1
|
Križić M, Popović M, Silovski T, Grbin D, Dedić Plavetić N. Real-world Outcomes of Dual HER2 Blockade Therapy in Metastatic HER2-Positive Breast Cancer: from Induction to Maintenance. Drugs Real World Outcomes 2024; 11:413-423. [PMID: 38879832 PMCID: PMC11365913 DOI: 10.1007/s40801-024-00438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Dual human epidermal growth factor receptor 2 (HER2) blockade with trastuzumab and pertuzumab combined with taxane-based chemotherapy (Cht) has been the standard first-line treatment for HER2-positive metastatic breast cancer (mBC) for years, due to the impressive results of the CLEOPATRA study. Real-world (RW) studies have become critical for assessing treatment effectiveness and safety in real-life circumstances. The aim of this study was to analyze the treatment outcomes of first-line therapy for HER2-positive mBC in RW clinical practice, specifically focusing on the use of maintenance endocrine therapy (ET) in hormone receptor positive (HR-positive) patients. METHODS This retrospective analysis included 106 HER2-positive mBC patients treated with trastuzumab and pertuzumab combined with taxane-based Cht from October 2015 to December 2020 at the University Hospital Centre Zagreb. RESULTS At a median follow-up of 30 months, median progression-free survival (PFS) was 25 months for the total population (95% confidence interval [CI] 16 - not analyzed). Patients with de novo mBC had longer median PFS than patients with recurrent disease (not reached vs. 18 months; hazard ratio 1.99; 95% CI 0.69-3.64, p<0.022). Age, hormone receptor positivity, visceral involvement, number of Cht cycles and previous adjuvant trastuzumab did not impact PFS. Most HR-positive patients (N=55, 88.7%) received maintenance ET after induction Cht. CONCLUSION This retrospective study provides additional data on patient characteristics, treatment and outcomes of RW HER2-positive mBC patients treated with pertuzumab and trastuzumab as first-line therapy. In our institution, maintenance ET after induction Cht has become standard clinical practice.
Collapse
Affiliation(s)
- Marija Križić
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia.
| | - Marina Popović
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Tajana Silovski
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dorotea Grbin
- Faculty of Science, Department of Biology, University of Zagreb, Zagreb, Croatia
| | - Natalija Dedić Plavetić
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
2
|
Jankowski K, Jagana V, Bisserier M, Hadri L. Switch-Independent 3A: An Epigenetic Regulator in Cancer with New Implications for Pulmonary Arterial Hypertension. Biomedicines 2023; 12:10. [PMID: 38275371 PMCID: PMC10813728 DOI: 10.3390/biomedicines12010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Epigenetic mechanisms, including DNA methylation, histone modifications, and non-coding RNA, play a crucial role in the regulation of gene expression and are pivotal in biological processes like apoptosis, cell proliferation, and differentiation. SIN3a serves as a scaffold protein and facilitates interactions with transcriptional epigenetic partners and specific DNA-binding transcription factors to modulate gene expression by adding or removing epigenetic marks. However, the activation or repression of gene expression depends on the factors that interact with SIN3a, as it can recruit both transcriptional activators and repressors. The role of SIN3a has been extensively investigated in the context of cancer, including melanoma, lung, and breast cancer. Our group is interested in defining the roles of SIN3a and its partners in pulmonary vascular disease. Pulmonary arterial hypertension (PAH) is a multifactorial disease often described as a cancer-like disease and characterized by disrupted cellular metabolism, sustained vascular cell proliferation, and resistance to apoptosis. Molecularly, PAH shares many common signaling pathways with cancer cells, offering the opportunity to further consider therapeutic strategies used for cancer. As a result, many signaling pathways observed in cancer were studied in PAH and have encouraged new research studying SIN3a's role in PAH due to its impact on cancer growth. This comparison offers new therapeutic options. In this review, we delineate the SIN3a-associated epigenetic mechanisms in cancer and PAH cells and highlight their impact on cell survival and proliferation. Furthermore, we explore in detail the role of SIN3a in cancer to provide new insights into its emerging role in PAH pathogenesis.
Collapse
Affiliation(s)
- Katherine Jankowski
- Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vineeta Jagana
- Department of Cell Biology and Anatomy & Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY 10595, USA; (V.J.); (M.B.)
| | - Malik Bisserier
- Department of Cell Biology and Anatomy & Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY 10595, USA; (V.J.); (M.B.)
| | - Lahouaria Hadri
- Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
3
|
GRHL2 Enhances Phosphorylated Estrogen Receptor (ER) Chromatin Binding and Regulates ER-Mediated Transcriptional Activation and Repression. Mol Cell Biol 2022; 42:e0019122. [PMID: 36036613 PMCID: PMC9584124 DOI: 10.1128/mcb.00191-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Phosphorylation of estrogen receptor α (ER) at serine 118 (pS118-ER) is induced by estrogen and is the most abundant posttranslational mark associated with a transcriptionally active receptor. Cistromic analysis of pS118-ER from our group revealed enrichment of the GRHL2 motif near pS118-ER binding sites. In this study, we used cistromic and transcriptomic analyses to interrogate the relationship between GRHL2 and pS118-ER. We found that GRHL2 is bound to chromatin at pS118-ER/GRHL2 co-occupancy sites prior to ligand treatment, and GRHL2 binding is required for maximal pS118-ER recruitment. pS118-ER/GRHL2 co-occupancy sites were enriched at active enhancers marked by H3K27ac and H3K4me1, along with FOXA1 and p300, compared to sites where each factor binds independently. Transcriptomic analysis yielded four subsets of ER/GRHL2-coregulated genes revealing that GRHL2 can both enhance and antagonize E2-mediated ER transcriptional activity. Gene ontology analysis indicated that coregulated genes are involved in cell migration. Accordingly, knockdown of GRHL2, combined with estrogen treatment, resulted in increased cell migration but no change in proliferation. These results support a model in which GRHL2 binds to selected enhancers and facilitates pS118-ER recruitment to chromatin, which then results in differential activation and repression of genes that control estrogen-regulated ER-positive breast cancer cell migration.
Collapse
|
4
|
Vijayan S, Loganathan C, Sakayanathan P, Thayumanavan P. Synthesis and Characterization of Plumbagin S-Allyl Cysteine Ester: Determination of Anticancer Activity In Silico and In Vitro. Appl Biochem Biotechnol 2022; 194:5827-5847. [PMID: 35819687 DOI: 10.1007/s12010-022-04079-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022]
Abstract
In recent years, derivatives of natural compounds are synthesized to increase the bioavailability, pharmacology, and pharmacokinetics properties. The naphthoquinone, plumbagin (PLU), is well known for its anticancer activity. However, the clinical use of PLU is hindered due to its toxicity. Previous reports have shown that modification of PLU at 5'-hydroxyl group has reduced its toxicity towards normal cell line. In accordance, in the present study, 5'-hydroxyl group of PLU was esterified with S-allyl cysteine (SAC) to obtain PLU-SAC ester. The drug-likeness of PLU-SAC was understood by in silico ADME analysis. PLU-SAC was characterized by UV-visible spectroscopy, mass spectroscopy, and nuclear magnetic resonance (NMR) spectroscopy. Molecular docking and dynamics simulation analysis revealed the interaction of PLU-SAC with proteins of interest in cancer therapy such as human estrogen receptor α, tumor protein p53 negative regulator mouse double minute 2, and cyclin-dependent kinase 2. MMGBSA calculation showed the favorable binding energy which in turn demonstrated the stable binding of PLU-SAC with these proteins. PLU-SAC showed apoptosis in breast cancer cell line (MCF-7) by inducing oxidative stress, disturbing mitochondrial function, arresting cells at G1 phase of cell cycle, and initiating DNA fragmentation. However, PLU-SAC did not show toxicity towards normal Vero cell line. PLU-SAC was synthesized and structurally characterized, and its anticancer activity was determined by in silico and in vitro analysis.
Collapse
Affiliation(s)
- Sudha Vijayan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, 636011, India
| | - Chitra Loganathan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, 636011, India.,Research and Development Center, Bioinnov Solutions LLP, Salem, Tamil Nadu, 636002, India
| | | | | |
Collapse
|
5
|
Viganò L, Locatelli A, Ulisse A, Galbardi B, Dugo M, Tosi D, Tacchetti C, Daniele T, Győrffy B, Sica L, Macchini M, Zambetti M, Zambelli S, Bianchini G, Gianni L. Modulation of the Estrogen/erbB2 Receptors Cross-talk by CDK4/6 Inhibition Triggers Sustained Senescence in Estrogen Receptor- and ErbB2-positive Breast Cancer. Clin Cancer Res 2022; 28:2167-2179. [PMID: 35254385 PMCID: PMC9595107 DOI: 10.1158/1078-0432.ccr-21-3185] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/24/2021] [Accepted: 03/03/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE The interplay between estrogen receptor (ER) and erbB tyrosine-kinase receptors (RTK) impacts growth and progression of ER-positive (ER+)/HER2-positive (HER2+) breast cancer and generates mitogenic signals converging onto the Cyclin-D1/CDK4/6 complex. We probed this cross-talk combining endocrine-therapy (fulvestrant), dual HER2-blockade (trastuzumab and pertuzumab), and CDK4/6-inhibition (palbociclib; PFHPert). EXPERIMENTAL DESIGN Cytotoxic drug effects, interactions, and pharmacodynamics were studied after 72 hours of treatment and over 6 more days of culture after drug wash-out in three ER+/HER2+, two HER2low, and two ER-negative (ER-)/HER2+ breast cancer cell lines. We assessed gene-expression dynamic and association with Ki67 downregulation in 28 patients with ER+/HER2+ breast cancer treated with neoadjuvant PFHPert in NA-PHER2 trial (NCT02530424). RESULTS In vitro, palbociclib and/or fulvestrant induced a functional activation of RTKs signalling. PFHPert had additive or synergistic antiproliferative activity, interfered with resistance mechanisms linked to the RTKs/Akt/MTORC1 axis and induced sustained senescence. Unexpected synergism was found in HER2low cells. In patients, Ki67 downregulation at week 2 and surgery were significantly associated to upregulation of senescence-related genes (P = 7.7E-4 and P = 1.8E-4, respectively). Activation of MTORC1 pathway was associated with high Ki67 at surgery (P = 0.019). CONCLUSIONS Resistance associated with the combination of drugs targeting ER and HER2 can be bypassed by cotargeting Rb, enhancing transition from quiescence to sustained senescence. MTORC1 pathway activation is a potential mechanism of escape and RTKs functional activation may be an alternative pathway for survival also in ER+/HER2low tumor. PFHPert combination is an effective chemotherapy-free regimen for ER+/HER2+ breast cancer, and the mechanistic elucidation of sensitivity/resistance patterns may provide insights for further treatment refinement.
Collapse
Affiliation(s)
- Lucia Viganò
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberta Locatelli
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adele Ulisse
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Barbara Galbardi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Diego Tosi
- Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Carlo Tacchetti
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tiziana Daniele
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Balázs Győrffy
- Department of Bioinformatics, Faculty of General Medicine, Semmelweis University, Budapest, Hungary
- 2nd Dept. of Pediatrics, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- TTK Oncology Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Lorenzo Sica
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Milvia Zambetti
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Zambelli
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
6
|
Zhu YS, Zhu J. Molecular and cellular functions of long non-coding RNAs in prostate and breast cancer. Adv Clin Chem 2022; 106:91-179. [PMID: 35152976 DOI: 10.1016/bs.acc.2021.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) are defined as noncoding RNA transcripts with a length greater than 200 nucleotides. Research over the last decade has made great strides in our understanding of lncRNAs, especially in the biology of their role in cancer. In this article, we will briefly discuss the biogenesis and characteristics of lncRNAs, then review their molecular and cellular functions in cancer by using prostate and breast cancer as examples. LncRNAs are abundant, diverse, and evolutionarily, less conserved than protein-coding genes. They are often expressed in a tumor and cell-specific manner. As a key epigenetic factor, lncRNAs can use a wide variety of molecular mechanisms to regulate gene expression at each step of the genetic information flow pathway. LncRNAs display widespread effects on cell behavior, tumor growth, and metastasis. They act intracellularly and extracellularly in an autocrine, paracrine and endocrine fashion. Increased understanding of lncRNA's role in cancer has facilitated the development of novel biomarkers for cancer diagnosis, led to greater understanding of cancer prognosis, enabled better prediction of therapeutic responses, and promoted identification of potential targets for cancer therapy.
