1
|
Sauer N, Matkowski I, Bodalska G, Murawski M, Dzięgiel P, Calik J. Prognostic Role of Prolactin-Induced Protein (PIP) in Breast Cancer. Cells 2023; 12:2252. [PMID: 37759471 PMCID: PMC10527336 DOI: 10.3390/cells12182252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Prolactin-inducible protein (PIP), also referred to as gross cystic disease fluid protein 15 (GCDFP-15), has been a trending topic in recent years due to its potential role as a specific marker in breast cancer. PIP binds to aquaporin-5 (AQP5), CD4, actin, fibrinogen, β-tubulin, serum albumin, hydroxyapatite, zinc α2-glycoprotein, and the Fc fragment of IgGs, and the expression of PIP has been demonstrated to be modulated by various cytokines, including IL4/13, IL1, and IL6. PIP gene expression has been extensively studied due to its captivating nature. It is influenced by various factors, with androgens, progesterone, glucocorticosteroids, prolactin, and growth hormone enhancing its expression while estrogens suppress it. The regulatory mechanisms involve important proteins such as STAT5A, STAT5B, Runx2, and androgen receptor, which collaborate to enhance PIP gene transcription and protein production. The expression level of PIP in breast cancer is dependent on the tumor stage and subtype. Higher expression is observed in early-stage tumors of the luminal A subtype, while lower expression is associated with luminal B, basal-like, and triple-negative subtypes, which have a poorer prognosis. PIP expression is also correlated with apocrine differentiation, hormone receptor positivity, and longer metastasis-free survival. PIP plays a role in supporting the immune system's antitumor response during the early stages of breast cancer development. However, as cancer progresses, the protective role of PIP may become less effective or diminished. In this work, we summarized the clinical significance of the PIP molecule in breast cancer and its potential role as a new candidate for cell-based therapies.
Collapse
Affiliation(s)
- Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Old Town Clinic, 50-127 Wroclaw, Poland
| | - Igor Matkowski
- Jan Mikulicz-Radecki University Teaching Hospital, Borowska 213, 50-556 Wroclaw, Poland;
| | - Grażyna Bodalska
- Faculty of Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Marek Murawski
- 1st Department and Clinic of Gynecology and Obstetrics, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368 Wroclaw, Poland;
- Department of Human Biology, Faculty of Physiotherapy, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland
| | - Jacek Calik
- Old Town Clinic, 50-127 Wroclaw, Poland
- Department of Clinical Oncology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
2
|
Urbaniak A, Jablonska K, Suchanski J, Partynska A, Szymczak-Kulus K, Matkowski R, Maciejczyk A, Ugorski M, Dziegiel P. Prolactin-induced protein (PIP) increases the sensitivity of breast cancer cells to drug-induced apoptosis. Sci Rep 2023; 13:6574. [PMID: 37085653 PMCID: PMC10121699 DOI: 10.1038/s41598-023-33707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/18/2023] [Indexed: 04/23/2023] Open
Abstract
We have previously shown that high expression of prolactin-induced protein (PIP) correlates with the response of breast cancer (BC) patients to standard adjuvant chemotherapy (doxorubicin and cyclophosphamide), which suggests that the absence of this glycoprotein is associated with resistance of tumor cells to chemotherapy. Therefore, in the present study, we analyzed the impact of PIP expression on resistance of BC cells to anti-cancer drugs and its biological role in BC progression. Expression of PIP and apoptotic genes in BC cell lines was analyzed using real-time PCR and Western blotting. PIP was detected in BC tissue specimens using immunohistochemistry. The tumorigenicity of cancer cells was analyzed by the in vivo tumor growth assay. Apoptotic cells were detected based on caspase-3 activation, Annexin V binding and TUNEL assay. The interaction of PIP with BC cells was analyzed using flow cytometry. Using two cellular models of BC (i.e. T47D cells with the knockdown of the PIP gene and MDA-MB-231 cells overexpressing PIP), we found that high expression of PIP resulted in (1) increased sensitivity of BC cells to apoptosis induced by doxorubicin (DOX), 4-hydroperoxycyclophosphamide (4-HC), and paclitaxel (PAX), and (2) improved efficacy of anti-cancer therapy with DOX in the xenograft mice model. Accordingly, a clinical study revealed that BC patients with higher PIP expression were characterized by longer 5-year overall survival and disease-free survival. Subsequent studies showed that PIP up-regulated the expression of the following pro-apoptotic genes: CRADD, DAPK1, FASLG, CD40 and BNIP2. This pro-apoptotic activity is mediated by secreted PIP and most probably involves the specific surface receptor. This study demonstrates that a high expression level of PIP sensitizes BC cells to anti-cancer drugs. Increased sensitivity to chemotherapy is the result of pro-apoptotic activity of PIP, which is evidenced by up-regulation of specific pro-apoptotic genes. As high expression of PIP significantly correlated with a better response of patients to anti-cancer drugs, this glycoprotein can be a marker for the prognostic evaluation of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Anna Urbaniak
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 31, 50-375, Wroclaw, Poland
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, 53-114, Wroclaw, Poland
| | - Karolina Jablonska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland
| | - Jaroslaw Suchanski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 31, 50-375, Wroclaw, Poland
| | - Aleksandra Partynska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland
| | - Katarzyna Szymczak-Kulus
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, 53-114, Wroclaw, Poland
| | - Rafal Matkowski
- Department of Oncology, Wroclaw Medical University, 50-368, Wroclaw, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413, Wroclaw, Poland
| | - Adam Maciejczyk
- Department of Oncology, Wroclaw Medical University, 50-368, Wroclaw, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413, Wroclaw, Poland
| | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 31, 50-375, Wroclaw, Poland.
| | - Piotr Dziegiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland.
- Department of Human Biology, Faculty of Physiotherapy, Wroclaw University of Health and Sport Sciences, 51-612, Wroclaw, Poland.
| |
Collapse
|
3
|
Hanamura T, Christenson JL, O'Neill KI, Rosas E, Spoelstra NS, Williams MM, Richer JK. Secreted indicators of androgen receptor activity in breast cancer pre-clinical models. Breast Cancer Res 2021; 23:102. [PMID: 34736512 PMCID: PMC8567567 DOI: 10.1186/s13058-021-01478-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/19/2021] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Accumulating evidence has attracted attention to the androgen receptor (AR) as a biomarker and therapeutic target in breast cancer. We hypothesized that AR activity within the tumor has clinical implications and investigated whether androgen responsive serum factors might serve as a minimally invasive indicator of tumor AR activity. METHODS Based on a comprehensive gene expression analysis of an AR-positive, triple negative breast cancer patient-derived xenograft (PDX) model, 163 dihydrotestosterone (DHT)-responsive genes were defined as an androgen responsive gene set. Among them, we focused on genes that were DHT-responsive that encode secreted proteins, namely KLK3, AZGP1 and PIP, that encode the secreted factors prostate specific antigen (PSA), zinc-alpha-2-glycoprotein (ZAG) and prolactin induced protein (PIP), respectively. Using AR-positive breast cancer cell lines representing all breast cancer subtypes, expression of candidate factors was assessed in response to agonist DHT and antagonist enzalutamide. Gene set enrichment analysis (GSEA) was performed on publically available gene expression datasets from breast cancer patients to analyze the relationship between genes encoding the secreted factors and other androgen responsive gene sets in each breast cancer subtype. RESULTS Anti-androgen treatment decreased proliferation in all cell lines tested representing various tumor subtypes. Expression of the secreted factors was regulated by AR activation in the majority of breast cancer cell lines. In GSEA, the candidate genes were positively correlated with an androgen responsive gene set across breast cancer subtypes. CONCLUSION KLK3, AZGP1 and PIP are AR regulated and reflect tumor AR activity. Further investigations are needed to examine the potential efficacy of these factors as serum biomarkers.
Collapse
Affiliation(s)
- Toru Hanamura
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Emmanuel Rosas
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Michelle M Williams
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
4
|
Hu T, Zhao G, Liu Y, Long M. A Machine Learning Approach to Differentiate Two Specific Breast Cancer Subtypes Using Androgen Receptor Pathway Genes. Technol Cancer Res Treat 2021; 20:15330338211027900. [PMID: 34159849 PMCID: PMC8226237 DOI: 10.1177/15330338211027900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Triple-negative breast cancer is a heterogeneous disease with different molecular
and histological subtypes. The Androgen receptor is expressed in a portion of
triple-negative breast cancer cases and the activation of the androgen receptor
pathway is thought to be a molecular subtyping signature as well as a
therapeutic target for triple-negative breast cancer. Thus, identification of
the androgen receptor pathway status is important for both molecular
characterization andclinical management. In this study, we investigate the
expression of the androgen receptor pathway in metaplastic breast cancer and
luminal androgen receptor subtypes of triple-negative breast cancer and found
that the androgen receptor pathway was downregulated in metaplastic breast
cancer compared to luminal androgen receptor subtype. Using random forest, we
found that the two subtypes of breast cancer can be molecularly classified with
the gene expression of the androgen receptor pathway.
Collapse
Affiliation(s)
- Taobo Hu
- Department of Breast Surgery, 71185Peking University People's Hospital, Beijing, China
| | - Guiyang Zhao
- Department of Oncology, Beijing Changping Hospital, Beijing, China
| | - Yiqiang Liu
- Department of Pathology, 71185Peking University Cancer Hospital, Beijing, China
| | - Mengping Long
- Department of Pathology, 71185Peking University Cancer Hospital, Beijing, China
| |
Collapse
|
5
|
Li X, Zhang Y, Jia L, Xing Y, Zhao B, Sui L, Liu D, Xu X. Downregulation of Prolactin-Induced Protein Promotes Osteogenic Differentiation of Periodontal Ligament Stem Cells. Med Sci Monit 2021; 27:e930610. [PMID: 34092782 PMCID: PMC8194291 DOI: 10.12659/msm.930610] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Periodontal ligament stem cells (PDLSCs) are promising seed cells for bone tissue engineering and periodontal regeneration applications. However, the mechanism underlying the osteogenic differentiation process remains largely unknown. Previous reports showed that prolactin-induced protein (PIP) was upregulated after PDLSCs osteogenic induction. However, few studies have reported on the function of PIP in osteogenic differentiation. The purpose of the present study was to investigate the effect of PIP on osteogenic differentiation of PDLSCs. Material/Methods The expression pattern of PIP during PDLSCs osteogenic differentiation was detected and the effect of each component in the osteogenic induction medium on PIP was also tested by qRT-PCR. Then, the PIP knockdown cells were established using lentivirus. The knockdown efficiency was measured and the proliferation, apoptosis, and osteogenic differentiation ability were examined to determine the functional role of PIP on PDLSCs. Results QRT-PCR showed that PIP was sustainedly upregulated during the osteogenic induction process and the phenomenon was mainly caused by the stimulation of dexamethasone in the induction medium. CCK-8 and flow cytometer showed that knocking down PIP had no influence on proliferation and apoptosis of PDLSCs. ALP staining and activity, Alizarin Red staining, and western blot analysis demonstrated PIP knockdown enhanced the osteogenic differentiation and mineralization of PDLSCs. Conclusions PIP was upregulated after osteogenic induction; however, PIP knockdown promoted PDLSCs osteogenic differentiation. PIP might be a by-product of osteogenic induction, and downregulating of PIP might be a new target in bone tissue engineering applications.
Collapse
Affiliation(s)
- Xiaomeng Li
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China (mainland).,Stomatological Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Yunpeng Zhang
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China (mainland).,Department of Oral Implantology, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Linglu Jia
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China (mainland)
| | - Yixiao Xing
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China (mainland)
| | - Bin Zhao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China (mainland)
| | - Lei Sui
- Stomatological Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Dayong Liu
- Stomatological Hospital, Tianjin Medical University, Tianjin, China (mainland)
| | - Xin Xu
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China (mainland)
| |
Collapse
|
6
|
Ashe H, Krakowiak P, Hasterok S, Sleppy R, Roller DG, Gioeli D. Role of the runt-related transcription factor (RUNX) family in prostate cancer. FEBS J 2021; 288:6112-6126. [PMID: 33682350 DOI: 10.1111/febs.15804] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) is a very complex disease that is a major cause of death in men worldwide. Currently, PCa dependence on the androgen receptor (AR) has resulted in use of AR antagonists and antiandrogen therapies that reduce endogenous steroid hormone production. However, within two to three years of receiving first-line androgen deprivation therapy, the majority of patients diagnosed with PCa progress to castration-resistant prostate cancer (CRPC). There is an urgent need for therapies that are more durable than antagonism of the AR axis. Studies of runt-related transcription factors (RUNX) and their heterodimerization partner, core-binding factor subunit b (CBFβ), are revealing that the RUNX family are drivers of CRPC. In this review, we describe what is presently understood about RUNX members in PCa, including what regulates and is regulated by RUNX proteins, and the role of RUNX proteins in the tumor microenvironment and AR signaling. We discuss the implications for therapeutically targeting RUNX, the potential for RUNX as PCa biomarkers, and the current pressing questions in the field.
