1
|
Sommariva M, Dolci M, Triulzi T, Ambrogi F, Dugo M, De Cecco L, Le Noci V, Bernardo G, Anselmi M, Montanari E, Pupa SM, Signorini L, Gagliano N, Sfondrini L, Delbue S, Tagliabue E. Impact of in vitro SARS-CoV-2 infection on breast cancer cells. Sci Rep 2024; 14:13134. [PMID: 38849411 PMCID: PMC11161491 DOI: 10.1038/s41598-024-63804-3] [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: 07/23/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
The pandemic of coronavirus disease 19 (COVID-19), caused by severe respiratory syndrome coronavirus 2 (SARS-CoV-2), had severe repercussions for breast cancer patients. Increasing evidence indicates that SARS-CoV-2 infection may directly impact breast cancer biology, but the effects of SARS-CoV-2 on breast tumor cells are still unknown. Here, we analyzed the molecular events occurring in the MCF7, MDA-MB-231 and HCC1937 breast cancer cell lines, representative of the luminal A, basal B/claudin-low and basal A subtypes, respectively, upon SARS-CoV-2 infection. Viral replication was monitored over time, and gene expression profiling was conducted. We found that MCF7 cells were the most permissive to viral replication. Treatment of MCF7 cells with Tamoxifen reduced the SARS-CoV-2 replication rate, suggesting an involvement of the estrogen receptor in sustaining virus replication in malignant cells. Interestingly, a metagene signature based on genes upregulated by SARS-CoV-2 infection in all three cell lines distinguished a subgroup of premenopausal luminal A breast cancer patients with a poor prognosis. As SARS-CoV-2 still spreads among the population, it is essential to understand the impact of SARS-CoV-2 infection on breast cancer, particularly in premenopausal patients diagnosed with the luminal A subtype, and to assess the long-term impact of COVID-19 on breast cancer outcomes.
Collapse
Affiliation(s)
- Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy.
| | - Maria Dolci
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, 20133, Milan, Italy
| | - Tiziana Triulzi
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Federico Ambrogi
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Via Celoria 22, 20133, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Loris De Cecco
- Integrated Biology of Rare Tumors, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Martina Anselmi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Elena Montanari
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Serenella M Pupa
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Lucia Signorini
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, 20133, Milan, Italy
| | - Nicoletta Gagliano
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Serena Delbue
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, 20133, Milan, Italy
| | - Elda Tagliabue
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| |
Collapse
|
2
|
Vathiotis IA, Trontzas I, Gavrielatou N, Gomatou G, Syrigos NK, Kotteas EA. Immune Checkpoint Blockade in Hormone Receptor-Positive Breast Cancer: Resistance Mechanisms and Future Perspectives. Clin Breast Cancer 2022; 22:642-649. [PMID: 35906130 DOI: 10.1016/j.clbc.2022.06.004] [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: 11/17/2021] [Revised: 05/21/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Anti-programmed cell death protein 1 immunotherapy has been incorporated in the treatment algorithm of triple-negative breast cancer (TNBC). However, clinical trial results for patients with hormone receptor (HR)-positive disease appear less compelling. HR-positive tumors exhibit lower levels of programmed death-ligand 1 expression in comparison with their triple-negative counterparts. Moreover, signaling through estrogen receptor alters the immune microenvironment, rendering such tumors immunologically "cold." To explain differential responses to immune checkpoint blockade, this review interrogates differences between HR-positive and TNBC. Starting from distinct genomic features, we further present disparities concerning the tumor microenvironment and finally, we summarize early-phase clinical trial results on promising novel immunotherapy combinations.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece; Department of Pathology, Yale University School of Medicine, New Haven, CT.
| | - Ioannis Trontzas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Niki Gavrielatou
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Georgia Gomatou
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Nikolaos K Syrigos
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Elias A Kotteas
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| |
Collapse
|
3
|
Tong D. Selective estrogen receptor modulators contribute to prostate cancer treatment by regulating the tumor immune microenvironment. J Immunother Cancer 2022; 10:jitc-2021-002944. [PMID: 35383112 PMCID: PMC8984050 DOI: 10.1136/jitc-2021-002944] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2022] [Indexed: 11/19/2022] Open
Abstract
Prostate cancer (PC) has previously been established as a cold tumor and develops in an inert immunosuppressive environment. Current research focuses on altering the immune microenvironment of PC from cold to hot; thus, in the present review, the diverse roles of estrogen and estrogen receptor (ER) signaling was examined in the tumor cell and tumor immune microenvironment (TIM). We hypothesized that ERα promotes PC progression and ERβ impedes epithelial-mesenchymal transition in PC cells, while in the TIM, ERβ mediates the immunosuppressive environment, and low levels of ERα is associated with disease development. Selective estrogen receptor modulators (SERMs) or selective ER degraders play diverse roles in the regulation of ER isoforms. Patients with PC may benefit from the use of SERMs, including raloxifene, in combination with anti-PD1/PD-L1 checkpoint immunotherapy, or TGF-β or Wnt antagonists. The present review demonstrated that immunotherapy-based strategies combined with SERMs may be an option for the future of PC-targeting therapy.
