1
|
Gharbi S, Mohammadi Z, Dezaki MS, Dokanehiifard S, Dabiri S, Korsching E. Characterization of the first microRNA in human CDH1 that affects cell cycle and apoptosis and indicates breast cancers progression. J Cell Biochem 2022; 123:657-672. [PMID: 34997630 DOI: 10.1002/jcb.30211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/26/2021] [Accepted: 12/21/2021] [Indexed: 11/12/2022]
Abstract
The E-cadherin protein (Cadherin 1, gene: CDH1), a master regulator of the human epithelial homeostasis, contributes to the epithelial-mesenchymal transition (EMT) which confers cell migratory features to the cells. The EMT is central to many pathophysiological changes in cancer. Therefore, a better understanding of this regulatory scenario is beneficial for therapeutic regiments. The CDH1 gene is approximately 100 kbp long and consists of 16 exons with a relatively large second intron. Since none microRNA (miRNA) has been identified in CDH1 up to now we screened the CDH1 gene for promising miRNA hairpin structures in silico. Out of the 27 hairpin structures we identified, one stable RNA fold with a promising sequence motive was selected for experimental verification. The exogenous validation of the hairpin sequence was performed by transfection of HEK293T cells and the mature miRNA sequences could be verified by quantitative polymerase chain reaction. The endogenous expression of the mature miRNA provisionally named CDH1-i2-miR-1 could be confirmed in two normal (HEK293T, HUVEK) and five cancer cell lines (MCF7, MDA-MB-231, SW480, HT-29, A549). The functional characterization by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay showed a suppression of HEK293T cell proliferation. A flow cytometry-based approach showed the ability of CDH1-i2-miR-1 to arrest transfected cells on a G2/M state while annexin staining exemplified an apoptotic effect. BAX and PTEN expression levels were affected following the overexpression with the new miRNA. The in vivo expression level was assessed in 35 breast tumor tissues and their paired nonmalignant marginal part. A fourfold downregulation in the tumor specimens compared to their marginal controls could be observed. It can be concluded that the sequence of the hub gene CDH1 harbors at least one miRNA but eventually even more relevant for the pathophysiology of breast cancer.
Collapse
Affiliation(s)
- Sedigheh Gharbi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Zahra Mohammadi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Maryam Saedi Dezaki
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Sadat Dokanehiifard
- Department of Human Genetics, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shahriar Dabiri
- Department of Pathology, Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Eberhard Korsching
- Institute of Bioinformatics, Faculty of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
2
|
Targeting mitochondrial dysfunction with small molecules in intervertebral disc aging and degeneration. GeroScience 2021; 43:517-537. [PMID: 33634362 PMCID: PMC8110620 DOI: 10.1007/s11357-021-00341-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/09/2021] [Indexed: 02/08/2023] Open
Abstract
The prevalence of rheumatic and musculoskeletal diseases (RMDs) including osteoarthritis (OA) and low back pain (LBP) in aging societies present significant cost burdens to health and social care systems. Intervertebral disc (IVD) degeneration, which is characterized by disc dehydration, anatomical alterations, and extensive changes in extracellular matrix (ECM) composition, is an important contributor to LBP. IVD cell homeostasis can be disrupted by mitochondrial dysfunction. Mitochondria are the main source of energy supply in IVD cells and a major contributor to the production of reactive oxygen species (ROS). Therefore, mitochondria represent a double-edged sword in IVD cells. Mitochondrial dysfunction results in oxidative stress, cell death, and premature cell senescence, which are all implicated in IVD degeneration. Considering the importance of optimal mitochondrial function for the preservation of IVD cell homeostasis, extensive studies have been done in recent years to evaluate the efficacy of small molecules targeting mitochondrial dysfunction. In this article, we review the pathogenesis of mitochondrial dysfunction, aiming to highlight the role of small molecules and a selected number of biological growth factors that regulate mitochondrial function and maintain IVD cell homeostasis. Furthermore, molecules that target mitochondria and their mechanisms of action and potential for IVD regeneration are identified. Finally, we discuss mitophagy as a key mediator of many cellular events and the small molecules regulating its function.