Collapse
Affiliation(s)
- Hannah Ashe
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Patryk Krakowiak
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Sylwia Hasterok
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Rosalie Sleppy
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Devin G Roller
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Daniel Gioeli
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
7
|
Sharif R, Bak-Nielsen S, Sejersen H, Ding K, Hjortdal J, Karamichos D. Prolactin-Induced Protein is a novel biomarker for Keratoconus. Exp Eye Res 2019; 179:55-63. [PMID: 30393162 PMCID: PMC6360109 DOI: 10.1016/j.exer.2018.10.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE The purpose of the study was to investigate the role of Prolactin-Induced Protein (PIP) as a predictive biomarker for Keratoconus (KC). PARTICIPANTS This study included one hundred and forty-seven patients with KC (105 male, 42 female), and sixty healthy controls (27 male, 33 female). METHODS Tears, plasma and saliva samples were collected from all participants. In both KC and healthy groups all collected samples were divided into four age subgroups (15-24y), (25-34y), (35-44y) and (45y and up). Samples were analyzed using western blot (WB) and enzyme-linked immunosorbent assay (ELISA). Areas under the receiver operating characteristic curves (AUROCs) were used to evaluate diagnostic accuracy for distinguishing between KC and healthy eyes. MAIN OUTCOME MEASURES Difference in PIP protein levels between patients with KC and healthy controls. RESULTS Results showed significant downregulation of PIP expression in all three biological fluids on KC patients when compared to healthy controls, independent of age, sex and severity. Since PIP is a hormonal-regulated protein, we also investigated the expression of major sex hormones. We detected significant upregulation in salivary and plasma Dehydroepiandrosterone sulfate (DHEA-S) levels and significant downregulation of estrone and estriol levels, in KC patients compared to healthy controls, independent of sex, age, and KC severity stage. ROC was used to determine the overall predictive accuracy of this protein in KC. Data showed an area under the curve (AUC) for PIP in tears of 0.937 (95%CI: 0.902-0.971), in plasma of 0.928 (95%CI: 0.890-0.968) and in saliva of 0.929 (95%CI: 0.890-0.968). CONCLUSIONS Conclusively, our results show that PIP levels are reduced in all three human biological fluids tested, and may independently or in combination with current imaging techniques aid in screening and diagnosis of KC. Our data revealed that PIP levels can potentially differentiate between disease and healthy cases, and PIP levels are stable in relation to KC severity, sex and age. Moreover, alterations in sex hormone levels in correlation with reduced PIP levels in KC provide an intriguing insight in the underlying KC pathophysiology and highlights the role of PIP as a KC biomarker.
Collapse
Affiliation(s)
- Rabab Sharif
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Sashia Bak-Nielsen
- Department of Ophthalmology, Aarhus University Hospital, Noerrebrogade 44, 8000, Aarhus, Denmark.
| | - Henrik Sejersen
- Department of Ophthalmology, Aarhus University Hospital, Noerrebrogade 44, 8000, Aarhus, Denmark.
| | - Kai Ding
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences, Oklahoma City, OK, 73104, USA
| | - Jesper Hjortdal
- Department of Ophthalmology, Aarhus University Hospital, Noerrebrogade 44, 8000, Aarhus, Denmark.
| | - Dimitrios Karamichos
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
8
|
Ihedioha O, Blanchard AA, Balhara J, Okwor I, Jia P, Uzonna J, Myal Y. The human breast cancer-associated protein, the prolactin-inducible protein (PIP), regulates intracellular signaling events and cytokine production by macrophages. Immunol Res 2019. [PMID: 29536339 DOI: 10.1007/s12026-018-8987-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The prolactin-inducible protein (PIP) is considered a valuable biomarker that is associated with both benign and malignant pathological conditions of the mammary gland. The function of PIP in breast tumorigenesis remains unknown; however, evidence from our laboratory and others suggest that it regulates host immunity. Studies with PIP-deficient (PIP-/-) mice demonstrated significantly lower numbers of CD4+ T cells in their secondary lymphoid organs, impaired Th1 response, and impaired nitric oxide (NO) production. To further delineate the immunoregulatory role of PIP, we compared the expression of IFN-γR and TLR4, pro-inflammatory cytokine production, and intracellular signaling events by IFN-γ and lipopolysaccharide (LPS)-stimulated macrophages from wild-type (WT) and PIP-/- mice. We showed that although the expressions of IFN-γR and TLR4 were comparable, productions of pro-inflammatory cytokines were decreased in PIP-/- macrophages. This was associated with decreased phosphorylation of mitogen-activated protein kinase (MAPK) and signal transducer of activation of transcription (STAT) proteins in macrophages from PIP-/- mice. Interestingly, the expression of suppressors of cytokine signaling (SOCS) 1 and 3 proteins, known to suppress IFN-γ and LPS signaling, was higher in PIP-/- macrophages compared to those from WT mice. Collectively, our studies show that deficiency of PIP significantly affects intracellular signaling events leading to decreased pro-inflammatory cytokine production, and further confirms a role for PIP as an important immunoregulatory protein. This direct link between PIP and cell-mediated immunity, a key component of the immune system that is critical for cancer control, may have significant therapeutic implications.
Collapse
Affiliation(s)
- Olivia Ihedioha
- Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anne A Blanchard
- Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jyoti Balhara
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Ifeoma Okwor
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ping Jia
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jude Uzonna
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Yvonne Myal
- Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
9
|
Urbaniak A, Jablonska K, Podhorska-Okolow M, Ugorski M, Dziegiel P. Prolactin-induced protein (PIP)-characterization and role in breast cancer progression. Am J Cancer Res 2018; 8:2150-2164. [PMID: 30555735 PMCID: PMC6291655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023] Open
Abstract
Prolactin-induced protein (PIP) is a small secreted glycoprotein carrying several N-linked carbohydrate chains. The expression of PIP is generally restricted to cells with apocrine properties. It was found in apocrine glands of the axilla, vulva, eyelid, ear canal, and seminal vesicle. Being a secretory protein, PIP is present in seminal plasma, saliva, lacrimal fluid, tears, sweat gland secretion. Little is known about the biological role of PIP. It binds to numerous proteins, however, in most cases the biological role of such interactions is poorly understood. A notable exception is its binding to CD4 receptors present on the surface of T lymphocytes, macrophages, and spermatozoa. The available data suggest that PIP can have immunomodulatory functions and plays an important role in cell-mediate adoptive immunity. PIP binds to bacteria from several genera, which suggests that this glycoprotein may participate also in innate immunity and protection of hosts against microbial infections. Increased levels of PIP were found in several types of human cancer (prostate, sweat and salivary gland cancers). It is especially common in breast cancer, however, data on the expression of PIP in normal and cancerous breast cancer tissues are to some degree conflicting. In early studies, it was shown that PIP is absent or its expression is very low in normal breast epithelium, whereas in breast cancers PIP is frequently expressed and present in large amounts. On the other hand, later study showed that expression of PIP is lower in advanced apocrine carcinomas and invasive carcinomas than in, respectively, in situ carcinomas and adjacent normal tissue. The most recent study revealed that PIP gene expression decreased gradually along with higher stage and grade of breast cancer. In agreement with these data, it was shown that that low levels or the lack of PIP expression are associated with a worse response of breast cancer cells to chemotherapy. It was proposed that PIP plays important role in the development and progression of breast cancer. However, its role in these processes is both unclear and controversial. In this review, the role of PIP in both physiological processes and carcinogenesis is discussed.
Collapse
Affiliation(s)
- Anna Urbaniak
- Laboratory of Glycobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWroclaw, Poland
- Department of Human Morphology, Division of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | - Karolina Jablonska
- Department of Human Morphology, Division of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | - Marzenna Podhorska-Okolow
- Department of Human Morphology, Division of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | - Maciej Ugorski
- Laboratory of Glycobiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWroclaw, Poland
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life SciencesWroclaw, Poland
| | - Piotr Dziegiel
- Department of Human Morphology, Division of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
- Department of Physiotherapy, University School of Physical EducationWroclaw, Poland
| |
Collapse
|
10
|
Al-Zahrani KN, Cook DP, Vanderhyden BC, Sabourin LA. Assessing the efficacy of androgen receptor and Sox10 as independent markers of the triple-negative breast cancer subtype by transcriptome profiling. Oncotarget 2018; 9:33348-33359. [PMID: 30279965 PMCID: PMC6161783 DOI: 10.18632/oncotarget.26072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/13/2018] [Indexed: 11/25/2022] Open
Abstract
The Androgen Receptor (AR) has recently garnered a lot of attention as a potential biomarker and therapeutic target in hormone-dependent cancers, including breast cancer. However, several inconsistencies exist within the literature as to which subtypes of breast cancer express AR or whether it can be used to define its own unique subtype. Here, we analyze 1246 invasive breast cancer samples from the Cancer Genome Atlas and show that human breast cancers that have been subtyped based on their HER2, ESR1, or PGR expression contain four clusters of genes that are differentially expressed across all subtypes. We demonstrate that Sox10 is highly expressed in approximately one-third of all HER2/ESR1/PGR-low tumors and is a candidate biomarker of the triple-negative subtype. Although AR expression is acquired in many breast cancer cases, its expression could not define a unique subtype. Despite several reports stating that AR expression is acquired in HER2/ESR1/PGR triple-negative cancers, here we show that a low percentage of these cancers express AR (~20%). In contrast, AR is highly expressed in HER2-positive or ESR1/PGR-positive cancers (> 95%). Although AR expression cannot be used as an independent subtype biomarker, our analysis shows that routine evaluation of AR expression in tumors which express HER2, ESR1 and/or PGR may identify a unique subset of tumors which would benefit from anti-androgen based therapies.
Collapse
Affiliation(s)
- Khalid N Al-Zahrani
- Ottawa Hospital Research Institute, Centre for Cancer Therapeutics, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David P Cook
- Ottawa Hospital Research Institute, Centre for Cancer Therapeutics, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barbara C Vanderhyden
- Ottawa Hospital Research Institute, Centre for Cancer Therapeutics, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Luc A Sabourin
- Ottawa Hospital Research Institute, Centre for Cancer Therapeutics, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
11
|
Prolactin Induced Protein (PIP) is a potential biomarker for early stage and malignant breast cancer. Breast 2018; 39:101-109. [DOI: 10.1016/j.breast.2018.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/18/2018] [Accepted: 03/29/2018] [Indexed: 11/21/2022] Open
|
12
|
MiR-23a transcriptional activated by Runx2 increases metastatic potential of mouse hepatoma cell via directly targeting Mgat3. Sci Rep 2018; 8:7366. [PMID: 29743543 PMCID: PMC5943354 DOI: 10.1038/s41598-018-25768-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/27/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) and aberrant glycosylation both play important roles in tumor metastasis. In this study, the role of miR-23a in N-glycosylation and the metastasis of mouse hepatocellular carcinoma (HCC) cells was investigated. The miRNA expression array profiles that were confirmed by qPCR and Western blot analyses revealed higher miR-23a expression levels in Hca-P cells (with lymphatic metastasis potential) than in Hepa1-6 cells (with no lymphatic metastasis potential), while the expression of mannoside acetylglucosaminyltransferase 3 (Mgat3) was negatively associated with metastasis potential. Mgat3 is a key glycosyltransferase in the synthesis of the bisecting (β1,4GlcNAc branching) N-glycan structure. Bioinformatics analysis indicated that Mgat3 may be a target of miR-23a, and this hypothesis was verified by dual-luciferase reporter gene assays. Furthermore, we found that the transcription factor Runx2 can directly bind to the miR-23a gene promoter and promote its expression, as shown in dual-luciferase reporter gene assays and ChIP assays. Collectively, these results indicate that miR-23a might increase the metastatic potential of mouse HCC by affecting the branch formation of N-glycan chains presented on the cell surface through the targeting of the glycosyltransferase Mgat3. These findings may provide insight into the relationship between abnormal miRNA expression and aberrant glycosylation during tumor lymphatic metastasis.
