1
|
Wiltshire E, de Moura MC, Piñeyro D, Joshi RS. Cellular and clinical impact of protein phosphatase enzyme epigenetic silencing in multiple cancer tissues. Hum Genomics 2024; 18:24. [PMID: 38475971 PMCID: PMC10935810 DOI: 10.1186/s40246-024-00592-x] [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: 11/03/2023] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Protein Phosphatase Enzymes (PPE) and protein kinases simultaneously control phosphorylation mechanisms that tightly regulate intracellular signalling pathways and stimulate cellular responses. In human malignancies, PPE and protein kinases are frequently mutated resulting in uncontrolled kinase activity and PPE suppression, leading to cell proliferation, migration and resistance to anti-cancer therapies. Cancer associated DNA hypermethylation at PPE promoters gives rise to transcriptional silencing (epimutations) and is a hallmark of cancer. Despite recent advances in sequencing technologies, data availability and computational capabilities, only a fraction of PPE have been reported as transcriptionally inactive as a consequence of epimutations. METHODS In this study, we examined promoter-associated DNA methylation profiles in Protein Phosphatase Enzymes and their Interacting Proteins (PPEIP) in a cohort of 705 cancer patients in five tissues (Large intestine, Oesophagus, Lung, Pancreas and Stomach) in three cell models (primary tumours, cancer cell lines and 3D embedded cancer cell cultures). As a subset of PPEIP are known tumour suppressor genes, we analysed the impact of PPEIP promoter hypermethylation marks on gene expression, cellular networks and in a clinical setting. RESULTS Here, we report epimutations in PPEIP are a frequent occurrence in the cancer genome and manifest independent of transcriptional activity. We observed that different tumours have varying susceptibility to epimutations and identify specific cellular signalling networks that are primarily affected by epimutations. Additionally, RNA-seq analysis showed the negative impact of epimutations on most (not all) Protein Tyrosine Phosphatase transcription. Finally, we detected novel clinical biomarkers that inform on patient mortality and anti-cancer treatment sensitivity. CONCLUSIONS We propose that DNA hypermethylation marks at PPEIP frequently contribute to the pathogenesis of malignancies and within the precision medicine space, hold promise as biomarkers to inform on clinical features such as patient survival and therapeutic response.
Collapse
Affiliation(s)
- Edward Wiltshire
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK
| | | | - David Piñeyro
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Ricky S Joshi
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, UK.
| |
Collapse
|
2
|
Kheraldine H, Gupta I, Cyprian FS, Vranic S, Al-Farsi HF, Merhi M, Dermime S, Al Moustafa AE. Targeting HER2-positive breast cancer cells by a combination of dasatinib and BMS-202: Insight into the molecular pathways. Cancer Cell Int 2024; 24:94. [PMID: 38431613 PMCID: PMC10909263 DOI: 10.1186/s12935-023-03195-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 12/26/2023] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Recent investigations have reported the benefits of using a tyrosine kinase inhibitor, dasatinib (DA), as well as programmed death-ligand 1 (PD-L1) inhibitors in the management of several solid tumors, including breast cancer. Nevertheless, the outcome of the combination of these inhibitors on HER2-positive breast cancer is not explored yet. METHODS Herein, we investigated the impact of DA and PD-L1 inhibitor (BMS-202) combination on HER2-positive breast cancer cell lines, SKBR3 and ZR75. RESULTS Our data reveal that the combination significantly inhibits cell viability of both cancer cell lines as compared to monotreatment. Moreover, the combination inhibits epithelial-mesenchymal transition (EMT) progression and reduces cancer cell invasion by restoring E-cadherin and β-catenin expressions and loss of vimentin, major biomarkers of EMT. Additionally, the combination reduces the colony formation of both cell lines in comparison with their matched control. Also, the combination considerably inhibits the angiogenesis of the chorioallantoic membrane model compared with monotreatment. Molecular pathway analysis of treated cells shows that this combination blocks HER2, AKT, β-catenin, and JNK1/2/3 activities. CONCLUSION Our findings implicate that a combination of DA and BMS-202 could have a significant impact on the management of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Hadeel Kheraldine
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Biomedical Research Centre, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Sidra Medicine, Doha, Qatar
| | - Farhan Sachal Cyprian
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Biomedical Research Centre, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Halema F Al-Farsi
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar.