Collapse
|
3
|
Dodda BR, Bondi CD, Hasan M, Clafshenkel WP, Gallagher KM, Kotlarczyk MP, Sethi S, Buszko E, Latimer JJ, Cline JM, Witt-Enderby PA, Davis VL. Co-administering Melatonin With an Estradiol-Progesterone Menopausal Hormone Therapy Represses Mammary Cancer Development in a Mouse Model of HER2-Positive Breast Cancer. Front Oncol 2019; 9:525. [PMID: 31355130 PMCID: PMC6636553 DOI: 10.3389/fonc.2019.00525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
Melatonin has numerous anti-cancer properties reported to influence cancer initiation, promotion, and metastasis. With the need for effective hormone therapies (HT) to treat menopausal symptoms without increasing breast cancer risk, co-administration of nocturnal melatonin with a natural, low-dose HT was evaluated in mice that develop primary and metastatic mammary cancer. Individually, melatonin (MEL) and estradiol-progesterone therapy (EPT) did not significantly affect mammary cancer development through age 14 months, but, when combined, the melatonin-estradiol-progesterone therapy (MEPT) significantly repressed tumor formation. This repression was due to effects on tumor incidence, but not latency. These results demonstrate that melatonin and the HT cooperate to decrease the mammary cancer risk. Melatonin and EPT also cooperate to alter the balance of the progesterone receptor (PR) isoforms by significantly increasing PRA protein expression only in MEPT mammary glands. Melatonin significantly suppressed amphiregulin transcripts in MEL and MEPT mammary glands, suggesting that amphiregulin together with the higher PRA:PRB balance and other factors may contribute to reducing cancer development in MEPT mice. Melatonin supplementation influenced mammary morphology by increasing tertiary branching in the mouse mammary glands and differentiation in human mammary epithelial cell cultures. Uterine weight in the luteal phase was elevated after long-term exposure to EPT, but not to MEPT, indicating that melatonin supplementation may reduce estrogen-induced uterine stimulation. Melatonin supplementation significantly decreased the incidence of grossly-detected lung metastases in MEL mice, suggesting that melatonin delays the formation of metastatic lesions and/or decreases aggressiveness in this model of HER2+ breast cancer. Mammary tumor development was similar in EPT and MEPT mice until age 8.6 months, but after 8.6 months, only MEPT continued to suppress cancer development. These data suggest that melatonin supplementation has a negligible effect in young MEPT mice, but is required in older mice to inhibit tumor formation. Since melatonin binding was significantly decreased in older mammary glands, irrespective of treatment, melatonin supplementation may overcome reduced melatonin responsiveness in the aged MEPT mice. Since melatonin levels are known to decline near menopause, nocturnal melatonin supplementation may also be needed in aging women to cooperate with HT to decrease breast cancer risk.
Collapse
Affiliation(s)
- Balasunder R Dodda
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Corry D Bondi
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Mahmud Hasan
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - William P Clafshenkel
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Katie M Gallagher
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Mary P Kotlarczyk
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Shalini Sethi
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Ethan Buszko
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Jean J Latimer
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - J Mark Cline
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Paula A Witt-Enderby
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.,UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Vicki L Davis
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Chi-miR-3031 regulates beta-casein via the PI3K/AKT-mTOR signaling pathway in goat mammary epithelial cells (GMECs). BMC Vet Res 2018; 14:369. [PMID: 30482199 PMCID: PMC6258393 DOI: 10.1186/s12917-018-1695-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MicroRNAs can regulate gene expression at the posttranscriptional level through translational repression or target degradation. Our previous investigations examined the differential expression levels of chi-miR-3031 in caprine mammary gland tissues in colostrum and common milk stages. RESULTS The present study detected the role of chi-miR-3031 in the lactation mechanisms of GMECs. High-throughput sequencing was used to analyze transcriptomic landscapes of GMECs transfected with chi-miR-3031 mimics (MC) and a mimic negative control (NC). In the MC and NC groups, we acquired 39,793,503 and 36,531,517 uniquely mapped reads, respectively, accounting for 85.85 and 81.66% of total reads. In the MC group, 180 differentially expressed unigenes were downregulated, whereas 157 unigenes were upregulated. KEGG pathway analyses showed that the prolactin, TNF and ErbB signaling pathways, including TGFα, PIK3R3, IGF2, ELF5, IGFBP5 and LHβ genes, played important roles in mammary development and milk secretion. Results from transcriptome sequencing, real-time PCR and western blotting showed that chi-miR-3031 suppressed the expression of IGFBP5 mRNA and protein. The expression levels of β-casein significantly increased in the MC and siRNA-IGFBP5 groups. We observed that the down-regulation of IGFBP5 activated mTOR at the Ser2448 site in GMECs transfected with MC and siRNA-IGFBP5. Previous findings and our results showed that chi-miR-3031 activated the PI3K-AKT-mTOR pathway and increased β-casein expression by down-regulating IGFBP5. CONCLUSIONS These findings will afford valuable information for improving milk quality and contribute the development of potential methods for amending lactation performance.
