1
|
McGrath MK, Abolhassani A, Guy L, Elshazly AM, Barrett JT, Mivechi NF, Gewirtz DA, Schoenlein PV. Autophagy and senescence facilitate the development of antiestrogen resistance in ER positive breast cancer. Front Endocrinol (Lausanne) 2024; 15:1298423. [PMID: 38567308 PMCID: PMC10986181 DOI: 10.3389/fendo.2024.1298423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Estrogen receptor positive (ER+) breast cancer is the most common breast cancer diagnosed annually in the US with endocrine-based therapy as standard-of-care for this breast cancer subtype. Endocrine therapy includes treatment with antiestrogens, such as selective estrogen receptor modulators (SERMs), selective estrogen receptor downregulators (SERDs), and aromatase inhibitors (AIs). Despite the appreciable remission achievable with these treatments, a substantial cohort of women will experience primary tumor recurrence, subsequent metastasis, and eventual death due to their disease. In these cases, the breast cancer cells have become resistant to endocrine therapy, with endocrine resistance identified as the major obstacle to the medical oncologist and patient. To combat the development of endocrine resistance, the treatment options for ER+, HER2 negative breast cancer now include CDK4/6 inhibitors used as adjuvants to antiestrogen treatment. In addition to the dysregulated activity of CDK4/6, a plethora of genetic and biochemical mechanisms have been identified that contribute to endocrine resistance. These mechanisms, which have been identified by lab-based studies utilizing appropriate cell and animal models of breast cancer, and by clinical studies in which gene expression profiles identify candidate endocrine resistance genes, are the subject of this review. In addition, we will discuss molecular targeting strategies now utilized in conjunction with endocrine therapy to combat the development of resistance or target resistant breast cancer cells. Of approaches currently being explored to improve endocrine treatment efficacy and patient outcome, two adaptive cell survival mechanisms, autophagy, and "reversible" senescence, are considered molecular targets. Autophagy and/or senescence induction have been identified in response to most antiestrogen treatments currently being used for the treatment of ER+ breast cancer and are often induced in response to CDK4/6 inhibitors. Unfortunately, effective strategies to target these cell survival pathways have not yet been successfully developed. Thus, there is an urgent need for the continued interrogation of autophagy and "reversible" senescence in clinically relevant breast cancer models with the long-term goal of identifying new molecular targets for improved treatment of ER+ breast cancer.
Collapse
Affiliation(s)
- Michael K. McGrath
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ali Abolhassani
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Luke Guy
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ahmed M. Elshazly
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - John T. Barrett
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Radiation Oncology, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Nahid F. Mivechi
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Radiation Oncology, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - David A. Gewirtz
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Patricia V. Schoenlein
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
- Department of Cellular Biology & Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
2
|
Nakadai T, Yang L, Kumegawa K, Maruyama R. Estrogen receptor α K303R mutation reorganizes its binding to forkhead box protein A1 regions and induces chromatin opening. Mol Biol Rep 2023; 50:1209-1220. [PMID: 36436079 PMCID: PMC9889408 DOI: 10.1007/s11033-022-08089-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Estrogen receptor alpha (ERα) is a frequently mutated gene in breast cancer (BC). While many studies have investigated molecular dysregulation by hotspot mutations at Y537 and D538, which exhibit an estrogen-independent constitutively active phenotype, the functional abnormalities of other mutations remain obscure. The K303R mutation in primary invasive BC has been implicated with endocrine resistance, tumor size, and lymph node positivity. However, the impact of the K303R mutation on the cell epigenome is yet unknown. METHODS AND RESULTS We introduced the K303R ERα mutant in ERα-negative MDA-MB-453 cells to monitor ERα-dependent transactivation and to perform epigenomic analyses. ATAC-seq and ChIP-Seq analyses indicated that both wild-type (WT) and the K303R mutant associated with Forkhead box (Fox) protein family motif regions at similar rates, even without an ERα-binding sequence, but only the K303R mutant induced chromatin opening at those regions. Biochemical analyses demonstrated that the WT and the K303R mutant can be tethered on DNA by FoxA1 indirectly, but only the K303R/FoxA1/DNA complex can induce associations with the nuclear receptor cofactor 2 (NCOA2). CONCLUSIONS These findings suggest that the K303R mutant induces chromatin opening at the Fox binding region through the FoxA1-dependent associations of the K303R mutant to NCOA2 and then probably disrupts the regulation of Fox-target genes, resulting in K303R-related BC events.
Collapse
Affiliation(s)
- Tomoyoshi Nakadai
- Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Liying Yang
- Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Kohei Kumegawa
- Cancer Cell Diversity Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Reo Maruyama
- Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
- Cancer Cell Diversity Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
3
|
Identification of a human estrogen receptor α tetrapeptidic fragment with dual antiproliferative and anti-nociceptive action. Sci Rep 2023; 13:1326. [PMID: 36693877 PMCID: PMC9873809 DOI: 10.1038/s41598-023-28062-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
The synthetic peptide ERα17p (sequence: PLMIKRSKKNSLALSLT), which corresponds to the 295-311 region of the human estrogen receptor α (ERα), induces apoptosis in breast cancer cells. In mice and at low doses, it promotes not only the decrease of the size of xenografted triple-negative human breast tumors, but also anti-inflammatory and anti-nociceptive effects. Recently, we have shown that these effects were due to its interaction with the seven-transmembrane G protein-coupled estrogen receptor GPER. Following modeling studies, the C-terminus of this peptide (sequence: NSLALSLT) remains compacted at the entrance of the GPER ligand-binding pocket, whereas its N-terminus (sequence: PLMI) engulfs in the depth of the same pocket. Thus, we have hypothesized that the PLMI motif could support the pharmacological actions of ERα17p. Here, we show that the PLMI peptide is, indeed, responsible for the GPER-dependent antiproliferative and anti-nociceptive effects of ERα17p. By using different biophysical approaches, we demonstrate that the NSLALSLT part of ERα17p is responsible for aggregation. Overall, the tetrapeptide PLMI, which supports the action of the parent peptide ERα17p, should be considered as a hit for the synthesis of new GPER modulators with dual antiproliferative and anti-nociceptive actions. This study highlights also the interest to modulate GPER for the control of pain.
Collapse
|
4
|
Tecalco-Cruz AC, Macías-Silva M, Ramírez-Jarquín JO, Ramírez-Jarquín UN. Decoding the Therapeutic Implications of the ERα Stability and Subcellular Distribution in Breast Cancer. Front Endocrinol (Lausanne) 2022; 13:867448. [PMID: 35498431 PMCID: PMC9044904 DOI: 10.3389/fendo.2022.867448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 01/22/2023] Open
Abstract
Approximately 70% of all breast cancer cases are estrogen receptor-alpha positive (ERα+) and any ERα signaling pathways deregulation is critical for the progression of malignant mammary neoplasia. ERα acts as a transcription factor that promotes the expression of estrogen target genes associated with pro-tumor activity in breast cancer cells. Furthermore, ERα is also part of extranuclear signaling pathways related to endocrine resistance. The regulation of ERα subcellular distribution and protein stability is critical to regulate its functions and, consequently, influence the response to endocrine therapies and progression of this pathology. This minireview highlights studies that have deciphered the molecular mechanisms implicated in controlling ERα stability and nucleo-cytoplasmic transport. These mechanisms offer information about novel biomarkers, therapeutic targets, and promising strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Angeles C. Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Mexico City, Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | - Uri Nimrod Ramírez-Jarquín
- Neural Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
- Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City, Mexico
| |
Collapse
|
5
|
Williams MM, Spoelstra NS, Arnesen S, O'Neill KI, Christenson JL, Reese J, Torkko KC, Goodspeed A, Rosas E, Hanamura T, Sams SB, Li Z, Oesterreich S, Riggins RB, Jacobsen BM, Elias A, Gertz J, Richer JK. Steroid Hormone Receptor and Infiltrating Immune Cell Status Reveals Therapeutic Vulnerabilities of ESR1-Mutant Breast Cancer. Cancer Res 2020; 81:732-746. [PMID: 33184106 DOI: 10.1158/0008-5472.can-20-1200] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
Mutations in ESR1 that confer constitutive estrogen receptor alpha (ER) activity in the absence of ligand are acquired by ≥40% of metastatic breast cancers (MBC) resistant to adjuvant aromatase inhibitor (AI) therapy. To identify targetable vulnerabilities in MBC, we examined steroid hormone receptors and tumor-infiltrating immune cells in metastatic lesions with or without ER mutations. ER and progesterone receptor (PR) were significantly lower in metastases with wild-type (WT) ER compared with those with mutant ER, suggesting that metastases that evade AI therapy by mechanism(s) other than acquiring ER mutations lose dependency on ER and PR. Metastases with mutant ER had significantly higher T regulatory and Th cells, total macrophages, and programmed death ligand-1 (PD-L1)-positive immune-suppressive macrophages than those with WT ER. Breast cancer cells with CRISPR-Cas9-edited ER (D538G, Y537S, or WT) and patient-derived xenografts harboring mutant or WT ER revealed genes and proteins elevated in mutant ER cells, including androgen receptor (AR), chitinase-3-like protein 1 (CHI3L1), and IFN-stimulated genes (ISG). Targeting these proteins blunted the selective advantage of ER-mutant tumor cells to survive estrogen deprivation, anchorage independence, and invasion. Thus, patients with mutant ER MBC might respond to standard-of-care fulvestrant or other selective ER degraders when combined with AR or CHI3L1 inhibition, perhaps with the addition of immunotherapy. SIGNIFICANCE: Targetable alterations in MBC, including AR, CHI3L1, and ISG, arise following estrogen-deprivation, and ER-mutant metastases may respond to immunotherapies due to elevated PD-L1+ macrophages.See related article by Arnesen et al., p. 539.
