative Platform (BCIP) to investigate the relationship between progression and prognosis of breast cancer with TYMS expression, and then verified the database analysis using immunohistochemical staining. Our results indicated TYMS expression was greater in breast cancer than adjacent normal tissues and greater in triple-negative breast cancer (TNBC) than non-TNBC tissues. TYMS expression also had significant positive correlations with histological grade, tumour size, and ER negativity, and PR negativity. The increased copy number of the TYMS gene appears to be the reason for its upregulation in breast cancer. Breast cancer patients with higher TYMS expression had poorer prognosis. Our data suggest that TYMS has potential use as a diagnostic and prognostic marker for breast cancer patients.
Collapse
Affiliation(s)
- Shaoran Song
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Bixia Tian
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Miao Zhang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xiaoqian Gao
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Liu Jie
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Peijun Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Juan Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
9
|
Nguyen QH, Le DH. Improving existing analysis pipeline to identify and analyze cancer driver genes using multi-omics data. Sci Rep 2020; 10:20521. [PMID: 33239644 PMCID: PMC7688645 DOI: 10.1038/s41598-020-77318-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
The cumulative of genes carrying mutations is vital for the establishment and development of cancer. However, this driver gene exploring research line has selected and used types of tools and models of analysis unsystematically and discretely. Also, the previous studies may have neglected low-frequency drivers and seldom predicted subgroup specificities of identified driver genes. In this study, we presented an improved driver gene identification and analysis pipeline that comprises the four most widely focused analyses for driver genes: enrichment analysis, clinical feature association with expression profiles of identified driver genes as well as with their functional modules, and patient stratification by existing advanced computational tools integrating multi-omics data. The improved pipeline's general usability was demonstrated straightforwardly for breast cancer, validated by some independent databases. Accordingly, 31 validated driver genes, including four novel ones, were discovered. Subsequently, we detected cancer-related significantly enriched gene ontology terms and pathways, probable drug targets, two co-expressed modules associated significantly with several clinical features, such as number of positive lymph nodes, Nottingham prognostic index, and tumor stage, and two biologically distinct groups of BRCA patients. Data and source code of the case study can be downloaded at https://github.com/hauldhut/drivergene.
Collapse
Affiliation(s)
- Quang-Huy Nguyen
- Department of Computational Biomedicine, Vingroup Big Data Institute, Hanoi, Vietnam.,Faculty of Pharmacy, Dainam University, Hanoi, Vietnam
| | - Duc-Hau Le
- Department of Computational Biomedicine, Vingroup Big Data Institute, Hanoi, Vietnam. .,College of Engineering and Computer Science, VinUniversity, Hanoi, Vietnam.
| |
Collapse
|
10
|
Sudhakaran M, Parra MR, Stoub H, Gallo KA, Doseff AI. Apigenin by targeting hnRNPA2 sensitizes triple-negative breast cancer spheroids to doxorubicin-induced apoptosis and regulates expression of ABCC4 and ABCG2 drug efflux transporters. Biochem Pharmacol 2020; 182:114259. [PMID: 33011162 DOI: 10.1016/j.bcp.2020.114259] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 01/14/2023]
Abstract