Collapse
Affiliation(s)
- Dali Tong
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Tamura Y, Tsutsumi S, Miyazono K, Koinuma D. PolyI:C attenuates transforming growth factor-β signaling to induce cytostasis of surrounding cells by secreted factors in triple-negative breast cancer. Cancer Sci 2021; 113:940-949. [PMID: 34897916 PMCID: PMC8898727 DOI: 10.1111/cas.15241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022] Open
Abstract
The activation of RIG‐I‐like receptor (RLR) signaling in cancer cells is widely recognized as a critical cancer therapy method. The expected mechanism of RLR ligand‐mediated cancer therapy involves the promotion of cancer cell death and strong induction of interferon (IFN)‐β that affects the tumor microenvironment. We have recently shown that activation of RLR signaling in triple‐negative breast cancer cells (TNBC) attenuates transforming growth factor‐β (TGF‐β) signaling, which partly contributes to the promotion of cancer cell pyroptosis. However, the consequences of suppression of TGF‐β signaling by RLR ligands with respect to IFN‐β‐mediated tumor suppression are not well characterized. This study showed that transfection of a typical RLR ligand polyI:C in cancer cells produces significant levels of IFN‐β, which inhibits the growth of the surrounding cancer cells. In addition, IFN‐β‐induced cell cycle arrest in surrounding cancer cells was inhibited by the expression of constitutively active Smad3. Constitutively active Smad3 suppresses IFN‐β expression through the alleviation of IFN regulatory factor 3 binding to the canonical target genes, as suggested by ChIP sequencing analysis. Based on these findings, a new facet of the protumorigenic function of TGF‐β that suppresses IFN‐β expression is suggested when RLR‐mediated cancer treatment is used in TNBC.
Collapse
Affiliation(s)
- Yusuke Tamura
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuichi Tsutsumi
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Wang K, Chen Y, Gao S, Wang M, Ge M, Yang Q, Liao M, Xu L, Chen J, Zeng Z, Chen H, Zhang XK, Lin T, Zhou H. Norlichexanthone purified from plant endophyte prevents postmenopausal osteoporosis by targeting ER α to inhibit RANKL signaling. Acta Pharm Sin B 2021; 11:442-455. [PMID: 33643823 PMCID: PMC7893202 DOI: 10.1016/j.apsb.2020.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/28/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
Although different types of drugs are available for postmenopausal osteoporosis, the limitations of the current therapies including drug resistances and adverse effects require identification of novel anti-osteoporosis agents. Here, we defined that norlichexanthone (NOR), a natural product, is a ligand of estrogen receptor-alpha (ERα) and revealed its therapeutic potential for postmenopausal osteoporosis. We used mammalian-one hybrid assay to screen for ERα modulators from crude extracts of several plant endophytes. As a result, NOR purified from the extract of endophyte ARL-13 was identified as a selective ERα modulator. NOR directly bound to ERα with an affinity in nanomolar range, revealing that it is a natural ligand of ERα. NOR induced osteoblast formation in MC3T3-E1 precursor cells. Conversely, NOR inhibited receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclast formation in both RAW264.7 macrophages and mouse primary monocytes. Mechanistically, NOR inhibited RANKL-induced association of ERα and TRAF6 to prevent ERα-mediated TRAF6 activation via Lys63-linked ubiquitination. Importantly, NOR exhibited potent anti-osteoporosis efficacy in an ovariectomized mouse model. Comparing to estrogen, NOR was of much less capability in stimulating endometrial hyperplasia and promoting mammalian cancer cell proliferation. Taken together, our study identified NOR as a natural and high affinity ligand of ERα with substantial anti-osteoporosis but less estrogenic activity.
