1
|
Fabre ML, Canzoneri R, Gurruchaga A, Lee J, Tatineni P, Kil H, Lacunza E, Aldaz CM, Abba MC. MALINC1 an Immune-Related Long Non-Coding RNA Associated with Early-Stage Breast Cancer Progression. Cancers (Basel) 2022; 14:cancers14122819. [PMID: 35740485 PMCID: PMC9221538 DOI: 10.3390/cancers14122819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Here we characterize the phenotypic and molecular effects of MALINC1, a long non-coding RNA (lncRNA) that we found significantly upregulated in premalignant ductal carcinoma in-situ lesions. We provide evidence that MALINC1 behaves as an oncogenic and immune-related lncRNA involved with early-stage breast cancer progression, showing prognostic and predictive value to immunotherapy in invasive breast carcinomas. Abstract Long non-coding RNAs are increasingly being recognized as cancer biomarkers in various malignancies, acting as either tumor suppressors or oncogenes. The long non-coding MALINC1 intergenic RNA was identified as significantly upregulated in breast ductal carcinoma in situ. The aim of this study was to characterize MALINC1 expression, localization, and phenotypic and molecular effects in non-invasive and invasive breast cancer cells. We determined that MALINC1 is an estrogen–estrogen receptor-modulated lncRNA enriched in the cytoplasmic fraction of luminal A/B breast cancer cells that is associated with worse overall survival in patients with primary invasive breast carcinomas. Transcriptomic studies in normal and DCIS cells identified the main signaling pathways modulated by MALINC1, which mainly involve bioprocesses related to innate and adaptive immune responses, extracellular matrix remodeling, cell adhesion, and activation of AP-1 signaling pathway. We determined that MALINC1 induces premalignant phenotypic changes by increasing cell migration in normal breast cells. Moreover, high MALINC1 expression in invasive carcinomas was associated with a pro-tumorigenic immune environment and a favorable predicted response to immunotherapy both in luminal and basal-like subtypes compared with low-MALINC1-expression tumors. We conclude that MALINC1 behaves as an oncogenic and immune-related lncRNA involved with early-stage breast cancer progression.
Collapse
Affiliation(s)
- María Laura Fabre
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina; (M.L.F.); (R.C.); (A.G.); (E.L.)
| | - Romina Canzoneri
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina; (M.L.F.); (R.C.); (A.G.); (E.L.)
| | - Agustina Gurruchaga
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina; (M.L.F.); (R.C.); (A.G.); (E.L.)
| | - Jaeho Lee
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (J.L.); (P.T.); (H.K.)
| | - Pradeep Tatineni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (J.L.); (P.T.); (H.K.)
| | - Hyunsuk Kil
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (J.L.); (P.T.); (H.K.)
| | - Ezequiel Lacunza
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina; (M.L.F.); (R.C.); (A.G.); (E.L.)
| | - C. Marcelo Aldaz
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; (J.L.); (P.T.); (H.K.)
- Correspondence: (C.M.A.); (M.C.A.)
| | - Martín Carlos Abba
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata 1900, Argentina; (M.L.F.); (R.C.); (A.G.); (E.L.)
- Correspondence: (C.M.A.); (M.C.A.)
| |
Collapse
|
2
|
Karakolevska-Ilova M, Zdravkovska M, Jasar D, Petrushevska G, Simeonovska-Joveva E. Can Estrogen Receptor, Progesterone Receptor, and Proliferative Index be Considered as Isolated Prognostic Factors of Overall Survival in Early luminal Breast Cancer? Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: There are a number of proven molecular and pathological markers important for the prognosis for OS of early luminal type breast cancer, but there are still some deficiencies mainly due to the non-linear relationship between the markers and the outcome of the disease.
METHODS AND PATIENTS: In this retro-prospective study, clinical and pathological data were provided from 336 patients with breast cancer who underwent breast surgery and treatment between January 2010 and December 2013, and followed until December 2018. The aim of the study was an evaluation of ER, PR, and Ki-67 as independent prognostic factors for OS of early luminal breast cancer.
RESULTS: Early luminal breast cancers were not predictive of ER expression status (p = 0.699, p = 0.356), whereas only early Luminal B was predictive for PR expression (>10%: 72.2%). Ki-67 in most of the cases of early Luminal B was with expression of >14–20% (p = 0.056). Patients with ER of 1–10% survived over 80 months (p = 0.0020) in early Luminal A, but ER expression status did not show prognostic significance for OS of early Luminal B (p = 0.775). PR status did not show prognostic significance for OS in early luminal types (p = 0.257, p = 0.622). ER >1%/PR >1% was protective in early Luminal B (p = 0.00043), but not in early Luminal A.
CONCLUSION: Our results suggest ER, but not PR as independent prognostic factor for OS but only of early Luminal A. We did not prove Ki-67 as independent prognostic factor for OS of highly proliferative early breast cancer.
Collapse
|
3
|
Suleiman RB, Muhammad A, Umara IA, Ibrahima MA, Erukainure OL, Forcados GE, Katsayal SB. Kolaviron Ameliorates 7, 12-Dimethylbenzanthracene - Induced Mammary Damage in Female Wistar Rats. Anticancer Agents Med Chem 2021; 22:181-192. [PMID: 34225638 DOI: 10.2174/1871520621666210322101232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/27/2020] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Kolaviron (KV) is a flavonoid rich portion obtained from Garcinia kola seeds with a number of reported pharmacological effects. However, its ameliorative effects on 7,12-Dimethylbenzanthracene (DMBA)-induced mammary damage has not been fully investigated, despite the reported use of the seeds in the treatment of inflammatory related disorders. OBJECTIVE To evaluate the ameliorative effects of KV on DMBA-induced mammary damage in female Wistar rats. METHODS Forty-nine (49) female Wistar rats were randomly assigned into seven groups of seven rats each. DMBA was administered orally to rats in five of the groups as a single dose of 80 mg/kg body wt while the remaining two groups received the vehicle. The rats were palpated weekly for 3 months to monitor tumor formation. After 3 months of DMBA administration, 1 ml of blood was collected to assay for estrogen receptor- α (ER-α) level. Thereafter, the vehicle (dimethyl sulfoxide) was daily administered to the negative control and positive control groups for the 14 days duration of the experiment while three groups were each given a daily oral dose of 50, 100 and 200 mg/kg body wt of KV for the duration of the experiment. The last DMBA-induced group received 10 mg/kg body wt of the standard drug tamoxifen twice in a week and the remaining DMBA-free group received 200 mg/kg body wt KV. Subsequently, the animals were humanly sacrificed and ER-α, sialic acids, sialidase, sialyltransferase levels were assay for in blood and mammary tissues followed by histopathological examinations. RESULTS Significantly higher levels of estrogen receptor-α (ER-α), formation of lobular neoplastic cells, epithelial hyperplasia, lymphocyte infiltration and increased sialylation were detected in DMBA-induced rats. Treatment with KV at 50, 100 and 200 mg/kg body weight resulted in a significant (p<0.05) decrease in ER-α level, significantly (p<0.05) lower free serum sialic acid (21.1%), total sialic acid level of the mammary tissue (21.57%), sialyltransferase activity (30.83%) as well as mRNA level of the sialyltransferase gene (ST3Gal1) were observed after KV interventions. CONCLUSION The findings suggest that KV could be further explored in targeting DMBA-induced mammary damage implicated in mammary carcinogenesis.
Collapse
Affiliation(s)
- Rabiatu B Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Ismaila A Umara
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mohammed A Ibrahima
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein 9300. South Africa
| | - Gilead E Forcados
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Sanusi B Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| |
Collapse
|
4
|
De Amicis F, Chiodo C, Morelli C, Casaburi I, Marsico S, Bruno R, Sisci D, Andò S, Lanzino M. AIB1 sequestration by androgen receptor inhibits estrogen-dependent cyclin D1 expression in breast cancer cells. BMC Cancer 2019; 19:1038. [PMID: 31684907 PMCID: PMC6829973 DOI: 10.1186/s12885-019-6262-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Background Androgens, through their own receptor, play a protective role on breast tumor development and progression and counterbalance estrogen-dependent growth stimuli which are intimately linked to breast carcinogenesis. Methods Cell counting by trypan blu exclusion was used to study androgen effect on estrogen-dependent breast tumor growth. Quantitative Real Time RT–PCR, western blotting, transient transfection, protein immunoprecipitation and chromatin immunoprecipitation assays were carried out to investigate how androgen treatment and/or androgen receptor overexpression influences the functional interaction between the steroid receptor coactivator AIB1 and the estrogen- or androgen receptor which, in turn affects the estrogen-induced cyclin D1 gene expression in MCF-7 breast cancer cells. Data were analyzed by ANOVA. Results Here we demonstrated, in estrogen receptor α (ERα)-positive breast cancer cells, an androgen-dependent mechanism through which ligand-activated androgen receptor (AR) decreases estradiol-induced cyclin D1 protein, mRNA and gene promoter activity. These effects involve the competition between AR and ERα for the interaction with the steroid receptor coactivator AIB1, a limiting factor in the functional coupling of the ERα with the cyclin D1 promoter. Indeed, AIB1 overexpression is able to reverse the down-regulatory effects exerted by AR on ERα-mediated induction of cyclin D1 promoter activity. Co-immunoprecipitation studies indicated that the preferential interaction of AIB1 with ERα or AR depends on the intracellular expression levels of the two steroid receptors. In addition, ChIP analysis evidenced that androgen administration decreased E2-induced recruitment of AIB1 on the AP-1 site containing region of the cyclin D1 gene promoter. Conclusions Taken together all these data support the hypothesis that AIB1 sequestration by AR may be an effective mechanism to explain the reduction of estrogen-induced cyclin D1 gene activity. In estrogen-dependent breast cancer cell proliferation, these findings reinforce the possibility that targeting AR signalling may potentiate the effectiveness of anti-estrogen adjuvant therapies.