Collapse
Affiliation(s)
- Michelle M Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Spencer Arnesen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jordan Reese
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kathleen C Torkko
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andrew Goodspeed
- Department of Pharmacology and University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emmanuel Rosas
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Toru Hanamura
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sharon B Sams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zheqi Li
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Women's Cancer Research Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center (HCC), Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rebecca B Riggins
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Britta M Jacobsen
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony Elias
- School of Medicine, Division of Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jason Gertz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
6
|
Jeffreys SA, Powter B, Balakrishnar B, Mok K, Soon P, Franken A, Neubauer H, de Souza P, Becker TM. Endocrine Resistance in Breast Cancer: The Role of Estrogen Receptor Stability. Cells 2020; 9:cells9092077. [PMID: 32932819 PMCID: PMC7564140 DOI: 10.3390/cells9092077] [Citation(s) in RCA: 14] [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: 08/04/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Therapy of hormone receptor positive breast cancer (BCa) generally targets estrogen receptor (ER) function and signaling by reducing estrogen production or by blocking its interaction with the ER. Despite good long-term responses, resistance to treatment remains a significant issue, with approximately 40% of BCa patients developing resistance to ET. Mutations in the gene encoding ERα, ESR1, have been identified in BCa patients and are implicated as drivers of resistance and disease recurrence. Understanding the molecular consequences of these mutations on ER protein levels and its activity, which is tightly regulated, is vital. ER activity is in part controlled via its short protein half-life and therefore changes to its stability, either through mutations or alterations in pathways involved in protein stability, may play a role in therapy resistance. Understanding these connections and how ESR1 alterations could affect protein stability may identify novel biomarkers of resistance. This review explores the current reported data regarding posttranslational modifications (PTMs) of the ER and the potential impact of known resistance associated ESR1 mutations on ER regulation by affecting these PTMs in the context of ET resistance.
Collapse
Affiliation(s)
- Sarah A. Jeffreys
- Centre for Circulating Tumour Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool NSW 2170, Australia; (B.P.); (P.S.); (A.F.); (P.d.S.); (T.M.B.)
- School of Medicine, Western Sydney University, Campbelltown NSW 2560, Australia
- Correspondence: ; Tel.: +61-2-873-89022
| | - Branka Powter
- Centre for Circulating Tumour Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool NSW 2170, Australia; (B.P.); (P.S.); (A.F.); (P.d.S.); (T.M.B.)
| | - Bavanthi Balakrishnar
- Department of Medical Oncology, Liverpool Hospital, Liverpool NSW 2170, Australia; (B.B.); (K.M.)
| | - Kelly Mok
- Department of Medical Oncology, Liverpool Hospital, Liverpool NSW 2170, Australia; (B.B.); (K.M.)
| | - Patsy Soon
- Centre for Circulating Tumour Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool NSW 2170, Australia; (B.P.); (P.S.); (A.F.); (P.d.S.); (T.M.B.)
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool NSW 2170, Australia
- Department of Surgery, Bankstown Hospital, Bankstown NSW 2200, Australia
| | - André Franken
- Centre for Circulating Tumour Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool NSW 2170, Australia; (B.P.); (P.S.); (A.F.); (P.d.S.); (T.M.B.)
- Department of Obstetrics and Gynaecology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Hans Neubauer
- Department of Obstetrics and Gynaecology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Paul de Souza
- Centre for Circulating Tumour Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool NSW 2170, Australia; (B.P.); (P.S.); (A.F.); (P.d.S.); (T.M.B.)
- School of Medicine, Western Sydney University, Campbelltown NSW 2560, Australia
- Department of Medical Oncology, Liverpool Hospital, Liverpool NSW 2170, Australia; (B.B.); (K.M.)
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool NSW 2170, Australia
- School of Medicine, University of Wollongong, Wollongong NSW 2522, Australia
| | - Therese M. Becker
- Centre for Circulating Tumour Cells Diagnostics & Research, Ingham Institute of Applied Medical Research, Liverpool NSW 2170, Australia; (B.P.); (P.S.); (A.F.); (P.d.S.); (T.M.B.)
- School of Medicine, Western Sydney University, Campbelltown NSW 2560, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool NSW 2170, Australia
| |
Collapse
|
7
|
Effects of estrogen receptor signaling on prostate cancer carcinogenesis. Transl Res 2020; 222:56-66. [PMID: 32413498 DOI: 10.1016/j.trsl.2020.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/11/2020] [Accepted: 04/07/2020] [Indexed: 12/25/2022]
Abstract
Management of advanced prostate cancer remains complex, with substantial changes in treatment options emerging in recent years having implications for treatment selection and sequencing. Recognition of the importance of androgen signaling has led to life-prolonging treatments, as well as "liquid biopsy" techniques to guide these treatments in some settings. Therapies that target estrogen receptor signaling are efficacious but infrequently used options for treatment of castration-resistant prostate cancer. It is possible that nuances of estrogen receptor (ER) signaling, or selective modulation of ER signaling, might favorably influence outcomes in castration-resistant prostate cancer. Expression of ERs and their variants has been investigated in other cancers such as breast. Constitutively activating gene alterations can potentially lead to ER activation and subsequently promote cancer progression. The identification of these aberrations may help identify cancer phenotypes that are susceptible or resistant to therapies involved in ER signaling. This review outlines the current literature regarding ER signaling in prostate cancer, and provides background for exploration of potentially useful ER signaling biomarkers in advanced prostate cancer.
Collapse
|
8
|
Dunphy KA, Black AL, Roberts AL, Sharma A, Li Z, Suresh S, Browne EP, Arcaro KF, Ser-Dolansky J, Bigelow C, Troester MA, Schneider SS, Makari-Judson G, Crisi GM, Jerry DJ. Inter-Individual Variation in Response to Estrogen in Human Breast Explants. J Mammary Gland Biol Neoplasia 2020; 25:51-68. [PMID: 32152951 PMCID: PMC7147970 DOI: 10.1007/s10911-020-09446-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Exposure to estrogen is strongly associated with increased breast cancer risk. While all women are exposed to estrogen, only 12% are expected to develop breast cancer during their lifetime. These women may be more sensitive to estrogen, as rodent models have demonstrated variability in estrogen sensitivity. Our objective was to determine individual variation in expression of estrogen receptor (ER) and estrogen-induced responses in the normal human breast. Human breast tissue from female donors undergoing reduction mammoplasty surgery were collected for microarray analysis of ER expression. To examine estrogen-induced responses, breast tissue from 23 female donors were cultured ex- vivo in basal or 10 nM 17β-estradiol (E2) media for 4 days. Expression of ER genes (ESR1 and ESR2) increased significantly with age. E2 induced consistent increases in global gene transcription, but expression of target genes AREG, PGR, and TGFβ2 increased significantly only in explants from nulliparous women. E2-treatment did not induce consistent changes in proliferation or radiation induced apoptosis. Responses to estrogen are highly variable among women and not associated with levels of ER expression, suggesting differences in intracellular signaling among individuals. The differences in sensitivity to E2-stimulated responses may contribute to variation in risk of breast cancer.
Collapse
Affiliation(s)
- Karen A Dunphy
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA.
| | - Amye L Black
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Amy L Roberts
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Aman Sharma
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Zida Li
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Sneha Suresh
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Eva P Browne
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Kathleen F Arcaro
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | | | - Carol Bigelow
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, MA, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sallie S Schneider
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Pioneer Valley Life Sciences, Springfield, MA, USA
| | - Grace Makari-Judson
- Division of Hematology-Oncology, University of Massachusetts Medical School/Baystate, Springfield, MA, USA
| | - Giovanna M Crisi
- Department of Pathology, University of Massachusetts Medical School/Baystate, Springfield, MA, USA
| | - D Joseph Jerry
- The Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Pioneer Valley Life Sciences, Springfield, MA, USA
| |
Collapse
|
9
|
Yang XM, Wu ZM, Huang H, Chu XY, Lou J, Xu LX, Chen YT, Wang LQ, Huang OP. Estrogen receptor 1 mutations in 260 cervical cancer samples from Chinese patients. Oncol Lett 2019; 18:2771-2776. [PMID: 31452755 PMCID: PMC6676661 DOI: 10.3892/ol.2019.10612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/04/2019] [Indexed: 01/10/2023] Open
Abstract
Cervical cancer is one of the leading causes of cancer-associated mortality among females; however, the underlying molecular mechanisms of its carcinogenesis remain largely unclear. Previous comprehensive genomic studies have revealed prevalent estrogen receptor 1 (ESR1) mutations in breast cancer, which are rare in certain other types of cancer. To the best of our knowledge, it is unknown whether ESR1 mutations also exist in cervical cancer. Considering the evidence that cervical cancer shares certain genetic aberrations with breast cancer, and that the progression of both breast and cervical cancers can be affected by estrogen, it is possible that cervical cancer may also harbor ESR1 mutations. In the present study, a total of 260 Chinese cervical cancer samples with distinct subtypes were tested for the presence of ESR1 mutations. A total of three heterozygous missense ESR1 mutations, p.K303R (c.908A>G), p.T311M (c.932C>T) and p.Y537C (c.1610A>G), were identified in 3/207 (1.4%) cervical squamous cell carcinoma samples, which were absent in 27 adenosquamous carcinomas and 26 adenocarcinomas samples. Of the three individuals with an ESR1mutation, 1 patient was also diagnosed with ovarian endometriosis and the other 2 patients were diagnosed with a uterine fibroid. A bioinformatics analysis suggested that these ESR1 mutations may be pathogenic by promoting the development of cervical cancer. Furthermore, a previous comprehensive study confirmed that individuals with cervical squamous cell carcinoma possessed ESR1 mutations. These combined studies indicate that ESR1 mutations may participate in the carcinogenesis of cervical squamous cell carcinoma, albeit at a low frequency. In conclusion, the present study identified three potentially pathogenic ESR1 mutations in Chinese cervical squamous cell carcinoma samples, but not in other subtypes.