Collapse
Affiliation(s)
- Yuan-Shan Zhu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Clinical and Translational Science Center, Weill Cornell Medicine, New York, NY, United States.
| | - Jifeng Zhu
- Clinical and Translational Science Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
7
|
Gianni L, Colleoni M, Bisagni G, Mansutti M, Zamagni C, Del Mastro L, Zambelli S, Bianchini G, Frassoldati A, Maffeis I, Valagussa P, Viale G. Effects of neoadjuvant trastuzumab, pertuzumab and palbociclib on Ki67 in HER2 and ER-positive breast cancer. NPJ Breast Cancer 2022; 8:1. [PMID: 35013314 PMCID: PMC8748500 DOI: 10.1038/s41523-021-00377-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/06/2021] [Indexed: 12/05/2022] Open
Abstract
The crosstalk between estrogen and HER2 receptors and cell-cycle regulation sustains resistance to endocrine therapy of HER2- and hormone receptor-positive breast cancer. We earlier reported that women with HER2 and ER-positive breast cancer receiving neoadjuvant dual HER2-block and palbociclib in the NA-PHER2 trial had Ki67 decrease and 27% pathological complete responses (pCR). We extended NA-PHER2 to Cohort B using dual HER2-block and palbociclib without fulvestrant and report here Ki67 drops at week-2 (mean change −25.7), at surgery (after 16 weeks, mean change −9.5), high objective response (88.5%) and pCR (19.2%). In Cohort C [Ki67 > 20% and HER2low (IHC 1+/2+ without gene amplification)], women also received fulvestrant, had dramatic Ki67 drop at week 2 (−29.5) persisting at surgery (−19.3), and objective responses in 78.3%. In view of the favorable tolerability and of the efficacy-predictive value of Ki67 drop at week-2, the chemotherapy-free approach of NA-PHER2 deserves further investigation in HER2 and ER-positive breast cancer. The trial is registered with ClinicalTrials.gov, number NCT02530424.
Collapse
Affiliation(s)
| | - Marco Colleoni
- IEO, European Institute of Oncology, IRCCS, Milano, Italy
| | | | - Mauro Mansutti
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Claudio Zamagni
- Addarii Medical Oncology IRCCS Azienda Ospedaliero-universitaria di Bologna, Bologna, Italy
| | - Lucia Del Mastro
- IRCCS Ospedale Policlinico San Martino, UO Breast Unit, Genova, Italy.,Università di Genova, Dipartimento di Medicina Interna e Specialità Mediche (Di.M.I.), Genova, Italy
| | - Stefania Zambelli
- Department of Medical Oncology, San Raffaele Scientific Institute, Milano, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Scientific Institute, Milano, Italy
| | - Antonio Frassoldati
- Department of Oncology, Azienda Ospedaliero Universitaria di Ferrara - Arcispedale Sant'Anna, Ferrara, Italy
| | | | | | - Giuseppe Viale
- IRCCS European Institute of Oncology, Milano, University of Milan, School of Medicine, Milano, Italy
| |
Collapse
|
8
|
Hühn D, Martí‐Rodrigo P, Mouron S, Hansel C, Tschapalda K, Porebski B, Häggblad M, Lidemalm L, Quintela‐Fandino M, Carreras‐Puigvert J, Fernandez‐Capetillo O. Prolonged estrogen deprivation triggers a broad immunosuppressive phenotype in breast cancer cells. Mol Oncol 2022; 16:148-165. [PMID: 34392603 PMCID: PMC8732350 DOI: 10.1002/1878-0261.13083] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/19/2021] [Accepted: 08/13/2021] [Indexed: 01/03/2023] Open
Abstract
Among others, expression levels of programmed cell death 1 ligand 1 (PD-L1) have been explored as biomarkers of the response to immune checkpoint inhibitors in cancer therapy. Here, we present the results of a chemical screen that interrogated how medically approved drugs influence PD-L1 expression. As expected, corticosteroids and inhibitors of Janus kinases were among the top PD-L1 downregulators. In addition, we identified that PD-L1 expression is induced by antiestrogenic compounds. Transcriptomic analyses indicate that chronic estrogen receptor alpha (ERα) inhibition triggers a broad immunosuppressive program in ER-positive breast cancer cells, which is subsequent to their growth arrest and involves the activation of multiple immune checkpoints together with the silencing of the antigen-presenting machinery. Accordingly, estrogen-deprived MCF7 cells are resistant to T-cell-mediated cell killing, in a manner that is independent of PD-L1, but which is reverted by estradiol. Our study reveals that while antiestrogen therapies efficiently limit the growth of ER-positive breast cancer cells, they concomitantly trigger a transcriptional program that favors their immune evasion.
Collapse
Affiliation(s)
- Daniela Hühn
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Pablo Martí‐Rodrigo
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Silvana Mouron
- Breast Cancer Clinical Research UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Catherine Hansel
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Kirsten Tschapalda
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Bartlomiej Porebski
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Maria Häggblad
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Louise Lidemalm
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Miguel Quintela‐Fandino
- Breast Cancer Clinical Research UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Jordi Carreras‐Puigvert
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Oscar Fernandez‐Capetillo
- Science for Life LaboratoryDivision of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
- Genomic Instability GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
9
|
The association between RAPSN methylation in peripheral blood and breast cancer in the Chinese population. J Hum Genet 2021; 66:1069-1078. [PMID: 33958711 DOI: 10.1038/s10038-021-00933-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 02/05/2023]
Abstract
DNA methylation in peripheral blood is associated with breast cancer (BC) but has mainly been studied in Caucasian populations. We investigated the association between blood-based methylation of receptor-associated protein of the synapse (RAPSN) and BC in Chinese population. The methylation levels of 12 RAPSN CpG sites were quantitatively evaluated by mass spectrometry in two case-control studies with 283 sporadic BC cases and 331 controls totally. The association was analyzed by logistic regression adjusted for covariants. The RAPSN methylation levels in patients with variant clinical characteristics were investigated by non-parametric tests. We found a significant association between BC and altered RAPSN methylation in blood in women at premenopausal and perimenopausal (age < 50 years old), but not in the elder women. This was approved by two independent case-control studies as well as by combining the subjects of the two studies (taken all subjects together, age < 50 years old, per 5% of methylation, odds ratio (OR) range from 1.17 to 1.30 for two CpG sites; OR = 0.75 for one CpG site; all p values < 0.02). This age-related RAPSN methylation was further modified by human epidermal growth factor receptor 2 (HER2) status (age < 50 years old, HER2 negative, per 5% of methylation, OR range from 1.27 to 1.48 for two CpG sites; OR = 0.76 for one CpG site; all p values < 0.02). We elucidated an association between BC and blood-based RAPSN methylation influenced by age and the status of HER2 in Chinese population.
Collapse
|
10
|
Portman N, Milioli HH, Alexandrou S, Coulson R, Yong A, Fernandez KJ, Chia KM, Halilovic E, Segara D, Parker A, Haupt S, Haupt Y, Tilley WD, Swarbrick A, Caldon CE, Lim E. MDM2 inhibition in combination with endocrine therapy and CDK4/6 inhibition for the treatment of ER-positive breast cancer. Breast Cancer Res 2020; 22:87. [PMID: 32787886 PMCID: PMC7425060 DOI: 10.1186/s13058-020-01318-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/21/2020] [Indexed: 01/21/2023] Open
Abstract
Background Resistance to endocrine therapy is a major clinical challenge in the management of oestrogen receptor (ER)-positive breast cancer. In this setting, p53 is frequently wildtype and its activity may be suppressed via upregulation of its key regulator MDM2. This underlies our rationale to evaluate MDM2 inhibition as a therapeutic strategy in treatment-resistant ER-positive breast cancer. Methods We used the MDM2 inhibitor NVP-CGM097 to treat in vitro and in vivo models alone and in combination with fulvestrant or palbociclib. We perform cell viability, cell cycle, apoptosis and senescence assays to evaluate anti-tumour effects in p53 wildtype and p53 mutant ER-positive cell lines (MCF-7, ZR75-1, T-47D) and MCF-7 lines resistant to endocrine therapy and to CDK4/6 inhibition. We further assess the drug effects in patient-derived xenograft (PDX) models of endocrine-sensitive and endocrine-resistant ER-positive breast cancer. Results We demonstrate that MDM2 inhibition results in cell cycle arrest and increased apoptosis in p53-wildtype in vitro and in vivo breast cancer models, leading to potent anti-tumour activity. We find that endocrine therapy or CDK4/6 inhibition synergises with MDM2 inhibition but does not further enhance apoptosis. Instead, combination treatments result in profound regulation of cell cycle-related transcriptional programmes, with synergy achieved through increased antagonism of cell cycle progression. Combination therapy pushes cell lines resistant to fulvestrant or palbociclib to become senescent and significantly reduces tumour growth in a fulvestrant-resistant patient-derived xenograft model. Conclusions We conclude that MDM2 inhibitors in combination with ER degraders or CDK4/6 inhibitors represent a rational strategy for treating advanced, endocrine-resistant ER-positive breast cancer, operating through synergistic activation of cell cycle co-regulatory programmes.
Collapse
Affiliation(s)
- Neil Portman
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Heloisa H Milioli
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Sarah Alexandrou
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Rhiannon Coulson
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Aliza Yong
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Kristine J Fernandez
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Kee Ming Chia
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Ensar Halilovic
- Novartis Institutes of Biomedical Research, Cambridge, MA, USA
| | - Davendra Segara
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Andrew Parker
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Sue Haupt
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ygal Haupt
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wayne D Tilley
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5001, Australia
| | - Alex Swarbrick
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - C Elizabeth Caldon
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Elgene Lim
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia. .,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia.
| |
Collapse
|
11
|
Gomez AL, Altamirano GA, Tschopp MV, Bosquiazzo VL, Muñoz-de-Toro M, Kass L. Exposure to a Glyphosate-based Herbicide Alters the Expression of Key Regulators of Mammary Gland Development on Pre-pubertal Male Rats. Toxicology 2020; 439:152477. [PMID: 32360609 DOI: 10.1016/j.tox.2020.152477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/03/2020] [Accepted: 04/24/2020] [Indexed: 12/17/2022]
Abstract
We previously reported that exposure during gestation and lactation to a low dose of glyphosate-based herbicide (GBH) reduced the area and perimeter of male offspring mammary gland at postnatal day 60 (PND60), whereas a higher dose increased the longitudinal growth of the gland. Here, our aim was to assess whether perinatal exposure to GBH exhibits endocrine disruptive action in male mammary gland at an early time point (pre-puberty), which could be related to the changes observed after puberty. We also wanted to explore whether an early evaluation of the male rat mammary gland is appropriate to assess exposure to potential endocrine disrupting chemicals (EDCs). Pregnant rats were orally exposed, through the diet, to vehicle (saline solution), 3.5 or 350 mg/kg/day of GBH from gestational day 9 until weaning. At PND21, the male offspring were euthanized, and mammary gland samples were collected. The histology and proliferation index of the mammary glands were evaluated, and the mRNA expression of estrogen (ESR1) and androgen (AR) receptors, cyclin D1 (Ccnd1), amphiregulin (Areg), insulin-like growth factor 1 (IGF1), epidermal growth factor receptor (EGFR) and IGF1 receptor (IGF1R) were assessed. Moreover, the phosphorylated-Erk1/2 (p-ERK1/2) protein expression was determined. No differences were observed in mammary epithelial structures and AR expression between experimental groups; however, the proliferation index was reduced in GBH3.5-exposed males. This result was associated with decreased ESR1, Ccnd1, Areg, IGF1, EGFR and IGF1R mRNA expressions, as well as reduced p-Erk1/2 protein expression in these animals. ESR1, Ccnd1, IGF1R and EGFR expressions were also reduced in GBH350-exposed males. In conclusion, the mammary gland development of pre-pubertal male rats is affected by perinatal exposure to GBH. Although further studies are still needed to understand the molecular mechanisms involved in GBH350 exposure, the present results may explain the alterations observed in mammary gland growth of post-pubertal males exposed to low doses of GBH. Our results also suggest that early evaluation of the male rat mammary gland is useful in assessing exposure to potential EDCs. However, analysis of EDCs effects at later time points should not be excluded.