Collapse
Affiliation(s)
- Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Chiara Chiodo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Ivan Casaburi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Rosalinda Bruno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Diego Sisci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy.
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| | - Marilena Lanzino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, CS, 87036, Arcavacata di Rende, Italy
| |
Collapse
|
5
|
Peterson A, Xia Z, Chen G, Lazarus P. In vitro metabolism of exemestane by hepatic cytochrome P450s: impact of nonsynonymous polymorphisms on formation of the active metabolite 17 β-dihydroexemestane. Pharmacol Res Perspect 2017; 5:e00314. [PMID: 28603633 PMCID: PMC5464343 DOI: 10.1002/prp2.314] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 11/29/2022] Open
Abstract
Exemestane (EXE) is an endocrine therapy commonly used by postmenopausal women with hormone‐responsive breast cancer due to its potency in inhibiting aromatase‐catalyzed estrogen synthesis. Preliminary in vitro studies sought to identify phase I EXE metabolites and hepatic cytochrome P450s (CYP450s) that participate in EXE biotransformation. Phase I metabolites were identified by incubating EXE with HEK293‐overexpressed CYP450s. CYP450s 1A2, 2C8, 2C9, 2C19, 2D6, 3A4, and 3A5 produce 17β‐dihydroexemestane (17β‐DHE), an active major metabolite, as well as two inactive metabolites. 17β‐DHE formation in pooled human liver microsomes subjected to isoform‐specific CYP450 inhibition was also monitored using tandem mass spectrometry. 17β‐DHE production in human liver microsomes was unaffected by isoform‐specific inhibition of CYP450s 2A6, 2B6, and 2E1 but decreased 12–39% following inhibition of drug‐metabolizing enzymes from CYP450 subfamilies 1A, 2C, 2D, and 3A. These results suggest that redundancy exists in the EXE metabolic pathway with multiple hepatic CYP450s catalyzing 17β‐DHE formation in vitro. To further expand the knowledge of phase I EXE metabolism, the impact of CYP450 genetic variation on 17β‐DHE formation was assessed via enzyme kinetic parameters. Affinity for EXE substrate and enzyme catalytic velocity were calculated for hepatic wild‐type CYP450s and their common nonsynonymous variants by monitoring the reduction of EXE to 17β‐DHE. Several functional polymorphisms in xenobiotic‐metabolizing CYP450s 1A2, 2C8, 2C9, and 2D6 resulted in deviant enzymatic activity relative to wild‐type enzyme. Thus, it is possible that functional polymorphisms in EXE‐metabolizing CYP450s contribute to inter‐individual variability in patient outcomes by mediating overall exposure to the drug and its active metabolite, 17β‐DHE.
Collapse
Affiliation(s)
- Amity Peterson
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| | - Zuping Xia
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| | - Gang Chen
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| | - Philip Lazarus
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| |
Collapse
|
6
|
Engel N, Adamus A, Schauer N, Kühn J, Nebe B, Seitz G, Kraft K. Synergistic Action of Genistein and Calcitriol in Immature Osteosarcoma MG-63 Cells by SGPL1 Up-Regulation. PLoS One 2017; 12:e0169742. [PMID: 28125641 PMCID: PMC5268493 DOI: 10.1371/journal.pone.0169742] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 12/21/2016] [Indexed: 11/18/2022] Open
Abstract
Background Phytoestrogens such as genistein, the most prominent isoflavone from soy, show concentration-dependent anti-estrogenic or estrogenic effects. High genistein concentrations (>10 μM) also promote proliferation of bone cancer cells in vitro. On the other hand, the most active component of the vitamin D family, calcitriol, has been shown to be tumor protective in vitro and in vivo. The purpose of this study was to examine a putative synergism of genistein and calcitriol in two osteosarcoma cell lines MG-63 (early osteoblast), Saos-2 (mature osteoblast) and primary osteoblasts. Methods Thus, an initial screening based on cell cycle phase alterations, estrogen (ER) and vitamin D receptor (VDR) expression, live cell metabolic monitoring, and metabolomics were performed. Results Exposure to the combination of 100 μM genistein and 10 nM calcitriol reduced the number of proliferative cells to control levels, increased ERß and VDR expression, and reduced extracellular acidification (40%) as well as respiratory activity (70%), primarily in MG-63 cells. In order to identify the underlying cellular mechanisms in the MG-63 cell line, metabolic profiling via GC/MS technology was conducted. Combined treatment significantly influenced lipids and amino acids preferably, whereas metabolites of the energy metabolism were not altered. The comparative analysis of the log2-ratios revealed that after combined treatment only the metabolite ethanolamine was highly up-regulated. This is the result: a strong overexpression (350%) of the enzyme sphingosine-1-phosphate lyase (SGPL1), which irreversibly degrades sphingosine-1-phosphate (S1P), thereby, generating ethanolamine. S1P production and secretion is associated with an increased capability of migration and invasion of cancer cells. Conclusion From these results can be concluded that the tumor promoting effect of high concentrations of genistein in immature osteosarcoma cells is reduced by the co-administration of calcitriol, primarily by the breakdown of S1P. It should be tested whether this anti-metastatic pathway can be stimulated by combined treatment also in metastatic xenograft mice models.
Collapse
Affiliation(s)
- Nadja Engel
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, Marburg, Germany
- Department of Cell Biology, Rostock University Medical Center, Schillingallee, Rostock, Germany
- * E-mail: ,
| | - Anna Adamus
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, Marburg, Germany
- Department of Cell Biology, Rostock University Medical Center, Schillingallee, Rostock, Germany
| | - Nicolas Schauer
- Metabolomic Discoveries GmbH, Am Mühlenberg, Potsdam-Golm, Germany
| | - Juliane Kühn
- Department of Cell Biology, Rostock University Medical Center, Schillingallee, Rostock, Germany
| | - Barbara Nebe
- Department of Cell Biology, Rostock University Medical Center, Schillingallee, Rostock, Germany
| | - Guido Seitz
- Department of Pediatric Surgery, University Hospital Marburg, Baldingerstraße, Marburg, Germany
| | - Karin Kraft
- Complementary Medicine, Center of Internal Medicine, Rostock University Medical Center, Ernst-Heydemann-Straße 6, Rostock, Germany
| |
Collapse
|
7
|
Browning HM, Gulland FMD, Hammond JA, Colegrove KM, Hall AJ. Common cancer in a wild animal: the California sea lion (Zalophus californianus) as an emerging model for carcinogenesis. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0228. [PMID: 26056370 DOI: 10.1098/rstb.2014.0228] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Naturally occurring cancers in non-laboratory species have great potential in helping to decipher the often complex causes of neoplasia. Wild animal models could add substantially to our understanding of carcinogenesis, particularly of genetic and environmental interactions, but they are currently underutilized. Studying neoplasia in wild animals is difficult and especially challenging in marine mammals owing to their inaccessibility, lack of exposure history, and ethical, logistical and legal limits on experimentation. Despite this, California sea lions (Zalophus californianus) offer an opportunity to investigate risk factors for neoplasia development that have implications for terrestrial mammals and humans who share much of their environment and diet. A relatively accessible California sea lion population on the west coast of the USA has a high prevalence of urogenital carcinoma and is regularly sampled during veterinary care in wildlife rehabilitation centres. Collaborative studies have revealed that genotype, persistent organic pollutants and a herpesvirus are all associated with this cancer. This paper reviews research to date on the epidemiology and pathogenesis of urogenital carcinoma in this species, and presents the California sea lion as an important and currently underexploited wild animal model of carcinogenesis.