Collapse
|
13
|
Naderi A. SRARP and HSPB7 are epigenetically regulated gene pairs that function as tumor suppressors and predict clinical outcome in malignancies. Mol Oncol 2018; 12:724-755. [PMID: 29577611 PMCID: PMC5928383 DOI: 10.1002/1878-0261.12195] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/27/2018] [Accepted: 03/10/2018] [Indexed: 12/16/2022] Open
Abstract
Deletions of chromosome 1p36 are common in cancers; however, despite extensive studies, there has been limited success for discovering candidate tumor suppressors in this region. SRARP has recently been identified as a novel corepressor of the androgen receptor (AR) and is located on chromosome 1p36. Here, bioinformatics analysis of large tumor datasets was performed to study SRARP and its gene pair, HSPB7. In addition, using cancer cell lines, mechanisms of SRARP and HSPB7 regulation and their molecular functions were investigated. This study demonstrated that SRARP and HSPB7 are a gene pair located 5.2 kb apart on 1p36.13 and are inactivated by deletions and epigenetic silencing in malignancies. Importantly, SRARP and HSPB7 have tumor suppressor functions in clonogenicity and cell viability associated with the downregulation of Akt and ERK. SRARP expression is inversely correlated with genes that promote cell proliferation and signal transduction, which supports its functions as a tumor suppressor. In addition, AR exerts dual regulatory effects on SRARP, and although an increased AR activity suppresses SRARP transcription, a minimum level of AR activity is required to maintain baseline SRARP expression in AR+ cancer cells. Furthermore, as observed with SRARP, HSPB7 interacts with the 14-3-3 protein, presenting a shared molecular feature between SRARP and HSPB7. Of note, genome- and epigenome-wide associations of SRARP and HSPB7 with survival strongly support their tumor suppressor functions. In particular, DNA hypermethylation, lower expression, somatic mutations, and lower copy numbers of SRARP are associated with worse cancer outcome. Moreover, DNA hypermethylation and lower expression of SRARP in normal adjacent tissues predict poor survival, suggesting that SRARP inactivation is an early event in carcinogenesis. In summary, SRARP and HSPB7 are tumor suppressors that are commonly inactivated in malignancies. SRARP inactivation is an early event in carcinogenesis that is strongly associated with worse survival, presenting potential translational applications.
Collapse
Affiliation(s)
- Ali Naderi
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| |
Collapse
|
14
|
Naderi A. C1orf64 is a novel androgen receptor target gene and coregulator that interacts with 14-3-3 protein in breast cancer. Oncotarget 2017; 8:57907-57933. [PMID: 28915724 PMCID: PMC5593696 DOI: 10.18632/oncotarget.17826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/11/2017] [Indexed: 12/12/2022] Open
Abstract
This study investigated the network of genes that are co-expressed with androgen receptor (AR) to discover novel AR targets in breast cancer. Bioinformatics analysis of two datasets from breast cancer cell lines resulted in the identification of an AR-gene signature constituted of 98 genes that highly correlated with AR expression. Notably, C1orf64 showed the highest positive correlation with AR across the datasets with a correlation coefficient (CC) of 0.737. In addition, C1orf64 closely correlated with AR expression in primary and metastatic breast tumors and C1orf64 expression was relatively higher in breast tumors with a lower grade and lobular histology. Furthermore, there is a functional interplay between AR and C1orf64 in breast cancer. In this process, AR activation directly represses C1orf64 transcription and C1orf64, in turn, interacts with AR as a corepressor and negatively regulates the AR-mediated induction of prolactin-induced protein (PIP) and AR reporter activity. Moreover, the corepressor effect of C1orf64 results in a reduction of AR binding to PIP promoter. The other aspect of this interplay involves a cross-talk between AR and estrogen receptor (ER) signaling in which C1orf64 silencing intensifies the AR-mediated down-regulation of ER target gene, progesterone receptor. Therefore, the repression of C1orf64 by AR provides an underlying mechanism for the AR inhibitory effects on ER signaling. To elucidate the biochemical mechanisms of C1orf64 function, this study demonstrates that C1orf64 is a phosphothreonine protein that interacts with the chaperone protein 14-3-3. In summary, C1orf64 is a novel AR coregulator and a 14-3-3 binding partner in breast cancer.
Collapse
Affiliation(s)
- Ali Naderi
- University of Hawaii Cancer Center, Cancer Biology Program, Honolulu, Hawaii 96813, USA
| |
Collapse
|
15
|
Shaoxian T, Baohua Y, Xiaoli X, Yufan C, Xiaoyu T, Hongfen L, Rui B, Xiangjie S, Ruohong S, Wentao Y. Characterisation of GATA3 expression in invasive breast cancer: differences in histological subtypes and immunohistochemically defined molecular subtypes. J Clin Pathol 2017; 70:926-934. [PMID: 28428285 DOI: 10.1136/jclinpath-2016-204137] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 11/04/2022]
Abstract
AIMS GATA-binding protein 3 (GATA3) is a sensitive and relatively specific marker in breast and urothelial carcinomas. Its diagnostic utility in primary and metastatic breast cancers has been explored and confirmed. However, the relationship between GATA3 expression and different breast carcinoma intrinsic subtypes has not been specifically defined in the literature despite a few reports with a small number of cases. The aim of the current investigation is to clarify GATA3 expression among different histological subtypes and surrogate molecular breast carcinoma subtypes in a large series of cases. METHODS Immunohistochemical staining of GATA3, GCDFP15 and mammaglobin was performed in a cohort of 1637 cases of primary invasive breast carcinoma. The association of GATA3 expression with different histological and surrogate intrinsic subtypes was assessed and compared with the expression of GCDFP15 and mammaglobin. RESULTS The overall positivity of GATA3 across the various immunohistochemistry-based surrogate intrinsic subtypes was 99.51% for luminal A-like, 97.70% for luminal B-like, 68.50% for HER2 overexpression and 20.16% for triple negative breast cancers. GATA3 expression was positively correlated with estrogen receptor (ER)-positive (luminal subtypes) breast carcinomas. For luminal-like and HER2 overexpression subtypes, GATA3 was much more sensitive than GCDFP15 and mammaglobin. For triple negative tumours, GATA3 was less sensitive than GCDFP15. CONCLUSIONS GATA3 exhibits a relatively high sensitivity for breast carcinomas. It is more sensitive than GCDFP15 and mammaglobin in luminal-like and HER2 overexpression subtypes. GATA3 expression is associated with breast carcinomas of luminal subtype and low histological grade.
Collapse
Affiliation(s)
- Tang Shaoxian
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Baohua
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xu Xiaoli
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Yufan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tu Xiaoyu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu Hongfen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bi Rui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sun Xiangjie
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shui Ruohong
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Wentao
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. J Hematol Oncol 2017; 10:89. [PMID: 28412963 PMCID: PMC5392969 DOI: 10.1186/s13045-017-0462-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022] Open
Abstract
Background Estrogen and progesterone are potent breast mitogens. In addition to steroid hormones, multiple signaling pathways input to estrogen receptor (ER) and progesterone receptor (PR) actions via posttranslational events. Protein kinases commonly activated in breast cancers phosphorylate steroid hormone receptors (SRs) and profoundly impact their activities. Methods To better understand the role of modified PRs in breast cancer, we measured total and phospho-Ser294 PRs in 209 human breast tumors represented on 2754 individual tissue spots within a tissue microarray and assayed the regulation of this site in human tumor explants cultured ex vivo. To complement this analysis, we assayed PR target gene regulation in T47D luminal breast cancer models following treatment with progestin (promegestone; R5020) and antiprogestins (mifepristone, onapristone, or aglepristone) in conditions under which the receptor is regulated by Lys388 SUMOylation (K388 intact) or is SUMO-deficient (via K388R mutation to mimic persistent Ser294 phosphorylation). Selected phospho-PR-driven target genes were validated by qRT-PCR and following RUNX2 shRNA knockdown in breast cancer cell lines. Primary and secondary mammosphere assays were performed to implicate phospho-Ser294 PRs, epidermal growth factor signaling, and RUNX2 in breast cancer stem cell biology. Results Phospho-Ser294 PR species were abundant in a majority (54%) of luminal breast tumors, and PR promoter selectivity was exquisitely sensitive to posttranslational modifications. Phospho-PR expression and target gene programs were significantly associated with invasive lobular carcinoma (ILC). Consistent with our finding that activated phospho-PRs undergo rapid ligand-dependent turnover, unique phospho-PR gene signatures were most prevalent in breast tumors clinically designated as PR-low to PR-null (luminal B) and included gene sets associated with cancer stem cell biology (HER2, PAX2, AHR, AR, RUNX). Validation studies demonstrated a requirement for RUNX2 in the regulation of selected phospho-PR target genes (SLC37A2). In vitro mammosphere formation assays support a role for phospho-Ser294-PRs via growth factor (EGF) signaling as well as RUNX2 as potent drivers of breast cancer stem cell fate. Conclusions We conclude that PR Ser294 phosphorylation is a common event in breast cancer progression that is required to maintain breast cancer stem cell fate, in part via cooperation with growth factor-initiated signaling pathways and key phospho-PR target genes including SLC37A2 and RUNX2. Clinical measurement of phosphorylated PRs should be considered a useful marker of breast tumor stem cell potential. Alternatively, unique phospho-PR target gene sets may provide useful tools with which to identify patients likely to respond to selective PR modulators that block PR Ser294 phosphorylation as part of rational combination (i.e., with antiestrogens) endocrine therapies designed to durably block breast cancer recurrence. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0462-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Todd P Knutson
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Thu H Truong
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Shihong Ma
- Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, J8.130C, Dallas, TX, 75390-9110, USA
| | - Nicholas J Brady
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Megan E Sullivan
- Department of Pathology, Evanston Hospital, University of Chicago, NorthShore University HealthSystem, Evanston, IL, 60201, USA
| | - Ganesh Raj
- Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, J8.130C, Dallas, TX, 75390-9110, USA
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Carol A Lange
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
17
|
Role of Runx2 in breast cancer-mediated bone metastasis. Int J Biol Macromol 2017; 99:608-614. [PMID: 28268169 DOI: 10.1016/j.ijbiomac.2017.03.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Breast cancer is one of the most prevalent forms of cancer in women. The currently available treatment for breast cancer is mostly curative except when it becomes metastatic. One of the major sites for metastasis of breast cancer is the bone. Homing of the circulating tumor cells is tightly regulated including a number of factors present in the cells and their microenvironment. Runx2, a transcription factor plays an important role in osteogenesis and breast cancer mediated bone metastases. One of the recent advances in molecular therapy includes the discovery of the small, non-coding microRNAs (miRNAs) and they target specific genes to reduce their expression at the post-transcriptional level. This review provides an outline of breast cancer mediated bone metastasis and summarizes the recent development on the regulation of Runx2 expression by miRNAs which can lead to novel molecular therapeutics for the same.
Collapse
|
18
|
Ihedioha OC, Shiu RP, Uzonna JE, Myal Y. Prolactin-Inducible Protein: From Breast Cancer Biomarker to Immune Modulator—Novel Insights from Knockout Mice. DNA Cell Biol 2016; 35:537-541. [DOI: 10.1089/dna.2016.3472] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
| | - Robert P.C. Shiu
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Jude E. Uzonna
- Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Yvonne Myal
- Department of Pathology, University of Manitoba, Winnipeg, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
19
|
Vuong W, Tew BY, Little GH, Frenkel B, Jones JO. High-Throughput Screen for Inhibitors of Androgen Receptor-RUNX2 Transcriptional Regulation in Prostate Cancer. J Pharmacol Exp Ther 2016; 359:256-261. [PMID: 27554677 DOI: 10.1124/jpet.116.234567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/12/2016] [Indexed: 01/03/2023] Open
Abstract
Runt-related transcription factor 2 (RUNX2) plays a critical role in prostate cancer progression. RUNX2 interacts with the androgen receptor (AR) and modulates its transcriptional activity in a locus-specific manner. RUNX2 and AR synergistically stimulate a subset of genes, including the pro-oncogene snail family zinc finger 2 (SNAI2). AR-RUNX2 signaling cooperatively induces invasiveness of prostate cancer cells via SNAI2; and coexpression of AR, RUNX2, and SNAI2 in prostate cancer biopsy samples predicts disease recurrence. Competitive inhibition of AR alone could not disrupt the synergistic activation of SNAI2. We therefore established a phenotypic cell-based screening assay for compounds that could inhibit AR-RUNX2 synergistic activity either directly or indirectly. This assay was used to screen 880 compounds as a proof of concept, resulting in identification of several compounds that disrupted the synergistic stimulation of genes. Further investigation suggested the involvement of epidermal growth factor receptor (EGFR) signaling in AR/RUNX2 synergistic activity. Our assay is amenable to high-throughput screening and can be used to identify inhibitors of the AR-RUNX2 interaction in prostate cancer cells.