Collapse
Affiliation(s)
- Ayelen L Gomez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gabriela A Altamirano
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - María V Tschopp
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Verónica L Bosquiazzo
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
12
|
Andò S, Naimo GD, Gelsomino L, Catalano S, Mauro L. Novel insights into adiponectin action in breast cancer: Evidence of its mechanistic effects mediated by ERα expression. Obes Rev 2020; 21:e13004. [PMID: 32067339 DOI: 10.1111/obr.13004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/11/2022]
Abstract
This review describes the multifaceted effects of adiponectin on breast cancer cell signalling, tumour metabolism, and microenvironment. It is largely documented that low adiponectin levels are associated with an increased risk of breast cancer. However, it needs to be still clarified what are the extents of the decrease of local/intra-tumoural adiponectin concentrations, which promote breast tumour malignancy. Most of the anti-proliferative and pro-apoptotic effects induced by adiponectin have been obtained in breast cancer cells not expressing estrogen receptor alpha (ERα). Here, we will highlight recent findings demonstrating the mechanistic effects through which adiponectin is able to fuel genomic and non-genomic estrogen signalling, inhibiting LKB1/AMPK/mTOR/S6K pathway and switching energy balance. Therefore, it emerges that the reduced adiponectin levels in patients with obesity work to sustain tumour growth and progression in ERα-positive breast cancer cells. All this may contribute to remove the misleading paradigm that adiponectin univocally inhibits breast cancer cell growth and progression independently on ERα status. The latter concept, here clearly provided by pre-clinical studies, may have translational relevance adopting adiponectin as a potential therapeutic tool. Indeed, the interfering role of ERα on adiponectin action addresses how a separate assessment of adiponectin treatment needs to be considered in novel therapeutic strategies for ERα-positive and ERα-negative breast cancer.
Collapse
Affiliation(s)
- Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy.,Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Loredana Mauro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
13
|
Xu X, Chen E, Mo L, Zhang L, Shao F, Miao K, Liu J, Su SM, Valecha M, Chan UI, Zheng H, Chen M, Chen W, Chen Q, Fu H, Aladjem MI, He Y, Deng CX. BRCA1 represses DNA replication initiation through antagonizing estrogen signaling and maintains genome stability in parallel with WEE1-MCM2 signaling during pregnancy. Hum Mol Genet 2020; 28:842-857. [PMID: 30445628 DOI: 10.1093/hmg/ddy398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/16/2018] [Accepted: 11/13/2018] [Indexed: 12/14/2022] Open
Abstract
The mammary gland undergoes fast cell proliferation during early pregnancy, yet the mechanism to ensure genome integrity during this highly proliferative stage is largely unknown. We show that pregnancy triggers replicative stresses leading to genetic instability in mice carrying a mammary specific disruption of breast cancer associated gene-1 (BRCA1). The fast cell proliferation was correlated with enhanced expression of most genes encoding replisomes, which are positively regulated by estrogen/ERα signaling but negatively regulated by BRCA1. Our further analysis revealed two parallel signaling pathways, which are mediated by ATR-CHK1 and WEE1-MCM2 and are responsible for regulating DNA replication checkpoint. Upon DNA damage, BRCA1 deficiency markedly enhances DNA replication initiation and preferably impairs DNA replication checkpoint mediated by ATR and CHK1. Meanwhile, DNA damage also activates WEE1-MCM2 signaling, which inhibits DNA replication initiation and enables BRCA1-deficient cells to avoid further genomic instability. Finally, we demonstrated that overriding this defense by WEE1 inhibition in combination with cisplatin, which causes DNA damage, serves as a promising therapeutic approach for killing BRCA1-deficient cancer cells.
Collapse
Affiliation(s)
- Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Eric Chen
- Genetics of Development and Disease Branch
| | - Lihua Mo
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lei Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR, China.,Department of Vascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Fangyuan Shao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Kai Miao
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jianlin Liu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Sek Man Su
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Monica Valecha
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Un In Chan
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | | | - Mark Chen
- Genetics of Development and Disease Branch
| | - Weiping Chen
- Gene Expression Core, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Qiang Chen
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Haiqing Fu
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yanzhen He
- Department of Vascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
14
|
Akula SM, Candido S, Abrams SL, Steelman LS, Lertpiriyapong K, Cocco L, Ramazzotti G, Ratti S, Follo MY, Martelli AM, Murata RM, Rosalen PL, Bueno-Silva B, Matias de Alencar S, Falasca M, Montalto G, Cervello M, Notarbartolo M, Gizak A, Rakus D, Libra M, McCubrey JA. Abilities of β-Estradiol to interact with chemotherapeutic drugs, signal transduction inhibitors and nutraceuticals and alter the proliferation of pancreatic cancer cells. Adv Biol Regul 2020; 75:100672. [PMID: 31685431 DOI: 10.1016/j.jbior.2019.100672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 06/10/2023]
Abstract
Improving the effects of chemotherapy and reducing the side effects are important goals in cancer research. Various approaches have been examined to enhance the effectiveness of chemotherapy. For example, signal transduction inhibitors or hormonal based approaches have been included with chemo- or radio-therapy. MIA-PaCa-2 and BxPC-3 pancreatic ductal adenocarcinoma (PDAC) cells both express the estrogen receptor (ER). The effects of β-estradiol on the growth of PDAC cells has not been examined yet the ER is expressed in PDAC cells. We have examined the effects of combining β-estradiol with chemotherapeutic drugs, signal transcription inhibitors, natural products and nutraceuticals on PDAC. In most cases, inclusion of β-estradiol with chemotherapeutic drugs increased chemosensitivity. These results indicate some approaches involving β-estradiol which may be used to increase the effectiveness of chemotherapeutic and other drugs on the growth of PDAC.
Collapse
Affiliation(s)
- Shaw M Akula
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Kvin Lertpiriyapong
- Center of Comparative Medicine and Pathology, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medicine and the Hospital for Special Surgery, New York City, New York, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Giulia Ramazzotti
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Matilde Y Follo
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Ramiro M Murata
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Bruno Bueno-Silva
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil; Dental Research Division, Guarulhos University, Guarulhos, Brazil
| | | | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia, 6102, Australia
| | - Giuseppe Montalto
- Dipartimento di Promozione Della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Monica Notarbartolo
- Department of Biological, Chemical and Pharmaceutical Science and Technology (STEBICEF), University of Palermo, Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA.
| |
Collapse
|
15
|
Tokgun PE, Tokgun O, Kurt S, Tomatir AG, Akca H. MYC-driven regulation of long non-coding RNA profiles in breast cancer cells. Gene 2019; 714:143955. [PMID: 31326549 DOI: 10.1016/j.gene.2019.143955] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 11/25/2022]
Abstract
AIM MYC deregulation contributes to breast cancer development and progression. Deregulated expression levels of long non-coding RNAs (lncRNA) have been demonstrated to be critical players in development and/or maintenance of breast cancer. In this study we aimed to evaluate lncRNA expressions depending on MYC overexpression and knockdown in breast cancer cells. MATERIALS AND METHODS Cells were infected with lentiviral vectors by either knockdown or overexpression of c-MYC. LncRNA cDNA was transcribed from total RNA samples and lncRNAs were evaluated by qRT-PCR. RESULTS Our results indicated that some of the lncRNAs having tumor suppressor (GAS5, MEG3, lincRNA-p21) and oncogenic roles (HOTAIR) are regulated by c-MYC. CONCLUSION We observed that c-MYC regulates lncRNAs that have important roles on proliferation, cell cycle and etc. Further studies will give us a light to identify molecular mechanisms related to MYC-lncRNA regulatory pathways in breast cancer.
Collapse
Affiliation(s)
- Pervin Elvan Tokgun
- Department of Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | - Onur Tokgun
- Department of Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | - Serap Kurt
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | - Ayse Gaye Tomatir
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | - Hakan Akca
- Department of Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| |
Collapse
|
16
|
Petrossian K, Kanaya N, Lo C, Hsu PY, Nguyen D, Yang L, Yang L, Warden C, Wu X, Pillai R, Bernal L, Huang CS, Kruper L, Yuan Y, Somlo G, Mortimer J, Chen S. ERα-mediated cell cycle progression is an important requisite for CDK4/6 inhibitor response in HR+ breast cancer. Oncotarget 2018; 9:27736-27751. [PMID: 29963233 PMCID: PMC6021239 DOI: 10.18632/oncotarget.25552] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/19/2018] [Indexed: 01/01/2023] Open
Abstract
While ER has multiple biological effects, ER-cyclin D1-CDK4/6-RB is a critical pathway for the action of estrogen on the cell cycle, especially for breast cancers that rely on estrogen for growth. The latest and most efficient CDK4/6 inhibitors target the phosphorylation of retinoblastoma (RB) tumor suppressor gene; thus, altering levels of many cell cycle molecules. Estrogen receptor (ER)+/HER2- breast cancers have shown great progression free survival when CDK4/6 inhibitors are combined with endocrine therapies. Here we report the mechanism of antiestrogen (fulvestrant) combination with CDK4/6 inhibitors is due to synergism in the suppression of ER-mediated cell cycle progression. Furthermore, we performed single cell analysis of cells from an estrogen dependent/hormone receptor-positive patient derived xenograft (PDX) tumor model treated with palbociclib. These single cells expressed various levels of ER and RB which are involved in cell cycle regulation; and the response to palbociclib treatment relies not only on the ER-cyclin D1-CDK4/6-RB pathway but it is also dependent on elevated levels of ER and/or RB. Our preclinical studies show that palbociclib response is dependent on cells with ER, which is directly involved in cell cycle progression in hormone receptor positive (HR+) breast cancer.