Collapse
Affiliation(s)
- Helen M Browning
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, St Andrews KY16 8LB, UK
| | | | | | - Kathleen M Colegrove
- Zoological Pathology Program, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Maywood, IL 60153, USA
| | - Ailsa J Hall
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, St Andrews KY16 8LB, UK
| |
Collapse
|
8
|
Spoerri M, Guscetti F, Hartnack S, Boos A, Oei C, Balogh O, Nowaczyk RM, Michel E, Reichler IM, Kowalewski MP. Endocrine control of canine mammary neoplasms: serum reproductive hormone levels and tissue expression of steroid hormone, prolactin and growth hormone receptors. BMC Vet Res 2015; 11:235. [PMID: 26370564 PMCID: PMC4570623 DOI: 10.1186/s12917-015-0546-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/21/2015] [Indexed: 01/17/2023] Open
Abstract
Background Neoplasms of the mammary gland are among the most common diseases in female domestic dogs (Canis familiaris). It is assumed that reproductive hormones influence tumorigenesis in this species, although the precise role of the endocrine milieu and reproductive state is subject to continuing discussion. In line with this, a recent systematic review of available data on the development of mammary neoplasms revealed weak evidence for risk reduction after neutering and an effect of age at neutering. Investigation of several hormone receptors has revealed decreased expression of estrogen receptor-alpha (ERα, ESR1), progesterone (P4) receptor (PGR), prolactin (PRL) receptor (PRLR) and growth hormone receptor (GHR) associated with neoplastic differentiation of mammary tissues. In other studies, increased levels of estrogens, progesterone and prolactin were found in serum and/or tissue homogenates of dogs with malignant neoplasms. However, the association between these entities within one animal population was never previously examined. Therefore, this study investigated the association between circulating serum concentrations of estradiol-17β, progesterone and prolactin, and gene expression of ERα (ESR1), ERβ (ESR2), PGR, PRLR, PRL and GHR, with respect to reproductive state (spayed vs. intact) and cycle stage (anestrus vs. diestrus). Additionally, the expression of E-cadherin (CDH-1) was evaluated as a possible indicator of metastatic potential. Results For all receptors, the lowest gene expression was found in malignant tumors compared to normal tissues of affected dogs. Steroid levels were not influenced by their corresponding receptor expression in mammary neoplasms, but increased PRL levels were negatively associated with low PRLR gene expression in malignant tumors. The expression of CDH-1 was influenced by tumor malignancy and cycle stage, i.e., the highest gene expression was found in benign mammary tumors in diestrous dogs compared to normal and malignant mammary tissues of anestrous and spayed dogs. Conclusions Herein, it has been confirmed that transformation towards malignant neoplasms is associated with significant reduction of gene expression of particular hormone receptors. Only PRLR in malignant tumors seems to be influenced by circulating PRL levels. In dogs, CDH-1 can be used as a prognostic factor; its expression, however, in benign tumors is influenced by cycle stage. Electronic supplementary material The online version of this article (doi:10.1186/s12917-015-0546-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michèle Spoerri
- Clinic of Reproductive Medicine, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland. .,Institute of Veterinary Anatomy, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland.
| | - Franco Guscetti
- Institute of Veterinary Pathology, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 268/272, Zurich, 8057, Switzerland.
| | - Sonja Hartnack
- Section of Epidemiology, Vetsuisse Faculty, Winterthurerstrasse 270, Zurich, 8057, Switzerland.
| | - Alois Boos
- Institute of Veterinary Anatomy, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland.
| | - Christine Oei
- Department of Animal Health, Faculty of Veterinary Medicine, Utrecht University, P.O Box 80125, Utrecht, 3508 TC, The Netherlands.
| | - Orsolya Balogh
- Clinic of Reproductive Medicine, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland.
| | - Renata M Nowaczyk
- Division of Animal Anatomy, Department of Animal Physiology and Biostructure, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland.
| | - Erika Michel
- Clinic of Reproductive Medicine, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland.
| | - Iris M Reichler
- Clinic of Reproductive Medicine, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland.
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty University of Zurich, Winterthurerstrasse 260, Zurich, 8057, Switzerland.
| |
Collapse
|
9
|
Basak P, Chatterjee S, Weger S, Bruce MC, Murphy LC, Raouf A. Estrogen regulates luminal progenitor cell differentiation through H19 gene expression. Endocr Relat Cancer 2015; 22:505-17. [PMID: 25944846 PMCID: PMC4498491 DOI: 10.1530/erc-15-0105] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although the role of estrogen signaling in breast cancer development has been extensively studied, the mechanisms that regulate the indispensable role of estrogen in normal mammary gland development have not been well studied. Because of the unavailability of culture system to maintain estrogen-receptor-positive (ERα(+)) cells in vitro, the molecular mechanisms that regulate estrogen/ERα signaling in the normal human breast are unknown. In the present study, we examined the effects of estrogen signaling on ERα(+) human luminal progenitors using a modified matrigel assay and found that estrogen signaling increased the expansion potential of these progenitors. Furthermore, we found that blocking ERα attenuated luminal progenitor expansion and decreased the luminal colony-forming potential of these progenitors. Additionally, blocking ERα decreased H19 expression in the luminal progenitors and led to the development of smaller luminal colonies. We further showed that knocking down the H19 gene in the luminal progenitors significantly decreased the colony-forming potential of the luminal progenitors, and this phenotype could not be rescued by the addition of estrogen. Lastly, we explored the clinical relevance of the estrogen-H19 signaling axis in breast tumors and found that ERα(+) tumors exhibited a higher expression of H19 as compared with ERα(-) tumors and that H19 expression showed a positive correlation with ERα expression in those tumors. Taken together, the present results indicate that the estrogen-ERα-H19 signaling axis plays a role in regulating the proliferation and differentiation potentials of the normal luminal progenitors and that this signaling network may also be important in the development of ER(+) breast cancer tumors.
Collapse
Affiliation(s)
- Pratima Basak
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Sumanta Chatterjee
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Steven Weger
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - M Christine Bruce
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Leigh C Murphy
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| | - Afshin Raouf
- Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2 Department of ImmunologyUniversity of Manitoba, 471 Apotex Centre 750 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0T5Manitoba Institute of Cell Biology675 McDermot Avenue, Winnipeg, Manitoba, Canada R3E 0V9Department of Biochemistry and Medical GeneticsUniversity of Manitoba, Winnipeg, Manitoba, Canada R3E 0W2
| |
Collapse
|
10
|
Lim H, Im K, Kim N, Kim H, Shin J, Sur J. Obesity, expression of adipocytokines, and macrophage infiltration in canine mammary tumors. Vet J 2015; 203:326-31. [DOI: 10.1016/j.tvjl.2015.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 01/28/2023]
|
11
|
Murray JI, West NR, Murphy LC, Watson PH. Intratumoural inflammation and endocrine resistance in breast cancer. Endocr Relat Cancer 2015; 22:R51-67. [PMID: 25404688 DOI: 10.1530/erc-14-0096] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is becoming clear that inflammation-associated mechanisms can affect progression of breast cancer and modulate responses to treatment. Estrogen receptor alpha (ERα (ESR1)) is the principal biomarker and therapeutic target for endocrine therapies in breast cancer. Over 70% of patients are ESR1-positive at diagnosis and are candidates for endocrine therapy. However, ESR1-positive tumours can become resistant to endocrine therapy. Multiple mechanisms of endocrine resistance have been proposed, including suppression of ESR1. This review discusses the relationship between intratumoural inflammation and endocrine resistance with a particular focus on inflammation-mediated suppression of ESR1.
Collapse
Affiliation(s)
- Jill I Murray
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Nathan R West
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Leigh C Murphy
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Peter H Watson
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Kim NH, Lim HY, Im KS, Shin JI, Kim HW, Sur JH. Evaluation of Clinicopathological Characteristics and Oestrogen Receptor Gene Expression in Oestrogen Receptor-negative, Progesterone Receptor-positive Canine Mammary Carcinomas. J Comp Pathol 2014; 151:42-50. [DOI: 10.1016/j.jcpa.2014.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 02/17/2014] [Accepted: 04/10/2014] [Indexed: 12/23/2022]
|
13
|
Aiad HA, Wahed MMAE, Asaad NY, El-Tahmody M, Elhosary E. Immunohistochemical expression of GPR30 in breast carcinoma of Egyptian patients: an association with immunohistochemical subtypes. APMIS 2014; 122:976-84. [PMID: 24628533 DOI: 10.1111/apm.12241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 12/02/2013] [Indexed: 11/27/2022]
Abstract
Breast carcinoma in Egyptian women is a biologically more aggressive disease than those diagnosed in Western women, although a substantial number of cases are hormone responsive. G protein-coupled receptor-30 (GPR30), a seven transmembrane domain protein, is currently recognized as an estrogen receptor. This study aimed at evaluating the expression of GPR30 in breast carcinomas of Egyptian patients and its association with clinicopathologic parameters and immunohistochemical subtypes of breast carcinoma. Immunohistochemical staining for GPR30 was applied on 51 archival formalin-fixed paraffin-embedded cases of invasive ductal carcinoma. Staining was assessed using a semiquantitative scoring system taking staining intensity and extent into consideration. GPR30 was observed in 33/51 (65%) of invasive ductal carcinoma cases. GPR30 was significantly associated with larger tumor size (p = 0.009), increased number of positive lymph nodes (p = 0.04), definite lymph-vascular invasion (LVI) (p = 0.002), peri-nodal invasion (p = 0.02), and the presence of coagulative tumor cell necrosis (p = 0.02). Moreover, a significant association between positive GPR30 expression and ER positivity (p = 0.02), as well as HER2/neu positivity (p = 0.03), were also observed. Most of the luminal A and B subtypes were GPR30 positive; however, all the triple negative cases were GPR30 negative (p = 0.010). GPR30 might contribute to the aggressive behavior of Egyptian breast carcinoma. Therefore, it could be useful in the therapeutic decision making in breast cancer patients.