Acquired resistance to doxorubicin is a major hurdle in triple-negative breast cancer (TNBC) therapy, emphasizing the need to identify improved strategies. Apigenin and other structurally related dietary flavones are emerging as potential chemo-sensitizers, but their effect on three-dimensional TNBC spheroid models has not been investigated. We previously showed that apigenin associates with heterogeneous ribonuclear protein A2/B1 (hnRNPA2), an RNA-binding protein involved in mRNA and co-transcriptional regulation. However, the role of hnRNPA2 in apigenin chemo-sensitizing activity has not been investigated. Here, we show that apigenin induced apoptosis in TNBC spheroids more effectively than apigenin-glycoside, owing to higher cellular uptake. Moreover, apigenin inhibited the growth of TNBC patient-derived organoids at an in vivo achievable concentration. Apigenin sensitized spheroids to doxorubicin-induced DNA damage, triggering caspase-9-mediated intrinsic apoptotic pathway and caspase-3 activity. Silencing of hnRNPA2 decreased apigenin-induced sensitization to doxorubicin in spheroids by diminishing apoptosis and partly abrogated apigenin-mediated reduction of ABCC4 and ABCG2 efflux transporters. Together these findings provide novel insights into the critical role of hnRNPA2 in mediating apigenin-induced sensitization of TNBC spheroids to doxorubicin by increasing the expression of efflux transporters and apoptosis, underscoring the relevance of using dietary compounds as a chemotherapeutic adjuvant.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 2/biosynthesis
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/metabolism
- Apigenin/administration & dosage
- Apigenin/metabolism
- Apoptosis/drug effects
- Apoptosis/physiology
- Cell Survival/drug effects
- Cell Survival/physiology
- Dose-Response Relationship, Drug
- Doxorubicin/administration & dosage
- Doxorubicin/metabolism
- Drug Delivery Systems/methods
- Female
- Gene Expression Regulation, Neoplastic
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/deficiency
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics
- Humans
- Mice
- Multidrug Resistance-Associated Proteins/biosynthesis
- Multidrug Resistance-Associated Proteins/genetics
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, United States
| | - Michael Ramirez Parra
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States
| | - Hayden Stoub
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, United States
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States.
| | - Andrea I Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
11
|
Liu Y, Li H, Liu F, Gao LB, Han R, Chen C, Ding X, Li S, Lu K, Yang L, Tian HM, Chen BB, Li X, Xu DH, Deng XL, Shi SL. Heterogeneous nuclear ribonucleoprotein A2/B1 is a negative regulator of human breast cancer metastasis by maintaining the balance of multiple genes and pathways. EBioMedicine 2020; 51:102583. [PMID: 31901866 PMCID: PMC6948170 DOI: 10.1016/j.ebiom.2019.11.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Background Heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 is an important RNA-binding protein that affects the RNA processing, splicing, transport and stability of many genes. hnRNPA2/B1 is expressed during proliferation and metastasis of various cancer types and promotes such processes. However, the precise role and mechanism of hnRNPA2/B1 in breast cancer remain unclear. Methods The association of hnRNPA2/B1 with breast cancer metastasis was assessed using tissue chips, mouse models and publicly available data. The role and mechanism of hnRNPA2/B1 in breast cancer metastasis were studied in cell lines and mouse models. Findings In contrast to other cancer research findings, hnRNPA2/B1 expression was negatively correlated with breast cancer metastasis. hnRNPA2/B1 inhibited MDA-MB-231 triple-negative breast cancer (TNBC) cell metastasis in vitro and in vivo. hnRNPA2/B1 knockout activated ERK-MAPK/Twist and GR-beta/TCF4 pathways but inhibited STAT3 and WNT/TCF4 signalling pathways. Profilin 2 (PFN2) promoted breast cancer cell migration and invasion, whereas hnRNPA2/B1 bound directly to the UAGGG locus in the 3′-untranslated region of PFN2 mRNA and reduced the stability of PFN2 mRNA. Interpretation Our data supported the role of hnRNPA2/B1 in tumour metastasis risk and survival prediction in patients with breast cancer. The inhibitory role of hnRNPA2/B1 in metastasis was a balance of downstream multiple genes and signalling pathways. PFN2 downregulation by hnRNPA2/B1 might partly explain the inhibitory mechanism of hnRNPA2/B1 in breast cancer metastasis. Therefore, hnRNPA2/B1 might be used as a new prognostic biomarker and valuable molecular target for breast cancer treatments.