Collapse
Affiliation(s)
- Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiao Lo
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Duc Nguyen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lixin Yang
- Molecular Pathology Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lu Yang
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Charles Warden
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Raju Pillai
- Molecular Pathology Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lauren Bernal
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiun-Sheng Huang
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Laura Kruper
- Department of Surgery, City of Hope Medical Center, Duarte, CA, United States
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - Joanne Mortimer
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
17
|
Horibata S, Rice EJ, Zheng H, Mukai C, Chu T, Marks BA, Coonrod SA, Danko CG. A bi-stable feedback loop between GDNF, EGR1, and ERα contribute to endocrine resistant breast cancer. PLoS One 2018; 13:e0194522. [PMID: 29614078 PMCID: PMC5882141 DOI: 10.1371/journal.pone.0194522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/05/2018] [Indexed: 01/06/2023] Open
Abstract
Discovering regulatory interactions between genes that specify the behavioral properties of cells remains an important challenge. We used the dynamics of transcriptional changes resolved by PRO-seq to identify a regulatory network responsible for endocrine resistance in breast cancer. We show that GDNF leads to endocrine resistance by switching the active state in a bi-stable feedback loop between GDNF, EGR1, and the master transcription factor ERα. GDNF stimulates MAP kinase, activating the transcription factors SRF and AP-1. SRF initiates an immediate transcriptional response, activating EGR1 and suppressing ERα. Newly translated EGR1 protein activates endogenous GDNF, leading to constitutive GDNF and EGR1 up-regulation, and the sustained down-regulation of ERα. Endocrine resistant MCF-7 cells are constitutively in the GDNF-high/ ERα-low state, suggesting that the state in the bi-stable feedback loop may provide a 'memory' of endocrine resistance. Thus, we identified a regulatory network switch that contributes to drug resistance in breast cancer.
Collapse
Affiliation(s)
- Sachi Horibata
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Edward J. Rice
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Hui Zheng
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Chinatsu Mukai
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Tinyi Chu
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Graduate Field of Computational Biology, Cornell University, Ithaca, NY, United States of America
| | - Brooke A. Marks
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Scott A. Coonrod
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| | - Charles G. Danko
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
18
|
Rong C, Meinert ÉFRC, Hess J. Estrogen Receptor Signaling in Radiotherapy: From Molecular Mechanisms to Clinical Studies. Int J Mol Sci 2018; 19:ijms19030713. [PMID: 29498642 PMCID: PMC5877574 DOI: 10.3390/ijms19030713] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/02/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Numerous studies have established a proof of concept that abnormal expression and function of estrogen receptors (ER) are crucial processes in initiation and development of hormone-related cancers and also affect the efficacy of anti-cancer therapy. Radiotherapy has been applied as one of the most common and potent therapeutic strategies, which is synergistic with surgical excision, chemotherapy and targeted therapy for treating malignant tumors. However, the impact of ionizing radiation on ER expression and ER-related signaling in cancer tissue, as well as the interaction between endocrine and irradiation therapy remains largely elusive. This review will discuss recent findings on ER and ER-related signaling, which are relevant for cancer radiotherapy. In addition, we will summarize pre-clinical and clinical studies that evaluate the consequences of anti-estrogen and irradiation therapy in cancer, including emerging studies on head and neck cancer, which might improve the understanding and development of novel therapeutic strategies for estrogen-related cancers.
Collapse
Affiliation(s)
- Chao Rong
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Étienne Fasolt Richard Corvin Meinert
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
- Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Jochen Hess
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
- Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
19
|
Abstract
HER2 and CDK4/6 are undoubted two most important biological targets for breast cancer. Anti-HER2 treatments enhance objective response and progression-free survival/disease-free survival as well as overall survival. Three CDK4/6 inhibitors consistently improve objective response and progression-free survival; however, overall survival data are waited. Optimization of chemotherapy and endocrine strategies remains an unmet need. Check point inhibitor-based immunotherapy combined with chemotherapy is a promising field, especially for triple-negative breast cancer.
Collapse
Affiliation(s)
- Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Wei Huang
- Roche Product Development in Asia Pacific.5F, Tower C, Parkview Green, No.9, Dongdaqiao Road, Chaoyang District, Beijing, 100020 People’s Republic of China
| | - Minhao Fan
- Hutchison MediPharma Limited, Building 4 917 Halei Road Zhangjiang Hi-Tech Park, Shanghai, 201203 China
| |
Collapse
|
20
|
Feola A, Ricci S, Kouidhi S, Rizzo A, Penon A, Formisano P, Giordano A, Di Carlo A, Di Domenico M. Multifaceted Breast Cancer: The Molecular Connection With Obesity. J Cell Physiol 2016; 232:69-77. [DOI: 10.1002/jcp.25475] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 06/30/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Antonia Feola
- Department of Biochemistry, Biophysics and General Pathology; Second University of Naples; Naples Italy
- IRCCS Malzoni Clinic; Avellino Italy
| | - Serena Ricci
- Department of Translational Medical Science; University of Naples “Federico II”; Naples Italy
- Department of Medico-Surgical Sciences and Biotechnologies; University of Rome “La Sapienza”; Rome Italy
| | - Soumaya Kouidhi
- Université de la Manouba, ISBST, BVBGR-LR11ES31; Biotechpole Sidi Thabet, 2020; Ariana Tunisia
| | - Antonietta Rizzo
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology; Second University of Naples; Naples Italy
| | - Antonella Penon
- Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
| | - Pietro Formisano
- Department of Translational Medical Science; University of Naples “Federico II”; Naples Italy
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience; University of Siena; Siena Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology; Temple University; Philadelphia Pennsylvania
| | - Angelina Di Carlo
- Department of Medico-Surgical Sciences and Biotechnologies; University of Rome “La Sapienza”; Rome Italy
| | - Marina Di Domenico
- Department of Biochemistry, Biophysics and General Pathology; Second University of Naples; Naples Italy
- IRCCS Malzoni Clinic; Avellino Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology; Temple University; Philadelphia Pennsylvania
| |
Collapse
|
21
|
Huang W, Nie W, Zhang W, Wang Y, Zhu A, Guan X. The expression status of TRX, AR, and cyclin D1 correlates with clinicopathological characteristics and ER status in breast cancer. Onco Targets Ther 2016; 9:4377-85. [PMID: 27499632 PMCID: PMC4959412 DOI: 10.2147/ott.s94703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The ER signaling pathway plays a critical role in breast cancer. ER signaling pathway-related proteins, such as TRX, AR, and cyclin D1, may have an important function in breast cancer. However, the ways that they influence breast cancer development and progression are still unclear. Patients and methods A total of 101 Chinese female patients diagnosed with invasive ductal breast adenocarcinoma were retrospectively enrolled in the study. The expression levels of TRX, AR, and cyclin D1 were detected by immunohistochemistry and analyzed via correlation with clinicopathological characteristics and the expression status of ER, PR, and HER2. Results The expression status of TRX, AR, and cyclin D1 was not associated with the patient’s age, menopausal status, tumor size, or histological differentiation (P>0.05), but was positively correlated with ER and PR (P<0.001, respectively). Most (66/76, 86.8) TRX-positive patients were also HER2-positive (P=0.003). Of AR- or cyclin D1-positive patients, most had relatively earlier I–II tumor stage (P=0.005 and P=0.047, respectively) and no metastatic lymph node involvement (P=0.008 and P=0.005, respectively). Conclusion TRX was found to be positively correlated with ER and PR expression, whereas it was negatively correlated with HER2 expression. In addition, we found that the positive expression of AR and cyclin D1 was correlated with lower TNM stage and fewer metastatic lymph nodes, and it was more common in ER-positive breast cancer than in the basal-like subtype. This may indicate that AR and cyclin D1 are good predictive and prognostic factors and closely interact with ER signaling pathway. Further studies will be necessary to investigate the response and clinical outcomes of treatment targeting TRX, AR, and cyclin D1.
Collapse
Affiliation(s)
- Weisun Huang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Weiwei Nie
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Wenwen Zhang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Yanru Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Aiyu Zhu
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Southern Medical University, Guangzhou, People's Republic of China; Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| |
Collapse
|
22
|
Holst F. Estrogen receptor alpha gene amplification in breast cancer: 25 years of debate. World J Clin Oncol 2016; 7:160-173. [PMID: 27081639 PMCID: PMC4826962 DOI: 10.5306/wjco.v7.i2.160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 01/05/2016] [Accepted: 02/16/2016] [Indexed: 02/06/2023] Open
Abstract
Twenty-five years ago, Nembrot and colleagues reported amplification of the estrogen receptor alpha gene (ESR1) in breast cancer, initiating a broad and still ongoing scientific debate on the prevalence and clinical significance of this genetic aberration, which affects one of the most important genes in breast cancer. Since then, a multitude of studies on this topic has been published, covering a wide range of divergent results and arguments. The reported prevalence of this alteration in breast cancer ranges from 0% to 75%, suggesting that ESR1 copy number analysis is hampered by technical and interpreter issues. To date, two major issues related to ESR1 amplification remain to be conclusively addressed: (1) The extent to which abundant amounts of messenger RNA can mimic amplification in standard fluorescence in situ hybridization assays in the analysis of strongly expressed genes like ESR1, and (2) the clinical relevance of ESR1 amplification: Such relevance is strongly disputed, with data showing predictive value for response as well as for resistance of the cancer to anti-estrogen therapies, or for subsequent development of cancers in the case of precursor lesions that display amplification of ESR1. This review provides a comprehensive summary of the various views on ESR1 amplification, and highlights explanations for the contradictions and conflicting data that could inform future ESR1 research.
Collapse
|
23
|
Lebok P, Kopperschmidt V, Kluth M, Hube-Magg C, Özden C, B T, Hussein K, Mittenzwei A, Lebeau A, Witzel I, Wölber L, Mahner S, Jänicke F, Geist S, Paluchowski P, Wilke C, Heilenkötter U, Simon R, Sauter G, Terracciano L, Krech R, von d Assen A, Müller V, Burandt E. Partial PTEN deletion is linked to poor prognosis in breast cancer. BMC Cancer 2015; 15:963. [PMID: 26672755 PMCID: PMC4682275 DOI: 10.1186/s12885-015-1770-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 10/10/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deletions of chromosome 10q23, including the PTEN (phosphatase and tensin homolog) locus, are known to occur in breast cancer, but systematic analyses of its clinical relevance are lacking. METHODS We thus analyzed a tissue microarray (TMA) with 2,197 breast cancers by fluorescence in-situ hybridization (FISH) using a PTEN-specific probe. RESULTS PTEN deletions were detected in 19% of no special type, 9% of lobular, 4% of tubular cancers and 46% in carcinomas with medullary features. 98.7% of deletions were heterozygous and only 1.3% were homozygous. PTEN deletion was significantly linked to advanced tumor stage (p=0.0054), high-grade (p<0.0001), high tumor cell proliferation (Ki67 Labeling Index; p<0.0001), and shortened overall survival (p=0.0090). PTEN deletions were inversely associated with features of luminal type breast cancers (ER/PR positivity; p<0.0001 each, and CCND1 amplification; p=0.0020). PTEN deletions were also strongly linked to amplification of genes involved in the PTEN/AKT pathway such as MYC (p=0.0430) and HER2 (p=0.0065). Remarkably the combined analysis of MYC, HER2, CCND1 and PTEN aberrations suggested that aberrations of multiple PTEN/AKT pathway genes have a strong additive effect on breast cancer prognosis. While cancers with one of these aberrations behaved only marginally different from cancers with none, disease outcome was markedly worse in cancers with two or more aberrations as compared to those with only one aberration (p=0.0002). In addition, the particularly poor prognosis of patients with HER2 amplification and PTEN deletions challenges the concept of PTEN deletions interfering with trastuzumab therapy. CONCLUSION PTEN deletion occurs in a relevant fraction of breast cancers, and is linked to aggressive tumor behavior. Reduced PTEN function cooperates with MYC and HER2 activation in conferring aggressive phenotype to cancer cells.