Collapse
Affiliation(s)
- Hayam A Aiad
- Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | | | | | | | | |
Collapse
|
14
|
Sundara Rajan S, Horgan K, Speirs V, Hanby AM. External validation of the ImmunoRatio image analysis application for ERα determination in breast cancer. J Clin Pathol 2013; 67:72-5. [DOI: 10.1136/jclinpath-2013-201680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
15
|
Habashy HO, Powe DG, Abdel-Fatah TM, Gee JMW, Nicholson RI, Green AR, Rakha EA, Ellis IO. A review of the biological and clinical characteristics of luminal-like oestrogen receptor-positive breast cancer. Histopathology 2011; 60:854-63. [PMID: 21906125 DOI: 10.1111/j.1365-2559.2011.03912.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Global gene expression profiling (GEP) studies of breast cancer have identified distinct biological classes with different clinical and therapeutic implications. Oestrogen receptor (ER) has been found to be a central marker of the molecular signature. GEP studies have consistently recognized a molecularly distinct class of tumours that is characterized by high-level expression of ER and other biomarkers recognized to be characteristic of normal luminal cells of the breast. This class is the largest of the GEP-defined molecular subclasses, comprising 60-70% of breast cancer cases. Moreover, it has been proposed that this group of tumours is composed of at least two subclasses distinguished by differing GEP profiles. At present, there is no consensus on the definition of the luminal subclasses and, in clinical practice, luminal-like tumours and ER-positive tumours are frequently considered to be the same. A better understanding of the biological features of luminal tumours could lead to their improved characterization and consistent identification. In this review, we explore the concept and definitions of the luminal-like class of breast carcinoma and their contribution to our understanding of their molecular features, clinical significance and therapeutic implications.
Collapse
Affiliation(s)
- Hany O Habashy
- Division of Pathology, School of Molecular Medical Sciences, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Overexpression of ligase defective E6-associated protein, E6-AP, results in mammary tumorigenesis. Breast Cancer Res Treat 2011; 132:97-108. [PMID: 21553290 DOI: 10.1007/s10549-011-1567-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 04/29/2011] [Indexed: 02/06/2023]
Abstract
E6-associated protein (E6-AP) is a dual function protein. It acts as an E3 ubiquitin-protein ligase enzyme and coactivator of steroid hormone receptors such as estrogen (ERα) and progesterone (PR) receptors. It promotes the degradation of ERα and PR through the ubiquitin-proteasome pathway. Furthermore, it has been shown that the levels of E6-AP are inversely associated with that of ERα in human breast tumors. But the role of wild-type human E6-AP and its ubiquitin-protein ligase activity in mammary tumorigenesis is still unknown. To investigate this role, the authors utilized transgenic mice lines that specifically overexpress either the wild-type human E6-AP (E6-AP(WT)) or the ubiquitin-protein ligase defective E6-AP that contains C833S mutation (E6-AP(C833S)) in the mammary gland. To further substantiate the role of E6-AP in the development of breast tumorigenesis, it was also examined the expression of E6-AP in a large cohort of human breast cancer samples. The transgenic mice that overexpress wild-type E6-AP (E6-AP(WT)) fail to develop mammary tumors. Unlike the E6-AP(WT) mice, the E6-AP(C833S) mice that overexpress ubiquitin-protein ligase defective E6-AP protein develop mammary hyperplasia with a median latency of 18 months. These observations suggest that the inactivation of the ubiquitin-protein ligase function of E6-AP is sufficient to initiate the process of mammary tumor development. Furthermore, the data also suggests that E6-AP exerts its effects on target cells by modulating the protein levels and functions of ERα and PR. In addition, it was found in human breast cancer patients that the level of E6-AP is decreased in invasive breast tumors compared to normal breast tissue. Moreover, the authors also show that the survival patterns for E6-AP negative patients were worse compared to E6-AP positive patients. Taken together, these data suggests that E6-AP may act as a tumor suppressor in breast.
Collapse
|
17
|
Habashy HO, Rakha EA, Aleskandarany M, Ahmed MA, Green AR, Ellis IO, Powe DG. FOXO3a nuclear localisation is associated with good prognosis in luminal-like breast cancer. Breast Cancer Res Treat 2011; 129:11-21. [PMID: 21336599 DOI: 10.1007/s10549-010-1161-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 09/02/2010] [Indexed: 12/20/2022]
Abstract
Oestrogen receptor (ER)-positive breast cancer (BC) constitutes a heterogeneous group of tumours with regard to outcome and response to therapy. Accurate stratification of ER-positive BC according to risk of relapse and response to therapy will be achieved through an improved understanding of ER and ER-related biological pathways. Recent studies have identified Forkhead box O3a (FOXO3a) transcription factor as an intracellular mediator of ERα expression and as an important downstream target of the Akt/PI3K pathway indicating a biological and potential clinical role for FOXO3a in ER-positive BC. In this study, we investigated the clinical relevance and biological associations of FOXO3a protein expression, using tissue microarrays and immunohistochemistry, in a large series of patients with invasive breast cancer. FOXO3a protein expression showed both nuclear and/or cytoplasmic staining patterns. FOXO3a predominant nuclear expression was positively associated with biomarkers of good prognosis including PgR, FOXA1 and p27 expression. There was an inverse association with mitotic counts, MIB1 growth fraction, C-MYC and PIK3CA expression. With respect to patient outcome, FOXO3a nuclear localisation was associated with longer BC specific survival (P < 0.001) and longer distant metastasis free interval (P = 0.001), independently of the well-established breast cancer prognostic factors. In conclusion, our results demonstrate the biological and prognostic role of FOXO3a protein expression and its subcellular localisation in ER-positive/luminal-like BC possibly through its involvement in controlling cell proliferation.
Collapse
Affiliation(s)
- Hany Onsy Habashy
- School of Molecular Medical Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | |
Collapse
|
18
|
Ramamoorthy S, Dhananjayan SC, Demayo FJ, Nawaz Z. Isoform-specific degradation of PR-B by E6-AP is critical for normal mammary gland development. Mol Endocrinol 2010; 24:2099-113. [PMID: 20829392 DOI: 10.1210/me.2010-0116] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
E6-associated protein (E6-AP), which was originally identified as an ubiquitin-protein ligase, also functions as a coactivator of estrogen (ER-α) and progesterone (PR) receptors. To investigate the in vivo role of E6-AP in mammary gland development, we generated transgenic mouse lines that either overexpress wild-type (WT) human E6-AP (E6-AP(WT)) or ubiquitin-protein ligase-defective E6-AP (E6-AP(C833S)) in the mammary gland. Here we show that overexpression of E6-AP(WT) results in impaired mammary gland development. In contrast, overexpression of E6-AP(C833S) or loss of E6-AP (E6-AP(KO)) increases lateral branching and alveolus-like protuberances in the mammary gland. We also show that the mammary phenotypes observed in the E6-AP transgenic and knockout mice are due, in large part, to the alteration of PR-B protein levels. We also observed alteration in ER-α protein level, which might contribute to the observed mammary phenotype by regulating PR expression. Furthermore, E6-AP regulates PR-B protein levels via the ubiquitin-proteasome pathway. Additionally, we also show that E6-AP impairs progesterone-induced Wnt-4 expression by decreasing the steady state level of PR-B in both mice and in human breast cancer cells. In conclusion, we present the novel observation that E6-AP controls mammary gland development by regulating PR-B protein turnover via the ubiquitin proteasome pathway. For the first time, we show that the E3-ligase activity rather than the coactivation function of E6-AP plays an important role in the mammary gland development, and the ubiquitin-dependent PR-B degradation is not required for its transactivation functions. This mechanism appears to regulate normal mammogenesis, and dysregulation of this process may be an important contributor to mammary cancer development and progression.
Collapse
Affiliation(s)
- Sivapriya Ramamoorthy
- Department of Biochemistry & Molecular Biology, Braman Breast Cancer Institute (M-877), University of Miami School of Medicine, Batchelor Building, Room 416, 1580 Northwest 10 Avenue, Miami, Florida 33136, USA
| | | | | | | |
Collapse
|
19
|
Lanzino M, Sisci D, Morelli C, Garofalo C, Catalano S, Casaburi I, Capparelli C, Giordano C, Giordano F, Maggiolini M, Andò S. Inhibition of cyclin D1 expression by androgen receptor in breast cancer cells--identification of a novel androgen response element. Nucleic Acids Res 2010; 38:5351-65. [PMID: 20421209 PMCID: PMC2938215 DOI: 10.1093/nar/gkq278] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cyclin D1 gene (CCND1) is a critical mitogen-regulated cell-cycle control element whose transcriptional modulation plays a crucial role in breast cancer growth and progression. Here we demonstrate that the non-aromatizable androgen 5-α-dihydrotestosterone (DHT) inhibits endogenous cyclin D1 expression, as evidenced by reduction of cyclin D1 mRNA and protein levels, and decrease of CCND1-promoter activity, in MCF-7 cells. The DHT-dependent inhibition of CCND1 gene activity requires the involvement and the integrity of the androgen receptor (AR) DNA-binding domain. Site directed mutagenesis, DNA affinity precipitation assay, electrophoretic mobility shift assay and chromatin immunoprecipitation analyses indicate that this inhibitory effect is ligand dependent and it is mediated by direct binding of AR to an androgen response element (CCND1-ARE) located at −570 to −556-bp upstream of the transcription start site, in the cyclin D1 proximal promoter. Moreover, AR-mediated repression of the CCND1 involves the recruitment of the atypical orphan nuclear receptor DAX1 as a component of a multiprotein repressor complex also embracing the participation of Histone Deacetylase 1. In conclusion, identification of the CCND1-ARE allows defining cyclin D1 as a specific androgen target gene in breast and might contribute to explain the molecular basis of the inhibitory role of androgens on breast cancer cells proliferation.