Collapse
Affiliation(s)
- Winston Vuong
- Beckman Research Institute, City of Hope, Duarte, California (W.V., B.Y.T., J.O.J.); Keck School of Medicine, University of Southern California, Los Angeles, California (G.H.L., B.F.)
| | - Ben Y Tew
- Beckman Research Institute, City of Hope, Duarte, California (W.V., B.Y.T., J.O.J.); Keck School of Medicine, University of Southern California, Los Angeles, California (G.H.L., B.F.)
| | - Gillian H Little
- Beckman Research Institute, City of Hope, Duarte, California (W.V., B.Y.T., J.O.J.); Keck School of Medicine, University of Southern California, Los Angeles, California (G.H.L., B.F.)
| | - Baruch Frenkel
- Beckman Research Institute, City of Hope, Duarte, California (W.V., B.Y.T., J.O.J.); Keck School of Medicine, University of Southern California, Los Angeles, California (G.H.L., B.F.)
| | - Jeremy O Jones
- Beckman Research Institute, City of Hope, Duarte, California (W.V., B.Y.T., J.O.J.); Keck School of Medicine, University of Southern California, Los Angeles, California (G.H.L., B.F.)
| |
Collapse
|
20
|
Jablonska K, Grzegrzolka J, Podhorska-Okolow M, Stasiolek M, Pula B, Olbromski M, Gomulkiewicz A, Piotrowska A, Rys J, Ambicka A, Ong SH, Zabel M, Dziegiel P. Prolactin-induced protein as a potential therapy response marker of adjuvant chemotherapy in breast cancer patients. Am J Cancer Res 2016; 6:878-893. [PMID: 27293986 PMCID: PMC4889707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/29/2016] [Indexed: 06/06/2023] Open
Abstract
Many studies are dedicated to exploring the molecular mechanisms of chemotherapy-resistance in breast cancer (BC). Some of them are focused on searching for candidate genes responsible for this process. The aim of this study was typing the candidate genes associated with the response to standard chemotherapy in the case of invasive ductal carcinoma. Frozen material from 28 biopsies obtained from IDC patients with different responses to chemotherapy were examined using gene expression microarray, Real-Time PCR (RT-PCR) and Western blot (WB). Based on the microarray results, further analysis of candidate gene expression was evaluated in 120 IDC cases by RT-PCR and in 224 IDC cases by immunohistochemistry (IHC). The results were correlated with clinical outcome and molecular subtype of the BC. Gene expression microarray revealed Prolactin-Induced Peptide (PIP) as a single gene differentially expressed in BC therapy responder or non-responder patients (p <0.05). The level of PIP expression was significantly higher in the BC therapy responder group than in the non-responder group at mRNA (p=0.0092) and protein level (p=0.0256). Expression of PIP mRNA was the highest in estrogen receptor positive (ER+) BC cases (p=0.0254) and it was the lowest in triple negative breast cancer (TNBC) (p=0.0336). Higher PIP mRNA expression was characterized by significantly longer disease free survival (DFS, p=0.0093), as well as metastasis free survival (MFS, p=0.0144). Additionally, PIP mRNA and PIP protein expression levels were significantly higher in luminal A than in other molecular subtypes and TNBC. Moreover significantly higher PIP expression was observed in G1, G2 vs. G3 cases (p=0.0027 and p=0.0013, respectively). Microarray analysis characterized PIP gene as a candidate for BC standard chemotherapy response marker. Analysis of clinical data suggests that PIP may be a good prognostic and predictive marker in IDC patients. Higher levels of PIP were related to longer DFS and MFS but not with OS.
Collapse
Affiliation(s)
- Karolina Jablonska
- Department of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | - Jedrzej Grzegrzolka
- Department of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | | | - Mariusz Stasiolek
- Department of Neurology, Polish Mother’s Memorial Hospital-Research InstituteLodz, Poland
| | - Bartosz Pula
- Department of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | - Mateusz Olbromski
- Department of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
| | | | | | - Janusz Rys
- Department of Tumor Pathology, Centre of Oncology, Maria Sklodowska-Curie Memorial InstituteCracow Branch, Cracow, Poland
| | - Aleksandra Ambicka
- Department of Tumor Pathology, Centre of Oncology, Maria Sklodowska-Curie Memorial InstituteCracow Branch, Cracow, Poland
| | - Siew Hwa Ong
- Acumen Research Laboratories, National University of SingaporeSingapore
| | - Maciej Zabel
- Department of Histology and EmbryologyPoznan, Poland
| | - Piotr Dziegiel
- Department of Histology and Embryology, Wroclaw Medical UniversityWroclaw, Poland
- Department of Physiotherapy, University School of Physical EducationWroclaw, Poland
| |
Collapse
|
21
|
Morimoto E, Li M, Khalid AB, Krum SA, Chimge NO, Frenkel B. Glucocorticoids Hijack Runx2 to Stimulate Wif1 for Suppression of Osteoblast Growth and Differentiation. J Cell Physiol 2016; 232:145-53. [PMID: 27061521 DOI: 10.1002/jcp.25399] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/22/2022]
Abstract
Inhibition of Runx2 is one of many mechanisms that suppress bone formation in glucocorticoid (GC)-induced osteoporosis (GIO). We profiled mRNA expression in ST2/Rx2(dox) cells after treatment with doxycycline (dox; to induce Runx2) and/or the synthetic GC dexamethasone (dex). As expected, dex typically antagonized Runx2-driven transcription. Select genes, however, were synergistic stimulated and this was confirmed by RT-qPCR. Among the genes synergistically stimulated by GCs and Runx2 was Wnt inhibitory Factor 1 (Wif1), and Wif1 protein was readily detectable in medium conditioned by cultures co-treated with dox and dex, but neither alone. Cooperation between Runx2 and GCs in stimulating Wif1 was also observed in primary preosteoblast cultures. GCs strongly inhibited dox-driven alkaline phosphatase (ALP) activity in control ST2/Rx2(dox) cells, but not in cells in which Wif1 was silenced. Unlike its anti-mitogenic activity in committed osteoblasts, induction of Runx2 transiently increased the percentage of cells in S-phase and accelerated proliferation in the ST2 mesenchymal pluripotent cell culture model. Furthermore, like the inhibition of Runx2-driven ALP activity, dex antagonized the transient mitogenic effect of Runx2 in ST2/Rx2(dox) cultures, and this inhibition eased upon Wif1 silencing. Plausibly, homeostatic feedback loops that rely on Runx2 activation to compensate for bone loss in GIO are thwarted, exacerbating disease progression through stimulation of Wif1. J. Cell. Physiol. 232: 145-153, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eri Morimoto
- Departments of Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Meng Li
- Bioinformatics Service Program, Norris Medical Library, University of Southern California, Los Angeles, California
| | - Aysha B Khalid
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Susan A Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Nyam-Osor Chimge
- Department of Medicine, Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Baruch Frenkel
- Departments of Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California. .,Department of Orthopedic Surgery, Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
22
|
Sowalsky AG, Sager R, Schaefer RJ, Bratslavsky G, Pandolfi PP, Balk SP, Kotula L. Loss of Wave1 gene defines a subtype of lethal prostate cancer. Oncotarget 2016; 6:12383-91. [PMID: 25906751 PMCID: PMC4494945 DOI: 10.18632/oncotarget.3564] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/13/2015] [Indexed: 12/15/2022] Open
Abstract
Genetic alterations involving TMPRSS2-ERG alterations and deletion of key tumor suppressor genes are associated with development and progression of prostate cancer (PCa). However, less defined are early events that may contribute to the development of high-risk metastatic prostate cancer. Bioinformatic analysis of existing tumor genomic data from PCa patients revealed that WAVE complex gene alterations are associated with a greater likelihood of prostate cancer recurrence. Further analysis of primary vs. castration resistant prostate cancer indicate that disruption of WAVE complex gene expression, and particularly WAVE1 gene (WASF1) loss, is also associated with castration resistance, where WASF1 is frequently co-deleted with PTEN and resists androgen deprivation therapy (ADT). Hence, we propose that WASF1 status defines a subtype of ADT-resistant patients. Better understanding of the effects of WAVE pathway disruption will lead to development of better diagnostic and treatment modalities.
Collapse
Affiliation(s)
- Adam G Sowalsky
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rebecca Sager
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Rachel J Schaefer
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Gennady Bratslavsky
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Pier Paolo Pandolfi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA
| | - Steven P Balk
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Cancer Research Institute, Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA 02215, USA
| | - Leszek Kotula
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.,Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
23
|
Heat shock protein 27 and gross cystic disease fluid protein 15 play critical roles in molecular apocrine breast cancer. Tumour Biol 2015; 37:8027-36. [PMID: 26711786 DOI: 10.1007/s13277-015-4712-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/20/2015] [Indexed: 10/22/2022] Open
Abstract
Molecular apocrine breast cancer (MABC) has a distinct hormonal profile, being estrogen receptor (ER) and progesterone receptor (PR) negative but androgen receptor (AR) positive. The clinical significance of MABC and its relative variables have not been absolutely clarified and remain to be determined. Five hundred cases of invasive breast carcinoma were randomly selected in this study, including 158 MABC cases and 342 nonMABC cases. Expression of ER, PR, epidermal growth factor receptor 2 (HER2), Ki67, AR, gross cystic disease fluid protein 15 (GCDFP15), and heat shock protein 27 (HSP27) were analyzed by immunohistochemistry. Differences of continuous variables between MABC and nonMABC subgroups were evaluated by the chi-square test. The Kaplan-Meier method was performed to evaluate disease-free survival (DFS) and overall survival (OS). The MABC subgroup had higher histological grade, bigger tumor size, more lymph node metastasis, and higher pTNM stage than the nonMABC subgroup (P < 0.05), and patients with MABC had poorer prognosis than those of the nonMABC subgroup (P < 0.05). Both GCDFP15 and HSP27 were expressed differently in the MABC and nonMABC subgroups (P < 0.05). Furthermore, in the MABC subgroup, positive HSP27 expression indicated higher risk of recurrence (P < 0.05) and positive GCDFP15 expression was also a poor marker for patient outcome (P < 0.05). MABC patients with HSP27 and GCDFP15 co-expression had worse outcome (P < 0.05). Our data suggested that MABC had a high risk of recurrence. Positive expression of both GCDFP15 and HSP27 were correlated with MABC malignancy. Targeting AR and HSP27 at the same time might offer a useful strategy to MABC.
Collapse
|
24
|
Melnik BC. Milk: an epigenetic amplifier of FTO-mediated transcription? Implications for Western diseases. J Transl Med 2015; 13:385. [PMID: 26691922 PMCID: PMC4687119 DOI: 10.1186/s12967-015-0746-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/04/2015] [Indexed: 12/14/2022] Open
Abstract
Single-nucleotide polymorphisms within intron 1 of the FTO (fat mass and obesity-associated) gene are associated with enhanced FTO expression, increased body weight, obesity and type 2 diabetes mellitus (T2DM). The N6-methyladenosine (m6A) demethylase FTO plays a pivotal regulatory role for postnatal growth and energy expenditure. The purpose of this review is to provide translational evidence that links milk signaling with FTO-activated transcription of the milk recipient. FTO-dependent demethylation of m6A regulates mRNA splicing required for adipogenesis, increases the stability of mRNAs, and affects microRNA (miRNA) expression and miRNA biosynthesis. FTO senses branched-chain amino acids (BCAAs) and activates the nutrient sensitive kinase mechanistic target of rapamycin complex 1 (mTORC1), which plays a key role in translation. Milk provides abundant BCAAs and glutamine, critical components increasing FTO expression. CpG hypomethylation in the first intron of FTO has recently been associated with T2DM. CpG methylation is generally associated with gene silencing. In contrast, CpG demethylation generally increases transcription. DNA de novo methylation of CpG sites is facilitated by DNA methyltransferases (DNMT) 3A and 3B, whereas DNA maintenance methylation is controlled by DNMT1. MiRNA-29s target all DNMTs and thus reduce DNA CpG methylation. Cow´s milk provides substantial amounts of exosomal miRNA-29s that reach the systemic circulation and target mRNAs of the milk recipient. Via DNMT suppression, milk exosomal miRNA-29s may reduce the magnitude of FTO methylation, thereby epigenetically increasing FTO expression in the milk consumer. High lactation performance with increased milk yield has recently been associated with excessive miRNA-29 expression of dairy cow mammary epithelial cells (DCMECs). Notably, the galactopoietic hormone prolactin upregulates the transcription factor STAT3, which induces miRNA-29 expression. In a retrovirus-like manner milk exosomes may transfer DCMEC-derived miRNA-29s and bovine FTO mRNA to the milk consumer amplifying FTO expression. There is compelling evidence that obesity, T2DM, prostate and breast cancer, and neurodegenerative diseases are all associated with increased FTO expression. Maximization of lactation performance by veterinary medicine with enhanced miRNA-29s and FTO expression associated with increased exosomal miRNA-29 and FTO mRNA transfer to the milk consumer may represent key epigenetic mechanisms promoting FTO/mTORC1-mediated diseases of civilization.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Sedanstrasse 115, 49090, Osnabrück, Germany.