Collapse
Affiliation(s)
- Xin-Min Yang
- College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi-Min Wu
- Department of Management Engineering, Shangrao Vocational and Technical College, Shangrao, Jiangxi 334109, P.R. China
| | - Huang Huang
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Yan Chu
- Department of Gynecological Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi 330029, P.R. China
| | - Jun Lou
- Department of Gynecological Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi 330029, P.R. China
| | - Li-Xian Xu
- College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Yuan-Ting Chen
- College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Qun Wang
- College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Ou-Ping Huang
- College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
10
|
Tecalco-Cruz AC, Ramírez-Jarquín JO, Cruz-Ramos E. Estrogen Receptor Alpha and its Ubiquitination in Breast Cancer Cells. Curr Drug Targets 2019; 20:690-704. [DOI: 10.2174/1389450119666181015114041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 12/23/2022]
Abstract
More than 70% of all breast cancer cases are estrogen receptor alpha-positive (ERα). ERα is a member of the nuclear receptor family, and its activity is implicated in the gene transcription linked to the proliferation of breast cancer cells, as well as in extranuclear signaling pathways related to the development of resistance to endocrine therapy. Protein-protein interactions and posttranslational modifications of ERα underlie critical mechanisms that modulate its activity. In this review, the relationship between ERα and ubiquitin protein (Ub), was investigated in the context of breast cancer cells. Interestingly, Ub can bind covalently or non-covalently to ERα resulting in either a proteolytic or non-proteolytic fate for this receptor. Thereby, Ub-dependent molecular pathways that modulate ERα signaling may play a central role in breast cancer progression, and consequently, present critical targets for treatment of this disease.
Collapse
Affiliation(s)
- Angeles C. Tecalco-Cruz
- Instituto de Investigaciones Biomedicas. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| | - Josué O. Ramírez-Jarquín
- Instituto de Fisiologia Celular. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| | - Eduardo Cruz-Ramos
- Instituto de Investigaciones Biomedicas. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| |
Collapse
|
11
|
Pejerrey SM, Dustin D, Kim JA, Gu G, Rechoum Y, Fuqua SAW. The Impact of ESR1 Mutations on the Treatment of Metastatic Breast Cancer. Discov Oncol 2018; 9:215-228. [PMID: 29736566 DOI: 10.1007/s12672-017-0306-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 08/31/2017] [Indexed: 12/25/2022] Open
Abstract
After nearly 20 years of research, it is now established that mutations within the estrogen receptor (ER) gene, ESR1, frequently occur in metastatic breast cancer and influence response to hormone therapy. Though early studies presented differing results, sensitive sequencing techniques now show that ESR1 mutations occur at a frequency between 20 and 40% depending on the assay method. Recent studies have focused on several "hot spot mutations," a cluster of mutations found in the hormone-binding domain of the ESR1 gene. Throughout the course of treatment, tumor evolution can occur, and ESR1 mutations emerge and become enriched in the metastatic setting. Sensitive techniques to continually monitor mutant burden in vivo are needed to effectively treat patients with mutant ESR1. The full impact of these mutations on tumor response to different therapies remains to be determined. However, recent studies indicate that mutant-bearing tumors may be less responsive to specific hormonal therapies, and suggest that aromatase inhibitor (AI) therapy may select for the emergence of ESR1 mutations. Additionally, different mutations may respond discretely to targeted therapies. The need for more preclinical mechanistic studies on ESR1 mutations and the development of better agents to target these mutations are urgently needed. In the future, sequential monitoring of ESR1 mutational status will likely direct personalized therapeutic regimens appropriate to each tumor's unique mutational landscape.
Collapse
Affiliation(s)
- Sasha M Pejerrey
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600, Houston, TX, 77030, USA
| | - Derek Dustin
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600, Houston, TX, 77030, USA
| | - Jin-Ah Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600, Houston, TX, 77030, USA
| | - Guowei Gu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600, Houston, TX, 77030, USA
| | - Yassine Rechoum
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600, Houston, TX, 77030, USA
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Jiang CF, Li DM, Shi ZM, Wang L, Liu MM, Ge X, Liu X, Qian YC, Wen YY, Zhen LL, Lin J, Liu LZ, Jiang BH. Estrogen regulates miRNA expression: implication of estrogen receptor and miR-124/AKT2 in tumor growth and angiogenesis. Oncotarget 2018; 7:36940-36955. [PMID: 27175587 PMCID: PMC5095050 DOI: 10.18632/oncotarget.9230] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 04/16/2016] [Indexed: 12/21/2022] Open
Abstract
It is currently known that estrogen plays an important role in breast cancer (BC) development, but the underlying molecular mechanism remains to be elucidated. Accumulating evidence has revealed important roles of microRNAs in various kinds of human cancers, including BC. In this study, we found that among the microRNAs regulated by estrogen, miR-124 was the most prominent downregulated miRNA. miR-124 was downregulated by estradiol (E2) treatment in estrogen receptor (ER) positive BC cells, miR-124 overexpression suppressed cell proliferation, migration and invasion in BC cells; while the suppression of miR-124 using Anti-miR-124 inhibitor had opposite cellular functions. Under the E2 treatment, miR-124 had stronger effect to inhibit cellular functions in MCF7 cells than that in MDA-MB-231 cells. In addition, we identified that ERα, but not ERβ, was required for E2-induced miR-124 downregulation. Furthermore, AKT2, a known oncogene, was a novel direct target of miR-124. AKT2 expression levels were inversely correlated with miR-124 expression levels in human breast cancer specimens. AKT2 was overexpressed in BC specimens, and its expression levels were much higher in ERα positive cancer tissues than those ERα negative cancer tissues. Consistent with miR-124 suppression, E2 treatment increased AKT2 expression levels in MCF7 cells via ERα. Finally, overexpression of miR-124 in MCF7 cells significantly suppressed tumor growth and angiogenesis by targeting AKT2. Our results provide a mechanistic insight into a functional role of new ERα/miR-124/AKT2 signaling pathway in BC development. miR-124 and AKT2 may be used as biomarkers for ERα positive BC and therapeutic effect in the future.
Collapse
Affiliation(s)
- Cheng-Fei Jiang
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Dong-Mei Li
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Zhu-Mei Shi
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Wang
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Min-Min Liu
- Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Xin Ge
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xue Liu
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ying-Chen Qian
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Yi-Yang Wen
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Lin-Lin Zhen
- Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Jie Lin
- Faculty of Software, Fujian Normal University, Fuzhou, China
| | - Ling-Zhi Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bing-Hua Jiang
- State Key Laboratory of Reproductive Medicine, Department of Pathology, Nanjing Medical University, Nanjing, China.,Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
13
|
Zheng L, Meng X, Li X, Zhang Y, Li C, Xiang C, Xing Y, Xia Y, Xi T. miR-125a-3p inhibits ERα transactivation and overrides tamoxifen resistance by targeting CDK3 in estrogen receptor-positive breast cancer. FASEB J 2018; 32:588-600. [PMID: 28939591 DOI: 10.1096/fj.201700461rr] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tamoxifen (TAM) is a major adjuvant therapy for patients who are diagnosed with estrogen receptor-α (ER)-positive breast cancer; however, TAM resistance occurs often during treatment and the underlying mechanism is unclear. Here, we report that miR-125a-3p inhibits ERα transcriptional activity and, thus, ER+ breast cancer cell proliferation, which causes cell-cycle arrest at the G1/S stage, inducing apoptosis and suppressing tumor growth by targeting cyclin-dependent kinase 3 (CDK3) in vitro and in vivo. In addition, CDK3 and miR-125a-3p expression levels were measured in 37 cancerous tissues paired with noncancerous samples, and their expression levels were negatively associated with miR-125a-3p level. Of interest, miR-125a-3p level is down-regulated in MCF-7 TAM-resistant (TamR) cells. Of more importance, up-regulation of miR-125a-3p resensitizes MCF-7 TamR cells to TAM, which is dependent on CDK3 expression. These results suggest that miR-125a-3p can function as a novel tumor suppressor in ER+ breast cancer by targeting CDK3, which may be a potential therapeutic approach for TamR breast cancer therapy.-Zheng, L., Meng, X., Li, X., Zhang, Y., Li, C., Xiang, C., Xing, Y., Xia, Y., Xi, T. miR-125a-3p inhibits ERα transactivation and overrides tamoxifen resistance by targeting CDK3 in estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology of Chinese Materia Medica, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Xia Meng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Cheng Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chenxi Xiang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yufeng Xia
- Department of Pharmacology of Chinese Materia Medica, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
14
|
Gelsomino L, Gu G, Rechoum Y, Beyer AR, Pejerrey SM, Tsimelzon A, Wang T, Huffman K, Ludlow A, Andò S, Fuqua SAW. ESR1 mutations affect anti-proliferative responses to tamoxifen through enhanced cross-talk with IGF signaling. Breast Cancer Res Treat 2016; 157:253-265. [PMID: 27178332 DOI: 10.1007/s10549-016-3829-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 01/06/2023]
Abstract
The purpose of this study was to address the role of ESR1 hormone-binding mutations in breast cancer. Soft agar anchorage-independent growth assay, Western blot, ERE reporter transactivation assay, proximity ligation assay (PLA), coimmunoprecipitation assay, silencing assay, digital droplet PCR (ddPCR), Kaplan-Meier analysis, and statistical analysis. It is now generally accepted that estrogen receptor (ESR1) mutations occur frequently in metastatic breast cancers; however, we do not yet know how to best treat these patients. We have modeled the three most frequent hormone-binding ESR1 (HBD-ESR1) mutations (Y537N, Y537S, and D538G) using stable lentiviral transduction in human breast cancer cell lines. Effects on growth were examined in response to hormonal and targeted agents, and mutation-specific changes were studied using microarray and Western blot analysis. We determined that the HBD-ESR1 mutations alter anti-proliferative effects to tamoxifen (Tam), due to cell-intrinsic changes in activation of the insulin-like growth factor receptor (IGF1R) signaling pathway and levels of PIK3R1/PIK3R3. The selective estrogen receptor degrader, fulvestrant, significantly reduced the anchorage-independent growth of ESR1 mutant-expressing cells, while combination treatments with the mTOR inhibitor everolimus, or an inhibitor blocking IGF1R, and the insulin receptor significantly enhanced anti-proliferative responses. Using digital drop (dd) PCR, we identified mutations at high frequencies ranging from 12 % for Y537N, 5 % for Y537S, and 2 % for D538G in archived primary breast tumors from women treated with adjuvant mono-tamoxifen therapy. The HBD-ESR1 mutations were not associated with recurrence-free or overall survival in response in this patient cohort and suggest that knowledge of other cell-intrinsic factors in combination with ESR1 mutation status will be needed determine anti-proliferative responses to Tam.