Collapse
Affiliation(s)
- Marilena Lanzino
- Dipartimento Farmaco-Biologico, University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Combinatorial biomarker expression in breast cancer. Breast Cancer Res Treat 2010; 120:293-308. [PMID: 20107892 DOI: 10.1007/s10549-010-0746-x] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 01/12/2010] [Indexed: 02/06/2023]
Abstract
Current clinical management of breast cancer relies on the availability of robust clinicopathological variables and few well-defined biological markers. Recent microarray-based expression profiling studies have emphasised the importance of the molecular portraits of breast cancer and the possibility of classifying breast cancer into biologically and molecularly distinct groups. Subsequent large scale immunohistochemical studies have demonstrated that the added value of studying the molecular biomarker expression in combination rather than individually. Oestrogen (ER) and progesterone (PR) receptors and HER2 are currently used in routine pathological assessment of breast cancer. Additional biomarkers such as proliferation markers and 'basal' markers are likely to be included in the future. A better understanding of the prognostic and predictive value of combinatorial assessment of biomarker expression could lead to improved breast cancer management in routine clinical practice and would add to our knowledge concerning the variation in behaviour and response to therapy. Here, we review the evidence on the value of assessing biomarker expression in breast cancer individually and in combination and its relation to the recent molecular classification of breast cancer.
Collapse
|
21
|
Putnik M, Zhao C, Gustafsson JÅ, Dahlman-Wright K. Effects of two common polymorphisms in the 3' untranslated regions of estrogen receptor beta on mRNA stability and translatability. BMC Genet 2009; 10:55. [PMID: 19754929 PMCID: PMC2759954 DOI: 10.1186/1471-2156-10-55] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 09/15/2009] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The present study represents the first attempt to functionally characterize two common single nucleotide polymorphisms (SNPs) in the 3'untranslated regions (3'UTRs) of estrogen receptor beta (ERbeta), focusing on the differences between alleles with regard to mRNA stability and translatability. These two ERbeta SNPs have been investigated for association with disease in a large number of reports. RESULTS Here we examined allelic expression in breast tumor samples from heterozygous individuals. A significant difference in mRNA levels of the two alleles was observed for one of the SNPs. A cell model system was employed to further investigate potential molecular effects of the two SNPs. We used a modified plasmid, containing the ERbeta promoter and ERbeta 3'UTRs which include the different alleles of investigated SNPs. Quantitative Real-Time PCR was used to determine mRNA levels after inhibition of transcription by actinomycin D, and a luciferase assay was used to determine protein levels. The obtained results suggested that there was no difference in mRNA stability or translatability between the alleles of investigated SNPs. CONCLUSION Our results indicate that observed associations between ERbeta 3'UTR SNPs and disease susceptibility are due to linkage disequilibrium with another gene variant, rather than the variant itself being the susceptibility factor.
Collapse
Affiliation(s)
- Milica Putnik
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, S-141 57 Huddinge, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, S-141 57 Huddinge, Sweden
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, S-141 57 Huddinge, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, S-141 57 Huddinge, Sweden
| |
Collapse
|
22
|
Hu DG, Mackenzie PI. Estrogen receptor alpha, fos-related antigen-2, and c-Jun coordinately regulate human UDP glucuronosyltransferase 2B15 and 2B17 expression in response to 17beta-estradiol in MCF-7 cells. Mol Pharmacol 2009; 76:425-39. [PMID: 19487245 DOI: 10.1124/mol.109.057380] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
UDP-glucuronosyltransferase 2B15 and 2B17 expression is up-regulated by 17beta-estradiol in MCF-7 breast cancer cells, as assessed by quantitative real-time polymerase chain reaction. Using 5'-deletion mapping and site-directed mutagenesis, we demonstrate that 17beta-estradiol activation of UGT2B15 gene transcription is mediated by a 282-base pair fragment positioned -454 to -172 nucleotides from the translation start site. This region contains two putative activator protein-1 (AP-1) elements, one imperfect estrogen response element (ERE), and two consensus ERE half-sites. We propose that these five sites act as an estrogen response unit (ERU), because mutation in any site reduces activation of the UGT2B15 promoter by 17beta-estradiol. Despite the presence of two AP-1 elements, the UGT2B15 promoter is not responsive to the AP-1 activator phorbol 12-myristate 13-acetate. Although electrophoretic mobility shift assays (EMSA) indicate that the AP-1 proteins c-Jun and Fos-related antigen 2 (Fra-2) bound to the distal AP-1 site, binding of Jun or Fos family members to the proximal AP-1 site was not detected by EMSA. Chromatin immunoprecipitation assays showed a 17beta-estradiol-induced recruitment of estrogen receptor (ER) alpha, c-Jun, and Fra-2 to the 282-bp ERU. The involvement of these three transcription factors in the stimulation of UGT2B15 gene expression by 17beta-estradiol was confirmed by siRNA silencing experiments. Mutagenesis and siRNA experiments indicate that UGT2B17 expression is also regulated by 17beta-estradiol via the ERU, which is fully conserved in both promoters. Because UGT2B15 and UGT2B17 inactivate steroid hormones by glucuronidation, the regulation of their genes by 17beta-estradiol may maintain steroid hormone homeostasis and prevent excessive estrogen signaling activity.
Collapse
Affiliation(s)
- Dong Gui Hu
- Department of Clinical Pharmacology, Flinders Medical Centre, Bedford Park, SA, Australia
| | | |
Collapse
|
23
|
Correlation between steroid receptors, angiogenic factors, and classical prognostic parameters in node-negative breast cancer patients. ARCH BIOL SCI 2009. [DOI: 10.2298/abs0904599v] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Breast cancer (BC) progression is an estrogen receptor (ER) signaling- and angiogenesis-dependent process. This study investigated relationships between classical prognostic factors and biomarkers ER, PR, VEGF, and bFGF in node-negative BC patients. Positive correlation between ER and both PR (p < 0.001) or FGF (p = 0.04) levels indicates ER-regulated expression of these factors and a potential synergistic effect of ER and bFGF in tumor progression. Aside from correlation of age with ER and bFGF levels (p = 0.003; p = 0.05; respectively), no correlation of biomarkers with classical prognostic parameters was found, indicating that those biomarkers could be independent prognostic factors.
Collapse
|
24
|
Novelli F, Milella M, Melucci E, Di Benedetto A, Sperduti I, Perrone-Donnorso R, Perracchio L, Venturo I, Nisticò C, Fabi A, Buglioni S, Natali PG, Mottolese M. A divergent role for estrogen receptor-beta in node-positive and node-negative breast cancer classified according to molecular subtypes: an observational prospective study. Breast Cancer Res 2008; 10:R74. [PMID: 18771580 PMCID: PMC2614505 DOI: 10.1186/bcr2139] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 07/09/2008] [Accepted: 09/04/2008] [Indexed: 11/20/2022] Open
Abstract
Introduction Estrogen receptor-alpha (ER-α) and progesterone receptor (PgR) are consolidated predictors of response to hormonal therapy (HT). In contrast, little information regarding the role of estrogen receptor-beta (ER-β) in various breast cancer risk groups treated with different therapeutic regimens is available. In particular, there are no data concerning ER-β distribution within the novel molecular breast cancer subtypes luminal A (LA) and luminal B (LB), HER2 (HS), and triple-negative (TN). Methods We conducted an observational prospective study using immunohistochemistry to evaluate ER-β expression in 936 breast carcinomas. Associations with conventional biopathological factors and with molecular subtypes were analyzed by multiple correspondence analysis (MCA), while univariate and multivariate Cox regression analysis and classification and regression tree analysis were applied to determine the impact of ER-β on disease-free survival in the 728 patients with complete follow-up data. Results ER-β evenly distributes (55.5%) across the four molecular breast cancer subtypes, confirming the lack of correlation between ER-β and classical prognosticators. However, the relationships among the biopathological factors, analyzed by MCA, showed that ER-β positivity is located in the quadrant containing more aggressive phenotypes such as HER2 and TN or ER-α/PgR/Bcl2- tumors. Kaplan-Meier curves and Cox regression analysis identified ER-β as a significant discriminating factor for disease-free survival both in the node-negative LA (P = 0.02) subgroup, where it is predictive of response to HT, and in the node-positive LB (P = 0.04) group, where, in association with PgR negativity, it conveys a higher risk of relapse. Conclusion Our data indicated that, in contrast to node-negative patients, in node-positive breast cancer patients, ER-β positivity appears to be a biomarker related to a more aggressive clinical course. In this context, further investigations are necessary to better assess the role of the different ER-β isophorms.