- Biomedical Research Centre, Qatar University, P. O. Box 2713, Doha, Qatar.
- Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Contribution of n-3 Long-Chain Polyunsaturated Fatty Acids to the Prevention of Breast Cancer Risk Factors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19137936. [PMID: 35805595 PMCID: PMC9265492 DOI: 10.3390/ijerph19137936] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 02/01/2023]
Abstract
Nowadays, diet and breast cancer are studied at different levels, particularly in tumor prevention and progression. Thus, the molecular mechanisms leading to better knowledge are deciphered with a higher precision. Among the molecules implicated in a preventive and anti-progressive way, n-3 long chain polyunsaturated fatty acids (n-3 LC-PUFAs) are good candidates. These molecules, like docosahexaenoic (DHA) and eicosapentaenoic (EPA) acids, are generally found in marine material, such as fat fishes or microalgae. EPA and DHA act as anti-proliferative, anti-invasive, and anti-angiogenic molecules in breast cancer cell lines, as well as in in vivo studies. A better characterization of the cellular and molecular pathways involving the action of these fatty acids is essential to have a realistic image of the therapeutic avenues envisaged behind their use. This need is reinforced by the increase in the number of clinical trials involving more and more n-3 LC-PUFAs, and this, in various pathologies ranging from obesity to a multitude of cancers. The objective of this review is, therefore, to highlight the new elements showing the preventive and beneficial effects of n-3 LC-PUFAs against the development and progression of breast cancer.
Collapse
|
4
|
Cai JW, Huang XM, Li XL, Qin S, Rong YM, Chen X, Weng JR, Zou YF, Lin XT. An 11-gene signature for the prediction of systemic recurrences in colon adenocarcinoma. Gastroenterol Rep (Oxf) 2021; 9:451-460. [PMID: 34733531 PMCID: PMC8560041 DOI: 10.1093/gastro/goab023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 05/16/2021] [Accepted: 05/16/2021] [Indexed: 11/16/2022] Open
Abstract
Background Prognosis varies among patients within the same colon adenocarcinoma (COAD) stage, indicating the need for reliable molecular markers to enable individualized treatment. This study aimed to investigate gene signatures that can be used for better prognostic prediction of COAD. Methods Gene-expression profiles of COAD patients were obtained from the Gene Expression Omnibus database (n = 332) and The Cancer Genome Atlas database (n = 431). The relationship between gene signature and relapse-free survival was analysed in the training set (n = 93) and validated in the internal validation set (n = 94) and external validation sets (n = 145 and 431). Results Overall, 11 genes (N-myc downstream regulated gene 1 [NDRG1], fms-like tyrosine kinase 1 [FLT1], lipopolysaccharide binding protein [LBP], fatty acid binding protein 4 [FABP4], adiponectin gene [ADIPOQ], angiotensinogen gene [AGT], activin A receptor, type II-like kinase 1 [ACVRL1], CC chemokine ligand 11 [CCL11], cell division cycle 42 [CDC42], T-cell receptor alpha variable 9_2 [TRAV9_2], and proopiomelanocortin [POMC]) were identified by univariable and least absolute shrinkage and selection operator (LASSO) Cox regression analyses. Based on the risk-score model, the patients were grouped into the high-risk or low-risk groups using the median risk score as the cut-off. The area under the curve (AUC) values for 1-, 3-, and 5-year recurrence were 0.970, 0.849, and 0.859, respectively. Patients in the high-risk group had significantly poorer relapse-free survival than did those in the low-risk group. The predictive accuracy of the 11-gene signature was proven in the validation sets. Our gene signature showed better predictive performance for 1-, 3-, and 5-year recurrence than did the other four models. Conclusions The 11-gene signature showed good performance in predicting recurrence in COAD. The accuracy of the signature for prognostic classification requires further confirmation.