Collapse
Affiliation(s)
- Luca Gelsomino
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Guowei Gu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yassine Rechoum
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Amanda R Beyer
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sasha M Pejerrey
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Anna Tsimelzon
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Tao Wang
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kenneth Huffman
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Ludlow
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sebastiano Andò
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Suzanne A W Fuqua
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Dan L Duncan Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
G9a-mediated methylation of ERα links the PHF20/MOF histone acetyltransferase complex to hormonal gene expression. Nat Commun 2016; 7:10810. [PMID: 26960573 PMCID: PMC4792926 DOI: 10.1038/ncomms10810] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/24/2016] [Indexed: 12/19/2022] Open
Abstract
The euchromatin histone methyltransferase 2 (also known as G9a) methylates histone H3K9 to repress gene expression, but it also acts as a coactivator for some nuclear receptors. The molecular mechanisms underlying this activation remain elusive. Here we show that G9a functions as a coactivator of the endogenous oestrogen receptor α (ERα) in breast cancer cells in a histone methylation-independent manner. G9a dimethylates ERα at K235 both in vitro and in cells. Dimethylation of ERαK235 is recognized by the Tudor domain of PHF20, which recruits the MOF histone acetyltransferase (HAT) complex to ERα target gene promoters to deposit histone H4K16 acetylation promoting active transcription. Together, our data suggest the molecular mechanism by which G9a functions as an ERα coactivator. Along with the PHF20/MOF complex, G9a links the crosstalk between ERα methylation and histone acetylation that governs the epigenetic regulation of hormonal gene expression. The histone methyltransferase G9a methylates histone H3K9 to repress gene expression, but it also acts as a coactivator for some nuclear receptors. Here, Zhang et al. show that methylation of ERα by G9a recruits the PHF20/MOF complex that deposits histone H4K16 acetylation promoting active transcription.
Collapse
|
16
|
Wang P, Bahreini A, Gyanchandani R, Lucas PC, Hartmaier RJ, Watters RJ, Jonnalagadda AR, Trejo Bittar HE, Berg A, Hamilton RL, Kurland BF, Weiss KR, Mathew A, Leone JP, Davidson NE, Nikiforova MN, Brufsky AM, Ambros TF, Stern AM, Puhalla SL, Lee AV, Oesterreich S. Sensitive Detection of Mono- and Polyclonal ESR1 Mutations in Primary Tumors, Metastatic Lesions, and Cell-Free DNA of Breast Cancer Patients. Clin Cancer Res 2015; 22:1130-7. [PMID: 26500237 DOI: 10.1158/1078-0432.ccr-15-1534] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 10/07/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE Given the clinical relevance of ESR1 mutations as potential drivers of resistance to endocrine therapy, this study used sensitive detection methods to determine the frequency of ESR1 mutations in primary and metastatic breast cancer, and in cell-free DNA (cfDNA). EXPERIMENTAL DESIGN Six ESR1 mutations (K303R, S463P, Y537C, Y537N, Y537S, D538G) were assessed by digital droplet PCR (ddPCR), with lower limits of detection of 0.05% to 0.16%, in primary tumors (n = 43), bone (n = 12) and brain metastases (n = 38), and cfDNA (n = 29). Correlations between ESR1 mutations in metastatic lesions and single (1 patient) or serial blood draws (4 patients) were assessed. RESULTS ESR1 mutations were detected for D538G (n = 13), Y537S (n = 3), and Y537C (n = 1), and not for K303R, S463P, or Y537N. Mutation rates were 7.0% (3/43 primary tumors), 9.1% (1/11 bone metastases), 12.5% (3/24 brain metastases), and 24.1% (7/29 cfDNA). Two patients showed polyclonal disease with more than one ESR1 mutation. Mutation allele frequencies were 0.07% to 0.2% in primary tumors, 1.4% in bone metastases, 34.3% to 44.9% in brain metastases, and 0.2% to 13.7% in cfDNA. In cases with both cfDNA and metastatic samples (n = 5), mutations were detected in both (n = 3) or in cfDNA only (n = 2). Treatment was associated with changes in ESR1 mutation detection and allele frequency. CONCLUSIONS ESR1 mutations were detected at very low allele frequencies in some primary breast cancers, and at high allele frequency in metastases, suggesting that in some tumors rare ESR1-mutant clones are enriched by endocrine therapy. Further studies should address whether sensitive detection of ESR1 mutations in primary breast cancer and in serial blood draws may be predictive for development of resistant disease. See related commentary by Gu and Fuqua, p. 1034.
Collapse
Affiliation(s)
- Peilu Wang
- School of Medicine, Tsinghua University, Beijing, People's Republic of China. Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania
| | - Amir Bahreini
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rekha Gyanchandani
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan J Hartmaier
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rebecca J Watters
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amruth R Jonnalagadda
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania
| | | | - Aaron Berg
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ronald L Hamilton
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brenda F Kurland
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kurt R Weiss
- Department of Orthopedic Surgery, University of Pittsburgh Medical Center (UPMC) Pittsburgh, Pittsburgh, Pennsylvania
| | - Aju Mathew
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | - Jose Pablo Leone
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | - Nancy E Davidson
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | | | - Adam M Brufsky
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | - Tadeu F Ambros
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | - Andrew M Stern
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shannon L Puhalla
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh, Pennsylvania
| | - Adrian V Lee
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Steffi Oesterreich
- Womens Cancer Research Center, Magee-Women Research Institute, Pittsburgh, Pennsylvania. Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
17
|
Shen R, Wang Y, Wang CX, Yin M, Liu HL, Chen JP, Han JQ, Wang WB. MiRNA-155 mediates TAM resistance by modulating SOCS6-STAT3 signalling pathway in breast cancer. Am J Transl Res 2015; 7:2115-2126. [PMID: 26692956 PMCID: PMC4656789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/11/2015] [Indexed: 06/05/2023]
Abstract
Breast cancer is the second leading cause of cancer induced death in women. Tamoxifen is an endocrine therapy which is administered to 70% of all breast cancer patients with estrogen receptor alpha (ERα) expression. Despite the initial response, most patients eventually acquire resistance to the drug. MicroRNAs (miRNAs) are a class of small non-coding RNAs which have the ability to post-transcriptionally regulate gene expression. Although the role of a few miRNAs has been described in tamoxifen resistance, little is known about how concerted actions of miRNAs targeting biological networks contribute to its resistance. In this study, we identified that miR-155 is frequently up-regulated in breast cancer with tamoxifen resistance. Ectopic expression of miR-155 induces cell survival and resistance to TAM, whereas inhibition of miR-155 causes cells to apoptosis and enhances TAM sensitivity. Further, we identified SOCS6 as a new direct target of miR-155. Sustained overexpression of miR-155 resulted in repression of SOCS6 protein and mRNA levels, and knockdown of miR-155 increased SOCS6 expression. Introduction of SOCS6 cDNA lacking the 3'-UTR abrogated miR-155-induced cell survival and chemoresistance. Finally, it was verified that SOCS6 or inhibition of STAT3 could inhibit miR-155 STAT3 activation and cell proliferation. In conclusion, our study reveals a molecular link between miR-155 and SOCS6-STAT3 and presents an evidence that miR-155 is a critical therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Rong Shen
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| | - Yu Wang
- Department of Radiotherapy, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| | - Cai-Xia Wang
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| | - Miao Yin
- College of Life Science, Shandong Normal UniversityJinan 250014, China
| | - Hong-Liang Liu
- Duke Cancer Institute, Duke University Medical CenterDurham, North Carolina, NC 27710, USA
| | - Jian-Peng Chen
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| | - Jun-Qing Han
- Department of Radiotherapy, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| | - Wei-Bo Wang
- Department of Chemotherapy, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250021, China
| |
Collapse
|
18
|
Therapeutic intervention based on circulating tumor cell phenotype in metastatic breast cancer: concept of the DETECT study program. Arch Gynecol Obstet 2015; 293:271-81. [DOI: 10.1007/s00404-015-3879-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 08/26/2015] [Indexed: 12/18/2022]
|
19
|
Abstract
Approximately 70% of breast cancers are oestrogen receptor α (ER) positive, and are, therefore, treated with endocrine therapies. However, about 25% of patients with primary disease and almost all patients with metastases will present with or eventually develop endocrine resistance. Despite the magnitude of this clinical challenge, the mechanisms underlying the development of resistance remain largely unknown. In the past 2 years, several studies unveiled gain-of-function mutations in ESR1, the gene encoding the ER, in approximately 20% of patients with metastatic ER-positive disease who received endocrine therapies, such as tamoxifen and aromatase inhibitors. These mutations are clustered in a 'hotspot' within the ligand-binding domain (LBD) of the ER and lead to ligand-independent ER activity that promotes tumour growth, partial resistance to endocrine therapy, and potentially enhanced metastatic capacity; thus, ER LBD mutations might account for a mechanism of acquired endocrine resistance in a substantial fraction of patients with metastatic disease. In general, the absence of detectable ESR1 mutations in patients with treatment-naive disease, and the correlation between the frequency of patients with tumours harbouring these mutations and the number of endocrine treatments received suggest that, under selective treatment pressure, clonal expansion of rare mutant clones occurs, leading to resistance. Preclinical and clinical development of rationale-based novel therapeutic strategies that inhibit these ER mutants has the potential to substantially improve treatment outcomes. We discuss the contribution of ESR1 mutations to the development of acquired resistance to endocrine therapy, and evaluate how mutated ER can be detected and targeted to overcome resistance and improve patient outcomes.