| |
Collapse
|
25
|
Prolactin-Induced Protein regulates cell adhesion in breast cancer. Biochem Biophys Res Commun 2015; 468:850-6. [PMID: 26585492 DOI: 10.1016/j.bbrc.2015.11.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
Prolactin-Induced Prolactin (PIP) is widely expressed in breast cancer and has key cellular functions in this disease that include promoting invasion and cell cycle progression. Notably, we have recently identified a strong association between PIP-binding partners and a number of cell functions that are involved in cell adhesion. Therefore in this study, we investigated the effect of PIP on the regulation of cell adhesion using PIP-silencing in breast cancer cell lines T-47D, BT-474, and MFM-223. Our findings suggest that PIP expression is necessary for cell adhesion in a process that shows variation in the pattern of PIP regulation of cell-matrix and cell-cell adhesions based on the types of adhesion surface and breast cancer cell line. In this respect, we observed that PIP-silencing markedly reduced cell adhesion to uncoated plates in all three cell lines. In addition, in T-47D and MFM-223 cells fibronectin matrix induced baseline adhesion and reversed the PIP-silencing mediated reduction of cell adhesion. However, in BT-474 cells we did not observe an induction of baseline adhesion by fibronectin and PIP-silencing led to a marked reduction in cell adhesion to both uncoated and fibronectin-coated plates. Furthermore, we observed a significant reduction in cell-cell adhesion of BT-474 cell line following PIP-silencing. To explain an underlying mechanism for PIP regulation of cell adhesion, we found that PIP expression is necessary for the formation of α-actinin/actin-rich podosomes at the adhesion-sites of breast cancer cells. In summary, this study suggests that PIP expression regulates the process of cell adhesion in breast cancer.
Collapse
|
26
|
Thalji NM, Hagler MA, Zhang H, Casaclang-Verzosa G, Nair AA, Suri RM, Miller JD. Nonbiased Molecular Screening Identifies Novel Molecular Regulators of Fibrogenic and Proliferative Signaling in Myxomatous Mitral Valve Disease. ACTA ACUST UNITED AC 2015; 8:516-28. [PMID: 25814644 DOI: 10.1161/circgenetics.114.000921] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 03/12/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathological processes underlying myxomatous mitral valve degeneration (MMVD) remain poorly understood. We sought to identify novel mechanisms contributing to the development of this condition. METHODS AND RESULTS Microarrays were used to measure gene expression in 11 myxomatous and 11 nonmyxomatous human mitral valves. Differential gene expression (thresholds P<0.05; fold-change >1.5) and pathway activation (Ingenuity) were confirmed using quantitative reverse transcriptase polymerase chain reaction and immunohistochemistry. Contributions of bone morphogenetic protein 4 and transforming growth factor (TGF)-β2 to differential gene expression were evaluated in vitro. Contributions of angiotensin II to differential pathway activation were examined in mice in vivo. A total of 2602 genes were differentially expressed between myxomatous and nonmyxomatous valves. Canonical TGF-β signaling was increased in MMVD because of increased ligand expression and derepression of SMA mothers against decapentaplegic 2/3 signaling and was confirmed with quantitative reverse transcriptase polymerase chain reaction and immunohistochemistry. Myxomatous valves demonstrated activation of canonical bone morphogenetic protein and Wnt/β-catenin signaling and upregulation of their common target runt-related transcription factor 2. Our data set provided transcriptional and immunohistochemical evidence for activated immune cell infiltration. In vitro treatment of mitral valve interstitial cells with TGF-β2 increased β-catenin signaling at mRNA and protein levels, suggesting interactions between TGF-β2 and Wnt signaling. In vivo infusion of mice with angiotensin II recaptured several changes in signaling pathways characteristic of human MMVD. CONCLUSIONS These data support a new disease framework whereby activation of TGF-β2, bone morphogenetic protein 4, Wnt/β-catenin, or immune signaling plays major roles in the pathogenesis of MMVD. We propose these pathways act in a context-dependent manner to drive phenotypic changes that fundamentally differ from those observed in aortic valve disease and open novel avenues guiding future research into the pathogenesis of MMVD.
Collapse
Affiliation(s)
- Nassir M Thalji
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Michael A Hagler
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Heyu Zhang
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Grace Casaclang-Verzosa
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Asha A Nair
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
| | - Rakesh M Suri
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN.
| | - Jordan D Miller
- From the Department of Surgery, Division of Cardiovascular Surgery (N.M.T., M.A.H., H.Z., G.C.-V., R.M.S., J.D.M.), Department of Biomedical Statistics and Informatics (A.A.N.), and Department of Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
27
|
Todorova K, Metodiev MV, Metodieva G, Zasheva D, Mincheff M, Hayrabedyan S. miR-204 is dysregulated in metastatic prostate cancer in vitro. Mol Carcinog 2015; 55:131-47. [PMID: 25630658 DOI: 10.1002/mc.22263] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 11/03/2015] [Accepted: 11/07/2015] [Indexed: 02/04/2023]
Abstract
During cancer progression, the genome instability incurred rearrangement could possibly turn some of the tumor suppressor micro-RNAs into pro-oncogenic ones. We aimed to investigate miR-204 in the context of prostate cancer progression using a cell line model of different levels of genome instability (LNCaP, PC3, VCaP and NCI H660), as demonstrated by the availability of ERG fusion. We studied the effect of miR-204 modulation on master transcription factors important for lineage development, cell differentiation and prostate cancer bone marrow metastasis. We followed c-MYB, ETS1 and RUNX2 transcript and protein expression and the miR-204 affected global proteome. We further investigated if these transcription factors exert an effect on miR-204 expression (qPCR, luciferase reporter assay) by silencing them using esiRNA. We found dualistic miR-204 effects, either acting as a tumor suppressor on c-MYB, or as an oncomiR on ETS1. RUNX2 and ETS1 regulation by miR-204 was ERG fusion dependent, demonstrating regulatory circuitry disruption in advanced metastatic models. miR-204 also differentially affected mRNA splicing and protein stability. miR-204 levels were found dependent on cancer hypermethylation and supported by positive feedback induced by all three transcription factors. In this regulatory circuitry among miR-204, c-MYB, RUNX2 and ETS1, the c-MYB was found to induce all three other members, but its expression was differentially affected by the methylation status in lymph node vs. bone metastasis. We demonstrate that not only tumor suppressor micro-RNA loss, but also significant genome rearrangement-driven regulatory loop perturbations play a role in the advanced cancer progression, conferring better pro-survival and metastatic potential.
Collapse
Affiliation(s)
- Krassimira Todorova
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | - Diana Zasheva
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Milcho Mincheff
- Cellular and Gene Therapy Ward, National Specialized Hematology Hospital, Sofia, Bulgaria
| | - Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
28
|
Prolactin-induced protein is required for cell cycle progression in breast cancer. Neoplasia 2015; 16:329-42.e1-14. [PMID: 24862759 DOI: 10.1016/j.neo.2014.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/06/2014] [Accepted: 03/24/2014] [Indexed: 11/21/2022] Open
Abstract
Prolactin-induced protein (PIP) is expressed in the majority of breast cancers and is used for the diagnostic evaluation of this disease as a characteristic biomarker; however, the molecular mechanisms of PIP function in breast cancer have remained largely unknown. In this study, we carried out a comprehensive investigation of PIP function using PIP silencing in a broad group of breast cancer cell lines, analysis of expression microarray data, proteomic analysis using mass spectrometry, and biomarker studies on breast tumors. We demonstrated that PIP is required for the progression through G1 phase, mitosis, and cytokinesis in luminal A, luminal B, and molecular apocrine breast cancer cells. In addition, PIP expression is associated with a transcriptional signature enriched with cell cycle genes and regulates key genes in this process including cyclin D1, cyclin B1, BUB1, and forkhead box M1 (FOXM1). It is notable that defects in mitotic transition and cytokinesis following PIP silencing are accompanied by an increase in aneuploidy of breast cancer cells. Importantly, we have identified novel PIP-binding partners in breast cancer and shown that PIP binds to β-tubulin and is necessary for microtubule polymerization. Furthermore, PIP interacts with actin-binding proteins including Arp2/3 and is needed for inside-out activation of integrin-β1 mediated through talin. This study suggests that PIP is required for cell cycle progression in breast cancer and provides a rationale for exploring PIP inhibition as a therapeutic approach in breast cancer that can potentially target microtubule polymerization.
Collapse
|
29
|
Ni YB, Tsang JYS, Chan SK, Tse GM. GATA-binding protein 3, gross cystic disease fluid protein-15 and mammaglobin have distinct prognostic implications in different invasive breast carcinoma subgroups. Histopathology 2015; 67:96-105. [DOI: 10.1111/his.12625] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/22/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Yun Bi Ni
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital; The Chinese University of Hong Kong; Hong Kong Hong Kong
| | - Julia Y S Tsang
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital; The Chinese University of Hong Kong; Hong Kong Hong Kong
| | - Siu Ki Chan
- Department of Pathology; Kwong Wah Hospital; Hong Kong Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital; The Chinese University of Hong Kong; Hong Kong Hong Kong
| |
Collapse
|
30
|
Sriram R, Lo V, Pryce B, Antonova L, Mears AJ, Daneshmand M, McKay B, Conway SJ, Muller WJ, Sabourin LA. Loss of periostin/OSF-2 in ErbB2/Neu-driven tumors results in androgen receptor-positive molecular apocrine-like tumors with reduced Notch1 activity. Breast Cancer Res 2015; 17:7. [PMID: 25592291 PMCID: PMC4355979 DOI: 10.1186/s13058-014-0513-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Periostin (Postn) is a secreted cell adhesion protein that activates signaling pathways to promote cancer cell survival, angiogenesis, invasion, and metastasis. Interestingly, Postn is frequently overexpressed in numerous human cancers, including breast, lung, colon, pancreatic, and ovarian cancer. METHODS Using transgenic mice expressing the Neu oncogene in the mammary epithelium crossed into Postn-deficient animals, we have assessed the effect of Postn gene deletion on Neu-driven mammary tumorigenesis. RESULTS Although Postn is exclusively expressed in the stromal fibroblasts of the mammary gland, Postn deletion does not affect mammary gland outgrowth during development or pregnancy. Furthermore, we find that loss of Postn in the mammary epithelium does not alter breast tumor initiation or growth in mouse mammary tumor virus (MMTV)-Neu expressing mice but results in an apocrine-like tumor phenotype. Surprisingly, we find that tumors derived from Postn-null animals express low levels of Notch protein and Hey1 mRNA but increased expression of androgen receptor (AR) and AR target genes. We show that tumor cells derived from wild-type animals do not proliferate when transplanted in a Postn-null environment but that this growth defect is rescued by the overexpression of active Notch or the AR target gene prolactin-induced protein (PIP/GCDFP-15). CONCLUSIONS Together our data suggest that loss of Postn in an ErbB2/Neu/HER2 overexpression model results in apocrine-like tumors that activate an AR-dependent pathway. This may have important implications for the treatment of breast cancers involving the therapeutic targeting of periostin or Notch signaling.
Collapse
Affiliation(s)
- Roshan Sriram
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Vivian Lo
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Benjamin Pryce
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Lilia Antonova
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| | - Alan J Mears
- Children's Hospital of Eastern Ontario, Research Institute, 501 Smyth Road, Ottawa, ON, K1H8L6, Canada.
| | - Manijeh Daneshmand
- Ottawa Hospital Research Institute, Cancer Therapeutics, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| | - Bruce McKay
- Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada.