Collapse
Affiliation(s)
- Keqi Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Yongyan Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Shuo Gao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Maosi Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Mengmeng Ge
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Qian Yang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Mingkai Liao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Junjie Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Haifeng Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Xiao-kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Ting Lin
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- Corresponding authors. Tel.: +86 592 2881105; fax: +86 592 2881105.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
- Corresponding authors. Tel.: +86 592 2881105; fax: +86 592 2881105.
| |
Collapse
|
6
|
Huang H, Zhou J, Chen H, Li J, Zhang C, Jiang X, Ni C. The immunomodulatory effects of endocrine therapy in breast cancer. J Exp Clin Cancer Res 2021; 40:19. [PMID: 33413549 PMCID: PMC7792133 DOI: 10.1186/s13046-020-01788-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023] Open
Abstract
Endocrine therapies with SERMs (selective estrogen receptor modulators) or SERDs (selective estrogen receptor downregulators) are standard therapies for patients with estrogen receptor (ER)-positive breast cancer. Multiple small molecule inhibitors targeting the PI3K-AKT-mTOR pathway or CDK4/6 have been developed to be used in combination with anti-estrogen drugs to overcome endocrine resistance. In addition to their direct antitumor effects, accumulating evidence has revealed the tumor immune microenvironment (TIM)-modulating effects of these therapeutic strategies, which have not been properly acknowledged previously. The immune microenvironment of breast tumors plays a crucial role in tumor development, metastasis and treatment response to endocrine therapy and immunotherapy. Therefore, in our current work, we comprehensively review the immunomodulatory effect of endocrine therapy and discuss its potential applications in combination with immune checkpoint inhibitors in breast cancer treatment.
Collapse
Affiliation(s)
- Huanhuan Huang
- Department of Breast Surgery, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Jun Zhou
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University, Zhejiang, 310006, Hangzhou, China
| | - Hailong Chen
- Department of Breast Surgery, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Jiaxin Li
- Department of Breast Surgery, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China
| | - Chao Zhang
- Department of Anatomy School of Medicine, Zhejiang University, Zhejiang, 310058, Hangzhou, China
| | - Xia Jiang
- School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610064, China.
- Department of Clinical Neuroscience Centre for Molecular Medicine, Karolinska Institute, Stockholm, 17176, Sweden.
| | - Chao Ni
- Department of Breast Surgery, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China.
- Key Laboratory of Tumour Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital Zhejiang University, Zhejiang, 310009, Hangzhou, China.
| |
Collapse
|
7
|
Liu W, Cui Y, Ren W, Irudayaraj J. Epigenetic biomarker screening by FLIM-FRET for combination therapy in ER+ breast cancer. Clin Epigenetics 2019; 11:16. [PMID: 30700309 PMCID: PMC6354376 DOI: 10.1186/s13148-019-0620-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/21/2019] [Indexed: 01/13/2023] Open
Abstract
Hormone-dependent gene expression involves dynamic and orchestrated regulation of epigenome leading to a cancerous state. Estrogen receptor (ER)-positive breast cancer rely on chromatin remodeling and association with epigenetic factors in inducing ER-dependent oncogenesis and thus cell over-proliferation. The mechanistic differences between epigenetic regulation and hormone signaling provide an avenue for combination therapy of ER-positive breast cancer. We hypothesized that epigenetic biomarkers within single nucleosome proximity of ER-dependent genes could serve as potential therapeutic targets. We described here a Fluorescence lifetime imaging-based Förster resonance energy transfer (FLIM-FRET) methodology for biomarker screening that could facilitate combination therapy based on our study. We screened 11 epigenetic-related markers which include oxidative forms of DNA methylation, histone modifications, and methyl-binding domain proteins. Among them, we identified H4K12acetylation (H4K12ac) and H3K27 acetylation (H3K27ac) as potential epigenetic therapeutic targets. When histone acetyltransferase inhibitor targeting H4K12ac and H3K27ac was combined with tamoxifen, an enhanced therapeutic outcome was observed against ER-positive breast cancer both in vitro and in vivo. Together, we demonstrate a single molecule approach as an effective screening tool for devising targeted epigenetic therapy.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
- Present Address: Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA
| | - Yi Cui
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
- Earth and Biological Science Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Wen Ren
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA
| | - Joseph Irudayaraj
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, 47907, USA.
- Bindley Bioscience Center, Purdue University, West Lafayette, 47907, IN, USA.