Collapse
|
20
|
Regulation of estrogen receptor signaling in breast carcinogenesis and breast cancer therapy. Cell Mol Life Sci 2014; 71:1549. [PMID: 25031550 PMCID: PMC3962223 DOI: 10.1007/s00018-013-1376-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Estrogen and estrogen receptors (ERs) are critical regulators of breast epithelial cell proliferation, differentiation, and apoptosis. Compromised signaling vis-à-vis the estrogen receptor is believed to be a major contributing factor in the malignancy of breast cells. Targeting the ER signaling pathway has been a focal point in the development of breast cancer therapy. Although approximately 75 % of breast cancer patients are classified as luminal type (ER(+)), which predicts for response to endocrine-based therapy; however, innate or acquired resistance to endocrine-based drugs remains a serious challenge. The complexity of regulation for estrogen signaling coupled with the crosstalk of other oncogenic signaling pathways is a reason for endocrine therapy resistance. Alternative strategies that target novel molecular mechanisms are necessary to overcome this current and urgent gap in therapy. A thorough analysis of estrogen-signaling regulation is critical. In this review article, we will summarize current insights into the regulation of estrogen signaling as related to breast carcinogenesis and breast cancer therapy.
Collapse
|
21
|
Fuqua SAW, Gu G, Rechoum Y. Estrogen receptor (ER) α mutations in breast cancer: hidden in plain sight. Breast Cancer Res Treat 2014; 144:11-9. [PMID: 24487689 DOI: 10.1007/s10549-014-2847-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/18/2014] [Indexed: 11/25/2022]
Abstract
The idea that somatic ERα mutations could play an important role in the evolution of hormone-dependent breast cancers was proposed some years ago (Fuqua J Mammary Gland Biol Neoplasia 6(4):407-417, 2001; Dasgupta et al. Annu Rev Med 65:279-292, 2013), but has remained controversial until recently. A significant amount of new data has confirmed these initial observations and shown their significance, along with much additional work relevant to the treatment of breast cancer. Thus, it is the purpose of this review to summarize the research to date on the existence and clinical consequences of ERα mutations in primary and metastatic breast cancer.
Collapse
Affiliation(s)
- Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Dan L Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA,
| | | | | |
Collapse
|
22
|
van Kruchten M, de Vries EGE, Brown M, de Vries EFJ, Glaudemans AWJM, Dierckx RAJO, Schröder CP, Hospers GAP. PET imaging of oestrogen receptors in patients with breast cancer. Lancet Oncol 2013; 14:e465-e475. [PMID: 24079874 DOI: 10.1016/s1470-2045(13)70292-4] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oestrogen receptors are overexpressed in around 70% of all breast cancers, and are a target for endocrine therapy. These receptors can be visualised on PET with use of 16α-[(18)F]-fluoro-17β-oestradiol ((18)F-FES) as a tracer. Compared with biopsy, which enables assessment of individual sites, whole-body (18)F-FES-PET enables quantification of oestrogen-receptor expression in all metastases. In several studies, measurement of tumour protein expression in oestrogen receptors by (18)F-FES-PET, concurrent with biopsy, detected oestrogen-receptor-positive tumour lesions with a sensitivity of 84% and specificity of 98%. Roughly 45% of patients with metastatic breast cancer have discordant oestrogen-receptor expression across lesions (ie, (18)F-FES-positive and (18)F-FES-negative metastases). Low tumour (18)F-FES uptake in metastases can predict failure of hormonal therapy in patients with oestrogen-receptor-positive primary tumours. Finally, (18)F-FES-PET has shown that oestrogen-receptor binding capacity changes after intervention with hormonal drugs, but findings need to be confirmed. Factors other than oestrogen-receptor expression, including menopausal status and concomitant therapies, that can affect tumour (18)F-FES uptake must be taken into account.
Collapse
Affiliation(s)
- Michel van Kruchten
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands.
| |
Collapse
|
23
|
Dynamic regulation of steroid hormone receptor transcriptional activity by reversible SUMOylation. VITAMINS AND HORMONES 2013; 93:227-61. [PMID: 23810010 DOI: 10.1016/b978-0-12-416673-8.00008-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Transcription complexes containing steroid hormone receptors (SRs) have been well characterized at selected canonical target genes. More recently, the advent of whole genome technologies has allowed for complete SR transcriptome analyses in diverse cell types and in response to a variety of cellular stimuli. These types of studies have revealed little overlap between the tissue or cell type-specific transcriptomes of a given SR, suggesting that all SRs are highly context-dependent transcription factors. However, the mechanisms controlling SR promoter selectivity have not been fully elucidated. Many factors may influence SR promoter selectivity, including chromatin structure, cofactor availability, and posttranslational modifications to SRs and/or their numerous coregulators; this review focuses on the impact that covalent attachment of small ubiquitin-like modifier (SUMO) moieties to SRs (i.e., SUMOylation) have on the transcriptional regulation of SR target genes.
Collapse
|
24
|
Modulation of estrogen receptor alpha activity and expression during breast cancer progression. VITAMINS AND HORMONES 2013; 93:135-60. [PMID: 23810005 DOI: 10.1016/b978-0-12-416673-8.00004-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Seventy percent of breast tumors express the estrogen receptor (ER), which is generally considered to predict a better outcome relative to ER-negative tumors, as they often respond to antiestrogen therapies. During cancer progression, mammary tumors can escape from estrogen control, resulting in the acquisition of invasive properties and resistance to treatment. ER expression is a dynamic phenomenon and is finely regulated at numerous levels, including the gene, mRNA, and protein levels. As a consequence, many molecular mechanisms have been implicated in modulating ER activity and estrogen signaling in mammary cancer. In fact, one-third of ER-positive breast cancer cells do not respond to first-line endocrine therapies, and a large subset of relapsing tumors retain ER expression. Increased knowledge of these mechanisms has led to the development of better prognostic methods and targeted therapies for patients; however, additional research is still needed to improve patient survival. In this chapter, we focus on the signaling pathways leading to changes in or loss of ER activity in breast cancer progression.
Collapse
|
25
|
Abbasi S, Rasouli M, Nouri M, Kalbasi S. Association of estrogen receptor-α A908G (K303R) mutation with breast cancer risk. Int J Clin Exp Med 2012; 6:39-49. [PMID: 23236557 PMCID: PMC3515972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 11/03/2012] [Indexed: 06/01/2023]
Abstract
Genetic mutations in premalignant breast lesions may have a role in malignancy progression or influence the behavior of subsequent disease. A point mutation in estrogen receptor-α (ER-α) as A908G (Lys303→Arg) was originally involved to hypersensitive to estrogen breast hyperplasia. We detected this mutation among Iranian women with invasive breast cancer. A population-based case-control study was conducted in 150 newly diagnosed invasive breast cancer and 147 healthy control individuals controls to screen for presence of the ER-α A908G mutation by using single-strand conformation polymorphism (SSCP) analysis and 33Pcycle DNA sequencing. We detected the 10.7% ER-α A908G mutation in the form of heterozygote genotype only among cancer patients (χ(2)=22.752, P=0.00). The allelic frequency of mutant allele AGG in codon 303 was significantly (χ(2)=29.709, P=0.001) higher in patients with the family history of breast cancer (28.9%) than those without the family history of breast cancer (1.9%). Our data suggest that ER-α codon 303 mutation is correlated with various aspects of breast cancer in Iran. ER-α genotype might represent a surrogate marker for predicting breast cancer developing later in life.