Collapse
Affiliation(s)
- P Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - V Kopperschmidt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - M Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - C Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - C Özden
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Taskin B
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - K Hussein
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - A Mittenzwei
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - A Lebeau
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - I Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - L Wölber
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - S Mahner
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - F Jänicke
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - S Geist
- Department of Gynecology, Regio Clinic Pinneberg, Pinneberg, Germany.
| | - P Paluchowski
- Department of Gynecology, Regio Clinic Pinneberg, Pinneberg, Germany.
| | - C Wilke
- Department of Gynecology, Regio Clinic Elmshorn, Elmshorn, Germany.
| | - U Heilenkötter
- Department of Gynecology, Clinical Centre Itzehoe, Itzehoe, Germany.
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - L Terracciano
- Department of Pathology, Basel University Clinics, Basel, Switzerland.
| | - R Krech
- Institute of Pathology, Clinical Centre Osnabrück, Osnabrück, Germany.
| | | | - V Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - E Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
24
|
Wang ZY, Yin L. Estrogen receptor alpha-36 (ER-α36): A new player in human breast cancer. Mol Cell Endocrinol 2015; 418 Pt 3:193-206. [PMID: 25917453 DOI: 10.1016/j.mce.2015.04.017] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 04/20/2015] [Accepted: 04/20/2015] [Indexed: 01/16/2023]
Abstract
Prevailing wisdom is that estrogen receptor (ER)-α mediated genomic estrogen signaling is responsible for estrogen-stimulated cell proliferation and development of ER-positive breast cancer. However, accumulating evidence indicates that another estrogen signaling pathway, non-genomic or rapid estrogen signaling, also plays an important role in mitogenic estrogen signaling. Previously, our laboratory cloned a 36 kDa variant of ER-α, ER-α36, and found that ER-α36 is mainly expressed in the cytoplasm and at the plasma membrane. ER-α36 mediates rapid estrogen signaling and inhibits genomic estrogen signaling. In this review, we review and update the biological function of ER-α36 in ER-positive and -negative breast cancer, breast cancer stem/progenitor cells and tamoxifen resistance, potential interaction and cross-talk of ER-α36 with other ERs and growth factor receptors, and intracellular pathways of ER-α36-mediated rapid estrogen signaling. The potential function and underlying mechanism of ER-α in development of ER-positive breast cancer will also be discussed.
Collapse
Affiliation(s)
- Zhao-Yi Wang
- Department of Medical Microbiology & Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE 68178, USA.
| | - Li Yin
- Department of Medical Microbiology & Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE 68178, USA
| |
Collapse
|
25
|
The Cyclin D/Cyclin-dependent Kinase 4/6 Complex as a Target in the Treatment of Breast Cancer. CURRENT BREAST CANCER REPORTS 2015. [DOI: 10.1007/s12609-015-0189-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Mauro L, Pellegrino M, Giordano F, Ricchio E, Rizza P, De Amicis F, Catalano S, Bonofiglio D, Panno ML, Andò S. Estrogen receptor-α drives adiponectin effects on cyclin D1 expression in breast cancer cells. FASEB J 2015; 29:2150-60. [PMID: 25657113 DOI: 10.1096/fj.14-262808] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/13/2015] [Indexed: 11/11/2022]
Abstract
Obesity is a risk factor for breast cancer, largely due to altered expression of various adipocytokines. As it concerns adiponectin, there are not univocal results regarding its role in breast cancer occurrence and progression. Here, we demonstrate that in animals injected with human estrogen receptor (ER)-α-negative MDA-MB-231 cells pretreated with adiponectin (1 and 5 µg/ml), a significant reduction (60 and 40%, respectively) in tumor volume is observed, whereas an increased tumor growth (54 and 109%, respectively) is evidenced in the animals receiving human ER-α-positive MCF-7 cells. Moreover, cyclin D1 (CD1) mRNA and protein levels are decreased in MDA-MB-231 cells, whereas they are up-regulated in ER-α-positive cells by adiponectin. These findings fit with the opposite effects of adiponectin on CD1 promoter: 0.44- and 0.34-fold decrease in MDA-MB-231 cells and 0.63- and 0.95-fold increase in MCF-7 cells, treated with 1 and 5 µg/ml, respectively. Functional studies indicate that these effects are mediated by the specific protein 1 motif located in the CD1 promoter. In the absence of ER-α, the adiponectin-mediated down-regulation of CD1 involves the recruitment of corepressors. In the presence of ER-α, the adiponectin-induced expression of CD1 requires the involvement of an activator complex. In conclusion, we propose that a possible mechanism through which adiponectin differently affects breast cancer growth is the opposite modulation of CD1 levels accordingly to ER-α expression.
Collapse
Affiliation(s)
- Loredana Mauro
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Pellegrino
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Francesca Giordano
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Emilia Ricchio
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Pietro Rizza
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Francesca De Amicis
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Stefania Catalano
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Daniela Bonofiglio
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Maria Luisa Panno
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| | - Sebastiano Andò
- *Department of Pharmacy, Health and Nutritional Sciences, and Centro Sanitario, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
27
|
Liu Y, Ao X, Jia Z, Bai XY, Xu Z, Hu G, Jiang X, Chen M, Wu H. FOXK2 transcription factor suppresses ERα-positive breast cancer cell growth through down-regulating the stability of ERα via mechanism involving BRCA1/BARD1. Sci Rep 2015; 5:8796. [PMID: 25740706 PMCID: PMC4350111 DOI: 10.1038/srep08796] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/04/2015] [Indexed: 01/05/2023] Open
Abstract
Estrogen receptors (ERs) are critical regulators of breast cancer development. Identification of molecules that regulate the function of ERs may facilitate the development of more effective breast cancer treatment strategies. In this study, we showed that the forkhead transcription factor FOXK2 interacted with ERα, and inhibited ERα-regulated transcriptional activities by enhancing the ubiquitin-mediated degradation of ERα. This process involved the interaction between FOXK2 and BRCA1/BARD1, the E3 ubiquitin ligase of ERα. FOXK2 interacted with BARD1 and acted as a scaffold protein for BRCA1/BARD1 and ERα, leading to enhanced degradation of ERα, which eventually accounted for its decreased transcriptional activity. Consistent with these observations, overexpression of FOXK2 inhibited the transcriptional activity of ERα, decreased the transcription of ERα target genes, and suppressed the proliferation of ERα-positive breast cancer cells. In contract, knockdown of FOXK2 in MCF-7 cells promoted cell proliferation. However, when ERα was also knocked down, knockdown of FOXK2 had no effect on cell proliferation. These findings suggested that FOXK2 might act as a negative regulator of ERα, and its association with both ERα and BRCA1/BARD1 could lead to the down-regulation of ERα transcriptional activity, effectively regulating the function of ERα.
Collapse
Affiliation(s)
- Ying Liu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Xiang Ao
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Zhaojun Jia
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Xiao-Yan Bai
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Zhaowei Xu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Gaolei Hu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Xiao Jiang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Min Chen
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Huijian Wu
- 1] School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China [2] School of Life Science and Medicine, Dalian University of Technology, Panjin 114221, Liaoning, China
| |
Collapse
|
28
|
Karam M, Bièche I, Legay C, Vacher S, Auclair C, Ricort JM. Protein kinase D1 regulates ERα-positive breast cancer cell growth response to 17β-estradiol and contributes to poor prognosis in patients. J Cell Mol Med 2014; 18:2536-52. [PMID: 25287328 PMCID: PMC4302658 DOI: 10.1111/jcmm.12322] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/07/2014] [Indexed: 12/21/2022] Open
Abstract
About 70% of human breast cancers express and are dependent for growth on estrogen receptor α (ERα), and therefore are sensitive to antiestrogen therapies. However, progression to an advanced, more aggressive phenotype is associated with acquisition of resistance to antiestrogens and/or invasive potential. In this study, we highlight the role of the serine/threonine-protein kinase D1 (PKD1) in ERα-positive breast cancers. Growth of ERα-positive MCF-7 and MDA-MB-415 human breast cancer cells was assayed in adherent or anchorage-independent conditions in cells overexpressing or depleted for PKD1. PKD1 induces cell growth through both an ERα-dependent manner, by increasing ERα expression and cell sensitivity to 17β-estradiol, and an ERα-independent manner, by reducing cell dependence to estrogens and conferring partial resistance to antiestrogen ICI 182,780. PKD1 knockdown in MDA-MB-415 cells strongly reduced estrogen-dependent and independent invasion. Quantification of PKD1 mRNA levels in 38 cancerous and non-cancerous breast cell lines and in 152 ERα-positive breast tumours from patients treated with adjuvant tamoxifen showed an association between PKD1 and ERα expression in 76.3% (29/38) of the breast cell lines tested and a strong correlation between PKD1 expression and invasiveness (P < 0.0001). In tamoxifen-treated patients, tumours with high PKD1 mRNA levels (n = 77, 50.66%) were significantly associated with less metastasis-free survival than tumours with low PKD1 mRNA expression (n = 75, 49.34%; P = 0.031). Moreover, PKD1 mRNA levels are strongly positively associated with EGFR and vimentin levels (P < 0.0000001). Thus, our study defines PKD1 as a novel attractive prognostic factor and a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Manale Karam
- Laboratoire de Biologie et de Pharmacologie Appliquée, UMR 8113 CNRS, Ecole Normale Supérieure de Cachan, Cachan, France
| | | | | | | | | | | |
Collapse
|
29
|
Estrogen induces Vav1 expression in human breast cancer cells. PLoS One 2014; 9:e99052. [PMID: 24905577 PMCID: PMC4048212 DOI: 10.1371/journal.pone.0099052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 05/09/2014] [Indexed: 12/21/2022] Open
Abstract
Vav1, a guanine nucleotide exchange factor (GEF) for Rho family GTPases, is a hematopoietic protein involved in a variety of cellular events. In recent years, aberrant expression of Vav1 has been reported in non-hematopoietic cancers including human breast cancer. It remains to be answered how Vav1 is expressed and what Vav1 does in its non-resident tissues. In this study, we aimed to explore the mechanism for Vav1 expression in breast cancer cells in correlation with estrogen-ER pathway. We not only verified the ectopic expression of Vav1 in human breast cancer cell lines, but also observed that Vav1 expression was induced by 17β-estradiol (E2), a typical estrogen receptor (ER) ligand, in ER-positive cell lines. On the other hand, Tamoxifen, a selective estrogen receptor modulator (SERM), and ICI 182,780, an ER antagonist, suppressed the expression of Vav1. The estrogen receptor modulating Vav1 expression was identified to be α form, not β. Furthermore, treatment of E2 increased the transcription of vav1 gene by enhancing the promoter activity, though there was no recognizable estrogen response element (ERE). Nevertheless, two regions at the vav1 gene promoter were defined to be responsible for E2-induced activation of vav1 promoter. Chromatin immunoprecipitation (ChIP) and co-immunoprecipitation (Co-IP) analyses suggested that ERα might access to the vav1 promoter via interacting with transcription factors, c-Myb and ELF-1. Consequently, the enhanced expression of Vav1 led to the elevation of Cyclin D1 and the progression of cell cycle. The present study implies that estrogen-ER modulates the transcription and expression of Vav1, which may contribute to the proliferation of cancerous cells.