- Present Address: Department of Bioengineering, Cancer Center at Illinois, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA.
| |
Collapse
|
8
|
Post AEM, Smid M, Nagelkerke A, Martens JWM, Bussink J, Sweep FCGJ, Span PN. Interferon-Stimulated Genes Are Involved in Cross-resistance to Radiotherapy in Tamoxifen-Resistant Breast Cancer. Clin Cancer Res 2018; 24:3397-3408. [PMID: 29661777 DOI: 10.1158/1078-0432.ccr-17-2551] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/07/2018] [Accepted: 04/10/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Treatment resistance is the main cause of adverse disease outcome in breast cancer patients. Here, we aimed to investigate common features in tamoxifen-resistant and radioresistant breast cancer, as tamoxifen-resistant breast cancer cells are cross-resistant to irradiation in vitroExperimental Design: RNA sequencing of tamoxifen-resistant and radioresistant breast cancer cells was performed and validated by quantitative PCR. Pathways were further investigated in vitro and in breast cancer patient cohorts to establish their relation with treatment resistance.Results: Both tamoxifen-resistant and radioresistant breast cancer cells had increased expression levels of genes involved in type I IFN signaling compared with nonresistant cells. IFN-stimulated genes (ISG) were induced in a dose-dependent and time-dependent manner after tamoxifen treatment and irradiation. Tamoxifen treatment also led to ssDNA presence in the cytoplasm, which is known to induce expression of ISGs, a phenomenon that has already been described for irradiation. Moreover, in a breast cancer patient cohort, high expression levels of ISGs were found in the primary tumor in around half of the patients. This was associated with a tumor-infiltrating lymphocyte (TIL) expression signature, although the ISGs were also expressed by the tumor cells themselves. Importantly, the expression of ISGs correlated with outcome in breast cancer patients treated with adjuvant tamoxifen or radiotherapy, but not in systemically untreated patients or chemotherapy-treated patients.Conclusions: Our data indicate that expression of ISGs by tumor cells is involved in acquired, treatment-induced resistance to tamoxifen and radiotherapy, and might play a role in intrinsic resistance via interaction with TILs. Clin Cancer Res; 24(14); 3397-408. ©2018 AACR.
Collapse
Affiliation(s)
- Annemarie E M Post
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands. .,Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Marcel Smid
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Anika Nagelkerke
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands.,Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Paul N Span
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
9
|
Fang Q, Yao S, Luo G, Zhang X. Identification of differentially expressed genes in human breast cancer cells induced by 4-hydroxyltamoxifen and elucidation of their pathophysiological relevance and mechanisms. Oncotarget 2017; 9:2475-2501. [PMID: 29416786 PMCID: PMC5788654 DOI: 10.18632/oncotarget.23504] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
While tamoxifen (TAM) is used for treating estrogen receptor (ER)a-positive breast cancer patients, its anti-breast cancer mechanisms are not completely elucidated. This study aimed to examine effects of 4-hydroxyltamoxifen (4-OH-TAM) on ER-positive (ER+) breast cancer MCF-7 cell growth and gene expression profiles. MCF-7 cell growth was inhibited by 4-OH-TAM dose-dependently with IC50 of 29 μM. 332 genes were up-regulated while 320 genes were down-regulated. The mRNA levels of up-regulated genes including STAT1, STAT2, EIF2AK2, TGM2, DDX58, PARP9, SASH1, RBL2 and USP18 as well as down-regulated genes including CCDN1, S100A9, S100A8, ANXA1 and PGR were confirmed by quantitative real-time PCR (qRT-PCR). In human breast tumor tissues, mRNA levels of EIF2Ak2, USP18, DDX58, RBL2, STAT2, PGR, S1000A9, and CCND1 were significantly higher in ER+- than in ER--breast cancer tissues. The mRNA levels of EIF2AK2, TGM2, USP18, DDX58, PARP9, STAT2, STAT1, PGR and CCND1 were all significantly higher in ER+-tumor tissues than in their corresponding tumor-adjacent tissues. These genes, except PGR and CCND1 which were down-regulated, were also up-regulated in ER+ MCF-7 cells by 4-OH-TAM. Total 14 genes mentioned above are involved in regulation of cell proliferation, apoptosis, cell cycles, and estrogen and interferon signal pathways. Bioinformatics analysis also revealed other novel and important regulatory factors that are associated with these genes and involved in the mentioned functional processes. This study has paved a foundation for elucidating TAM anti-breast cancer mechanisms in E2/ER-dependent and independent pathways.