Collapse
Affiliation(s)
- Sakineh Abbasi
- Department of Biotechnology, School of Allied Medicine, Tehran University of Medical SciencesTehran, 1985717443, Iran
| | - Mina Rasouli
- Laboratory of Vaccines and Immunotherapeutics,Institute of Bioscience, University Putra MalaysiaUPM, Serdang,43400, Selangor, Malaysia
| | - Mehrnaz Nouri
- Department of Biology, Lund University Sölvegatan 35SE-223 62 Lund, Sweden
| | - Samira Kalbasi
- DVM, Faculty of Veterinary, University of TehranTehran, 1417965463, Iran
| |
Collapse
|
26
|
Anbalagan M, Huderson B, Murphy L, Rowan BG. Post-translational modifications of nuclear receptors and human disease. NUCLEAR RECEPTOR SIGNALING 2012; 10:e001. [PMID: 22438791 PMCID: PMC3309075 DOI: 10.1621/nrs.10001] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 08/19/2011] [Indexed: 12/12/2022]
Abstract
Nuclear receptors (NR) impact a myriad of physiological processes including homeostasis, reproduction, development, and metabolism. NRs are regulated by post-translational modifications (PTM) that markedly impact receptor function. Recent studies have identified NR PTMs that are involved in the onset and progression of human diseases, including cancer. The majority of evidence linking NR PTMs with disease has been demonstrated for phosphorylation, acetylation and sumoylation of androgen receptor (AR), estrogen receptor α (ERα), glucocorticoid receptor (GR) and peroxisome proliferator activated receptor γ (PPARγ). Phosphorylation of AR has been associated with hormone refractory prostate cancer and decreased disease-specific survival. AR acetylation and sumoylation increased growth of prostate cancer tumor models. AR phosphorylation reduced the toxicity of the expanded polyglutamine AR in Kennedy's Disease as a consequence of reduced ligand binding. A comprehensive evaluation of ERα phosphorylation in breast cancer revealed several sites associated with better clinical outcome to tamoxifen therapy, whereas other phosphorylation sites were associated with poorer clinical outcome. ERα acetylation and sumoylation may also have predictive value for breast cancer. GR phosphorylation and acetylation impact GR responsiveness to glucocorticoids that are used as anti-inflammatory drugs. PPARγ phosphorylation can regulate the balance between growth and differentiation in adipose tissue that is linked to obesity and insulin resistance. Sumoylation of PPARγ is linked to repression of inflammatory genes important in patients with inflammatory diseases. NR PTMs provide an additional measure of NR function that can be used as both biomarkers of disease progression, and predictive markers for patient response to NR-directed treatments.
Collapse
Affiliation(s)
- Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | |
Collapse
|
27
|
Barone I, Catalano S, Gelsomino L, Marsico S, Giordano C, Panza S, Bonofiglio D, Bossi G, Covington KR, Fuqua SAW, Andò S. Leptin mediates tumor-stromal interactions that promote the invasive growth of breast cancer cells. Cancer Res 2012; 72:1416-27. [PMID: 22282662 DOI: 10.1158/0008-5472.can-11-2558] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Obesity confers risks to cancer development and progression but the mechanisms underlying these risks remain unclear. In this study, we identify a role for the obesity cytokine leptin, which has been implicated previously in breast cancer development, as a determinant for the tumor-promoting activity of cancer-associated fibroblasts (CAF) in both wild-type (WT) and K303R mutant estrogen receptor-α (ERα)-expressing breast cancer cells. Human CAFs stimulated a greater increase in the proliferation and migration of breast cancer cells expressing the K303R-ERα hyperactive receptor than WT-ERα-expressing cells. A concomitant increase was seen in leptin receptor isoform expression and activation of the leptin signaling pathway in cells expressing K303R-ERα compared with WT-ERα, correlating with leptin effects on cell growth, motility, and invasiveness in mutant cells. Epidermal growth factor and other factors secreted by K303R-ERα cells stimulated CAF proliferation, migration, and subsequent leptin secretion. Moreover, K303R-ERα expression generated a leptin hypersensitive phenotype in vivo. Together, our results reveal a bidirectional cross-talk between breast cancer cells and "educated" CAFs that drives tumor progression via leptin signaling. In elucidating a mechanism that connects obesity and cancer, these findings reinforce the concept that blocking cancer-stromal cell communication may represent an effective strategy for targeted therapy of breast cancer.
Collapse
Affiliation(s)
- Ines Barone
- Centro Sanitario, University of Calabria, Rende, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhang Y, Li Z, Sacks DB, Ames JB. Structural basis for Ca2+-induced activation and dimerization of estrogen receptor α by calmodulin. J Biol Chem 2012; 287:9336-44. [PMID: 22275375 DOI: 10.1074/jbc.m111.334797] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The estrogen receptor α (ER-α) regulates expression of target genes implicated in development, metabolism, and breast cancer. Calcium-dependent regulation of ER-α is critical for activating gene expression and is controlled by calmodulin (CaM). Here, we present the NMR structures for the two lobes of CaM each bound to a localized region of ER-α (residues 287-305). A model of the complete CaM·ER-α complex was constructed by combining these two structures with additional data. The two lobes of CaM both compete for binding at the same site on ER-α (residues 292, 296, 299, 302, and 303), which explains why full-length CaM binds two molecules of ER-α in a 1:2 complex and stabilizes ER-α dimerization. Exposed glutamate residues in CaM (Glu(11), Glu(14), Glu(84), and Glu(87)) form salt bridges with key lysine residues in ER-α (Lys(299), Lys(302), and Lys(303)), which are likely to prevent ubiquitination at these sites and inhibit degradation of ER-α. Mutants of ER-α at the CaM-binding site (W292A and K299A) weaken binding to CaM, and I298E/K299D disrupts estrogen-induced transcription. CaM facilitates dimerization of ER-α in the absence of estrogen, and stimulation of ER-α by either Ca(2+) and/or estrogen may serve to regulate transcription in a combinatorial fashion.
Collapse
Affiliation(s)
- Yonghong Zhang
- Department of Chemistry, University of California, Davis, California 95616, USA
| | | | | | | |
Collapse
|
29
|
Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM, Corbo L. Cracking the estrogen receptor's posttranslational code in breast tumors. Endocr Rev 2011; 32:597-622. [PMID: 21680538 DOI: 10.1210/er.2010-0016] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estrogen signaling pathways, because of their central role in regulating the growth and survival of breast tumor cells, have been identified as suitable and efficient targets for cancer therapies. Agents blocking estrogen activity are already widely used clinically, and many new molecules have entered clinical trials, but intrinsic or acquired resistance to treatment limits their efficacy. The basic molecular studies underlying estrogen signaling have defined the critical role of estrogen receptors (ER) in many aspects of breast tumorigenesis. However, important knowledge gaps remain about the role of posttranslational modifications (PTM) of ER in initiation and progression of breast carcinogenesis. Whereas major attention has been focused on the phosphorylation of ER, many other PTM (such as acetylation, ubiquitination, sumoylation, methylation, and palmitoylation) have been identified as events modifying ER expression and stability, subcellular localization, and sensitivity to hormonal response. This article will provide an overview of the current and emerging knowledge on ER PTM, with a particular focus on their deregulation in breast cancer. We also discuss their clinical relevance and the functional relationship between PTM. A thorough understanding of the complete picture of these modifications in ER carcinogenesis might not only open new avenues for identifying new markers for prognosis or prediction of response to endocrine therapy but also could promote the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Muriel Le Romancer
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Bâtiment Cheney D, 28 rue Laennec, 69373 Lyon Cedex 08, France.
| | | | | | | | | | | |
Collapse
|
30
|
La Rosa P, Pesiri V, Marino M, Acconcia F. 17β-Estradiol-induced cell proliferation requires estrogen receptor (ER) α monoubiquitination. Cell Signal 2011; 23:1128-35. [PMID: 21356307 DOI: 10.1016/j.cellsig.2011.02.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 02/20/2011] [Indexed: 12/21/2022]
Abstract
Protein monoubiquitination (monoUbq) (i.e., the attachment of one single ubiquitin to the substrate) is a non-proteolytic reversible modification that controls protein functions. Among other proteins, the estrogen receptor α (ERα), which mediates the pleiotropic effects of the cognate hormone 17β-estradiol (E2), is a monoubiquitinated protein. Although it has been demonstrated that E2 rapidly reduces ERα monoUbq in breast cancer cells, the impact of monoUbq in the regulation of the ERα activities is poorly appreciated. Here, we show that mutation of the ERα monoUbq sites prevents the E2-induced ERα phosphorylation in the serine residue 118 (S118), reduces ERα transcriptional activity, and precludes the ERα-mediated extranuclear activation of signaling pathways (i.e., AKT activation) thus impeding the E2-induced cyclin D1 promoter activation and consequently cell proliferation. In addition, the interference with ERα monoUbq deregulates E2-induced association of ERα to the insulin like growth factor receptor (IGF-1-R). Altogether these data demonstrate an inherent role for monoUbq in ERα signaling and point to the physiological function of ERα monoUbq in the regulation of E2-induced cell proliferation.