Collapse
|
30
|
Potential activity of fevicordin-A from Phaleria macrocarpa (Scheff) Boerl. seeds as estrogen receptor antagonist based on cytotoxicity and molecular modelling studies. Int J Mol Sci 2014; 15:7225-49. [PMID: 24776765 PMCID: PMC4057669 DOI: 10.3390/ijms15057225] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/10/2014] [Accepted: 01/15/2014] [Indexed: 12/28/2022] Open
Abstract
Fevicordin-A (FevA) isolated from Phaleria macrocarpa (Scheff) Boerl. seeds was evaluated for its potential anticancer activity by in vitro and in silico approaches. Cytotoxicity studies indicated that FevA was selective against cell lines of human breast adenocarcinoma (MCF-7) with an IC50 value of 6.4 μM. At 11.2 μM, FevA resulted in 76.8% cell death of T-47D human breast cancer cell lines. Critical pharmacophore features amongst human Estrogen Receptor-α (hERα) antagonists were conserved in FevA with regard to a hypothesis that they could make notable contributions to its pharmacological activity. The binding stability as well as the dynamic behavior of FevA towards the hERα receptor in agonist and antagonist binding sites were probed using molecular dynamics (MD) simulation approach. Analysis of MD simulation suggested that the tail of FevA was accountable for the repulsion of the C-terminal of Helix-11 (H11) in both agonist and antagonist receptor forms. The flexibility of loop-534 indicated the ability to disrupt the hydrogen bond zipper network between H3 and H11 in hERα. In addition, MM/GBSA calculation from the molecular dynamic simulations also revealed a stronger binding affinity of FevA in antagonistic action as compared to that of agonistic action. Collectively, both the experimental and computational results indicated that FevA has potential as a candidate for an anticancer agent, which is worth promoting for further preclinical evaluation.
Collapse
|
31
|
Mir M, Bergamaschi A, Katzenellenbogen BS, Popescu G. Highly sensitive quantitative imaging for monitoring single cancer cell growth kinetics and drug response. PLoS One 2014; 9:e89000. [PMID: 24558461 PMCID: PMC3928317 DOI: 10.1371/journal.pone.0089000] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/13/2014] [Indexed: 12/18/2022] Open
Abstract
The detection and treatment of cancer has advanced significantly in the past several decades, with important improvements in our understanding of the fundamental molecular and genetic basis of the disease. Despite these advancements, drug-screening methodologies have remained essentially unchanged since the introduction of the in vitro human cell line screen in 1990. Although the existing methods provide information on the overall effects of compounds on cell viability, they are restricted by bulk measurements, large sample sizes, and lack capability to measure proliferation kinetics at the individual cell level. To truly understand the nature of cancer cell proliferation and to develop personalized adjuvant therapies, there is a need for new methodologies that provide quantitative information to monitor the effect of drugs on cell growth as well as morphological and phenotypic changes at the single cell level. Here we show that a quantitative phase imaging modality known as spatial light interference microscopy (SLIM) addresses these needs and provides additional advantages over existing proliferation assays. We demonstrate these capabilities through measurements on the effects of the hormone estradiol and the antiestrogen ICI182,780 (Faslodex) on the growth of MCF-7 breast cancer cells. Along with providing information on changes in the overall growth, SLIM provides additional biologically relevant information. For example, we find that exposure to estradiol results in rapidly growing cells with lower dry mass than the control population. Subsequently blocking the estrogen receptor with ICI results in slower growing cells, with lower dry masses than the control. This ability to measure changes in growth kinetics in response to environmental conditions provides new insight on growth regulation mechanisms. Our results establish the capabilities of SLIM as an advanced drug screening technology that provides information on changes in proliferation kinetics at the cellular level with greater sensitivity than any existing method.
Collapse
Affiliation(s)
- Mustafa Mir
- Quantitative Light Imaging Laboratory, Department of Electrical and Computer Engineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Anna Bergamaschi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Benita S. Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Gabriel Popescu
- Quantitative Light Imaging Laboratory, Department of Electrical and Computer Engineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
32
|
Hah N, Kraus WL. Hormone-regulated transcriptomes: lessons learned from estrogen signaling pathways in breast cancer cells. Mol Cell Endocrinol 2014; 382:652-664. [PMID: 23810978 PMCID: PMC3844033 DOI: 10.1016/j.mce.2013.06.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/15/2013] [Accepted: 06/17/2013] [Indexed: 12/21/2022]
Abstract
Recent rapid advances in next generation sequencing technologies have expanded our understanding of steroid hormone signaling to a genome-wide level. In this review, we discuss the use of a novel genomic approach, global nuclear run-on coupled with massively parallel sequencing (GRO-seq), to explore new facets of the steroid hormone-regulated transcriptome, especially estrogen responses in breast cancer cells. GRO-seq is a high throughput sequencing method adapted from conventional nuclear run-on methodologies, which is used to obtain a map of the position and orientation of all transcriptionally engaged RNA polymerases across the genome with extremely high spatial resolution. GRO-seq, which is an excellent tool for examining transcriptional responses to extracellular stimuli, has been used to comprehensively assay the effects of estrogen signaling on the transcriptome of ERα-positive MCF-7 human breast cancer cells. These studies have revealed new details about estrogen-dependent transcriptional regulation, including effects on transcription by all three RNA polymerases, complex transcriptional dynamics in response to estrogen signaling, and identification novel, unannotated non-coding RNAs. Collectively, these studies have been useful in discerning the molecular logic of the estrogen-regulated mitogenic response.
Collapse
Affiliation(s)
- Nasun Hah
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, United States.
| | - W Lee Kraus
- The Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States; Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
33
|
Abstract
Germline mutations of human breast cancer-associated gene 1 (BRCA1) predispose women to breast and ovarian cancers. In mice, over 20 distinct mutations, including null, hypomorphic, isoform, conditional, and point mutations, have been created to study functions of Brca1 in mammary development and tumorigenesis. Analyses using these mutant mice have yielded an enormous amount of information that greatly facilitates our understanding of the gender- and tissue-specific tumor suppressor functions of BRCA1, as well as enriches our insights into applying these preclinical models of disease to breast cancer research. Here, we review features of these mutant mice and their applications to cancer prevention and therapeutic treatment.
Collapse
|
34
|
Meher AP, Joshi AA, Joshi SR. Preconceptional omega-3 fatty acid supplementation on a micronutrient-deficient diet improves the reproductive cycle in Wistar rats. Reprod Fertil Dev 2013; 25:1085-94. [DOI: 10.1071/rd12210] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 10/04/2012] [Indexed: 11/23/2022] Open
Abstract
Folic acid and vitamin B12 deficiencies are associated with high reproductive risks ranging from infertility to fetal structural defects. The aim of the present study was to examine the effects of preconceptional omega-3 fatty acid supplementation (eicosapentaenoic acid and docosahexaenoic acid) to a micronutrient-deficient diet on the reproductive cycle in Wistar rats. Female rats were divided into five groups from birth and throughout pregnancy: a control group, a folic acid-deficient (FD) group, a vitamin B12-deficient (BD) group, a folic acid-deficient + omega-3 fatty acid-supplemented (FDO) group and a vitamin B12 deficient + omega-3 fatty acid-supplemented (BDO) group. Dams were killed on gestation Day 20 and their ovaries and mammary glands were dissected out and subjected to histological examination. Maternal micronutrient deficiency (FD and BD groups) resulted in an abnormal oestrous cycle (P < 0.001), whereas omega-3 fatty acid supplementation (FDO and BDO groups) restored the oestrous cycle to normal. There were fewer corpora lutea in the ovaries of FD rats compared with controls. In addition, rats in both the FD and BD groups exhibited an absence of lactating ducts in their mammary glands compared with controls. The findings of the present study indicate, for the first time, that maternal micronutrient deficiency affects the oestrous cycle and morphology of the ovary and mammary glands. Omega-3 fatty acid supplementation ameliorated these effects. This may have implications for infertility and pregnancy outcomes.
Collapse
|
35
|
Park E, Gong EY, Romanelli MG, Lee K. Suppression of estrogen receptor-alpha transactivation by thyroid transcription factor-2 in breast cancer cells. Biochem Biophys Res Commun 2012; 421:532-7. [PMID: 22521644 DOI: 10.1016/j.bbrc.2012.04.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 04/09/2012] [Indexed: 10/28/2022]
Abstract
Estrogen receptors (ERs), which mediate estrogen actions, regulate cell growth and differentiation of a variety of normal tissues and hormone-responsive tumors through interaction with cellular factors. In this study, we show that thyroid transcription factor-2 (TTF-2) is expressed in mammary gland and acts as ERα co-repressor. TTF-2 inhibited ERα transactivation in a dose-dependent manner in MCF-7 breast cancer cells. In addition, TTF-2 directly bound to and formed a complex with ERα, colocalizing with ERα in the nucleus. In MCF-7/TTF-2 stable cell lines, TTF-2 repressed the expression of endogenous ERα target genes such as pS2 and cyclin D1 by interrupting ERα binding to target promoters and also significantly decreased cell proliferation. Taken together, these data suggest that TTF-2 may modulate the function of ERα as a corepressor and play a role in ER-dependent proliferation of mammary cells.
Collapse
Affiliation(s)
- Eunsook Park
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | |
Collapse
|
36
|
Abstract
Cancer cells silence autosomal tumor suppressor genes by Knudson's two-hit mechanism in which loss-of-function mutations and then loss of heterozygosity occur at the tumor suppressor gene loci. However, the identification of X-linked tumor suppressor genes has challenged the traditional theory of 'two-hit inactivation' in tumor suppressor genes, introducing the novel concept that a single genetic hit can cause loss of tumor suppressor function. The mechanism through which these genes are silenced in human cancer is unclear, but elucidating the details will greatly enhance our understanding of the pathogenesis of human cancer. Here, we review the identification of X-linked tumor suppressor genes and discuss the potential mechanisms of their inactivation. In addition, we also discuss how the identification of X-linked tumor suppressor genes can potentially lead to new approaches in cancer therapy.
Collapse
Affiliation(s)
- Runhua Liu
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Genetics, School of Medicine, University of Alabama at Birmingham and Comprehensive Cancer Center, Birmingham, AL, USA
| | - Mandy Kain
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Lizhong Wang
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Genetics, School of Medicine, University of Alabama at Birmingham and Comprehensive Cancer Center, Birmingham, AL, USA
| |
Collapse
|
37
|
Sun M, Isaacs GD, Hah N, Heldring N, Fogarty EA, Kraus WL. Estrogen regulates JNK1 genomic localization to control gene expression and cell growth in breast cancer cells. Mol Endocrinol 2012; 26:736-47. [PMID: 22446103 DOI: 10.1210/me.2011-1158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Steroid hormone and MAPK signaling pathways functionally intersect, but the molecular mechanisms of this cross talk are unclear. Here, we demonstrate a functional convergence of the estrogen and c-Jun N-terminal kinase 1 (JNK1) signaling pathways at the genomic level in breast cancer cells. We find that JNK1 binds to many promoters across the genome. Although most of the JNK1-binding sites are constitutive, a subset is estrogen regulated (either induced on inhibited). At the estrogen-induced sites, estrogen receptor (ER)α is required for the binding of JNK1 by promoting its recruitment to estrogen response elements or other classes of DNA elements through a tethering mechanism, which in some cases involves activating protein-1. At estrogen-regulated promoters, JNK1 functions as a transcriptional coregulator of ERα in a manner that is dependent on its kinase activity. The convergence of ERα and JNK1 at target gene promoters regulates estrogen-dependent gene expression outcomes, as well as downstream estrogen-dependent cell growth responses. Analysis of existing gene expression profiles from breast cancer biopsies suggests a role for functional interplay between ERα and JNK1 in the progression and clinical outcome of breast cancers.