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, 705 Riley Hospital Drive, Indianapolis, IN, 46202, USA.
| | - William J Muller
- Department of Biochemistry and Goodman Cancer Research Center, McGill University, 1200 Pine Avenue West, Montreal, QC, H3G 1A1, Canada.
| | - Luc A Sabourin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada. .,Ottawa Hospital Research Institute, Cancer Therapeutics, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
31
|
Naderi A. Prolactin-induced protein in breast cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 846:189-200. [PMID: 25472539 DOI: 10.1007/978-3-319-12114-7_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Prolactin-induced protein (PIP) is a 17-kDa single polypeptide chain that is secreted by a number of normal apocrine cells, such as milk, saliva, and seminal fluid. PIP is widely expressed in breast cancer and is commonly used as a diagnostic biomarker for the histopathological diagnosis of this disease. Expression of PIP in breast cancer is regulated by androgen and prolactin through a number of transcription factors and signaling cross-talks, including STAT5, Runx2, and CREB1. Notably, PIP is induced by a positive feedback loop between androgen receptor (AR) and extracellular signal-regulated kinase (ERK). The available data indicate that PIP has an aspartyl protease activity that can degrade fibronectin. Importantly, PIP is necessary for outside-in activation of integrin-β1 signaling pathway and regulation of key downstream signaling targets of this pathway such as interaction of integrin-β1 with integrin-linked kinase 1 (ILK1) and ErbB2. Furthermore, the importance of PIP in cell proliferation has been demonstrated by the fact that purified PIP promotes growth of breast cancer cells and PIP expression is necessary for the proliferation of T-47D and MDA-MB-453 cell lines. In addition to cell proliferation, PIP mediates invasion of breast cancer cells in a process that partially depends on the degradation of fibronectin by this protein. Therefore, PIP is a breast cancer-related protein that is expressed in a majority of breast tumors and has a significant function in the biology of this disease.
Collapse
Affiliation(s)
- Ali Naderi
- Holden Comprehensive Cancer Center, University of Iowa, 3202 MERF, 375 Newton Road, 52242, Iowa City, IA, USA,
| |
Collapse
|
32
|
Coagulation factor VII is regulated by androgen receptor in breast cancer. Exp Cell Res 2014; 331:239-250. [PMID: 25447311 DOI: 10.1016/j.yexcr.2014.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 11/22/2022]
Abstract
Androgen receptor (AR) is widely expressed in breast cancer; however, there is limited information on the key molecular functions and gene targets of AR in this disease. In this study, gene expression data from a cohort of 52 breast cancer cell lines was analyzed to identify a network of AR co-expressed genes. A total of 300 genes, which were significantly enriched for cell cycle and metabolic functions, showed absolute correlation coefficients (|CC|) of more than 0.5 with AR expression across the dataset. In this network, a subset of 35 "AR-signature" genes were highly co-expressed with AR (|CC|>0.6) that included transcriptional regulators PATZ1, NFATC4, and SPDEF. Furthermore, gene encoding coagulation factor VII (F7) demonstrated the closest expression pattern with AR (CC=0.716) in the dataset and factor VII protein expression was significantly associated to that of AR in a cohort of 209 breast tumors. Moreover, functional studies demonstrated that AR activation results in the induction of factor VII expression at both transcript and protein levels and AR directly binds to a proximal region of F7 promoter in breast cancer cells. Importantly, AR activation in breast cancer cells induced endogenous factor VII activity to convert factor X to Xa in conjunction with tissue factor. In summary, F7 is a novel AR target gene and AR activation regulates the ectopic expression and activity of factor VII in breast cancer cells. These findings have functional implications in the pathobiology of thromboembolic events and regulation of factor VII/tissue factor signaling in breast cancer.
Collapse
|
33
|
Koromila T, Baniwal SK, Song YS, Martin A, Xiong J, Frenkel B. Glucocorticoids antagonize RUNX2 during osteoblast differentiation in cultures of ST2 pluripotent mesenchymal cells. J Cell Biochem 2014; 115:27-33. [PMID: 23943595 DOI: 10.1002/jcb.24646] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 11/10/2022]
Abstract
The efficacy of glucocorticoids (GCs) in treating a wide range of autoimmune and inflammatory conditions is blemished by severe side effects, including osteoporosis. The chief mechanism leading to GC-induced osteoporosis is inhibition of bone formation, but the role of RUNX2, a master regulator of osteoblast differentiation and bone formation, has not been well studied. We assessed effects of the synthetic GC dexamethasone (dex) on transcription of RUNX2-stimulated genes during the differentiation of mesenchymal pluripotent cells into osteoblasts. Dex inhibited a RUNX2 reporter gene and attenuated locus-dependently RUNX2-driven expression of several endogenous target genes. The anti-RUNX2 activity of dex was not attributable to decreased RUNX2 expression, but rather to physical interaction between RUNX2 and the GC receptor (GR), demonstrated by co-immunoprecipitation assays and co-immunofluorescence imaging. Investigation of the RUNX2/GR interaction may lead to the development of bone-sparing GC treatment modalities for the management of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Theodora Koromila
- Department of Biochemistry and Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
While it has been known for decades that androgen hormones influence normal breast development and breast carcinogenesis, the underlying mechanisms have only been recently elucidated. To date, most studies have focused on androgen action in breast cancer cell lines, yet these studies represent artificial systems that often do not faithfully replicate/recapitulate the cellular, molecular and hormonal environments of breast tumours in vivo. It is critical to have a better understanding of how androgens act in the normal mammary gland as well as in in vivo systems that maintain a relevant tumour microenvironment to gain insights into the role of androgens in the modulation of breast cancer development. This in turn will facilitate application of androgen-modulation therapy in breast cancer. This is particularly relevant as current clinical trials focus on inhibiting androgen action as breast cancer therapy but, depending on the steroid receptor profile of the tumour, certain individuals may be better served by selectively stimulating androgen action. Androgen receptor (AR) protein is primarily expressed by the hormone-sensing compartment of normal breast epithelium, commonly referred to as oestrogen receptor alpha (ERa (ESR1))-positive breast epithelial cells, which also express progesterone receptors (PRs) and prolactin receptors and exert powerful developmental influences on adjacent breast epithelial cells. Recent lineage-tracing studies, particularly those focussed on NOTCH signalling, and genetic analysis of cancer risk in the normal breast highlight how signalling via the hormone-sensing compartment can influence normal breast development and breast cancer susceptibility. This provides an impetus to focus on the relationship between androgens, AR and NOTCH signalling and the crosstalk between ERa and PR signalling in the hormone-sensing component of breast epithelium in order to unravel the mechanisms behind the ability of androgens to modulate breast cancer initiation and growth.
Collapse
Affiliation(s)
- Gerard A Tarulli
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories (DRMCRL)Faculty of Health Sciences, School of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
35
|
A genome-wide RNAi screen identifies FOXO4 as a metastasis-suppressor through counteracting PI3K/AKT signal pathway in prostate cancer. PLoS One 2014; 9:e101411. [PMID: 24983969 PMCID: PMC4077825 DOI: 10.1371/journal.pone.0101411] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 06/05/2014] [Indexed: 11/21/2022] Open
Abstract
Activation of the PI3K/AKT signal pathway is a known driving force for the progression to castration-recurrent prostate cancer (CR-CaP), which constitutes the major lethal phenotype of CaP. Here, we identify using a genomic shRNA screen the PI3K/AKT-inactivating downstream target, FOXO4, as a potential CaP metastasis suppressor. FOXO4 protein levels inversely correlate with the invasive potential of a panel of human CaP cell lines, with decreased mRNA levels correlating with increased incidence of clinical metastasis. Knockdown (KD) of FOXO4 in human LNCaP cells causes increased invasion in vitro and lymph node (LN) metastasis in vivo without affecting indices of proliferation or apoptosis. Increased Matrigel invasiveness was found by KD of FOXO1 but not FOXO3. Comparison of differentially expressed genes affected by FOXO4-KD in LNCaP cells in culture, in primary tumors and in LN metastases identified a panel of upregulated genes, including PIP, CAMK2N1, PLA2G16 and PGC, which, if knocked down by siRNA, could decrease the increased invasiveness associated with FOXO4 deficiency. Although only some of these genes encode FOXO promoter binding sites, they are all RUNX2-inducible, and RUNX2 binding to the PIP promoter is increased in FOXO4-KD cells. Indeed, the forced expression of FOXO4 reversed the increased invasiveness of LNCaP/shFOXO4 cells; the forced expression of FOXO4 did not alter RUNX2 protein levels, yet it decreased RUNX2 binding to the PIP promoter, resulting in PIP downregulation. Finally, there was a correlation between FOXO4, but not FOXO1 or FOXO3, downregulation and decreased metastasis-free survival in human CaP patients. Our data strongly suggest that increased PI3K/AKT-mediated metastatic invasiveness in CaP is associated with FOXO4 loss, and that mechanisms to induce FOXO4 re-expression might suppress CaP metastatic aggressiveness.
Collapse
|
36
|
Little GH, Baniwal SK, Adisetiyo H, Groshen S, Chimge NO, Kim SY, Khalid O, Hawes D, Jones JO, Pinski J, Schones DE, Frenkel B. Differential effects of RUNX2 on the androgen receptor in prostate cancer: synergistic stimulation of a gene set exemplified by SNAI2 and subsequent invasiveness. Cancer Res 2014; 74:2857-68. [PMID: 24648349 DOI: 10.1158/0008-5472.can-13-2003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Changes to androgen signaling during prostate carcinogenesis are associated with both inhibition of cellular differentiation and promotion of malignant phenotypes. The androgen receptor (AR)-binding transcription factor RUNX2 has been linked to prostate cancer progression but the underlying mechanisms have not been fully defined. In this study, we investigated the genome-wide influence of RUNX2 on androgen-induced gene expression and AR DNA binding in prostate cancer cells. RUNX2 inhibited the androgen response partly by promoting the dissociation of AR from its target genes such as the tumor suppressor NKX3-1. However, AR activity persists in the presence of RUNX2 at other AR target genes, some of which are cooperatively stimulated by androgen and RUNX2 signaling. These genes are associated with putative enhancers co-occupied by AR and RUNX2. One such gene, the invasion-promoting Snail family transcription factor SNAI2, was co-activated by AR and RUNX2. Indeed, these two transcription factors together, but neither alone stimulated prostate cancer cell invasiveness, which could be abolished by SNAI2 silencing. Furthermore, an immunohistochemical analysis of SNAI2 in archived primary prostate cancer specimens revealed a correlation with the RUNX2 histoscore, and simultaneous strong staining for SNAI2, RUNX2, and AR (but not any pair alone) was associated with disease recurrence. Overall, our findings suggest cooperation between AR and RUNX in the stimulation of oncogenes such as SNAI2, which might be targeted for individualized prostate cancer therapy.
Collapse
Affiliation(s)
- Gillian H Little
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Sanjeev K Baniwal
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Helty Adisetiyo
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Susan Groshen
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Nyam-Osor Chimge
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Sun Young Kim
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Omar Khalid
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Debra Hawes
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Jeremy O Jones
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Jacek Pinski
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Dustin E Schones
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Baruch Frenkel
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CaliforniaAuthors' Affiliations: Departments of Biochemistry and Molecular Biology, Orthopedic Surgery, Preventive Medicine, and Medicine; Institute for Genetic Medicine; USC/Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles; Departments of Molecular Pharmacology and Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California
| |
Collapse
|
37
|
Parris TZ, Kovács A, Aziz L, Hajizadeh S, Nemes S, Semaan M, Forssell-Aronsson E, Karlsson P, Helou K. Additive effect of the AZGP1, PIP, S100A8 and UBE2C molecular biomarkers improves outcome prediction in breast carcinoma. Int J Cancer 2013; 134:1617-29. [PMID: 24114735 DOI: 10.1002/ijc.28497] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/12/2013] [Indexed: 01/03/2023]
Abstract
The deregulation of key cellular pathways is fundamental for the survival and expansion of neoplastic cells, which in turn can have a detrimental effect on patient outcome. To develop effective individualized cancer therapies, we need to have a better understanding of which cellular pathways are perturbed in a genetically defined subgroup of patients. Here, we validate the prognostic value of a 13-marker signature in independent gene expression microarray datasets (n = 1,141) and immunohistochemistry with full-faced FFPE samples (n = 71). The predictive performance of individual markers and panels containing multiple markers was assessed using Cox regression analysis. In the external gene expression dataset, six of the 13 genes (AZGP1, NME5, S100A8, SCUBE2, STC2 and UBE2C) retained their prognostic potential and were significantly associated with disease-free survival (p < 0.001). Protein analyses refined the signature to a four-marker panel [AZGP1, Prolactin-inducible protein (PIP), S100A8 and UBE2C] significantly correlated with cycling, high grade tumors and lower disease-specific survival rates. AZGP1 and PIP were found in significantly lower levels in invasive breast tissue as compared with adjacent normal tissue, whereas elevated levels of S100A8 and UBE2C were observed. A predictive model containing the four-marker panel in conjunction with established clinical variables outperformed a model containing the clinical variables alone. Our findings suggest that deregulated AZGP1, PIP, S100A8 and UBE2C are critical for the aggressive breast cancer phenotype, which may be useful as novel therapeutic targets for drug development to complement established clinical variables.