Collapse
Affiliation(s)
- Qi Fang
- Department of Breast Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Shuang Yao
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Guanghua Luo
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| | - Xiaoying Zhang
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, P.R. China
| |
Collapse
|
10
|
Dunnick JK, Morgan DL, Elmore SA, Gerrish K, Pandiri A, Ton TV, Shockley KR, Merrick BA. Tetrabromobisphenol A activates the hepatic interferon pathway in rats. Toxicol Lett 2017; 266:32-41. [PMID: 27914987 PMCID: PMC5791538 DOI: 10.1016/j.toxlet.2016.11.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Accepted: 11/25/2016] [Indexed: 11/25/2022]
Abstract
Tetrabromobisphenol A (TBBPA) is a widely used flame retardant in printed circuit boards, paper, and textiles. In a two-year study, TBBPA showed evidence of uterine tumors in female Wistar-Han rats and liver and colon tumors in B6C3F1 mice. In order to gain further insight into early gene and pathway changes leading to cancer, we exposed female Wistar Han rats to TBBPA at 0, 25, 250, or 1000mg/kg (oral gavage in corn oil, 5×/week) for 13 weeks. Because at the end of the TBBPA exposure period, there were no treatment-related effects on body weights, liver or uterus lesions, and liver and uterine organ weights were within 10% of controls, only the high dose animals were analyzed. Analysis of the hepatic and uterine transcriptomes showed TBBPA-induced changes primarily in the liver (1000mg/kg), with 159 transcripts corresponding to 132 genes differentially expressed compared to controls (FDR=0.05). Pathway analysis showed activation of interferon (IFN) and metabolic networks. TBBPA induced few molecular changes in the uterus. Activation of the interferon pathway in the liver occurred after 13-weeks of TBBPA exposure, and with longer term TBBPA exposure this may lead to immunomodulatory changes that contribute to carcinogenic processes.
Collapse
Affiliation(s)
- J K Dunnick
- Toxicology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| | - D L Morgan
- NTP Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - S A Elmore
- Cellular and Molecular Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - K Gerrish
- Molecular Genomics Core, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - A Pandiri
- Cellular and Molecular Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - T V Ton
- Cellular and Molecular Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - K R Shockley
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - B A Merrick
- Biomolecular Screening Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
11
|
LIU MEI, YANG YAJUN, ZHENG HONG, ZHONG XIAORONG, WANG YU, WANG ZHU, WANG YAOGENG, WANG YANPING. Membrane-bound complement regulatory proteins are prognostic factors of operable breast cancer treated with adjuvant trastuzumab: A retrospective study. Oncol Rep 2014; 32:2619-27. [DOI: 10.3892/or.2014.3496] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/20/2014] [Indexed: 11/06/2022] Open
|
12
|
Hilborn E, Sivik T, Fornander T, Stål O, Nordenskjöld B, Jansson A. C-X-C ligand 10 and C-X-C receptor 3 status can predict tamoxifen treatment response in breast cancer patients. Breast Cancer Res Treat 2014; 145:73-82. [PMID: 24715380 PMCID: PMC3984417 DOI: 10.1007/s10549-014-2933-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 11/24/2022]
Abstract
To investigate the expression levels of CXCL10 and CXCR3 in tumors from breast cancer patients randomized to adjuvant tamoxifen treatment or no endocrine treatment, in order to further study the connection to prognosis and prediction of tamoxifen treatment outcome. Immunohistochemistry on tissue microarrays from 912 breast cancer patients randomized to tamoxifen or no endocrine treatment. CXCR3 status was found to be a prognostic tool in predicting distant recurrence, as well as reduced breast cancer-specific survival. In patients with estrogen receptor (ER)-positive tumors, tumors with strong CXCL10 levels had improved effect of tamoxifen treatment in terms of local recurrence-free survival [risk ratio (RR) 0.46 (95 % CI 0.25–0.85, P = 0.01)] compared with patients with tumors expressing weak CXCL10 expression. Further, patients with ER-positive tumors with strong CXCR3 expression had an improved effect of tamoxifen in terms of breast cancer-specific survival [RR 0.34 (95 % CI 0.19–0.62, P < 0.001)] compared with the group with weak CXCR3 levels [RR 1.33 (95 % CI 0.38–4.79, P = 0.65)]. We show here for the first time that CXCL10 and CXCR3 expression are both predictors of favorable outcome in patients treated with tamoxifen.