Collapse
|
5
|
Petersen I, Dietel M, Geilenkeuser WJ, Mireskandari M, Weichert W, Steiger K, Scheel AH, Büttner R, Schirmacher P, Warth A, Lasitschka F, Schildhaus HU, Kirchner T, Reu S, Kreipe H, Länger F, Tiemann M, Schulte C, Jöhrens K. EGFR immunohistochemistry as biomarker for antibody-based therapy of squamous NSCLC – Experience from the first ring trial of the German Quality Assurance Initiative for Pathology (QuIP ® ). Pathol Res Pract 2017; 213:1530-1535. [DOI: 10.1016/j.prp.2017.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/16/2017] [Accepted: 09/16/2017] [Indexed: 10/18/2022]
|
6
|
Bohrer LR, Chaffee TS, Chuntova P, Brady NJ, Witschen PM, Kemp SE, Nelson AC, Walcheck B, Schwertfeger KL. ADAM17 in tumor associated leukocytes regulates inflammatory mediators and promotes mammary tumor formation. Genes Cancer 2016; 7:240-253. [PMID: 27738494 PMCID: PMC5059114 DOI: 10.18632/genesandcancer.115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The presence of inflammatory cells within the tumor microenvironment has been tightly linked to mammary tumor formation and progression. Specifically, interactions between tumor cells and infiltrating macrophages can contribute to the generation of a pro-tumorigenic microenvironment. Understanding the complex mechanisms that drive tumor cell-macrophage cross-talk will ultimately lead to the development of approaches to prevent or treat early stage breast cancers. As described here, we demonstrate that the cell surface protease a disintegrin and metalloproteinase 17 (ADAM17) is expressed by macrophages in mammary tumors and contributes to regulating the expression of pro-inflammatory mediators, including inflammatory cytokines and the inflammatory mediator cyclooxygenase-2 (Cox-2). Furthermore, we demonstrate that ADAM17 is expressed on leukocytes, including macrophages, within polyoma middle T (PyMT)-derived mammary tumors. Genetic deletion of ADAM17 in leukocytes resulted in decreased onset of mammary tumor growth, which was associated with reduced expression of the Cox-2 within the tumor. These findings demonstrate that ADAM17 regulates key inflammatory mediators in macrophages and that leukocyte-specific ADAM17 is an important promoter of mammary tumor initiation. Understanding the mechanisms associated with early stage tumorigenesis has implications for the development of preventive and/or treatment strategies for early stage breast cancers.
Collapse
Affiliation(s)
- Laura R Bohrer
- Department of Lab Medicine and Pathology, University of Minnesota, MN, USA
| | - Thomas S Chaffee
- Department of Lab Medicine and Pathology, University of Minnesota, MN, USA
| | - Pavlina Chuntova
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, MN, USA
| | - Nicholas J Brady
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, MN, USA
| | - Patrice M Witschen
- Department of Veterinary and Biomedical Sciences, University of Minnesota, MN, USA
| | - Sarah E Kemp
- Department of Lab Medicine and Pathology, University of Minnesota, MN, USA
| | - Andrew C Nelson
- Department of Lab Medicine and Pathology, University of Minnesota, MN, USA
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, MN, USA
| | - Kathryn L Schwertfeger
- Department of Lab Medicine and Pathology, University of Minnesota, MN, USA.,Masonic Cancer Center, University of Minnesota, MN, USA
| |
Collapse
|
7
|
Abstract
Breast cancer is no longer considered a single disease, but instead is made up of multiple subtypes with genetically and most likely epigenetically heterogeneous tumors composed of numerous clones. Both the hierarchical cancer stem cell and clonal evolution models have been invoked to help explain this intratumoral heterogeneity. Several recent studies have helped define the functional interactions among the different cellular subpopulations necessary for the evolution of this complex ecosystem. These interactions involve paracrine interactions that include locally acting Wnt family members, reminiscent of the signaling pathways important for normal mammary gland development and stem cell self-renewal. In this review, we discuss the interactions among various cell populations in both normal and tumor tissues. A better understanding of these interactions, especially in the metastatic setting, will be important for the development of improved combinatorial therapies designed to prevent relapse and to ultimately decrease mortality.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Developmental Biology, University of Pittsburgh, 204 Craft Ave., Pittsburgh, PA, 15213, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
8
|
Holdman XB, Welte T, Rajapakshe K, Pond A, Coarfa C, Mo Q, Huang S, Hilsenbeck SG, Edwards DP, Zhang X, Rosen JM. Upregulation of EGFR signaling is correlated with tumor stroma remodeling and tumor recurrence in FGFR1-driven breast cancer. Breast Cancer Res 2015; 17:141. [PMID: 26581390 PMCID: PMC4652386 DOI: 10.1186/s13058-015-0649-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/03/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction Despite advances in early detection and adjuvant targeted therapies, breast cancer is still the second most common cause of cancer mortality among women. Tumor recurrence is one of the major contributors to breast cancer mortality. However, the mechanisms underlying this process are not completely understood. In this study, we investigated the mechanisms of tumor dormancy and recurrence in a preclinical mouse model of breast cancer. Methods To elucidate the mechanisms driving tumor recurrence, we employed a transplantable Wnt1/inducible fibroblast growth factor receptor (FGFR) 1 mouse mammary tumor model and utilized an FGFR specific inhibitor, BGJ398, to study the recurrence after treatment. Histological staining was performed to analyze the residual tumor cells and tumor stroma. Reverse phase protein array was performed to compare primary and recurrent tumors to investigate the molecular mechanisms leading to tumor recurrence. Results Treatment with BGJ398 resulted in rapid tumor regression, leaving a nonpalpable