Collapse
Affiliation(s)
- Jia-Wei Cai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiao-Ming Huang
- Department of Hepatobiliary Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiao-Lan Li
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Reproductive Medicine, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Si Qin
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Medical Ultrasonics, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yu-Ming Rong
- Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Xi Chen
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Jing-Rong Weng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yi-Feng Zou
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xu-Tao Lin
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
5
|
Farnoosh G, Saeedi-Boroujeni A, Jalali A, Keikhaei B, Mahmoudian-Sani MR. Polymorphisms in genes involved in breast cancer among Iranian patients. Per Med 2021; 18:153-169. [PMID: 33565318 DOI: 10.2217/pme-2020-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review gives a summary of the important genetic polymorphisms in breast cancer with a focus on people in Iran. Several single nucleotide polymorphisms were considered as breast cancer susceptibility polymorphisms within genes (STK15, ERRs, ESR1, p53, SEP15, AURKA, SHBG, SRC, FAS, VEGF, XRCC1, GST, NFκB1, XPC, XRCC3, sirtuin-3, NKG2D). Cytosine-adenine repeat (IGF-I), rs3877899, G-2548A, GGC (eRF3a/GSPT1), IVS2nt-124A/G have shown an increased risk of breast cancers and a decreased risk has been observed in 4G/5G (PAI-1), rs6505162, tri-nucleotide (GCG TGFBR1). We observed that the signaling pathways and antioxidant related genes are the main molecular processes associated with breast cancer progression. Further studies on types of polymorphisms in breast cancer could validate the prognostic value of biomarkers.
Collapse
Affiliation(s)
- Gholamreza Farnoosh
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Saeedi-Boroujeni
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Immunology Today, Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Akram Jalali
- Department of Molecular Medicine & Genetics, School of Medicine Hamadan University of Medical Sciences
| | - Bijan Keikhaei
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
6
|
Al-Othman N, Ahram M, Alqaraleh M. Role of androgen and microRNA in triple-negative breast cancer. Breast Dis 2020; 39:15-27. [PMID: 31839601 DOI: 10.3233/bd-190416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancer (BC) is the most frequent type of malignancy affecting females worldwide. Molecular-based studies resulted in an identification of at least four subtypes of breast carcinoma, including luminal A and luminal B, Human growth factor receptor (HER-2)-enriched and triple-negative tumors (basal-like and normal breast-like). A proportion of BC cases are of the triple-negative breast cancer (TNBC) type. TNBC lacks the expression of estrogen receptor (ER), progesterone receptor (PR), and HER-2, and is known to express androgen receptor (AR) at considerable levels. AR has been shown to promote the progression of TNBC. However, the exact mechanisms have yet to be unraveled. One of these mechanisms could be through regulating the expression of microRNA (miRNA) molecules, which play an important regulatory role in BC through post-transcriptional gene silencing. Activation of AR controls the expression of miRNA molecules, which target selective mRNAs, consequently, affecting protein expression. In this review we attempt to elucidate the relations between AR and miRNA in TNBC.