Collapse
Affiliation(s)
- Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Laquer VT, Hevezi PA, Albrecht H, Chen TS, Zlotnik A, Kelly KM. Microarray analysis of port wine stains before and after pulsed dye laser treatment. Lasers Surg Med 2013; 45:67-75. [PMID: 23440713 DOI: 10.1002/lsm.22087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Neither the pathogenesis of port wine stain (PWS) birthmarks nor tissue effects of pulsed dye laser (PDL) treatment of these lesions is fully understood. There are few published reports utilizing gene expression analysis in human PWS skin. We aim to compare gene expression in PWS before and after PDL, using DNA microarrays that represent most, if not all, human genes to obtain comprehensive molecular profiles of PWS lesions and PDL-associated tissue effects. MATERIALS AND METHODS Five human subjects had PDL treatment of their PWS. One week later, three biopsies were taken from each subject: normal skin (N); untreated PWS (PWS); PWS post-PDL (PWS + PDL). Samples included two lower extremity lesions, two facial lesions, and one facial nodule. High-quality total RNA isolated from skin biopsies was processed and applied to Affymetrix Human gene 1.0ST microarrays for gene expression analysis. We performed a 16 pair-wise comparison identifying either up- or down-regulated genes between N versus PWS and PWS versus PWS + PDL for four of the donor samples. The PWS nodule (nPWS) was analyzed separately. RESULTS There was significant variation in gene expression profiles between individuals. By doing pair-wise comparisons between samples taken from the same donor, we were able to identify genes that may participate in the formation of PWS lesions and PDL tissue effects. Genes associated with immune, epidermal, and lipid metabolism were up-regulated in PWS skin. The nPWS exhibited more profound differences in gene expression than the rest of the samples, with significant differential expression of genes associated with angiogenesis, tumorigenesis, and inflammation. CONCLUSION In summary, gene expression profiles from N, PWS, and PWS + PDL demonstrated significant variation within samples from the same donor and between donors. By doing pair-wise comparisons between samples taken from the same donor and comparing these results between donors, we were able to identify genes that may participate in formation of PWS and PDL effects. Our preliminary results indicate changes in gene expression of angiogenesis-related genes, suggesting that dysregulation of angiogenic signals and/or components may contribute to PWS pathology.
Collapse
Affiliation(s)
- Vivian T Laquer
- Department of Dermatology, University of California, Irvine, Irvine, California, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Baniwal SK, Chimge NO, Jordan VC, Tripathy D, Frenkel B. Prolactin-induced protein (PIP) regulates proliferation of luminal A type breast cancer cells in an estrogen-independent manner. PLoS One 2013; 8:e62361. [PMID: 23755096 PMCID: PMC3670933 DOI: 10.1371/journal.pone.0062361] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 03/20/2013] [Indexed: 11/18/2022] Open
Abstract
Prolactin-induced Protein (PIP), an aspartyl protease unessential for normal mammalian cell function, is required for the proliferation and invasion of some breast cancer (BCa) cell types. Because PIP expression is particularly high in the Luminal A BCa subtype, we investigated the roles of PIP in the related T47D BCa cell line. Nucleic acid and antibody arrays were employed to screen effects of PIP silencing on global gene expression and activation of receptor tyrosine kinases (RTKs), respectively. Expression of PIP-stimulated genes, as defined in the T47D cell culture model, was well correlated with the expression of PIP itself across a cohort of 557 mRNA profiles of diverse BCa tumors, and bioinformatics analysis revealed cJUN and cMYC as major nodes in the PIP-dependent gene network. Among 71 RTKs tested, PIP silencing resulted in decreased phosphorylation of focal adhesion kinase (FAK), ephrin B3 (EphB3), FYN, and hemopoietic cell kinase (HCK). Ablation of PIP also abrogated serum-induced activation of the downstream serine/threonine kinases AKT, ERK1/2, and JNK1. Consistent with these results, PIP-depleted cells exhibited defects in adhesion to fibronectin, cytoskeletal stress fiber assembly and protein secretion. In addition, PIP silencing abrogated the mitogenic response of T47D BCa cells to estradiol (E2). The dependence of BCa cell proliferation was unrelated, however, to estrogen signaling because: 1) PIP silencing did not affect the transcriptional response of estrogen target genes to hormone treatment, and 2) PIP was required for the proliferation of tamoxifen-resistant BCa cells. Pharmacological inhibition of PIP may therefore serve the bases for both augmentation of existing therapies for hormone-dependent tumors and the development of novel therapeutic approaches for hormone-resistant BCa.
Collapse
Affiliation(s)
- Sanjeev K Baniwal
- Department of Orthopedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America.
| | | | | | | | | |
Collapse
|
40
|
Lehmann-Che J, Hamy AS, Porcher R, Barritault M, Bouhidel F, Habuellelah H, Leman-Detours S, de Roquancourt A, Cahen-Doidy L, Bourstyn E, de Cremoux P, de Bazelaire C, Albiter M, Giacchetti S, Cuvier C, Janin A, Espié M, de Thé H, Bertheau P. Molecular apocrine breast cancers are aggressive estrogen receptor negative tumors overexpressing either HER2 or GCDFP15. Breast Cancer Res 2013; 15:R37. [PMID: 23663520 PMCID: PMC4053236 DOI: 10.1186/bcr3421] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 05/11/2013] [Indexed: 02/05/2023] Open
Abstract
Introduction Molecular apocrine (MA) tumors are estrogen receptor (ER) negative breast cancers characterized by androgen receptor (AR) expression. We analyzed a group of 58 transcriptionally defined MA tumors and proposed a new tool to identify these tumors. Methods We performed quantitative reverse transcription PCR (qRT-PCR) for ESR1, AR, FOXA1 and AR-related genes, and immunohistochemistry (IHC) for ER, PR, Human Epidermal Growth Factor Receptor 2 (HER2), CK5/6, CK17, EGFR, Ki67, AR, FOXA1 and GCDFP15 and we analyzed clinical features. Results MA tumors were all characterized by ESR1(-) AR(+) FOXA1(+) and AR-related genes positive mRNA profile. IHC staining on these tumors showed 93% ER(-), only 58% AR(+) and 90% FOXA1(+). 67% and 57% MA tumors were HER2(3+) and GCDFP15(+), respectively. Almost all MA tumors (94%) had the IHC signature HER2(3+) or GCDFP15(+) but none of the 13 control basal-like (BL) tumors did. Clinically, MA tumors were rather aggressive, with poor prognostic factors. Conclusion MA tumors could be better defined by their qRT-PCR-AR profile than by AR IHC. In addition, we found that HER2 or GCDFP15 protein overexpression is a sensitive and specific tool to differentiate MA from BL in the context of ER negative tumors. A composite molecular and IHC signature could, therefore, help to identify MA tumors in daily practice.
Collapse
|
41
|
Halbach S, Rigbolt KT, Wöhrle FU, Diedrich B, Gretzmeier C, Brummer T, Dengjel J. Alterations of Gab2 signalling complexes in imatinib and dasatinib treated chronic myeloid leukaemia cells. Cell Commun Signal 2013; 11:30. [PMID: 23607741 PMCID: PMC3640961 DOI: 10.1186/1478-811x-11-30] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/25/2013] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The Gab2 docking protein acts as an important signal amplifier downstream of various growth factor receptors and Bcr-Abl, the driver of chronic myeloid leukaemia (CML). Despite the success of Bcr-Abl tyrosine kinase inhibitors (TKI) in the therapy of CML, TKI-resistance remains an unsolved problem in the clinic. We have recently shown that Gab2 signalling counteracts the efficacy of four distinct Bcr-Abl inhibitors. In the course of that project, we noticed that two clinically relevant drugs, imatinib and dasatinib, provoke distinct alterations in the electrophoretic mobility of Gab2, its signalling output and protein interactions. As the signalling potential of the docking protein is highly modulated by its phosphorylation status, we set out to obtain more insights into the impact of TKIs on Gab2 phosphorylation. FINDINGS Using stable isotope labelling by amino acids in cell culture (SILAC)-based quantitative mass spectrometry (MS), we show now that imatinib and dasatinib provoke distinct effects on the phosphorylation status and interactome of Gab2. This study identifies several new phosphorylation sites on Gab2 and confirms many sites previously known from other experimental systems. At equimolar concentrations, dasatinib is more effective in preventing Gab2 tyrosine and serine/threonine phosphorylation than imatinib. It also affects the phosphorylation status of more residues than imatinib. In addition, we also identify novel components of the Gab2 signalling complex, such as casein kinases, stathmins and PIP1 as well as known interaction partners whose association with Gab2 is disrupted by imatinib and/or dasatinib. CONCLUSIONS By using MS-based proteomics, we have identified new and confirmed known phosphorylation sites and interaction partners of Gab2, which may play an important role in the regulation of this docking protein. Given the growing importance of Gab2 in several tumour entities we expect that our results will help to understand the complex regulation of Gab2 and how this docking protein can contribute to malignancy.
Collapse
Affiliation(s)
- Sebastian Halbach
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, Albert-Ludwigs-University Freiburg, Stefan-Meier-Str, 17, Freiburg 79104, Germany.
| | | | | | | | | | | | | |
Collapse
|
42
|
Purcell DJ, Khalid O, Ou CY, Little GH, Frenkel B, Baniwal SK, Stallcup MR. Recruitment of coregulator G9a by Runx2 for selective enhancement or suppression of transcription. J Cell Biochem 2012; 113:2406-14. [PMID: 22389001 DOI: 10.1002/jcb.24114] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Runx2, best known for its role in regulating osteoblast-specific gene expression, also plays an increasingly recognized role in prostate and breast cancer metastasis. Using the C4-2B/Rx2(dox) prostate cancer cell line that conditionally expressed Runx2 in response to doxycycline treatment, we identified and characterized G9a, a histone methyltransferase, as a novel regulator for Runx2 activity. G9a function was locus-dependent. Whereas depletion of G9a reduced expression of many Runx2 target genes, including MMP9, CSF2, SDF1, and CST7, expression of others, such as MMP13 and PIP, was enhanced. Physical association between G9a and Runx2 was indicated by co-immunoprecipitation, GST-pulldown, immunofluorescence, and fluorescence recovery after photobleaching (FRAP) assays. Since G9a makes repressive histone methylation marks and is primarily known as a corepressor, we further investigated the mechanism by which G9a functioned as a positive regulator for Runx2 target genes. Transient reporter assays indicated that the histone methyltransferase activity of G9a was not required for transcriptional activation by Runx2. Chromatin immunoprecipitation assays for Runx2 and G9a showed that G9a was recruited to endogenous Runx2 binding sites. We conclude that a subset of cancer-related Runx2 target genes require recruitment of G9a for their expression, but do not depend on its histone methyltransferase activity.
Collapse
Affiliation(s)
- Daniel J Purcell
- Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, California 90089-9176, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Sancisi V, Borettini G, Maramotti S, Ragazzi M, Tamagnini I, Nicoli D, Piana S, Ciarrocchi A. Runx2 isoform I controls a panel of proinvasive genes driving aggressiveness of papillary thyroid carcinomas. J Clin Endocrinol Metab 2012; 97:E2006-15. [PMID: 22821892 PMCID: PMC3462932 DOI: 10.1210/jc.2012-1903] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CONTEXT The ability of tumor cells to invade adjacent tissues is governed by a complicated network of molecular signals, most of which have not yet been identified. In a recent work, we reported that the transcriptional regulator Id1 contributes to thyroid cancer progression by powering the invasion capacity of tumor cells. OBJECTIVE The intent of this work was to further investigate the biology of invasive thyroid tumors, through the analysis of the molecular interactions existing between Id1 and some of its target genes and through the characterization of the function of these factors in the progression of thyroid tumors. RESULTS We showed that Id1 controls the expression of the Runx2 isoform I and that this transcription factor plays a central role in mediating the Id1 proinvasive function in thyroid tumor cells. We demonstrated that Runx2 regulates proliferation, migration, and invasiveness by activating a panel of genes involved in matrix degradation and cellular invasion, which we previously identified as Id1 target genes in thyroid tumor cells. Finally, we show that Runx2 is strongly expressed in metastatic human thyroid tumors both at the primary site and in metastases. CONCLUSION Overall, our experiments demonstrate the existence of a previously unknown molecular axis that controls thyroid tumor invasiveness by altering the ability of tumor cells to interact with the surrounding microenvironment. These factors could prove to be valuable markers that permit early diagnosis of aggressive thyroid tumors.