mass of dormant tumor cells organized into a luminal and basal epithelial layer similar to the normal mammary gland, but surrounded by dense stroma with markedly reduced levels of myeloid-derived tumor suppressor cells (MDSCs) and decreased tumor vasculature. Following cessation of treatment the tumors recurred over a period of 1 to 4 months. The recurrent tumors displayed dense stroma with increased collagen, tenascin-C expression, and MDSC infiltration. Activation of the epidermal growth factor receptor (EGFR) pathway was observed in recurrent tumors, and inhibition of EGFR with lapatinib in combination with BGJ398 resulted in a significant delay in tumor recurrence accompanied by reduced stroma, yet there was no difference observed in initial tumor regression between the groups treated with BGJ398 alone or in combination with lapatinib. Conclusion These studies have revealed a correlation between tumor recurrence and changes of stromal microenvironment accompanied by altered EGFR signaling. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0649-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xue B Holdman
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Thomas Welte
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Adam Pond
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Qianxing Mo
- Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Dean P Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Xiang Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Farooqui M, Bohrer LR, Brady NJ, Chuntova P, Kemp SE, Wardwell CT, Nelson AC, Schwertfeger KL. Epiregulin contributes to breast tumorigenesis through regulating matrix metalloproteinase 1 and promoting cell survival. Mol Cancer 2015. [PMID: 26215578 PMCID: PMC4517352 DOI: 10.1186/s12943-015-0408-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The epidermal growth factor (EGF) family of ligands has been implicated in promoting breast cancer initiation, growth and progression. The contributions of EGF family ligands and their receptors to breast cancer are complex, and the specific mechanisms through which different ligands regulate breast tumor initiation and growth are not well-defined. These studies focus on the EGF family member epiregulin (EREG) as a mediator of early stage breast tumorigenesis. METHODS EREG expression levels were assessed in both cell lines and human samples of ductal carcinoma in situ (DCIS) using quantitative RT-PCR, ELISA and immunohistochemistry. Gene knock-down approaches using shRNA-based strategies were used to determine the requirement of EREG for growth of MCF10DCIS cells in vivo, and for identifying mechanisms through which EREG promotes tumor cell survival. Experiments were performed using a combination of two-dimensional culture, three-dimensional culture and tumor growth in vivo. RESULTS In comparison with other EGF family members, EREG was induced in MCF10DCIS cells compared with MCF10A and MCF10AT cells and its expression was partially regulated by fibroblast growth factor receptor (FGFR) activity. Reduced EREG expression in MCF10DCIS cells led to decreased tumor growth in vivo, which was associated with reduced cell survival. Furthermore, treatment of MCF10A cells with exogenous EREG enhanced cell survival both in three-dimensional culture and in response to chemotherapeutic agents. Examination of EREG-induced signaling pathways demonstrated that EREG promoted survival of MCF10A cells through regulating expression of matrix metalloproteinase-1 (MMP-1). To determine the relevance of these findings in human tumors, samples of DCIS were analyzed for EREG and MMP-1 expression. EREG was induced in DCIS lesions compared to normal breast epithelium, and EREG and MMP-1 were correlated in a subset of DCIS samples. CONCLUSIONS Together, these studies lead to identification of a novel pathway involving EREG and MMP-1 that contributes to the formation of early stage breast cancer. Understanding these complex pathways could ultimately lead to the development of novel biomarkers of neoplastic progression and/or new therapeutic strategies for patients with early stage cancer.
Collapse
Affiliation(s)
- Mariya Farooqui
- Department of Lab Medicine and Pathology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Laura R Bohrer
- Department of Lab Medicine and Pathology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Nicholas J Brady
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Pavlina Chuntova
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sarah E Kemp
- Department of Lab Medicine and Pathology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - C Taylor Wardwell
- College of Veterinary Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Andrew C Nelson
- Department of Lab Medicine and Pathology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Kathryn L Schwertfeger
- Department of Lab Medicine and Pathology, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
10
|
Yeh ES, Vernon-Grey A, Martin H, Chodosh LA. Tetracycline-regulated mouse models of cancer. Cold Spring Harb Protoc 2014; 2014:pdb.top069823. [PMID: 25275112 DOI: 10.1101/pdb.top069823] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Genetically engineered mouse models (GEMMs) have proven essential to the study of mammalian gene function in both development and disease. However, traditional constitutive transgenic mouse model systems are limited by the temporal and spatial characteristics of the experimental promoter used to drive transgene expression. To address this limitation, considerable effort has been dedicated to developing conditional and inducible mouse model systems. Although a number of approaches to generating inducible GEMMs have been pursued, several have been restricted by toxic or undesired physiological side effects of the compounds used to activate gene expression. The development of tetracycline (tet)-dependent regulatory systems has allowed for circumvention of these issues resulting in the widespread adoption of these systems as an invaluable tool for modeling the complex nature of cancer progression.