Collapse
Affiliation(s)
- Yu Liu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China; School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai 200240, PR China
| | - Huan Li
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Fan Liu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Li-Bin Gao
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Rong Han
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Chen Chen
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Xue Ding
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Shuang Li
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Kun Lu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Ling Yang
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Hui-Min Tian
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Bin-Bin Chen
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Xiao Li
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Dong-Hui Xu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Xiao-Ling Deng
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Song-Lin Shi
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China.
| |
Collapse
|
12
|
Li P, Zhou C, Yan Y, Li J, Liu J, Zhang Y, Liu P. Crumbs protein homolog 3 (CRB3) expression is associated with oestrogen and progesterone receptor positivity in breast cancer. Clin Exp Pharmacol Physiol 2019; 46:837-844. [PMID: 31087799 PMCID: PMC6772053 DOI: 10.1111/1440-1681.13104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/11/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022]
Abstract
The crumbs protein homolog 3 (CRB3) regulates the tight junction to help maintain epithelial polarity. Altered CRB3 expression was associated with carcinogenesis of epithelial cells. This study detected CRB3 expression in 192 cases of breast cancer tissues and in the Molecular Taxonomy of Breast Cancer International Consortium (Metabric) and The Cancer Genome Atlas (TCGA) datasets for association with triple negative breast cancer (TNBC) phenotypes. The in vitro experiments confirm the ex vivo data. The data showed that levels of both CRB3 mRNA and protein were associated with TNBC phenotypes, ie, 41.1% (39/95) of ER+ breast cancer was CRB3-positive, whereas 26.9% (25/93) ER- tumour was CRB3-positive (P = 0.046). Moreover, 47.6% (30/63) of PR+ breast cancer was CRB3-positive vs 28.4% (33/116) PR- tumours positive for CRB3 (P = 0.013). In addition, 40.1% (27/66) of ER+/PR+ tumour was CRB3-positive, but only 22.4% (19/85) of TNBC showed CRB3 expression (P = 0.048). Indeed, levels of CRB3 mRNA were higher in non-TNBC than TNBC in both Metabric (P = 3.682e-10) and TCGA datasets (P = 2.501e-07). The in vitro data showed that CRB3 expression was higher in luminal (MCF7 and T47D) than in HER2 (MDA-MB-453 and SK-BR-3) and basal (MDA-MB-231 and BT-549) breast cancer cell lines. More interestingly, ERα regulated expression of CRB3 protein in MCF7 and BT-549 cells and ERα expression was associated with CRB3 expression in breast cancer tissues specimens. This study demonstrated that ERα could be a novel regulator for CRB3 expression in breast cancer.
Collapse
Affiliation(s)
- Pingping Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Can Zhou
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yu Yan
- Department of Breast SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Juan Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jie Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yan Zhang
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Peijun Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
13
|
Loss of PICH promotes chromosome instability and cell death in triple-negative breast cancer. Cell Death Dis 2019; 10:428. [PMID: 31160555 PMCID: PMC6547724 DOI: 10.1038/s41419-019-1662-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/24/2019] [Accepted: 05/13/2019] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC), defined by the lack of expression of estrogen, progesterone, and ERBB2 receptors, has the worst prognosis of all breast cancers. It is difficult to treat owing to a lack of effective molecular targets. Here, we report that the growth of TNBC cells is exceptionally dependent on PICH, a DNA-dependent ATPase. Clinical samples analysis showed that PICH is highly expressed in TNBC compared to other breast cancer subtypes. Importantly, its high expression correlates with higher risk of distal metastasis and worse clinical outcomes. Further analysis revealed that PICH depletion selectively impairs the proliferation of TNBC cells, but not that of luminal breast cancer cells, in vitro and in vivo. In addition, knockdown of PICH in TNBC cells induces the formation of chromatin bridges and lagging chromosomes in anaphase, frequently resulting in micronucleation or binucleation, finally leading to mitotic catastrophe and apoptosis. Collectively, our findings show the dependency of TNBC cells on PICH for faithful chromosome segregation and the clinical potential of PICH inhibition to improve treatment of patients with high-risk TNBC.