Collapse
Affiliation(s)
- Erik Hilborn
- Division of Surgery and Clinical Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, 581 85, Linköping, Sweden,
| | | | | | | | | | | |
Collapse
|
13
|
Sayeed A, Luciani-Torres G, Meng Z, Bennington JL, Moore DH, Dairkee SH. Aberrant regulation of the BST2 (Tetherin) promoter enhances cell proliferation and apoptosis evasion in high grade breast cancer cells. PLoS One 2013; 8:e67191. [PMID: 23840623 PMCID: PMC3688682 DOI: 10.1371/journal.pone.0067191] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/16/2013] [Indexed: 11/18/2022] Open
Abstract
Normal cellular phenotypes that serve an oncogenic function during tumorigenesis are potential candidates for cancer targeting drugs. Within a subset of invasive primary breast carcinoma, we observed relatively abundant expression of Tetherin, a cell surface protein encoded by the Bone Marrow Stromal Cell Antigen (BST2) known to play an inhibitory role in viral release from infected immune cells of the host. Using breast cancer cell lines derived from low and intermediate histopathologic grade invasive primary tumors that maintain growth-suppressive TGFβ signaling, we demonstrate that BST2 is negatively regulated by the TGFβ axis in epithelial cells. Binding of the transcription factor AP2 to the BST2 promoter was attenuated by inhibition of the TGFβ pathway thereby increasing BST2 expression in tumor cells. In contrast, inherent TGFβ resistance characteristic of high grade breast tumors is a key factor underlying compromised BST2 regulation, and consequently its constitutive overexpression relative to non-malignant breast epithelium, and to most low and intermediate grade cancer cells. In both 2-dimensional and 3-dimensional growth conditions, BST2-silenced tumor cells displayed an enhancement in tamoxifen or staurosporine-induced apoptotic cell death together with a reduction in the S-phase fraction compared to BST2 overexpressing counterparts. In a subset of breast cancer patients treated with pro apoptotic hormonal therapy, BST2 expression correlated with a trend for poor clinical outcome, further supporting its role in conferring an anti apoptotic phenotype. Similar to the effects of gene manipulation, declining levels of endogenous BST2 induced by the phytoalexin – resveratrol, restored apoptotic function, and curbed cell proliferation. We provide evidence for a direct approach that diminishes aberrant BST2 expression in cancer cells as an early targeting strategy to assist in surmounting resistance to pro apoptotic therapies.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antineoplastic Agents, Hormonal/pharmacology
- Apoptosis
- Base Sequence
- Binding Sites
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Cell Line, Tumor
- Cell Proliferation
- Drug Synergism
- Fatty Acid-Binding Proteins/metabolism
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Molecular Sequence Data
- Promoter Regions, Genetic
- Proportional Hazards Models
- Protein Binding
- Resveratrol
- Stilbenes/pharmacology
- Tamoxifen/pharmacology
- Transforming Growth Factor beta/physiology
Collapse
Affiliation(s)
- Aejaz Sayeed
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - Gloria Luciani-Torres
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - Zhenhang Meng
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - James L. Bennington
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - Dan H. Moore
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - Shanaz H. Dairkee
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
14
|
Brody JS. Transcriptome alterations induced by cigarette smoke. Int J Cancer 2012; 131:2754-62. [PMID: 22961494 DOI: 10.1002/ijc.27829] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/16/2012] [Indexed: 12/24/2022]
Abstract
Cigarette smoke alters the transcriptome of multiple tissues; those directly exposed to toxic products and those exposed to circulating components and metabolic products of tobacco smoke. In most tissues and organs that have been studied, the smoking transcriptome is characterized by increased expression of antioxidant and xenobiotic genes as well as a wide spectrum of inflammation-related genes, and potential oncogenic genes. Smoking is associated with an increased incidence of cancer in a number of organs both those directly exposed (lungs and airways) and those indirectly exposed (bladder, liver, pancreas). Individual transcriptomic responses vary, based to some degree on as yet to be clarified genetic factors, and likely how and what the individual has smoked. The complexity of individual responses to tobacco exposure and of smoking-related cancers in various organs is beginning to be revealed in transcriptomic and whole genome sequencing studies, of both tumors and cytologically normal appearing cells that have been exposed to cigarette smoke or its products creating a genomic "field of injury." The recent application of next generation sequencing to defining the transcriptome alterations induced by cigarette smoke holds the promise of discovering new approaches to personalized prevention and treatment of smoking-related lung diseases in the future.
Collapse
Affiliation(s)
- Jerome S Brody
- Boston University School of Medicine, Pulmonary Center (R-3), Boston, MA 02118, USA.