Collapse
Affiliation(s)
- Elizabeth S Yeh
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Ann Vernon-Grey
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Heather Martin
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| | - Lewis A Chodosh
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|
11
|
Ćirović S, Vještica J, Mueller CA, Tatić S, Vasiljević J, Milenković S, Mueller GA, Marković-Lipkovski J. NCAM and FGFR1 coexpression and colocalization in renal tumors. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1402-1414. [PMID: 24817936 PMCID: PMC4014220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 02/05/2014] [Indexed: 06/03/2023]
Abstract
Neural cell adhesion molecule (NCAM) and fibroblast growth factor receptor (FGFR) have a role in epithelial-mesenchymal transformation during tumor genesis. Interplay between both molecules activates FGFR signaling and it could be responsible for tumor development. Renal epithelial tumors were analyzed for FGFR1 and NCAM coexpression by immunohistochemistry and for colocalization of these molecules on the particular tumor cells by triple immunofluorescence. Detection of NCAM isoforms in renal tumors was evaluated by RT-PCR. Applying immunohistochemistry we revealed that the majority of analyzed renal neoplasms, including renal cell carcinoma (RCC) and oncocytoma coexpressed NCAM and FGFR1. Triple immunofluorescent technique confirmed that both markers are commonly colocalized on the same tumor cells. Interestingly, it seemed that different position of NCAM and FGFR1 expression on renal tumor cells is related to renal tumor type or grade: exclusively membranous FGFR1/NCAM expression occurred in low grade clear cell RCC (cRCC); cytoplasmatic and membranous expression was present in high grade cRCC and other RCC types; oncocytoma showed only cytoplasmatic staining of both markers. NCAM-140 and NCAM-120 were detected in almost all analyzed renal neoplasms. Expression of both molecules on different cell compartments in various kidney tumors indicated that NCAM/FGFR1 interaction could play distinct roles in renal tumor genesis.
Collapse
MESH Headings
- Adenoma/metabolism
- Adenoma/pathology
- Adenoma, Oxyphilic/metabolism
- Adenoma, Oxyphilic/pathology
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Kidney/metabolism
- Kidney/pathology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- Middle Aged
- Neural Cell Adhesion Molecules/genetics
- Neural Cell Adhesion Molecules/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
Collapse
Affiliation(s)
- Sanja Ćirović
- Institute of Pathology, Faculty of Medicine, University of BelgradeSerbia
| | - Jelena Vještica
- Institute of Pathology, Faculty of Medicine, University of BelgradeSerbia
| | - Claudia A Mueller
- Department of Nephrology and Rheumatology, Georg-August-UniversityGöttingen, Germany
| | - Svetislav Tatić
- Institute of Pathology, Faculty of Medicine, University of BelgradeSerbia
| | - Jovan Vasiljević
- Institute of Pathology, Faculty of Medicine, University of BelgradeSerbia
| | | | - Gerhard A Mueller
- Department of Nephrology and Rheumatology, Georg-August-UniversityGöttingen, Germany
| | | |
Collapse
|
12
|
Rosen JM, Roarty K. Paracrine signaling in mammary gland development: what can we learn about intratumoral heterogeneity? Breast Cancer Res 2014; 16:202. [PMID: 24476463 PMCID: PMC3978850 DOI: 10.1186/bcr3610] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Paracrine signaling mechanisms play a critical role in both normal mammary gland development and breast cancer. Dissection of these mechanisms using genetically engineered mouse models has provided significant insight into our understanding of the mechanisms that guide intratumoral heterogeneity. In the following perspective, we briefly review some of the emerging concepts in this field and emphasize why elucidation of these pathways will be important for future progress in devising new and improved combinatorial therapeutic approaches for breast and other solid cancers.
Collapse
|
13
|
Bohrer LR, Chuntova P, Bade LK, Beadnell TC, Leon RP, Brady NJ, Ryu Y, Goldberg JE, Schmechel SC, Koopmeiners JS, McCarthy JB, Schwertfeger KL. Activation of the FGFR-STAT3 pathway in breast cancer cells induces a hyaluronan-rich microenvironment that licenses tumor formation. Cancer Res 2013; 74:374-86. [PMID: 24197137 DOI: 10.1158/0008-5472.can-13-2469] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Aberrant activation of fibroblast growth factor receptors (FGFR) contributes to breast cancer growth, progression, and therapeutic resistance. Because of the complex nature of the FGF/FGFR axis, and the numerous effects of FGFR activation on tumor cells and the surrounding microenvironment, the specific mechanisms through which aberrant FGFR activity contributes to breast cancer are not completely understood. We show here that FGFR activation induces accumulation of hyaluronan within the extracellular matrix and that blocking hyaluronan synthesis decreases proliferation, migration, and therapeutic resistance. Furthermore, FGFR-mediated hyaluronan accumulation requires activation of the STAT3 pathway, which regulates expression of hyaluronan synthase 2 (HAS2) and subsequent hyaluronan synthesis. Using a novel in vivo model of FGFR-dependent tumor growth, we demonstrate that STAT3 inhibition decreases both FGFR-driven tumor growth and hyaluronan levels within the tumor. Finally, our results suggest that combinatorial therapies inhibiting both FGFR activity and hyaluronan synthesis is more effective than targeting either pathway alone and may be a relevant therapeutic approach for breast cancers associated with high levels of FGFR activity. In conclusion, these studies indicate a novel targetable mechanism through which FGFR activation in breast cancer cells induces a protumorigenic microenvironment.