Collapse
Affiliation(s)
- Miao Sun
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8511, USA
| | | | | | | | | | | |
Collapse
|
38
|
Steroidal Metabolites Transformed by Marchantia polymorpha Cultures Block Breast Cancer Estrogen Biosynthesis. Cell Biochem Biophys 2012; 63:85-96. [DOI: 10.1007/s12013-012-9343-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Miermont AM, Cabrera MC, Frech SM, Nakles RE, Diaz-Cruz ES, Shiffert MT, Furth PA. Association of Over-Expressed Estrogen Receptor Alpha with Development of Tamoxifen Resistant Hyperplasia and Adenocarcinomas in Genetically Engineered Mice. ACTA ACUST UNITED AC 2012; Suppl 12. [PMID: 24575359 PMCID: PMC3932557 DOI: 10.4172/2161-0940.s12-001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Estrogen receptor alpha (ERα) and cyclin D1 are frequently co-expressed in human breast cancer. Some, but not all, studies link tamoxifen resistance to co-expression of cyclin D1 and ERα. In mice over-expression of either cyclin D1 or ERα in mammary epithelial cells is sufficient to induce mammary hyperplasia. Cyclin D1 over-expression in mice leads to mammary adenocarcinoma associated with activated estrogen signaling pathways. ERα over-expression in mice leads to mammary hyperplasia and cancer. Significantly, disease development in these mice is abrogated by loss of cyclin D1. METHODS Genetically engineered mouse models were used to determine whether or not ERα over-expression demonstrated cooperativity with cyclin D1 over-expression in cancer development, reaction to the chemical carcinogen DMBA, or tamoxifen response. RESULTS Adding ERα over-expression to cyclin D1 over-expression increased the prevalence of hyperplasia but not cancer. Single dose DMBA exposure did not increase cancer prevalence in any of the genotypes although cyclin D1 over-expressing mice demonstrated a significant increase in hyperplasia. Tamoxifen treatment was initiated at both young and older ages to test for genotype-specific differences in response. Although normal ductal structures regressed in all genotypes at both younger and older ages, tamoxifen did not significantly reduce the prevalence of either hyperplasia or cancer in any of the genotypes. All of the cancers that developed were hormone receptor positive, including those that developed on tamoxifen, and all showed expression of nuclear-localized cyclin D1. In summary, development of tamoxifen resistant hyperplasia and cancer was associated with expression of ERα and cyclin D1. CONCLUSION These preclinical models will be useful to test strategies for overcoming tamoxifen resistance, perhaps by simultaneously targeting cell cycle regulatory pathways associated with cyclin D1.
Collapse
Affiliation(s)
- Anne M Miermont
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Marina Carla Cabrera
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Silvina M Frech
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Rebecca E Nakles
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Edgar S Diaz-Cruz
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Maddalena Tilli Shiffert
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA ; Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Priscilla A Furth
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA ; Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA ; Department of Nanobiomedical Science and WCU Research Center of Nanobiomedical Science, Dankook University, Chungnam 330-714, Korea
| |
Collapse
|
40
|
Abstract
MYC is a key regulator of cell growth, proliferation, metabolism, differentiation, and apoptosis. MYC deregulation contributes to breast cancer development and progression and is associated with poor outcomes. Multiple mechanisms are involved in MYC deregulation in breast cancer, including gene amplification, transcriptional regulation, and mRNA and protein stabilization, which correlate with loss of tumor suppressors and activation of oncogenic pathways. The heterogeneity in breast cancer is increasingly recognized. Breast cancer has been classified into 5 or more subtypes based on gene expression profiles, and each subtype has distinct biological features and clinical outcomes. Among these subtypes, basal-like tumor is associated with a poor prognosis and has a lack of therapeutic targets. MYC is overexpressed in the basal-like subtype and may serve as a target for this aggressive subtype of breast cancer. Tumor suppressor BRCA1 inhibits MYC's transcriptional and transforming activity. Loss of BRCA1 with MYC overexpression leads to the development of breast cancer-especially, basal-like breast cancer. As a downstream effector of estrogen receptor and epidermal growth factor receptor family pathways, MYC may contribute to resistance to adjuvant therapy. Targeting MYC-regulated pathways in combination with inhibitors of other oncogenic pathways may provide a promising therapeutic strategy for breast cancer, the basal-like subtype in particular.
Collapse
Affiliation(s)
- Jinhua Xu
- Center for Clinical Cancer Genetics, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
41
|
Lange CA, Yee D. Killing the second messenger: targeting loss of cell cycle control in endocrine-resistant breast cancer. Endocr Relat Cancer 2011; 18:C19-24. [PMID: 21613412 PMCID: PMC3924782 DOI: 10.1530/erc-11-0112] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The majority (∼70%) of breast cancers are steroid hormone receptor (SR) positive at the time of diagnosis. Endocrine therapies that target estrogen receptor α (ERα) action (tamoxifen, toremifene, fulvestrant) or estrogen synthesis (aromatase inhibitors: letrozole, anastrozole, exemestane; or ovarian suppression) are a clinical mainstay. However, up to 50% of SR+ breast cancers exhibit de novo or acquired resistance to these clinical interventions. Mechanisms of resistance to endocrine therapies often include upregulation and/or activation of signal transduction pathways that input to cell cycle regulation. Cyclin D1, the regulatory subunit of cyclin-dependent protein kinases four and six (CDK4/6) serves as a convergence point for multiple signaling pathways. In a recent paper entitled 'Therapeutically Activating Retinoblastoma (RB): Reestablishing Cell Cycle Control in Endocrine Therapy-Resistant Breast Cancer', Thangavel et al. reported maintenance of cyclin D1 expression and RB phosphorylation in the face of ER ablation in multiple breast cancer cell line models of endocrine resistance. RB-dysfunction defined a unique gene signature that was associated with luminal B-type breast cancer and predictive of poor response to endocrine therapies. Notably, a new CDK4/6 inhibitor (PD-0332991) was capable of inducing growth arrest by a mechanism that was most consistent with cellular senescence. In this review, these findings are discussed in the context of SRs as important mediators of cell cycle progression, and the frequent loss of cell cycle checkpoint control that typifies breast cancer progression. These studies provide renewed hope of effectively stabilizing endocrine-resistant breast cancers using available complementary (to endocrine-based therapies) cytostatic agents in the form of CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Carol A Lange
- Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, 420 Delaware Street South East, MMC 806, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
42
|
Huang Y, Li X, Muyan M. Estrogen receptors similarly mediate the effects of 17β-estradiol on cellular responses but differ in their potencies. Endocrine 2011; 39:48-61. [PMID: 21069581 PMCID: PMC3683410 DOI: 10.1007/s12020-010-9411-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 10/04/2010] [Indexed: 02/06/2023]
Abstract
17β-estradiol (E2), as the main circulating estrogen hormone, plays critical roles in the physiology and pathophysiology of various tissues. The E2 information is primarily conveyed by the transcription factors, estrogen receptors (ERs) α and β. ERs share similar structural and functional features. Experimental studies indicate that upon binding to E2, ERs directly or indirectly interact with DNA and regulate gene expressions with ERα being more potent transregulator than ERβ. However, studies also showed that ERβ induces alterations in phenotypic features of cancer cell lines independent of E2. These observations suggested that the manner in which the unliganded ERβ induces phenotypic alterations in cancer cell models differs from that of ERα. Studies demonstrated that while requiring E2 for function at low levels of synthesis, the unliganded ERα at augmented concentrations modulates gene expressions and cellular growth. We, therefore, anticipated that heightened levels of ERβ synthesis could similarly circumvent the dependency on E2 leading to gene transcriptions and cellular proliferation. To test this prediction, we used adenovirus-infected cancer cell lines in which ERs were shown to induce genomic and cellular responses. We found that while ERβ at low levels of synthesis was dependent upon E2 for function, the receptor at high levels regulated gene expression and cellular proliferation independent of E2. We then addressed whether ERs at comparable levels that require E2 for function differentially alter gene expressions and cellular responses. We found that ERs mediate the effects of E2 on gene expression, cellular proliferation, apoptosis, and motility with an overlapping pattern. However, ERα was more potent regulator than ERβ in inducing cellular responses. Our results suggest that differences in potencies to regulate the expression of genes are a critical feature of the ER subtypes in mediating E2 signaling in cancer cell lines.
Collapse
Affiliation(s)
- Yanfang Huang
- Department of Biochemistry and Biophysics, University of Rochester Medical School, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
43
|
Tedesco I, Russo M, Russo GL. Commentary on ‘Resveratrol commonly displays hormesis: Occurrence and biomedical significance’. Hum Exp Toxicol 2010; 29:1029-31. [DOI: 10.1177/0960327110383628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The review by Calabrese et al. describes the hormetic dose responses induced by phytoalexin resveratrol in a wide range of biological models. We agree and support the authors’ strategy to present an impressive number of experiments furnished with an exhaustive bibliography to emphasize that ‘many effects induced by resveratrol are dependent on dose and that opposite effects occur at low and high doses, being indicative of a hormetic dose response.’ We also highly appreciate the holistic view of the hormetic behavior of resveratrol provided by the authors spanning from tumor and non-tumor cell lines to human and parasitic diseases. In our comments, we touched minor points whose discussion would have strengthened the work of Calabrese, such as contradictions on the role of resveratrol in the ‘French Paradox,’ its effect on aromatase activity, glutamate cysteine ligase expression and glutathione levels. Overall, we encourage colleagues working in this field to read the present review and consider its relevant biological implications. The vision of Calabrese et al. is far too important to be ignored.
Collapse
Affiliation(s)
- Idolo Tedesco
- Institute of Food Sciences, National Research Council, Avellino, Italy,
| | - Maria Russo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Gian Luigi Russo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| |
Collapse
|
44
|
Ellison-Zelski SJ, Alarid ET. Maximum growth and survival of estrogen receptor-alpha positive breast cancer cells requires the Sin3A transcriptional repressor. Mol Cancer 2010; 9:263. [PMID: 20920219 PMCID: PMC2956731 DOI: 10.1186/1476-4598-9-263] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 09/29/2010] [Indexed: 11/16/2022] Open
Abstract
Background Sin3A is an evolutionarily conserved transcriptional repressor which regulates gene expression as part of the multi-protein Sin3 repressive complex. It functions as a scaffold upon which proteins with enzymatic activity dock, including chromatin modifying histone deacetylases. Although regulation of transcription by Sin3A has been studied in detail, little is understood about the function of Sin3A in cancer cells. We previously showed that Sin3A is expressed in breast cancer cells and is a repressor of estrogen receptor-alpha (ERα, ESR1) gene expression. Here, we expand our previous studies to elucidate the function of Sin3A in the control of gene expression and growth of breast cancer cells. Results Analysis of gene expression following knockdown of Sin3A revealed changes in both basal and regulated gene transcription. Genes of known importance in breast cancer and estrogen signaling, including ERBB2, PGR, MYC, CLU, and NCOA2, were among those identified as Sin3A-responsive. The mechanism of Sin3A action varied among genes and was found to be mediated through both HDAC1/2 -dependent and -independent activities. Loss of Sin3A inhibited breast cancer cell growth by increasing apoptosis without affecting cell cycle progression. Analysis of both ERα-positive and ERα-negative cell lines revealed that the effects of Sin3A on growth were cell-type specific, as Sin3A expression promoted maximum growth of only the ERα-positive cells, and, notably, Sin3A protein itself was increased by estrogen. Further gene expression experiments revealed that Sin3A repressed expression of key apoptotic genes, including TRAIL, TRAILR1, CASP10, and APAF1, in ERα-positive, but not ERα-negative, cell lines, which could provide a mechanistic explanation for cell-type differences in growth. Conclusions This study identifies Sin3A as a regulator of gene expression, survival, and growth in ERα-positive breast cancer cells. Sin3A regulates the transcription of genes involved in breast cancer and apoptosis and acts through multiple mechanisms not limited to histone deacetylase function. These findings reveal previously undescribed functions of Sin3A in breast cancer and provide evidence for an important role of this transcriptional repressor in ERα-positive tumor cell growth.