| |
Collapse
|
15
|
Klymiuk I, Kenner L, Adler T, Busch DH, Boersma A, Irmler M, Gailus-Durner V, Fuchs H, Leitner N, Müller M, Kühn R, Schlederer M, Treise I, de Angelis MH, Beckers J. In vivo functional requirement of the mouse Ifitm1 gene for germ cell development, interferon mediated immune response and somitogenesis. PLoS One 2012; 7:e44609. [PMID: 23115618 PMCID: PMC3480353 DOI: 10.1371/journal.pone.0044609] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 08/03/2012] [Indexed: 01/19/2023] Open
Abstract
The mammalian Interferon induced transmembrane protein 1 (Ifitm1) gene was originally identified as a member of a gene family highly inducible by type I and type II interferons. Based on expression analyses, it was suggested to be required for normal primordial germ cell migration. The knockdown of Ifitm1 in mouse embryos provided evidence for a role in somitogenesis. We generated the first targeted knockin allele of the Ifitm1 gene to systematically reassess all inferred functions. Sperm motility and the fertility of male and female mutant mice are as in wild type littermates. Embryonic somites and the adult vertebral column appear normal in homozygous Ifitm1 knockout mice, demonstrating that Ifitm1 is not essential for normal segmentation of the paraxial mesoderm. Proportions of leucocyte subsets, including granulocytes, monocytes, B-cells, T-cells, NK-cells, and NKT-cells, are unchanged in mutant mice. Based on a normal immune response to Listeria monocytogenes infection, there is no evidence for a dysfunction in downstream IFNγ signaling in Ifitm1 mutant mice. Expression from the Ifitm1 locus from E8.5 to E14.5 is highly dynamic. In contrast, in adult mice, Ifitm1 expression is highly restricted and strong in the bronchial epithelium. Intriguingly, IFITM1 is highly overexpressed in tumor epithelia cells of human squamous cell carcinomas and in adenocarcinomas of NSCLC patients. These analyses underline the general importance of targeted in vivo studies for the functional annotation of the mammalian genome. The first comprehensive description of the Ifitm1 expression pattern provides a rational basis for the further examination of Ifitm1 gene functions. Based on our data, the fact that IFITM1 can function as a negative regulator of cell proliferation, and because the gene maps to chromosome band 11p15.5, previously associated with NSCLC, it is likely that IFITM1 in man has a key role in tumor formation.
Collapse
Affiliation(s)
- Ingeborg Klymiuk
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- * E-mail: (IK); (JB)
| | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research and Institute for Clinical Pathology, Medical University Vienna, Vienna, Austria
| | - Thure Adler
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Auke Boersma
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- Institute of Laboratory Animal Science and Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martin Irmler
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Valérie Gailus-Durner
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Nicole Leitner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ralf Kühn
- Institute of Developmental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Michaela Schlederer
- Ludwig Boltzmann Institute for Cancer Research, Ludwig Boltzmann Gesellschaft, Vienna, Austria
| | - Irina Treise
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- Experimental Genetics, Technische Universität München, Freising-Weihenstephan, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München GmbH, Neuherberg, Germany
- Experimental Genetics, Technische Universität München, Freising-Weihenstephan, Germany
- * E-mail: (IK); (JB)
| |
Collapse
|
16
|
Gabriel U, Li L, Bolenz C, Steidler A, Kränzlin B, Saile M, Gretz N, Trojan L, Michel MS. New insights into the influence of cigarette smoking on urothelial carcinogenesis: Smoking-induced gene expression in tumor-free urothelium might discriminate muscle-invasive from nonmuscle-invasive urothelial bladder cancer. Mol Carcinog 2011; 51:907-15. [DOI: 10.1002/mc.20860] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 08/29/2011] [Accepted: 09/08/2011] [Indexed: 11/07/2022]
|
17
|
Lee MH, Kim JW, Kim JH, Kang KS, Kong G, Lee MO. Gene expression profiling of murine hepatic steatosis induced by tamoxifen. Toxicol Lett 2010; 199:416-24. [PMID: 20937368 DOI: 10.1016/j.toxlet.2010.10.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 10/04/2010] [Accepted: 10/05/2010] [Indexed: 11/24/2022]
Abstract
Tamoxifen is an antiestrogenic agent used widely in the treatment of estrogen receptor-positive breast cancer. However, hepatic steatosis has been reported during clinical trials of tamoxifen. To explore the mechanism responsible for this tamoxifen-induced hepatic steatosis, we used microarray analysis to profile the gene expression pattern of mouse liver after tamoxifen treatment. Tamoxifen was administered orally as a single dose of 10mg/kg (low dose), 50mg/kg (medium dose), or 100mg/kg (high dose) to C57BL/6 mice, and the livers were removed 2h, 4h, 8h, and 24h later. From microarray data obtained from the liver samples, 414 genes were selected as tamoxifen-responsive genes (P<0.05, two-way ANOVA; cutoff ≥ 1.5-fold response). These genes were classified into three groups: 308 of the 414 genes showed a time-dependent response, nine genes showed a dose-dependent response, and 97 genes showed a time- and dose-dependent response. Most of the 308 time-dependent-responsive genes were associated predominantly with the biological processes involved in lipid metabolism. Overrepresented transcription factor binding site analysis showed that the following nuclear receptors that are important in lipid and carbohydrate metabolism were overrepresented: the androgen receptor (AR), nuclear receptor subfamily 2 group F member 1 (NR2F1), hepatocyte nuclear factor 4α (HNF4α), and retinoic acid receptor-related orphan receptor alpha 1 (RORα1). Reporter gene analysis further revealed that tamoxifen repressed the 5α-dihydrotestosterone-induced activation of the AR and the intrinsic transactivation function of RORα1, HNF4α, and NR2F1. Taken together, these data provide a better understanding of the molecular mechanism underlying tamoxifen-induced steatogenic hepatotoxicity and useful information for predicting steatogenic hepatotoxicity.