Collapse
Affiliation(s)
- Valentina Sancisi
- Laboratory of Molecular Biology, Department of Oncology, Azienda Ospedaliera Arcispedale S. Maria Nuova, Istituto di Ricovero e Cura a Carattere Scientifico, viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
La Montagna R, Caligiuri I, Maranta P, Lucchetti C, Esposito L, Paggi MG, Toffoli G, Rizzolio F, Giordano A. Androgen receptor serine 81 mediates Pin1 interaction and activity. Cell Cycle 2012; 11:3415-20. [PMID: 22894932 DOI: 10.4161/cc.21730] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hormone-dependent tumors are characterized by deregulated activity of specific steroid receptors, allowing aberrant expression of many genes involved in cancer initiation, progression and metastasis. In prostate cancer, the androgen receptor (AR) protein has pivotal functions, and over the years it has been the target of different drugs. AR is a nuclear receptor whose activity is regulated by a phosphorylation mechanism controlled by hormone and growth factors. Following phosphorylation, AR interacts with many cofactors that closely control its function. Among such cofactors, Pin1 is a peptidyl-prolyl isomerase that is involved in the control of protein phosphorylation and has a prognostic value in prostate cancer. In the present study, we demonstrate that ARSer81 is involved in the interaction with Pin1, and that this interaction is important for the transcriptional activity of AR. Since Pin1 expression positively correlates with tumor grade, our results suggest that Pin1 can participate in this process by modulating AR function.
Collapse
Affiliation(s)
- Raffaele La Montagna
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Naderi A, Meyer M. Prolactin-induced protein mediates cell invasion and regulates integrin signaling in estrogen receptor-negative breast cancer. Breast Cancer Res 2012; 14:R111. [PMID: 22817771 PMCID: PMC3680918 DOI: 10.1186/bcr3232] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/20/2012] [Indexed: 12/11/2022] Open
Abstract
Introduction Molecular apocrine is a subtype of estrogen receptor (ER)-negative breast cancer that is characterized by a steroid-response gene signature. We have recently identified a positive feedback loop between androgen receptor (AR) and extracellular signal-regulated kinase (ERK) signaling in this subtype. In this study, we investigated the transcriptional regulation of molecular apocrine genes by the AR-ERK feedback loop. Methods The transcriptional effects of AR and ERK inhibition on molecular apocrine genes were assessed in cell lines. The most regulated gene in this process, prolactin-induced protein (PIP), was further studied using immunohistochemistry of breast tumors and xenograft models. The transcriptional regulation of PIP was assessed by luciferase reporter assay and chromatin immunoprecipitation. The functional significance of PIP in cell invasion and viability was assessed using siRNA knockdown experiments and the mechanism of PIP effect on integrin-β1 signaling was studied using immunoblotting and immunoprecipitation. Results We found that PIP is the most regulated molecular apocrine gene by the AR-ERK feedback loop and is overexpressed in ER-/AR+ breast tumors. In addition, PIP expression is regulated by AR-ERK signaling in xenograft models. These observations are explained by the fact that PIP is a target gene of the ERK-CREB1 pathway and is also induced by AR activation. Furthermore, we demonstrated that PIP has a significant functional role in maintaining cell invasion and viability of molecular apocrine cells because of a positive regulatory effect on the Integrin-ERK and Integrin-Akt signaling pathways. In fact, PIP-knockdown markedly decreases the phosphorylation of ERK, Akt, and CREB1. Importantly, PIP knockdown leads to a marked reduction of integrin-β1 binding to ILK1 and ErbB2 that can be reversed by the addition of fibronectin fragments. Conclusions We have identified a novel feedback loop between PIP and CREB1 mediated through the Integrin signaling pathway. In this process, PIP cleaves fibronectin to release fragments that activate integrin signaling, which in turn increases PIP expression through the ERK-CREB1 pathway. In addition, we demonstrated that PIP expression has a profound effect on cell invasion and the viability of molecular apocrine cells. Therefore, PIP signaling may be a potential therapeutic target in molecular apocrine breast cancer.
Collapse
|
46
|
Muraoka S, Kume H, Watanabe S, Adachi J, Kuwano M, Sato M, Kawasaki N, Kodera Y, Ishitobi M, Inaji H, Miyamoto Y, Kato K, Tomonaga T. Strategy for SRM-based verification of biomarker candidates discovered by iTRAQ method in limited breast cancer tissue samples. J Proteome Res 2012; 11:4201-10. [PMID: 22716024 DOI: 10.1021/pr300322q] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since LC-MS-based quantitative proteomics has become increasingly applied to a wide range of biological applications over the past decade, numerous studies have performed relative and/or absolute abundance determinations across large sets of proteins. In this study, we discovered prognostic biomarker candidates from limited breast cancer tissue samples using discovery-through-verification strategy combining iTRAQ method followed by selected reaction monitoring/multiple reaction monitoring analysis (SRM/MRM). We identified and quantified 5122 proteins with high confidence in 18 patient tissue samples (pooled high-risk (n=9) or low-risk (n=9)). A total of 2480 proteins (48.4%) of them were annotated as membrane proteins, 16.1% were plasma membrane and 6.6% were extracellular space proteins by Gene Ontology analysis. Forty-nine proteins with >2-fold differences in two groups were chosen for further analysis and verified in 16 individual tissue samples (high-risk (n=9) or low-risk (n=7)) using SRM/MRM. Twenty-three proteins were differentially expressed among two groups of which MFAP4 and GP2 were further confirmed by Western blotting in 17 tissue samples (high-risk (n=9) or low-risk (n=8)) and Immunohistochemistry (IHC) in 24 tissue samples (high-risk (n=12) or low-risk (n=12)). These results indicate that the combination of iTRAQ and SRM/MRM proteomics will be a powerful tool for identification and verification of candidate protein biomarkers.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Ibaraki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Little GH, Noushmehr H, Baniwal SK, Berman BP, Coetzee GA, Frenkel B. Genome-wide Runx2 occupancy in prostate cancer cells suggests a role in regulating secretion. Nucleic Acids Res 2011; 40:3538-47. [PMID: 22187159 PMCID: PMC3333873 DOI: 10.1093/nar/gkr1219] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Runx2 is a metastatic transcription factor (TF) increasingly expressed during prostate cancer (PCa) progression. Using PCa cells conditionally expressing Runx2, we previously identified Runx2-regulated genes with known roles in epithelial-mesenchymal transition, invasiveness, angiogenesis, extracellular matrix proteolysis and osteolysis. To map Runx2-occupied regions (R2ORs) in PCa cells, we first analyzed regions predicted to bind Runx2 based on the expression data, and found that recruitment to sites upstream of the KLK2 and CSF2 genes was cyclical over time. Genome-wide ChIP-seq analysis at a time of maximum occupancy at these sites revealed 1603 high-confidence R2ORs, enriched with cognate motifs for RUNX, GATA and ETS TFs. The R2ORs were distributed with little regard to annotated transcription start sites (TSSs), mainly in introns and intergenic regions. Runx2-upregulated genes, however, displayed enrichment for R2ORs within 40 kb of their TSSs. The main annotated functions enriched in 98 Runx2-upregulated genes with nearby R2ORs were related to invasiveness and membrane trafficking/secretion. Indeed, using SDS-PAGE, mass spectrometry and western analyses, we show that Runx2 enhances secretion of several proteins, including fatty acid synthase and metastasis-associated laminins. Thus, combined analysis of Runx2's transcriptome and genomic occupancy in PCa cells lead to defining its novel role in regulating protein secretion.
Collapse
Affiliation(s)
- Gillian H Little
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Chimge NO, Baniwal SK, Little GH, Chen YB, Kahn M, Tripathy D, Borok Z, Frenkel B. Regulation of breast cancer metastasis by Runx2 and estrogen signaling: the role of SNAI2. Breast Cancer Res 2011; 13:R127. [PMID: 22151997 PMCID: PMC3326569 DOI: 10.1186/bcr3073] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 10/25/2011] [Accepted: 12/09/2011] [Indexed: 12/20/2022] Open
Abstract
Introduction In contrast to its role in breast cancer (BCa) initiation, estrogen signaling has a protective effect in later stages, where estrogen receptor (ER)α loss associates with aggressive metastatic disease. We asked whether the beneficial effect of estrogen signaling in late-stage BCa is attributable to the recently reported estrogen-mediated antagonism of the pro-metastatic transcription factor Runx2. Methods MCF7/Rx2dox breast cancer cells were engineered with a lentivirus expressing Runx2 in response to doxycycline (dox). Cells treated with dox and/or estradiol (E2) were subjected to genome-wide expression profiling, RT-qPCR analysis of specific genes, and Matrigel™ invasion assays. Knockdown of genes of interest was performed using lentiviruses expressing appropriate shRNAs, either constitutively or in response to dox. Gene expression in BCa tumors was investigated using a cohort of 557 patients compiled from publicly available datasets. Association of gene expression with clinical metastasis was assessed by dichotomizing patients into those expressing genes of interest at either high or low levels, and comparing the respective Kaplan-Meier curves of metastasis-free survival. Results Runx2 induced epithelial-mesenchymal transition (EMT) evidenced by acquisition of a fibroblastic morphology, decreased expression of E-cadherin, increased expression of vimentin and invasiveness. Runx2 stimulated SNAI2 expression in a WNT- and transforming growth factor (TGF)β-dependent manner, and knockdown of SNAI2 abrogated the pro-metastatic activities of Runx2. E2 antagonized the pro-metastatic activities of Runx2, including SNAI2 upregulation. In primary BCa tumors, Runx2 activity, SNAI2 expression, and metastasis were positively correlated, and SNAI2 expression was negatively correlated with ERα. However, the negative correlation between SNAI2 and ERα in bone-seeking BCa cells was weaker than the respective negative correlation in tumors seeking lung. Furthermore, the absence of ERα in primary tumors was associated with lung- and brain- but not with bone metastasis, and tumor biopsies from bone metastatic sites displayed the unusual combination of high Runx2/SNAI2 and high ERα expression. Conclusions E2 antagonizes Runx2-induced EMT and invasiveness of BCa cells, partly through attenuating expression of SNAI2, a Runx2 target required for mediating its pro-metastatic property. That ERα loss promotes non-osseous metastasis by unleashing Runx2/SNAI2 is supported by the negative correlation observed in corresponding tumors. Unknown mechanisms in bone-seeking BCa allow high Runx2/SNAI2 expression despite high ERα level
Collapse
Affiliation(s)
- Nyam-Osor Chimge
- Department of Biochemistry & Molecular Biology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Los Angeles, CA 90033, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Chimge NO, Baniwal SK, Luo J, Coetzee S, Khalid O, Berman BP, Tripathy D, Ellis MJ, Frenkel B. Opposing effects of Runx2 and estradiol on breast cancer cell proliferation: in vitro identification of reciprocally regulated gene signature related to clinical letrozole responsiveness. Clin Cancer Res 2011; 18:901-11. [PMID: 22147940 DOI: 10.1158/1078-0432.ccr-11-1530] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE To assess the clinical significance of the interaction between estrogen and Runx2 signaling, previously shown in vitro. EXPERIMENTAL DESIGN MCF7/Rx2(dox) breast cancer cells were treated with estradiol and/or doxycycline to induce Runx2, and global gene expression was profiled to define genes regulated by estradiol, Runx2, or both. Anchorage-independent growth was assessed by soft-agar colony formation assays. Expression of gene sets defined using the MCF7/Rx2(dox) system was analyzed in pre- and on-treatment biopsies from hormone receptor-positive patients undergoing neoadjuvant letrozole treatment in two independent studies, and short-term changes in gene expression were correlated with tumor size reduction or Ki67 index at surgery. RESULTS Reflecting its oncogenic property, estradiol strongly promoted soft-agar colony formation, whereas Runx2 blocked this process suggesting tumor suppressor property. Transcriptome analysis of MCF7/Rx2(dox) cells treated with estradiol and/or doxycycline showed reciprocal attenuation of Runx2 and estrogen signaling. Correspondingly in breast cancer tumors, expression of estradiol- and Runx2-regulated genes was inversely correlated, and letrozole increased expression of Runx2-stimulated genes, as defined in the MCF7/Rx2(dox) model. Of particular interest was a gene set upregulated by estradiol and downregulated by Runx2 in vitro; its short-term response to letrozole treatment associated with tumor size reduction and Ki67 index at surgery better than other estradiol-regulated gene sets. CONCLUSION This work provides clinical evidence for the importance of antagonism between Runx2 and E2 signaling in breast cancer. Likely sensing the tension between them, letrozole responsiveness of a genomic node, positively regulated by estradiol and negatively regulated by Runx2 in vitro, best correlated with the clinical efficacy of letrozole treatment.
Collapse
Affiliation(s)
- Nyam-Osor Chimge
- Department of Biochemistry, Institute for Genetic Medicine, USC Epigenome Center, Keck School of Medicine of the University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|