Collapse
|
45
|
Nair BC, Nair SS, Chakravarty D, Challa R, Manavathi B, Yew PR, Kumar R, Tekmal RR, Vadlamudi RK. Cyclin-dependent kinase-mediated phosphorylation plays a critical role in the oncogenic functions of PELP1. Cancer Res 2010; 70:7166-75. [PMID: 20807815 DOI: 10.1158/0008-5472.can-10-0628] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Estrogen receptor (ER) signaling plays an important role in breast cancer progression, and ER functions are influenced by coregulatory proteins. PELP1 (proline-, glutamic acid-, and leucine-rich protein 1) is a nuclear receptor coregulator that plays an important role in ER signaling. Its expression is deregulated in hormonal cancers. We identified PELP1 as a novel cyclin-dependent kinase (CDK) substrate. Using site-directed mutagenesis and in vitro kinase assays, we identified Ser(477) and Ser(991) of PELP1 as CDK phosphorylation sites. Using the PELP1 Ser(991) phospho-specific antibody, we show that PELP1 is hyperphosphorylated during cell cycle progression. Model cells stably expressing the PELP1 mutant that lack CDK sites had defects in estradiol (E2)-mediated cell cycle progression and significantly affected PELP1-mediated oncogenic functions in vivo. Mechanistic studies showed that PELP1 modulates transcription factor E2F1 transactivation functions, that PELP1 is recruited to pRb/E2F target genes, and that PELP1 facilitates ER signaling cross talk with cell cycle machinery. We conclude that PELP1 is a novel substrate of interphase CDKs and that its phosphorylation is important for the proper function of PELP1 in modulating hormone-driven cell cycle progression and also for optimal E2F transactivation function. Because the expression of both PELP1 and CDKs is deregulated in breast tumors, CDK-PELP1 interactions will have implications in breast cancer progression.
Collapse
Affiliation(s)
- Binoj C Nair
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Floyd Curl Drive, Mail Code 7836, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dalvai M, Bystricky K. Cell cycle and anti-estrogen effects synergize to regulate cell proliferation and ER target gene expression. PLoS One 2010; 5:e11011. [PMID: 20543978 PMCID: PMC2882356 DOI: 10.1371/journal.pone.0011011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 05/06/2010] [Indexed: 12/03/2022] Open
Abstract
Antiestrogens are designed to antagonize hormone induced proliferation and ERalpha target gene expression in mammary tumor cells. Commonly used drugs such as OH-Tamoxifen and ICI 182780 (Fulvestrant) block cell cycle progression in G0/G1. Inversely, the effect of cell cycle stage on ER regulated gene expression has not been tested directly. We show that in ERalpha-positive breast cancer cells (MCF-7) the estrogen receptor gene and downstream target genes are cell cycle regulated with expression levels varying as much as three-fold between phases of the cell cycle. Steroid free culture conditions commonly used to assess the effect of hormones or antiestrogens on gene expression also block MCF-7 cells in G1-phase when several ERalpha target genes are overexpressed. Thus, cell cycle effects have to be taken into account when analyzing the impact of hormonal treatments on gene transcription. We found that antiestrogens repress transcription of several ERalpha target genes specifically in S phase. This observation corroborates the more rapid and strong impact of antiestrogen treatments on cell proliferation in thymidine, hydroxyurea or aphidicolin arrested cells and correlates with an increase of apoptosis compared to similar treatments in lovastatin or nocodazol treated cells. Hence, cell cycle effects synergize with the action of antiestrogens. An interesting therapeutic perspective could be to enhance the action of anti-estrogens by associating hormone-therapy with specific cell cycle drugs.
Collapse
Affiliation(s)
- Mathieu Dalvai
- Université de Toulouse, UPS, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
- CNRS, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
| | - Kerstin Bystricky
- Université de Toulouse, UPS, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
- CNRS, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
| |
Collapse
|
47
|
Urbinati G, Audisio D, Marsaud V, Plassat V, Arpicco S, Sola B, Fattal E, Renoir JM. Therapeutic potential of new 4-hydroxy-tamoxifen-loaded pH-gradient liposomes in a multiple myeloma experimental model. Pharm Res 2009; 27:327-39. [PMID: 20033476 DOI: 10.1007/s11095-009-0023-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 12/01/2009] [Indexed: 11/28/2022]
Abstract
PURPOSE To determine the better liposomal formulation incorporating the active metabolite of tamoxifen, 4-hydroxy-tamoxifen (4HT) and the biological impact of 4HT-pH-gradient liposomes on response to in vivo treatment. METHODS Several pegylated liposomes were formulated by varying the composition of lipids, increasing external pH from 7.4 to 9.0 and doubling the lipid concentration. Dipalmitoylphosphatidylcholine / cholesterol / distearoylphosphoethanolamine poly(ethylene)glycol liposomes (DL-9 liposomes) were chosen for their physico-chemical properties. Toxicity and release kinetics were assessed in breast cancer MCF-7 as well as in multiple myeloma (MM) cells. In vivo antitumor activity and bio-distribution were measured in the RPMI8226 MM model. RESULTS Compared to conventional non-pH-gradient liposomes, 4HT-DL-9 liposomes resulted in concentration of up to 1 mM 4HT, greater stability, relative toxicity and slow 4HT release. Intravenous injections of 4HT-DL-9 liposomes at 4 mg/kg/week blocked MM tumor growth. Ki67 and CD34 labeling decreased in treated tumors, concomitantly with increase of activated caspase-3 supporting a cell proliferation arrest, a decrease of tumor vasculature and the induction of tumor cell death. CONCLUSION This antitumor effect was assumed to be the result of a modified biodistribution of 4HT once trapped in DL-9 liposomes. Such 4HT-containing pH-gradient Stealth nanocarriers could be helpful for MM treatment.
Collapse
Affiliation(s)
- Giorgia Urbinati
- CNRS, UMR 8612, Physico-Chimie, Pharmacotechnie, Biopharmacie, Laboratoire Pharmacologie Cellulaire et Moléculaire des Anticancéreux, Faculté de Pharmacie, 5 rue J.B. Clément, Châtenay-Malabry, F-92296, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Chan MP, Chan MM, Tahan SR. Melanocytic nevi in pregnancy: histologic features and Ki-67 proliferation index. J Cutan Pathol 2009; 37:843-51. [PMID: 20015190 DOI: 10.1111/j.1600-0560.2009.01491.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Changes in the clinical appearance of benign dermal nevi during pregnancy may be concerning for malignant transformation. Because the hormonal milieu of pregnancy has not proven to alter their banal behavior, histologic characterization is needed to prevent over-diagnosis and unnecessary treatment. METHODS Dermal nevi excised from pregnant women (n = 16) were compared with nevi from location- and aged-matched control patients (n = 15). Histologic features and Ki-67 proliferation index were evaluated. RESULTS Nevi in pregnancy were more likely to have dermal mitotic figures (62.5% vs. 13.3%, p = 0.028) and higher mitotic rates (1.44 vs. 0.20 mitoses/mm(2), p = 0.0027) than control nevi. A distinctive histologic entity, termed superficial micronodules of pregnancy (SMOPs), was observed more frequently in the nevi of pregnancy (81.3% vs. 26.7%, p = 0.040), and showed consistent immunoreactivity for HMB45. There was a trend toward higher Ki-67 proliferation index in the nevi of pregnancy (3.0% vs. 1.0%, p = 0.073). Prominent multinucleated melanocytes were seen only in controls. There was no significant difference in pigmentation or irritation changes between groups. CONCLUSIONS Dermal nevi removed during pregnancy show characteristic histologic features including increased dermal mitoses, superficial micronodules of pregnancy (SMOPs), and trend toward increased Ki-67 proliferation index.
Collapse
Affiliation(s)
- May P Chan
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | |
Collapse
|
49
|
Green AR, Young P, Krivinskas S, Rakha EA, Claire Paish E, Powe DG, Ellis IO. The expression of ERalpha, ERbeta and PR in lobular carcinoma in situ of the breast determined using laser microdissection and real-time PCR. Histopathology 2009; 54:419-27. [PMID: 19309393 DOI: 10.1111/j.1365-2559.2009.03233.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS To determine the status of oestrogen receptor (ER) subtypes (ERalpha and ERbeta) in lobular carcinoma in situ (LCIS) of the breast. METHODS AND RESULTS Forty-seven cases of LCIS and six cases with normal breast lobules were subjected to immunohistochemistry and evaluated for ERalpha, ERbeta and progesterone receptor (PR) expression. mRNA for ERalpha, ERbeta1 and ERbeta2 were quantified in LCIS and normal lobules using laser microdissection coupled with real-time polymerase chain reaction. LCIS showed a higher level of steroid receptor protein expression than normal lobules. There was no difference in ERbeta1 gene or ERbeta protein expression between normal lobules, pure LCIS, or LCIS associated with invasive breast cancer. No significant difference in expression of either ERalpha or ERbeta was found between pure LCIS and LCIS associated with invasive cancer. However, PR was significantly lower in those cases of LCIS with associated invasive than in those without synchronous invasive disease. CONCLUSIONS Increased expression of steroid receptors in LCIS suggests their possible role in the biology of LCIS and, for PR, could influence the predisposition of women diagnosed with LCIS to develop invasive breast carcinoma.
Collapse
Affiliation(s)
- Andrew R Green
- Division of Pathology, School of Molecular Medical Sciences, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, UK.
| | | | | | | | | | | | | |
Collapse
|
50
|
Chen Y, Guggisberg N, Jorda M, Gonzalez-Angulo A, Hennessy B, Mills GB, Tan CK, Slingerland JM. Combined Src and aromatase inhibition impairs human breast cancer growth in vivo and bypass pathways are activated in AZD0530-resistant tumors. Clin Cancer Res 2009; 15:3396-405. [PMID: 19451593 DOI: 10.1158/1078-0432.ccr-08-3127] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Antiestrogens are used to treat estrogen receptor (ER)-alpha-positive breast cancers and cause a p27-dependent G(1) arrest. Estrogen-bound ER recruits Src to mediate proteolysis of p27 and drive cell proliferation. Here, we tested the antitumor efficacy of combined Src and aromatase inhibition for ER-positive breast cancer. EXPERIMENTAL DESIGN Antiproliferative effects of the aromatase inhibitor, anastrozole, and Src inhibitor, AZD0530, alone or in combination were tested in vitro and in vivo on aromatase-transfected MCF-7Arom5 xenografts. Xenografts were analyzed by immunohistochemistry and proteomic analysis to identify potential biomarkers of drug response and resistance. RESULTS AZD0530 and anastrozole together increased p27 and caused greater G(1) cell cycle arrest than either drug alone. AZD0530 monotherapy initially retarded xenograft growth in vivo, but drug resistance rapidly emerged. Combined anastrozole/AZD0530 reduced drug resistance and showed greater antitumor efficacy in vivo with greater Src and epidermal growth factor receptor inhibition and a greater increase in p27 and reduction of Ki-67 than either drug alone, supporting further evaluation of these putative predictors of response to combined Src/aromatase inhibition in vivo. Anastrozole alone stimulated Src activity both in vitro and in vivo. AZD0530-resistant tumors showed activation of bypass pathways including MEK and phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin, raising the possibility that MEK, mammalian target of rapamycin (mTOR), or PI3K inhibitors may augment Src inhibitor efficacy. CONCLUSIONS These data support clinical investigation of anastrozole-AZD0530 therapy for postmenopausal ER-positive breast cancer. Loss of p27 and increased Ki-67 may predict response and further clinical studies should evaluate for activation of bypass pathways including MEK and PI3K pathways during Src inhibitor therapy.
Collapse
Affiliation(s)
- Yi Chen
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | | | |
Collapse
|