Collapse
Affiliation(s)
- Laura R Bohrer
- Authors' Affiliations: Department of Lab Medicine and Pathology; Masonic Cancer Center; Biostatistics and Bioinformatics Core, Masonic Cancer Center; Microbiology, Immunology and Cancer Biology Graduate Program; Graduate Program in Molecular, Cellular, Developmental Biology, and Genetics; BioNet, Academic Health Center, University of Minnesota, Minneapolis; Hamline University, Biology Department, Saint Paul, Minnesota; and Department of Pathology, University of Washington, Seattle, Washington
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Brady N, Chuntova P, Bade LK, Schwertfeger KL. The FGF/FGFR axis as a therapeutic target in breast cancer. Expert Rev Endocrinol Metab 2013; 8:391-402. [PMID: 25400686 PMCID: PMC4228698 DOI: 10.1586/17446651.2013.811910] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factor receptor (FGFR) signaling is a vital component of both embryonic and postnatal mammary gland development, which has prompted researchers to investigate both its relevance to breast cancer and its potential as a therapeutic target. Deregulated FGFR signaling during breast cancer occurs through various mechanisms, including amplification of the receptor genes, aberrant ligand expression, receptor mutations and translocations. Recent experimental outcomes involving both animal models and human breast cancer cell lines have led to the initiation of multiple early clinical trials investigating the safety and efficacy of small molecule FGFR inhibitors. In this article we review both the most recent discoveries and the need for further investigation of the mechanisms through which FGF/FGFR signaling has emerged as an oncogenic driver.
Collapse
Affiliation(s)
- Nicholas Brady
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| | - Polly Chuntova
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| | - Lindsey K Bade
- Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| |
Collapse
|
15
|
Pond AC, Bin X, Batts T, Roarty K, Hilsenbeck S, Rosen JM. Fibroblast growth factor receptor signaling is essential for normal mammary gland development and stem cell function. Stem Cells 2013; 31:178-89. [PMID: 23097355 DOI: 10.1002/stem.1266] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 09/25/2012] [Indexed: 12/30/2022]
Abstract
Fibroblast growth factor (FGF) signaling plays an important role in embryonic stem cells and adult tissue homeostasis, but the function of FGFs in mammary gland stem cells is less well defined. Both FGFR1 and FGFR2 are expressed in basal and luminal mammary epithelial cells (MECs), suggesting that together they might play a role in mammary gland development and stem cell dynamics. Previous studies have demonstrated that the deletion of FGFR2 resulted only in transient developmental defects in branching morphogenesis. Using a conditional deletion strategy, we investigated the consequences of FGFR1 deletion alone and then the simultaneous deletion of both FGFR1 and FGFR2 in the mammary epithelium. FGFR1 deletion using a keratin 14 promoter-driven Cre-recombinase resulted in an early, yet transient delay in development. However, no reduction in functional outgrowth potential was observed following limiting dilution transplantation analysis. In contrast, a significant reduction in outgrowth potential was observed upon the deletion of both FGFR1 and FGFR2 in MECs using adenovirus-Cre. Additionally, using a fluorescent reporter mouse model to monitor Cre-mediated recombination, we observed a competitive disadvantage following transplantation of both FGFR1/R2-null MECs, most prominently in the basal epithelial cells. This correlated with the complete loss of the mammary stem cell repopulating population in the FGFR1/R2-attenuated epithelium. FGFR1/R2-null MECs were partially rescued in chimeric outgrowths containing wild-type MECs, suggesting the potential importance of paracrine mechanisms involved in the maintenance of the basal epithelial stem cells. These studies document the requirement for functional FGFR signaling in mammary stem cells during development.