Collapse
Affiliation(s)
- Piergiorgio La Rosa
- Department of Biology, University Roma Tre, Viale Guglielmo Marconi 446, Rome, Italy
| | | | | | | |
Collapse
|
31
|
Cittelly DM, Das PM, Spoelstra NS, Edgerton SM, Richer JK, Thor AD, Jones FE. Downregulation of miR-342 is associated with tamoxifen resistant breast tumors. Mol Cancer 2010; 9:317. [PMID: 21172025 PMCID: PMC3024251 DOI: 10.1186/1476-4598-9-317] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 12/20/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tumor resistance to the selective estrogen receptor modulator tamoxifen remains a serious clinical problem especially in patients with tumors that also overexpress HER2. We have recently demonstrated that the clinically important isoform of HER2, HERΔ16, promotes therapeutically refractory breast cancer including resistance to endocrine therapy. Likewise additional breast tumor cell models of tamoxifen resistance have been developed that do not involve HER2 overexpression. However, a unifying molecular mechanism of tamoxifen resistance has remained elusive. RESULTS Here we analyzed multiple cell models of tamoxifen resistance derived from MCF-7 cells to examine the influence of microRNAs (miRNAs) on tamoxifen resistance. We compared miRNA expression profiles of tamoxifen sensitive MCF-7 cells and tamoxifen resistant MCF-7/HER2Δ16 cells. We observed significant and dramatic downregulation of miR-342 in the MCF-7/HER2Δ16 cell line as well as the HER2 negative but tamoxifen resistant MCF-7 variants TAMR1 and LCC2. Restoring miR-342 expression in the MCF-7/HER2Δ16 and TAMR1 cell lines sensitized these cells to tamoxifen-induced apoptosis with a dramatic reduction in cell growth. Expression of miR-342 was also reduced in a panel of tamoxifen refractory human breast tumors, underscoring the potential clinical importance of miR-342 downregulation. Towards the goal of identifying direct and indirect targets of miR-342 we restored miR-342 expression in MCF-7/HER2Δ16 cells and analyzed changes in global gene expression by microarray. The impact of miR-342 on gene expression in MCF-7/HER2Δ16 cells was not limited to miR-342 in silica predicted targets. Ingenuity Pathways Analysis of the dataset revealed a significant influence of miR-342 on multiple tumor cell cycle regulators. CONCLUSIONS Our findings suggest that miR-342 regulates tamoxifen response in breast tumor cell lines and our clinical data indicates a trend towards reduced miR-342 expression and tamoxifen resistance. In addition, our results suggest that miR-342 regulates expression of genes involved in tamoxifen mediated tumor cell apoptosis and cell cycle progression. Restoring miR-342 expression may represent a novel therapeutic approach to sensitizing and suppressing the growth of tamoxifen refractory breast tumors.
Collapse
Affiliation(s)
- Diana M Cittelly
- Department of Pathology, University of Colorado Denver, School of Medicine, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Ghimenti C, Mello-Grand M, Regolo L, Zambelli A, Chiorino G. Absence of the K303R estrogen receptor α mutation in breast cancer patients exhibiting different responses to aromatase inhibitor anastrozole neoadjuvant treatment. Exp Ther Med 2010; 1:939-942. [PMID: 22993622 DOI: 10.3892/etm.2010.151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 09/09/2010] [Indexed: 01/30/2023] Open
Abstract
Aromatase inhibitors, such as anastrozole, are established in the treatment of hormone-dependent breast cancer. However, approximately 20% of patients treated with anastrozole do not respond, and it remains impossible to accurately predict sensitivity. Thus, novel markers to predict response are required. The K303R estrogen receptor (ER)α mutation confers resistance to tamoxifen treatment. Moreover, K303R-expressing MCF-7 cells, transfected with an aromatase expression vector and stimulated with androstenedione (an aromatase substrate), were found to be resistant to the inhibitory effect of anastrozole. The aim of this study was to verify whether the presence of the K303R ERα mutation is associated with response to 3-month neoadjuvant treatment with anastrozole (Arimidex) in a cohort of post-menopausal breast cancer patients. Of 37 patients with ER(+) tumors, 19 showed a clinical response to anastrozole and 18 were resistant. Biopsies were obtained from tumors responding to the therapy or from non-responding tumors. None carried the K303R ERα mutation. To our knowledge, this is the first study to search for K303R ERα mutations in tumors clinically responsive or resistant to an aromatase inhibitor. Lack of the mutation leads us to believe that this mutation has in vivo biological significance in only a subset of breast cancers.
Collapse
Affiliation(s)
- Chiara Ghimenti
- Cancer Genomics Laboratory, Fondazione 'Edo ed Elvo Tempia Valenta', Biella
| | | | | | | | | |
Collapse
|
33
|
Poulard C, Bouchekioua-Bouzaghou K, Sentis S, Corbo L, Le Romancer M. Les modifications post-traductionnelles orchestrent l’action du récepteur des œstrogènes εRα dans les tumeurs mammaires. Med Sci (Paris) 2010; 26:636-40. [DOI: 10.1051/medsci/2010266-7636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
34
|
Barone I, Brusco L, Fuqua SAW. Estrogen receptor mutations and changes in downstream gene expression and signaling. Clin Cancer Res 2010; 16:2702-8. [PMID: 20427689 DOI: 10.1158/1078-0432.ccr-09-1753] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Estrogens play a crucial role in regulating the growth and differentiation of breast cancers, with approximately two thirds of all breast tumors expressing the estrogen receptor alpha (ERalpha). Therefore, therapeutic strategies directed at inhibiting the action of ERalpha by using anti-estrogens such as tamoxifen, or reducing estrogens levels by using aromatase inhibitors, such as letrozole, anastrozole, or exemestane, are the standard treatments offered to women with ERalpha-positive cancer. However, not all patients respond to endocrine therapies (termed de novo resistance), and a large number of patients who do respond will eventually develop disease progression or recurrence while on therapy (acquired resistance). Recently, variant forms of the receptor have been identified owing to alternative splicing or gene mutation. This article reviews these variant receptors and their clinical relevance in resistance to endocrine therapy, by addressing their molecular cross-talk with growth factor receptors and signaling components. Understanding the complexity of receptor-mediated signaling has promise for new combined therapeutic options that focus on more efficient blockade of receptor cross-talk.
Collapse
Affiliation(s)
- Ines Barone
- Centro Sanitario and Department of Cellular Biology, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | | | | |
Collapse
|
35
|
Phosphorylation of the mutant K303R estrogen receptor alpha at serine 305 affects aromatase inhibitor sensitivity. Oncogene 2010; 29:2404-14. [PMID: 20101208 PMCID: PMC2922934 DOI: 10.1038/onc.2009.520] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We previously identified a lysine to arginine transition at residue 303 (K303R) in ERα in invasive breast cancers, which confers resistance to the aromatase inhibitor (AI) anastrozole (Ana) when expressed in MCF-7 breast cancer cells. Here we show that AI resistance arises through an enhanced cross-talk of the IGF-1R/IRS-1/Akt pathway with ERα, and the serine (S) residue 305 adjacent to the K303R mutation plays a key role in mediating this cross-talk. The ERα S305 residue is an important site that modifies response to tamoxifen; thus, we questioned whether this site could also influence AI response. We generated stable transfectants expressing wild-type (WT), K303R ERα, or a double K303R/S305A mutant receptor, and found that the AI-resistant phenotype associated with expression of the K303R mutation was dependent on activation of S305 within the receptor. Ana significantly reduced growth in K303R/S305A-expressing cells. Preventing S305 phosphorylation with a blocking peptide inhibited IGF-1R/IRS-1/Akt activation, and also restored AI sensitivity. Our data suggest that the K303R mutation and the S305 ERα residue may be a novel determinant of aromatase inhibitor response in breast cancer, and blockade of S305 phosphorylation represents a new therapeutic strategy for treating tumors resistant to hormone therapy.
Collapse
|
36
|
Ma Y, Fan S, Hu C, Meng Q, Fuqua SA, Pestell RG, Tomita YA, Rosen EM. BRCA1 regulates acetylation and ubiquitination of estrogen receptor-alpha. Mol Endocrinol 2010; 24:76-90. [PMID: 19887647 PMCID: PMC2802901 DOI: 10.1210/me.2009-0218] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 09/30/2009] [Indexed: 11/19/2022] Open
Abstract
Inherited mutations of the breast cancer susceptibility gene BRCA1 confer a high risk for breast cancer development. The (300)RXKK and (266)KXK motifs have been identified previously as sites for acetylation of the estrogen receptor-alpha (ER-alpha), and (302)K was also found to be a site for BRCA1-mediated mono-ubiquitination of ER-alpha in vitro. Here we show that ER-alpha proteins with single or double lysine mutations of these motifs (including K303R, a cancer-associated mutant) are resistant to inhibition by BRCA1, even though the mutant ER-alpha proteins retain the ability to bind to BRCA1. We also found that BRCA1 overexpression reduced and knockdown increased the level of acetylated wild-type ER-alpha, without changing the total ER-alpha protein level. Increased acetylation of ER-alpha due to BRCA1 small interfering RNA was dependent upon phosphatidylinositol 3-kinase/Akt signaling and on up-regulation of the coactivator p300. In addition, using an in vitro acetylation assay, we found that in vitro-translated wild-type BRCA1 but not a cancer-associated point mutant (C61G) inhibited p300-mediated acetylation of ER-alpha. Furthermore, BRCA1 overexpression increased the levels of mono-ubiquitinated ER-alpha protein, and a BRCA1 mutant that is defective for ubiquitin ligase activity but retains other BRCA1 functions (I26A) did not ubiquitinate ER-alpha or repress its activity in vivo. Finally, ER-alpha proteins with mutations of the (300)RXKK or (266)KXK motifs showed modest or no BRCA1-induced ubiquitination. We propose a model in which BRCA1 represses ER-alpha activity, in part, by regulating the relative degree of acetylation vs. ubiquitination of ER-alpha.