Collapse
Affiliation(s)
- Flavia Novelli
- Pathology Department, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Insulin receptor substrate 1 modulates the transcriptional activity and the stability of androgen receptor in breast cancer cells. Breast Cancer Res Treat 2008; 115:297-306. [DOI: 10.1007/s10549-008-0079-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 05/22/2008] [Indexed: 01/25/2023]
|
26
|
Skliris GP, Leygue E, Watson PH, Murphy LC. Estrogen receptor alpha negative breast cancer patients: estrogen receptor beta as a therapeutic target. J Steroid Biochem Mol Biol 2008; 109:1-10. [PMID: 18243688 DOI: 10.1016/j.jsbmb.2007.12.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Clinical management of breast cancer is increasingly guided by assessment of tumor phenotypic parameters. One of these is estrogen receptor (ER) status, currently defined by ERalpha expression. However with the discovery of a second ER, ERbeta and its variant isoforms, the definition of ER status is potentially more complex. In breast tumors there are two ERbeta expression cohorts. One where ERbeta is co-expressed with ERalpha and the other expressing ERbeta alone. In the latter subgroup of currently defined ER negative patients ERbeta has the potential to be a therapeutic target. Characterization of the nature and role of ERbeta in ERalpha negative tumors is essentially unexplored but available data suggest that the role of ERbeta may be different when co-expressed with ERalpha and when expressed alone. This review summarizes available data and explores the possibility that ERbeta signaling may be a therapeutic target in these tumors. Evidence so far supports the idea that the role of ERbeta in breast cancer is different in ERalpha negative compared to ERalpha positive tumors. However, cohort size and numbers of independent studies are small to date, and more studies are needed with better standardization of antibodies and protocols. Also, the ability to determine the role of ERbeta in ERalpha negative breast cancer and therefore assess ERbeta signaling pathways as therapeutic targets would be greatly facilitated by identification of specific downstream markers of ERbeta activity in breast cancer.
Collapse
Affiliation(s)
- George P Skliris
- Manitoba Institute of Cell Biology, Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada R3E OV9
| | | | | | | |
Collapse
|
27
|
Liu M, Liberzon A, Kong SW, Lai WR, Park PJ, Kohane IS, Kasif S. Network-based analysis of affected biological processes in type 2 diabetes models. PLoS Genet 2007; 3:e96. [PMID: 17571924 PMCID: PMC1904360 DOI: 10.1371/journal.pgen.0030096] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Accepted: 05/01/2007] [Indexed: 11/19/2022] Open
Abstract
Type 2 diabetes mellitus is a complex disorder associated with multiple genetic, epigenetic, developmental, and environmental factors. Animal models of type 2 diabetes differ based on diet, drug treatment, and gene knockouts, and yet all display the clinical hallmarks of hyperglycemia and insulin resistance in peripheral tissue. The recent advances in gene-expression microarray technologies present an unprecedented opportunity to study type 2 diabetes mellitus at a genome-wide scale and across different models. To date, a key challenge has been to identify the biological processes or signaling pathways that play significant roles in the disorder. Here, using a network-based analysis methodology, we identified two sets of genes, associated with insulin signaling and a network of nuclear receptors, which are recurrent in a statistically significant number of diabetes and insulin resistance models and transcriptionally altered across diverse tissue types. We additionally identified a network of protein–protein interactions between members from the two gene sets that may facilitate signaling between them. Taken together, the results illustrate the benefits of integrating high-throughput microarray studies, together with protein–protein interaction networks, in elucidating the underlying biological processes associated with a complex disorder. Type 2 diabetes mellitus currently affects millions of people. It is clinically characterized by insulin resistance in addition to an impaired glucose response and associated with numerous complications including heart disease, stroke, neuropathy, and kidney failure, among others. Accurate identification of the underlying molecular mechanisms of the disease or its complications is an important research problem that could lead to novel diagnostics and therapy. The main challenge stems from the fact that insulin resistance is a complex disorder and affects a multitude of biological processes, metabolic networks, and signaling pathways. In this report, the authors develop a network-based methodology that appears to be more sensitive than previous approaches in detecting deregulated molecular processes in a disease state. The methodology revealed that both insulin signaling and nuclear receptor networks are consistently and differentially expressed in many models of insulin resistance. The positive results suggest such network-based diagnostic technologies hold promise as potentially useful clinical and research tools in the future.
Collapse
Affiliation(s)
- Manway Liu
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail: (ML); (SK)
| | - Arthur Liberzon
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Sek Won Kong
- Department of Cardiology, Children's Hospital, Boston, Massachusetts, United States of America
- Children's Hospital Informatics Program at the Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts, United States of America
| | - Weil R Lai
- Harvard-Partners Center for Genetics and Genomics, Boston, Massachusetts, United States of America
| | - Peter J Park
- Children's Hospital Informatics Program at the Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts, United States of America
- Harvard-Partners Center for Genetics and Genomics, Boston, Massachusetts, United States of America
| | - Isaac S Kohane
- Children's Hospital Informatics Program at the Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts, United States of America
- Center of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Simon Kasif
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- Children's Hospital Informatics Program at the Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts, United States of America
- Center for Advanced Genomic Technology, Boston University, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail: (ML); (SK)
| |
Collapse
|
28
|
Cherlet T, Murphy LC. Estrogen receptors inhibit Smad3 transcriptional activity through Ap-1 transcription factors. Mol Cell Biochem 2007; 306:33-42. [PMID: 17660955 DOI: 10.1007/s11010-007-9551-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 07/12/2007] [Indexed: 02/07/2023]
Abstract
Breast tumorigenesis and breast cancer progression involves the deregulation or hyperactivation of intracellular signaling proteins that leads to uncontrolled cellular proliferation, invasion and metastasis. For example, the expression and cellular responses to estogen receptor (ER) and transforming growth factor beta (TGFbeta) signaling pathways change during breast tumorigenesis and breast cancer progression. While the expression and activity of ER and TGFbeta maybe significant in the development of breast cancer, alterations in the cross-talk between these pathways may be equally important. Autocrine and paracrine effects of TGFbeta on breast cancer cell growth have been known for some time, but only recently have direct interactions between ER and TGFbeta been described. The purpose of this article was to further characterize the cross-talk between ER and TGFbeta, by examining ER interaction with Smad3, a downstream mediator of TGFbeta signaling. Transient transfection of Cos1 cells with p3TP-lux, demonstrate that ERalpha and ERbeta(1) repress Smad3 transcriptional activity in an estradiol-dependent manner and that this effect is inhibited by antiestrogen treatment. The ERbeta variants, ERbeta(2) and ERbeta(5), did not have any effect on Smad3 transcriptional activity. Further experiments attempted to characterize the molecular mechanism by which activated ER inhibits Smad3 transcriptional activity. Results indicate that ligand-bound ER does not affect Smad3 protein expression levels and that ER does not form direct protein interactions with Smad3. Transient transfection of Cos1 cells with the Ap-1 transcription factor c-Jun but not c-Fos was able to rescue the inhibitory effect of estrogen on Smad3 transcriptional activity. Based on these results, a model is proposed whereby c-Jun is limiting in its ability to act as a Smad3 co-activator in the presence of E(2)-bound ER, possibly due to ER sequestering c-Jun away from the Smad3 responsive promoter.
Collapse
Affiliation(s)
- Tracy Cherlet
- Manitoba Institute of Cell Biology, Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB, Canada R3E 0V9
| | | |
Collapse
|
29
|
Ayed-Boussema I, Ouanes Z, Bacha H, Abid S. Toxicities induced in cultured cells exposed to zearalenone: Apoptosis or mutagenesis? J Biochem Mol Toxicol 2007; 21:136-44. [PMID: 17623888 DOI: 10.1002/jbt.20171] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Zearalenone (ZEN) is a fusarotoxin produced mainly by Fusarium graminearum in temperate and warm countries. ZEN has several adverse effects on humans and animals. It has a strong estrogenic activity associated with hyperestrogenism and leads to several physiological alterations in the reproductive tract. Even though the mutagenic and genotoxic proprieties of ZEN have been described recently, its molecular mechanisms of action are not completely understood. The aim of this study was to determine the involvement of other possible mechanisms in ZEN-induced toxicities. Each of the following toxicities, cytotoxicity, cell cycle perturbation, genotoxicity, and mutagenicity, was monitored in Vero cells exposed to ZEN. Our results showed that ZEN-reduced cell viability correlated to cell cycle perturbation-induced DNA fragmentation, resulting in DNA-laddering patterns on agar gel electrophoresis. This observation is consistent with apoptosis, which was confirmed by induction of apoptotic bodies. Moreover, ZEN induced in a concentration-dependant manner the formation of micronuclei and chromosome aberrations. This apparent contradiction between the apoptotic and mutagenic effects of ZEN can be explained by the modification of normal cellular regulation inducing apoptotic or antiapoptotic factors resulting from a lack of or an incorrect DNA-reparation in relation to cell exposure to the toxin.