Collapse
Affiliation(s)
- Adam C Pond
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
16
|
Latasa MU, Salis F, Urtasun R, Garcia-Irigoyen O, Elizalde M, Uriarte I, Santamaria M, Feo F, Pascale RM, Prieto J, Berasain C, Avila MA. Regulation of amphiregulin gene expression by β-catenin signaling in human hepatocellular carcinoma cells: a novel crosstalk between FGF19 and the EGFR system. PLoS One 2012; 7:e52711. [PMID: 23285165 PMCID: PMC3527604 DOI: 10.1371/journal.pone.0052711] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/20/2012] [Indexed: 01/20/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent liver tumor and a deadly disease with limited therapeutic options. Dysregulation of cell signaling pathways is a common denominator in tumorigenesis, including hepatocarcinogenesis. The epidermal growth factor receptor (EGFR) signaling system is commonly activated in HCC, and is currently being evaluated as a therapeutic target in combination therapies. We and others have identified a central role for the EGFR ligand amphiregulin (AR) in the proliferation, survival and drug resistance of HCC cells. AR expression is frequently up-regulated in HCC tissues and cells through mechanisms not completely known. Here we identify the β-catenin signaling pathway as a novel mechanism leading to transcriptional activation of the AR gene in human HCC cells. Activation of β-catenin signaling, or expression of the T41A β-catenin active mutant, led to the induction of AR expression involving three specific β-catenin-Tcf responsive elements in its proximal promoter. We demonstrate that HCC cells expressing the T41A β-catenin active mutant show enhanced proliferation that is dependent in part on AR expression and EGFR signaling. We also demonstrate here a novel cross-talk of the EGFR system with fibroblast growth factor 19 (FGF19). FGF19 is a recently identified driver gene in hepatocarcinogenesis and an activator of β-catenin signaling in HCC and colon cancer cells. We show that FGF19 induced AR gene expression through the β-catenin pathway in human HCC cells. Importantly, AR up-regulation and EGFR signaling participated in the induction of cyclin D1 and cell proliferation elicited by FGF19. Finally, we demonstrate a positive correlation between FGF19 and AR expression in human HCC tissues, therefore supporting in clinical samples our experimental observations. These findings identify the AR/EGFR system as a key mediator of FGF19 responses in HCC cells involving β-catenin signaling, and suggest that combined targeting of FGF19 and AR/EGFR may enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Maria U. Latasa
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Fabiana Salis
- Division of Experimental Pathology and Oncology, Department of Clinical and Experimental Medicine & Oncology, University of Sassari, Sassari, Italy
| | - Raquel Urtasun
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Oihane Garcia-Irigoyen
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Maria Elizalde
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Iker Uriarte
- Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas, University Clinic, University of Navarra, Pamplona, Spain
| | - Monica Santamaria
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Francesco Feo
- Division of Experimental Pathology and Oncology, Department of Clinical and Experimental Medicine & Oncology, University of Sassari, Sassari, Italy
| | - Rosa M. Pascale
- Division of Experimental Pathology and Oncology, Department of Clinical and Experimental Medicine & Oncology, University of Sassari, Sassari, Italy
| | - Jesús Prieto
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas, University Clinic, University of Navarra, Pamplona, Spain
| | - Carmen Berasain
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas, University Clinic, University of Navarra, Pamplona, Spain
- * E-mail: (CB); (MAA)
| | - Matías A. Avila
- Division of Hepatology and Gene Therapy, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas, University Clinic, University of Navarra, Pamplona, Spain
- * E-mail: (CB); (MAA)
| |
Collapse
|
17
|
Reed JR, Stone MD, Beadnell TC, Ryu Y, Griffin TJ, Schwertfeger KL. Fibroblast growth factor receptor 1 activation in mammary tumor cells promotes macrophage recruitment in a CX3CL1-dependent manner. PLoS One 2012; 7:e45877. [PMID: 23029290 PMCID: PMC3454319 DOI: 10.1371/journal.pone.0045877] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 08/22/2012] [Indexed: 12/15/2022] Open
Abstract
Tumor formation is an extensive process requiring complex interactions that involve both tumor cell-intrinsic pathways and soluble mediators within the microenvironment. Tumor cells exploit the intrinsic functions of many soluble molecules, including chemokines and their receptors, to regulate pro-tumorigenic phenotypes that are required for growth and progression of the primary tumor. Previous studies have shown that activation of inducible FGFR1 (iFGFR1) in mammary epithelial cells resulted in increased proliferation, migration, and invasion in vitro and tumor formation in vivo. These studies also demonstrated that iFGFR1 activation stimulated recruitment of macrophages to the epithelium where macrophages contributed to iFGFR1-mediated epithelial cell proliferation and angiogenesis. The studies presented here further utilize this model to identify the mechanisms that regulate FGFR1-induced macrophage recruitment. Results from this study elucidate a novel role for the inflammatory chemokine CX3CL1 in FGFR1-induced macrophage migration. Specifically, we illustrate that activation of both the inducible FGFR1 construct in mouse mammary epithelial cells and endogenous FGFR in the triple negative breast cancer cell line, HS578T, leads to expression of the chemokine CX3CL1. Furthermore, we demonstrate that FGFR-induced CX3CL1 is sufficient to recruit CX3CR1-expressing macrophages in vitro. Finally, blocking CX3CR1 in vivo leads to decreased iFGFR1-induced macrophage recruitment, which correlates with decreased angiogenesis. While CX3CL1 is a known target of FGF signaling in the wound healing environment, these studies demonstrate that FGFR activation also leads to induction of CX3CL1 in a tumor setting. Furthermore, these results define a novel role for CX3CL1 in promoting macrophage recruitment during mammary tumor formation, suggesting that the CX3CL1/CX3CR1 axis may represent a potential therapeutic approach for targeting breast cancers associated with high levels of tumor-associated macrophages.