Collapse
Affiliation(s)
- Yongxian Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057-1469, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Barone I, Cui Y, Herynk MH, Corona-Rodriguez A, Giordano C, Selever J, Beyer A, Andò S, Fuqua SAW. Expression of the K303R estrogen receptor-alpha breast cancer mutation induces resistance to an aromatase inhibitor via addiction to the PI3K/Akt kinase pathway. Cancer Res 2009; 69:4724-32. [PMID: 19487288 DOI: 10.1158/0008-5472.can-08-4194] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aromatase inhibitors (AI) are rapidly becoming the first choice for hormonal treatment of estrogen receptor-alpha (ERalpha)-positive breast cancer in postmenopausal women. However, de novo and acquired resistance frequently occurs. We have previously identified a lysine to arginine transition at residue 303 (K303R) in ERalpha in premalignant breast lesions and invasive breast cancers, which confers estrogen hypersensitivity and resistance to tamoxifen treatment. Thus, we questioned whether resistance to AIs could arise in breast cancer cells expressing the ERalpha mutation. As preclinical models to directly test this possibility, we generated K303R-overexpressing MCF-7 cells stably transfected with an aromatase expression vector. Cells were stimulated with the aromatase substrate, androstenedione, with or without the AI anastrozole (Ana). We found that Ana decreased androstenedione-stimulated growth of wild-type cells, whereas K303R-expressing cells were resistant to the inhibitory effect of Ana on growth. We propose that a mechanism of resistance involves an increased binding between the mutant receptor and the p85alpha regulatory subunit of phosphatidylinositol-3-OH kinase (PI3K), leading to increased PI3K activity and activation of protein kinase B/Akt survival pathways. Inhibition of the selective "addiction" to the PI3K/Akt pathway reversed AI resistance associated with expression of the mutant receptor. Our findings suggest that the K303R ERalpha mutation might be a new predictive marker of response to AIs in mutation-positive breast tumors, and that targeting the PI3K/Akt pathway may be a useful strategy for treating patients with tumors resistant to hormone therapy.
Collapse
Affiliation(s)
- Ines Barone
- Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Søiland H, Skaland I, Varhaug JE, Kørner H, Janssen EA, Gudlaugsson E, Baak JP, Søreide JA. Co-expression of estrogen receptor alpha and Apolipoprotein D in node positive operable breast cancer--possible relevance for survival and effects of adjuvant tamoxifen in postmenopausal patients. Acta Oncol 2009; 48:514-21. [PMID: 19107621 DOI: 10.1080/02841860802620613] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Estrogen receptor-alpha (ERalpha) is an important prognostic and predictive marker in breast cancer. ERalpha signaling normally down-regulates expression of Apolipoprotein D (ApoD), a lipocalin that binds, transports or chelates lipophilic ligands, including tamoxifen (TAM). Hence, the co-expression of ApoD may therefore identify clinical relevant subgroups of ERalpha positive breast cancer patients. MATERIAL AND METHODS ApoD, ERalpha, and progesterone receptor (PR) protein expressions were determined by immunohistochemistry (IHC) in primary tumors of 290 patients with operable breast cancer. The median follow-up was 12 years. Patients were stratified according to age, nodal stage and the expression of ERalpha and the combined cytoplasm and nuclear staining of ApoD (ApoD(CN)). RESULTS In elderly women (> or =70 years) (n = 76), ApoD(CN) expression identified different prognostic subgroups in ERalpha positive patients (Trend: p < 0.0001). Multivariate analysis in this age group (n = 72), showed that the ERalpha-positive /ApoD(CN)-negative subgroup had a better breast cancer specific survival (BCSS) compared with the ERalpha-positive/ApoD(CN)-positive group (hazard ratio (HR) = 4.3; 95% CI = 1.6-11.9; p = 0.005). This difference was predominantly seen in the node positive patients (n = 30) (HR = 10.5; 95% CI = 2.3-47.6; p = 0.002). In a subset of postmenopausal ERalpha-positive/node positive patients (n = 60) previously enrolled in a trial on 2 year adjuvant TAM 20 mg vs. placebo, a better BCSS was observed in ApoD(CN) negative patients compared to placebo (p = 0.02). In ApoD(CN) positive patients, adjuvant TAM did not provide any survival benefit. DISCUSSION ERalpha and ApoD(CN) co-expression seems to be of prognostic importance in node positive elderly patients with operable breast cancer. In addition, we hypothesize that ApoD(CN) expression may be a novel marker and/or mechanism of TAM resistance in postmenopausal node positive patients. Thus, when targeting the ERalpha pathway in these patients, the ApoD status of the tumor may be of clinical relevance.
Collapse
|
39
|
Accelerated mammary maturation and differentiation, and delayed MMTVneu-induced tumorigenesis of K303R mutant ERalpha transgenic mice. Oncogene 2009; 28:3177-87. [PMID: 19561644 DOI: 10.1038/onc.2009.174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We identified a somatic mutation in estrogen receptor-alpha (ERalpha) in breast cancer causing a lysine to arginine transition (K303R) resulting in hypersensitivity to estrogen, altered associations with coactivators and corepressors and altered posttranslational modifications of ERalpha. We have developed a transgenic mouse expressing the K303R mutant ERalpha under control of the mouse mammary tumor virus (MMTV) promoter. At 4 months of age, K303R ERalpha transgenic animals demonstrate precocious alveolar budding compared with wild-type ERalpha transgenic mice or nontransgenic littermates. Despite these morphologic differences, K303R ERalpha transgenic mice displayed no differences in levels of ERalpha, progesterone receptor or proliferation at this time-point. Pregnancy or chronic estrogen plus progesterone exposure in K303R ERalpha transgenic mice also resulted in significantly more alveolar budding, increased beta-casein production and dilated ducts when compared with nontransgenic littermates. To examine the effects of mutant expression on tumorigenesis, mutant ERalpha mice were crossed with FVB-MMTVneu mice and significantly delayed time to neu-mediated tumorigenesis in bigenic animals. In contrast, mutant expression did not affect carcinogen-induced tumorigenesis. Collectively, these data demonstrate that aberrant estrogenic signaling through the K303R ERalpha mutation may lead to precocious alveolar budding in virgin mice, and to an expedited maturation and differentiation phenotype in the mammary glands of hormonally stimulated animals.
Collapse
|
40
|
Giordano C, Cui Y, Barone I, Ando S, Mancini MA, Berno V, Fuqua SAW. Growth factor-induced resistance to tamoxifen is associated with a mutation of estrogen receptor alpha and its phosphorylation at serine 305. Breast Cancer Res Treat 2009; 119:71-85. [PMID: 19205871 DOI: 10.1007/s10549-009-0334-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Accepted: 01/30/2009] [Indexed: 02/06/2023]
Abstract
Estrogens play a crucial role in breast tumor growth, which is the rationale for the use of antiestrogens, such as tamoxifen, in women with estrogen receptor (ER)-alpha-positive breast cancer. However, hormone resistance is a major clinical problem. Altered growth factor signaling to the ERalpha pathway has been shown to be associated with the development of clinical resistance. We previously have identified a mutation that replaces arginine for lysine at residue 303 (K303R) of ERalpha, which confers hypersensitive growth in low levels of estrogen. To determine if the K303R mutation could participate in the evolution of hormone resistance, we generated MCF-7 breast cancer cells stably transfected with either wild-type (WT) or K303R ERalpha. We found that the mutation confers decreased sensitivity to tamoxifen in the presence of the growth factor heregulin, using anchorage-independent growth assays. K303R ERalpha-expressing cells were hypersensitive to growth factor signals. Our data suggest that phosphorylation of serine 305 within the hinge domain of ERalpha might play a key role in increasing ligand-independent activity of the mutant receptor. We hypothesize that the mutation adapts the receptor for enhanced bidirectional cross-talk with the HER2 growth factor receptor pathway, which then impacts on responsiveness to tamoxifen.
Collapse
Affiliation(s)
- Cinzia Giordano
- Department of Pharmaco-Biology, University of Calabria, Rende, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Subramanian K, Jia D, Kapoor-Vazirani P, Powell DR, Collins RE, Sharma D, Peng J, Cheng X, Vertino PM. Regulation of estrogen receptor alpha by the SET7 lysine methyltransferase. Mol Cell 2008; 30:336-47. [PMID: 18471979 DOI: 10.1016/j.molcel.2008.03.022] [Citation(s) in RCA: 231] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 12/04/2007] [Accepted: 03/04/2008] [Indexed: 02/07/2023]
Abstract
Estrogen receptor alpha (ER) is a ligand-dependent transcription factor. Upon binding estrogen, ER recruits coactivator complexes with histone acetyltransferase or methyltransferase activities to activate downstream target genes. In addition to histones, coactivators can modify ER itself and other proteins in the transactivation complex. Here, we show that ER is directly methylated at lysine 302 (K302) by the SET7 methyltransferase. SET7-mediated methylation stabilizes ER and is necessary for the efficient recruitment of ER to its target genes and for their transactivation. The SET7-ER complex structure reveals the molecular basis for ER peptide recognition and predicts that modifications or mutations of nearby residues would affect K302 methylation. Indeed, a breast cancer-associated mutation at K303 (K303R) alters methylation at K302 in vitro and in vivo. These findings raise the possibility that generation, recognition, and removal of modifications within the ER hinge region generate "ER modification cassettes" that yield distinct patterns for signaling downstream events.
Collapse
Affiliation(s)
- Krithika Subramanian
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|