Collapse
Affiliation(s)
- Imen Ayed-Boussema
- Laboratory of Research on Biologically Compatible Compounds, Faculty of Dental Medicine, Monastir University, Rue Avicenne, 5019 Monastir, Tunisia
| | | | | | | |
Collapse
|
30
|
Mo R, Zhu YT, Zhang Z, Rao SM, Zhu YJ. GAS6 is an estrogen-inducible gene in mammary epithelial cells. Biochem Biophys Res Commun 2006; 353:189-94. [PMID: 17174935 PMCID: PMC1820842 DOI: 10.1016/j.bbrc.2006.12.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Accepted: 12/04/2006] [Indexed: 11/21/2022]
Abstract
To identify estrogen-responsive genes in mammary glands, microarray assays were performed. Twenty genes were found to be up-regulated while 16 genes were repressed in the 9h estrogen treated glands. The induction of GAS6, one of the genes up-regulated by estrogen, was confirmed by RNase protection assay. Furthermore, GAS6 was also demonstrated to be induced by estrogen in ER positive breast cancer cells. Analysis of GAS6 promoter revealed that GAS6 promoter was regulated by estrogen. An estrogen response element (ERE) was identified in the GAS6 promoter. Electrophoretic mobility shift assay revealed that ERalpha interacted with the ERE in the GAS6 promoter. Chromatin immunoprecipitation demonstrated that ERalpha was recruited to the GAS6 promoter upon estrogen stimulation. These results suggested that GAS6 is an estrogen target gene in mammary epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | - Yi-Jun Zhu
- To whom correspondence should be addressed: Dept. of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611-3008, Tel.: 312-503-3113, Fax: 312-503-8249, E-mail:
| |
Collapse
|
31
|
Kumaraguruparan R, Prathiba D, Nagini S. Of humans and canines: Immunohistochemical analysis of PCNA, Bcl-2, p53, cytokeratin and ER in mammary tumours. Res Vet Sci 2006; 81:218-24. [PMID: 16740286 DOI: 10.1016/j.rvsc.2005.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2005] [Revised: 07/09/2005] [Accepted: 08/12/2005] [Indexed: 11/26/2022]
Abstract
Mammary tumours are the most common neoplasms in humans and canines. Human and canine mammary tumours share several important epidemiological, clinicopathological and biochemical features. Development of mammary tumours involves accumulation of mutant cells caused by excessive proliferation and insufficient apoptosis or dysregulation of cellular differentiation. The present study was therefore designed to investigate the expression of proliferation, differentiation, and apoptosis associated proteins together with expression of estrogen receptors (ER) in both human and canine mammary tumours. Thirty breast cancer patients categorized as pre- and postmenopausal, and 30 mammary gland tumours obtained from bitches were included in this study. The expression of proliferating cell nuclear antigen (PCNA), Bcl-2, p53, cytokeratin and ER in tumour tissues and adjacent tissues were investigated using immunohistochemical staining. While the expression of PCNA, Bcl-2, p53 and ER was significantly increased, expression of cytokeratin was significantly lower in both human as well as canine mammary tumours compared to corresponding adjacent tissues. The magnitude of the changes was however more pronounced in premenopausal patients compared to postmenopausal patients. The changes in proliferation, apoptosis and differentiation associated proteins in human and canine mammary tumours validate use of the canine model to understand the molecular mechanisms of mammary carcinogenesis.
Collapse
Affiliation(s)
- R Kumaraguruparan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608 002, Chidambaram, Tamil Nadu, India
| | | | | |
Collapse
|
32
|
Plants with antitumor properties: from biologically active molecules to drugs. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/s1572-557x(05)02004-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
33
|
Lanzino M, De Amicis F, McPhaul MJ, Marsico S, Panno ML, Andò S. Endogenous coactivator ARA70 interacts with estrogen receptor alpha (ERalpha) and modulates the functional ERalpha/androgen receptor interplay in MCF-7 cells. J Biol Chem 2005; 280:20421-30. [PMID: 15772083 DOI: 10.1074/jbc.m413576200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Overexpression of androgen receptor (AR) decreases estrogen receptor alpha (ERalpha) transactivation, which plays a basic role in hormone-dependent breast cancer. This transcriptional interference can be due to shared coactivators. Here we demonstrated that in MCF-7 cells ARA70, an AR-specific coactivator, interacted with endogenous ERalpha, increasing its transcriptional activity, and it was recruited to the pS2 gene promoter. Moreover, a dominant negative ARA70 down-regulated ERalpha transcriptional activity as well as pS2 mRNA. ARA70 overexpression reversed the AR down-regulatory effect on ERalpha signaling. However, in the presence of a progressive increase of transfected AR, ARA70 switched into enhancing the inhibitory effect of AR on ERalpha signaling. These opposite effects of ARA70 were further evidenced by coimmunoprecipitation assay in MCF-7wt, MCF-7-overexpressing AR, and HeLa cells, exogenously expressing an excess of ERalpha with respect to AR or an excess of AR with respect to ERalpha. Thus, ARA70 is a coactivator for ERalpha and may represent a functional link between ERalpha/AR modulating their cross-talk in models of estrogen signaling in MCF-7 and HeLa cells.
Collapse
Affiliation(s)
- Marilena Lanzino
- Department of Pharmacobiology, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | | | | | | | | | | |
Collapse
|
34
|
Tsakountakis N, Sanidas E, Stathopoulos E, Kafousi M, Anogiannaki N, Georgoulias V, Tsiftsis DD. Correlation of breast cancer risk factors with HER-2/neu protein overexpression according to menopausal and estrogen receptor status. BMC WOMENS HEALTH 2005; 5:1. [PMID: 15694000 PMCID: PMC549187 DOI: 10.1186/1472-6874-5-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/16/2004] [Accepted: 02/04/2005] [Indexed: 11/23/2022]
Abstract
Background Several researchers have claimed that classification of tumours on the basis of HER-2/neu overexpression or amplification may define a subset of breast cancer in which the net effect of a risk factor could be rather more obvious and its impact on breast cancer development more clear. We decided to investigate, in a group of patients from a geographical area with a low incidence of breast cancer, whether HER-2/neu positive tumours are correlated with established or suspected risk factors for breast cancer and thus to identify distinct subgroups of high risk women. Methods This study analysed data from patients who attended the Breast Unit at the University Hospital of Heraklion, Crete, Greece between 1996 and 2002. 384 women with primary invasive breast cancer were compared with 566 screened women who were referred to the Unit and had not developed breast neoplasm by the time the data were analysed. Risk factor data were obtained from each subject by personal interviews using a structured questionnaire. The detection and scoring of the HER-2/neu protein, estrogen and progesterone receptor expression were performed using immunochemistry. Odds ratios and 95% confidence intervals were determined by chi-square test and logistic regression analysis. Case-case odds ratios were calculated in order to measure the risk heterogeneity between HER-2/neu+ and HER-2/neu-tumours. Separate analyses were performed for premenopausal and postmenopausal women and according to estrogen receptor status. Results In multivariate analysis without HER-2/neu stratification, an increased breast cancer risk was associated with only four of the factors examined: use of oral contraceptives (OR = 4.40, 95%C.I: 1.46–13.28), use of HRT (OR = 7.34, 95%C.I: 2.03–26.53), an age at first full pregnancy more than 23 years (OR = 1.91, 95%C.I: 1.29–2.83) and body mass index more than 29 kg/m2 (OR = 3.13, 95%C.I: 2.02–4.84). Additionally, a history of abortion or miscarriage (OR = 0.56, 95%C.I: 0.38–0.82) was correlated with a decreased risk of breast cancer. In the case to case comparison only BMI >29 kg/m2 revealed a relative connection that was stronger with positive than with negative HER-2/neu tumours (ratio of OR's = 2.23, 95%C.I: 1.20–4.15, p = 0.011). This may indicate evidence of heterogeneity of a rather significant degree for this factor. In the ER negative group an age at first full pregnancy >23 years and a BMI >29 kg/m2 were associated with an increased risk in both HER-2/neu groups, but the association was significantly stronger for the latter factor in the positive HER-2/neu tumours (ratio of OR's = 2.46, 95%CI: 0.97–6.21). Conclusions Our study did not confirm that the established or putative hormonal breast cancer risk factors differ regarding their relations with HER-2/neu+ versus HER-2/neu-breast tumours, with the exception of increased BMI. Further innovative studies with larger sample sizes are needed to examine how the status of these potentially modifiable breast cancer risk factors interacts with biological markers such as HER-2/neu oncoprotein.