Collapse
|
14
|
Lee J, Yesilkanal AE, Wynne JP, Frankenberger C, Liu J, Yan J, Elbaz M, Rabe DC, Rustandy FD, Tiwari P, Grossman EA, Hart PC, Kang C, Sanderson SM, Andrade J, Nomura DK, Bonini MG, Locasale JW, Rosner MR. Effective breast cancer combination therapy targeting BACH1 and mitochondrial metabolism. Nature 2019; 568:254-258. [PMID: 30842661 PMCID: PMC6698916 DOI: 10.1038/s41586-019-1005-x] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 02/06/2019] [Indexed: 12/20/2022]
Abstract
Mitochondrial metabolism is an attractive target for cancer therapy1,2. Reprogramming metabolic pathways could improve the ability of metabolic inhibitors to suppress cancers with limited treatment options, such as triple-negative breast cancer (TNBC)1,3. Here we show that BTB and CNC homology1 (BACH1)4, a haem-binding transcription factor that is increased in expression in tumours from patients with TNBC, targets mitochondrial metabolism. BACH1 decreases glucose utilization in the tricarboxylic acid cycle and negatively regulates transcription of electron transport chain (ETC) genes. BACH1 depletion by shRNA or degradation by hemin sensitizes cells to ETC inhibitors such as metformin5,6, suppressing growth of both cell line and patient-derived tumour xenografts. Expression of a haem-resistant BACH1 mutant in cells that express a short hairpin RNA for BACH1 rescues the BACH1 phenotype and restores metformin resistance in hemin-treated cells and tumours7. Finally, BACH1 gene expression inversely correlates with ETC gene expression in tumours from patients with breast cancer and in other tumour types, which highlights the clinical relevance of our findings. This study demonstrates that mitochondrial metabolism can be exploited by targeting BACH1 to sensitize breast cancer and potentially other tumour tissues to mitochondrial inhibitors.
Collapse
Affiliation(s)
- Jiyoung Lee
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Ali E Yesilkanal
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Joseph P Wynne
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Casey Frankenberger
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Juan Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Jielin Yan
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Mohamad Elbaz
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Daniel C Rabe
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Felicia D Rustandy
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Payal Tiwari
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Elizabeth A Grossman
- Department of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA.,Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Peter C Hart
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Christie Kang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Sydney M Sanderson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Jorge Andrade
- Center for Research Informatics, University of Chicago, Chicago, IL, USA
| | - Daniel K Nomura
- Department of Chemistry, Molecular and Cell Biology, and Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA.,Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA.,Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA, USA
| | - Marcelo G Bonini
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
15
|
Kim HY, Choi HJ, Lee JY, Kong G. Cancer Target Gene Screening: a web application for breast cancer target gene screening using multi-omics data analysis. Brief Bioinform 2019; 21:663-675. [DOI: 10.1093/bib/bbz003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 12/22/2022] Open
Abstract
Abstract
Breast cancer comprises several molecular subtypes with distinct clinical features and treatment responses, and a substantial portion of each subtype remains incurable. A comprehensive analysis of multi-omics data and clinical profiles is required in order to better understand the biological complexity of this cancer type and to identify new prognostic and therapeutic markers. Thus, there arises a need for useful analytical tools to assist in the investigation and clinical management of the disease. We developed Cancer Target Gene Screening (CTGS), a web application that provides rapid and user-friendly analysis of multi-omics data sets from a large number of primary breast tumors. It allows the investigation of genomic and epigenomic aberrations, evaluation of transcriptomic profiles and performance of survival analyses and of bivariate correlations between layers of omics data. Notably, the genome-wide screening function of CTGS prioritizes candidate genes of clinical and biological significance among genes with copy number alteration, DNA methylation and dysregulated expression by the integrative analysis of different types of omics data in customized subgroups of breast cancer patients. These features may help in the identification of druggable cancer driver genes in a specific subtype or the clinical condition of human breast cancer. CTGS is available at http://ctgs.biohackers.net.
Collapse
Affiliation(s)
- Hyung-Yong Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hee-Joo Choi
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
| | - Jeong-Yeon Lee
- Department of Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Gu Kong
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|