Collapse
Affiliation(s)
- Min-Ho Lee
- College of Pharmacy and Bio-MAX Institute, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | |
Collapse
|
18
|
Biological reprogramming in acquired resistance to endocrine therapy of breast cancer. Oncogene 2010; 29:6071-83. [PMID: 20711236 DOI: 10.1038/onc.2010.333] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endocrine therapies targeting the proliferative effect of 17β-estradiol through estrogen receptor α (ERα) are the most effective systemic treatment of ERα-positive breast cancer. However, most breast tumors initially responsive to these therapies develop resistance through molecular mechanisms that are not yet fully understood. The long-term estrogen-deprived (LTED) MCF7 cell model has been proposed to recapitulate acquired resistance to aromatase inhibitors in postmenopausal women. To elucidate this resistance, genomic, transcriptomic and molecular data were integrated into the time course of MCF7-LTED adaptation. Dynamic and widespread genomic changes were observed, including amplification of the ESR1 locus consequently linked to an increase in ERα. Dynamic transcriptomic profiles were also observed that correlated significantly with genomic changes and were predicted to be influenced by transcription factors known to be involved in acquired resistance or cell proliferation (for example, interferon regulatory transcription factor 1 and E2F1, respectively) but, notably, not by canonical ERα transcriptional function. Consistently, at the molecular level, activation of growth factor signaling pathways by EGFR/ERBB/AKT and a switch from phospho-Ser118 (pS118)- to pS167-ERα were observed during MCF7-LTED adaptation. Evaluation of relevant clinical settings identified significant associations between MCF7-LTED and breast tumor transcriptome profiles that characterize ERα-negative status, early response to letrozole and tamoxifen, and recurrence after tamoxifen treatment. In accordance with these profiles, MCF7-LTED cells showed increased sensitivity to inhibition of FGFR-mediated signaling with PD173074. This study provides mechanistic insight into acquired resistance to endocrine therapies of breast cancer and highlights a potential therapeutic strategy.
Collapse
|
19
|
He X, Dong DD, Yie SM, Yang H, Cao M, Ye SR, Li K, Liu J, Chen J. HLA-G expression in human breast cancer: implications for diagnosis and prognosis, and effect on allocytotoxic lymphocyte response after hormone treatment in vitro. Ann Surg Oncol 2010; 17:1459-69. [PMID: 20052552 DOI: 10.1245/s10434-009-0891-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The aim of this study is to investigate clinical implications of human leukocyte antigen-G (HLA-G) expression in breast cancer. METHODS HLA-G expression in 235 primary breast cancer tissues was investigated using immunohistochemistry, and plasma soluble HLA-G (sHLA-G) was measured in 44 breast cancer patients using a specific HLA-G enzyme-linked immunosorbent assay (ELISA). Effects of estradiol/progesterone and their antagonists tamoxifen/RU486 on HLA-G expression in cultured breast cancer MCF-7 cells were determined by real-time polymerase chain reaction (PCR) and the ELISA. Alterations of HLA-G expression by the hormone treatments on subsequent allocytotoxic lymphocyte (allo-CTL) response were also examined. RESULTS In the study, approximately 66% of neoplasm lesions were identified to have positive HLA-G expression. This expression was significantly correlated with tumor size, nodal status, and clinical disease stage (P = 0.0001, 0.012, and 0.0001, respectively). Patients with positive HLA-G expression had a lower survival rate than those with negative expression (P < 0.028). Plasma sHLA-G levels were significantly higher in breast cancer patients than in healthy controls (P < 0.001), with the area under the receiver-operating characteristic (ROC) curve being 0.95. HLA-G expression in breast cancer MCF-7 cells was enhanced by estradiol/progesterone but reduced by their antagonists. Cytotoxicity studies showed that allo-CTL response in MCF-7 cells was inhibited by prior treatment with estradiol/progesterone, but was amplified by their antagonists. The effects could be restored or further strengthened by the addition of anti-HLA-G antibodies. CONCLUSION Our findings suggest that HLA-G may have potential clinical implications in diagnosis, prognosis, and immunotherapy of patients with breast cancer.
Collapse
Affiliation(s)
- Xu He
- Core Laboratory, Sichuan Academy of Medical Sciences, Chengdu, Sichuan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|