Collapse
Affiliation(s)
- Nikos Tsakountakis
- Dept of Family and Social Medicine, Heraklion Medical School, University of Crete, Greece
| | - Elias Sanidas
- Dept of Surgical Oncology, Heraklion Medical School, University of Crete, Greece
| | | | - Maria Kafousi
- Dept of Pathology, Heraklion Medical School, University of Crete, Greece
| | | | - Vasilis Georgoulias
- Dept of Medical Oncology, Heraklion Medical School, University of ≥Crete, Greece
| | - Dimitris D Tsiftsis
- Dept of Surgical Oncology, Heraklion Medical School, University of Crete, Greece
| |
Collapse
|
35
|
Jia Y, Qi C, Zhang Z, Zhu YT, Rao SM, Zhu YJ. Peroxisome proliferator-activated receptor-binding protein null mutation results in defective mammary gland development. J Biol Chem 2005; 280:10766-73. [PMID: 15647257 DOI: 10.1074/jbc.m413331200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A conditional null mutation of peroxisome proliferator-activated receptor-binding protein (PBP) gene was generated to understand its role in mammary gland development. PBP-deficient mammary glands exhibited retarded ductal elongation during puberty, and decreased alveolar density during pregnancy and lactation. PBP-deficient mammary glands could not produce milk to nurse pups during lactation. Both the mammary ductal elongation in response to estrogen treatment and the mammary lobuloalveolar proliferation stimulated by estrogen plus progesterone were attenuated in PBP-deficient mammary glands. The proliferation index was decreased in PBP-deficient mammary glands. PBP-deficient mammary epithelial cells expressed abundant beta-casein, whey acidic protein, and WDNM1 mRNA, indicating a relatively intact differentiated function. PBP-deficient epithelial cells were unable to form mammospheres, which were considered to be derived from mammary progenitor/stem cells. We conclude that PBP plays a pivotal role in the normal mammary gland development.
Collapse
Affiliation(s)
- Yuzhi Jia
- Department of Pathology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Ave., Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
36
|
Murphy L, Cherlet T, Adeyinka A, Niu Y, Snell L, Watson P. Phospho-serine-118 estrogen receptor-alpha detection in human breast tumors in vivo. Clin Cancer Res 2004; 10:1354-9. [PMID: 14977836 DOI: 10.1158/1078-0432.ccr-03-0112] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine whether estrogen receptor (ER)-alpha specifically phosphorylated at Ser(118) is detectable in multiple human breast cancer biopsy samples. To gain insight into possible roles for P-Ser(118)-ERalpha in human breast cancer in vivo. EXPERIMENTAL DESIGN A specific antibody for P-Ser(118)-ERalpha was validated for immunohistochemistry (IHC), and Western blot analysis confirmed IHC results. IHC was used to determine the relationship of P-Ser(118)-ERalpha to known prognostic markers and active mitogen-activated protein kinase (MAPK; erk1/2) expression. RESULTS P-Ser(118)-ERalpha was significantly correlated with the expression of total ER, determined by ligand binding assay (r = 0.442, P = 0.002), but not with progesterone receptor expression or nodal status. P-Ser(118)-ERalpha was inversely correlated with histological grade (r = -0.34, P = 0.023), reflecting a similar trend for total ER (r = -0.287, P = 0.056). Categorical contingency analysis confirmed that P-Ser(118)-ERalpha was more frequently associated with lower than higher grade breast tumors (P = 0.038). In addition P-Ser(118)-ERalpha was significantly associated with detection of active MAPK (Erk1/2; Spearman r = 0.649, P < 0.0001; Fisher's exact test, P = 0.0004). CONCLUSIONS P-Ser(118)-ERalpha detection is associated with a more differentiated phenotype and other markers of good prognosis in human breast cancer. P-Ser(118)-ERalpha is correlated with active MAPK in human breast tumor biopsies, suggesting the possibility that active MAPK either directly or indirectly has a role in the regulation of P-Ser(118)-ERalpha expression in vivo. These data provide evidence for a role of P-Ser(118)-ERalpha in human breast cancer in vivo.
Collapse
Affiliation(s)
- Leigh Murphy
- Manitoba Institute of Cell Biology, Department of Biochemistry, University of Manitoba, Winnipeg, Manitoba, Canada.
| | | | | | | | | | | |
Collapse
|
37
|
Qi C, Kashireddy P, Zhu YT, Rao SM, Zhu YJ. Null Mutation of Peroxisome Proliferator-activated Receptor-interacting Protein in Mammary Glands Causes Defective Mammopoiesis. J Biol Chem 2004; 279:33696-701. [PMID: 15161927 DOI: 10.1074/jbc.m401266200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To investigate the role of nuclear receptor coactivator peroxisome proliferator-activated receptor-interacting protein (PRIP) in mammary gland development, we generated a conditional null mutation of PRIP in mammary glands. In PRIP-deficient mammary glands, the elongation of ducts during puberty was not affected, but the numbers of ductal branches were decreased, a condition that persisted long after puberty, indicating that the potential of ductal branching was impaired. During pregnancy, PRIP-deficient mammary glands exhibited decreased alveolar density. The lactating PRIP-deficient glands contained scant lobuloalveoli with many adipocytes, whereas the wild type glands were composed of virtually no adipocytes but mostly lobuloalveoli. As a result, PRIP mammary-deficient glands could not produce enough milk to nurse all the pups during lactation. The ductal branching of mammary glands in response to estrogen treatment was attenuated in PRIP mutant glands. Whereas the proliferation index was similar between wild type and PRIP-deficient glands, increased apoptosis was observed in PRIP-deficient glands. PRIP-deficient glands expressed increased amphiregulin, transforming growth factor-alpha, and betacellulin mRNA as compared with wild type glands. The differentiated function of PRIP-deficient mammary epithelial cells was largely intact, as evidenced by the expression of abundant beta-casein, whey acidic protein (WAP), and WDNM1 mRNA. We conclude that PRIP is important for normal mammary gland development.
Collapse
Affiliation(s)
- Chao Qi
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | | | |
Collapse
|
38
|
Bu H, Kashireddy P, Chang J, Zhu YT, Zhang Z, Zheng W, Rao SM, Zhu YJ. ERBP, a novel estrogen receptor binding protein enhancing the activity of estrogen receptor. Biochem Biophys Res Commun 2004; 317:54-9. [PMID: 15047147 DOI: 10.1016/j.bbrc.2004.02.179] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2004] [Indexed: 01/12/2023]
Abstract
To understand the mechanism by which estrogen receptor (ER) activates transcription in a tissue specific fashion, we isolated ERalpha binding protein (ERBP) by performing yeast two-hybrid screening with human mammary gland cDNA library. ERBP is a nuclear protein and its mRNA is ubiquitously expressed. The in vitro interaction of ERBP with ERalpha was demonstrated by GST pull-down assay and this interaction was enhanced by estrogen. In addition, ERBP also bound to PPARgamma, RXRalpha, and ERbeta. ERBP interacted with the DNA binding domain and the hinge region of ERalpha. There are two ERalpha binding regions on ERBP. The binding of ERBP region at C-terminus to ERalpha is increased by estrogen while the binding of ERBP region at N-terminus is not affected by estrogen. The interaction of ERBP with ERalpha was further confirmed in vivo by immunoprecipitation. Transient transfection experiment demonstrated that ERBP enhanced the transcriptional activity of ERalpha.
Collapse
Affiliation(s)
- Hengfu Bu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Murphy L, Cherlet T, Lewis A, Banu Y, Watson P. New insights into estrogen receptor function in human breast cancer. Ann Med 2003; 35:614-31. [PMID: 14708971 DOI: 10.1080/07853890310014579] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
An important new concept associated with estrogen receptor (ER) function in breast cancer is that ER status/ phenotype is multifaceted. In particular, the two full-length, ligand binding ERs (ER-alpha and ER-beta) and possibly multiple variant isoforms of ER must be considered. In addition, cross-talk factors that can influence ER activity in a ligand independent fashion and factors downstream of the ER, including coactivators and corepressors, clearly have important roles in ER function. Their careful evaluation in addition to ER status will be necessary to more fully understand the etiology of breast cancer and the changes occurring in estrogen signaling during breast tumorigenesis and breast cancer progression. Such knowledge is necessary to have a significant impact on better prevention and treatment strategies for human breast cancer.
Collapse
Affiliation(s)
- Leigh Murphy
- Manitoba Institute of Cell Biology, Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada R3E 0V9.
| | | | | | | | | |
Collapse
|
40
|
Piva R, Gambari R. Transcription factor decoy (TFD) in breast cancer research and treatment. Technol Cancer Res Treat 2002; 1:405-16. [PMID: 12625767 DOI: 10.1177/153303460200100512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Synthetic oligonucleotides have recently been the object of many investigations aimed to develop sequence-selective compounds able to modulate, either positively or negatively, transcription of eukaryotic and viral genes. Alteration of transcription could be obtained by using synthetic oligonucleotides mimicking target sites of transcription factors (the transcription factor decoy -TFD- approach). This could lead to either inhibition or activation of gene expression, depending on the biological functions of the target transcription factors. Since several transcription factors are involved in tumor onset and progression, this issue is of great interest in order to design anti-tumor compounds. In addition to oligonucleotides, peptide nucleic acids (PNA) can be proposed for the modulation of gene expression. In this respect, double-stranded PNA-DNA chimeras have been shown to be capable to exhibit strong decoy activity. In the case of treatment of breast cancer cells, decoy oligonucleotides mimicking CRE binding sites, promoter region of estrogen receptor alpha gene, NF-kB binding sites have been used with promising results. Therefore, the transcription factor decoy approach could be object of further studies to develop protocols for the treatment of breast cancer. In the future, transcription factors regulating cell cycle, hormone-dependent differentiation, tumor invasion and metastasis are expected to be suitable targets for transcription factor decoy.
Collapse
Affiliation(s)
- Roberta Piva
- Department of Biochemistry and Molecular Biology, Ferrara University, Via Luigi Borsari, 46, 44100 Ferrara, Italy
| | | |
Collapse
|