Collapse
MESH Headings
- Animals
- Breast Neoplasms
- CX3C Chemokine Receptor 1
- Cell Line, Tumor
- Cell Movement
- Cell Transformation, Neoplastic
- Chemokine CX3CL1/metabolism
- Chemokine CX3CL1/physiology
- Epithelial Cells/metabolism
- Epithelial Cells/physiology
- Female
- Humans
- Macrophages/metabolism
- Macrophages/physiology
- Mammary Glands, Animal/blood supply
- Mammary Glands, Animal/pathology
- Mice
- Mice, Transgenic
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptors, Chemokine/metabolism
Collapse
Affiliation(s)
- Johanna R. Reed
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Matthew D. Stone
- Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Thomas C. Beadnell
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Yungil Ryu
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Timothy J. Griffin
- Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kathryn L. Schwertfeger
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
18
|
Bohrer LR, Schwertfeger KL. Macrophages promote fibroblast growth factor receptor-driven tumor cell migration and invasion in a CXCR2-dependent manner. Mol Cancer Res 2012; 10:1294-305. [PMID: 22893608 DOI: 10.1158/1541-7786.mcr-12-0275] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Infiltration of immune cells, specifically macrophages, into the tumor microenvironment has been linked to increased mammary tumor formation and progression. Activation of growth factor receptor signaling pathways within mammary epithelial cells, such as the fibroblast growth factor receptor 1 (FGFR1) pathway, induces recruitment of macrophages to the mammary epithelium. These macrophages promote increased epithelial cell proliferation and angiogenesis. However, the specific mechanisms by which these macrophages are regulated by the preneoplastic epithelial cells and the mechanisms of action of the macrophages within the developing FGFR1-driven tumor microenvironment remain unknown. In this study, we show that activation of inducible FGFR1 in mammary glands leads to decreased activity of the TGFβ/Smad3 pathway in macrophages associated with early stage lesions. Further studies show that macrophages have increased expression of inflammatory chemokines that bind Cxcr2 following exposure to conditioned media from mammary epithelial and tumor cells in which the FGF pathway had been activated. The increase in these ligands is inhibited following activation of the TGFβ pathway, suggesting that decreased TGFβ signaling contributes to the upregulation of these chemokines. Using coculture studies, we further show that macrophages are capable of promoting epithelial and tumor cell migration and invasion through activation of Cxcr2. These results indicate that macrophage-derived Cxcr2 ligands may be important for promoting mammary tumor formation regulated by FGFR signaling. Furthermore, these results suggest that targeting Cxcr2 may represent a novel therapeutic strategy for breast cancers that are associated with high levels of infiltrating macrophages.
Collapse
Affiliation(s)
- Laura R Bohrer
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
19
|
Jain VK, Turner NC. Challenges and opportunities in the targeting of fibroblast growth factor receptors in breast cancer. Breast Cancer Res 2012; 14:208. [PMID: 22731805 PMCID: PMC3446326 DOI: 10.1186/bcr3139] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Activation of the fibroblast growth factor receptor pathway is a common event in many cancer types. Here we review the role of fibroblast growth factor receptor signalling in breast cancer, from SNPs in FGFR2 that influence breast cancer risk and SNPs in FGFR4 that associate with breast cancer prognosis, and potential therapeutic targets such as receptor amplification and aberrant autocrine and paracrine ligand expression. We discuss the multiple therapeutic strategies in preclinical and clinical development and the current and future challenges to successfully targeting this pathway in cancer.
Collapse
MESH Headings
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/therapy
- Female
- Gene Amplification
- Humans
- Molecular Targeted Therapy
- Polymorphism, Single Nucleotide
- Prognosis
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Vikram K Jain
- GI Unit, Royal Marsden Hospital, Downs Road, Sutton, Surrey SM2 5PT, UK
| | - Nicholas C Turner
- Breast Unit, Royal Marsden Hospital, Fulham Road, London SW3 6JJ, UK
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| |
Collapse
|
20
|
Kim S, Roopra A, Alexander CM. A phenotypic mouse model of basaloid breast tumors. PLoS One 2012; 7:e30979. [PMID: 22347416 PMCID: PMC3276569 DOI: 10.1371/journal.pone.0030979] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 12/30/2011] [Indexed: 12/23/2022] Open
Abstract
Chemotherapeutic strategies that target basal-like breast tumors are difficult to design without understanding their cellular and molecular basis. Here, we induce tumors in mice by carcinogen administration, creating a phenocopy of tumors with the diagnostic and functional aspects of human triple negative disease (including EGFR expression/lack of erbB, estrogen-independent growth and co-clustering of the transcriptome with other basaloid models). These tumor strains are a complement to established mouse models that are based on mutations in Brca1 and/or p53. Tumors comprise two distinct cell subpopulations, basal and luminal epithelial cells. These cell fractions were purified by flow cytometry, and only basal cell fractions found to have tumor initiating activity (cancer stem cells). The phenotype of serially regenerated tumors was stable, and irrespective of tumor precursor cell. Tumors were passaged entirely in vivo and serial generations tested for their phenotypic stability. The relative chemo-sensitivity of basal and luminal cells were evaluated. Upon treatment with anthracycline, tumors were effectively de-bulked, but recurred; this correlated with maintenance of a high rate of basal cell division throughout the treatment period. Thus, these tumors grow as robust cell mixtures of basal bipotential tumor initiating cells alongside a luminal majority, and the cellular response to drug administration is dominated by the distinct biology of the two cell types. Given the ability to separate basal and luminal cells, and the discovery potential of this approach, we propose that this mouse model could be a convenient one for preclinical studies.
Collapse
Affiliation(s)
- Soyoung Kim
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|