Collapse
Affiliation(s)
- Nihad Al-Othman
- Division of Anatomy, Biochemistry and Genetic, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Moath Alqaraleh
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| |
Collapse
|
7
|
Androgens Induce Invasiveness of Triple Negative Breast Cancer Cells Through AR/Src/PI3-K Complex Assembly. Sci Rep 2019; 9:4490. [PMID: 30872694 PMCID: PMC6418124 DOI: 10.1038/s41598-019-41016-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 02/25/2019] [Indexed: 12/22/2022] Open
Abstract
Breast cancer (BC) is still characterized by high morbidity and mortality. A specific BC subtype named triple negative BC (TNBC) lacks estrogen and progesterone receptors (ER and PR, respectively) and is characterized by the absence of overexpression/amplification of human epidermal growth factor receptor 2 (HER2). The androgen receptor (AR) is expressed in TNBC, although its function in these cancers is still debated. Moreover, few therapeutic options are currently available for the treatment of TNBC. In this study, we have used TNBC-derived MDA-MB231 and MDA-MB453 cells that, albeit at different extent, both express AR. Androgen challenging induces migration and invasiveness of these cells. Use of the anti-androgen bicalutamide or AR knockdown experiments show that these effects depend on AR. Furthermore, the small peptide, S1, which mimics the AR proline-rich motif responsible for the interaction of AR with SH3-Src, reverses the effects in both cell lines, suggesting that the assembly of a complex made up of AR and Src drives the androgen-induced motility and invasiveness. Co-immunoprecipitation experiments in androgen-treated MDA-MB231 and MDA-MB453 cells show that the AR/Src complex recruits p85α, the regulatory subunit of PI3-K. In such a way, the basic machinery leading to migration and invasiveness is turned-on. The S1 peptide inhibits motility and invasiveness of TNBC cells and disrupts the AR/Src/p85α complex assembly in MDA-MB231 cells. This study shows that the rapid androgen activation of Src/PI3-K signaling drives migration and invasiveness of TNBC cells and suggests that the S1 peptide is a promising therapeutic option for these cancers.
Collapse
|
8
|
Lamar JM, Xiao Y, Norton E, Jiang ZG, Gerhard GM, Kooner S, Warren JSA, Hynes RO. SRC tyrosine kinase activates the YAP/TAZ axis and thereby drives tumor growth and metastasis. J Biol Chem 2018; 294:2302-2317. [PMID: 30559289 PMCID: PMC6378979 DOI: 10.1074/jbc.ra118.004364] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/27/2018] [Indexed: 01/02/2023] Open
Abstract
When properly employed, targeted therapies are effective cancer treatments. However, the development of such therapies requires the identification of targetable drivers of cancer development and metastasis. The expression and nuclear localization of the transcriptional coactivators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are increased in many human cancers, and experimental evidence indicates that aberrant YAP or TAZ activation drives tumor formation and metastasis. Although these findings make YAP and TAZ appealing therapeutic targets, both have important functions in adult tissues, so directly targeting them could cause adverse effects. The identification of pathways active in cancer cells and required for YAP/TAZ activity could provide a way to inhibit YAP and TAZ. Here, we show that SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) is an important driver of YAP/TAZ activity in human breast cancer and melanoma cells. SRC activation increased YAP/TAZ activity and the expression of YAP/TAZ-regulated genes. In contrast, SRC inhibition or knockdown repressed both YAP/TAZ activity and the expression of YAP/TAZ-regulated genes. We also show that SRC increases the activity of YAP and TAZ by repressing large tumor suppressor homolog (LATS), and we identify the GTPase-activating protein GIT ArfGAP 1 (GIT1) as an SRC effector that regulates both YAP and TAZ. Importantly, we demonstrate that SRC-mediated YAP/TAZ activity promotes tumor growth and enhances metastasis and that SRC-dependent tumor progression depends, at least in part, on YAP and TAZ. Our findings suggest that therapies targeting SRC could help manage some YAP/TAZ-dependent cancers.
Collapse
Affiliation(s)
- John M Lamar
- From the Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York 12208 and .,the Koch Institute for Integrative Cancer Research
| | - Yuxuan Xiao
- From the Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York 12208 and
| | - Emily Norton
- From the Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York 12208 and
| | - Zhi-Gang Jiang
- the Koch Institute for Integrative Cancer Research.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Genevieve M Gerhard
- From the Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York 12208 and
| | - Simrin Kooner
- From the Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York 12208 and
| | - Janine S A Warren
- From the Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York 12208 and
| | - Richard O Hynes
- the Koch Institute for Integrative Cancer Research, .,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.,Department of Biology, and
| |
Collapse
|
9
|
Warren JSA, Xiao Y, Lamar JM. YAP/TAZ Activation as a Target for Treating Metastatic Cancer. Cancers (Basel) 2018; 10:cancers10040115. [PMID: 29642615 PMCID: PMC5923370 DOI: 10.3390/cancers10040115] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
Yes-Associated Protein (YAP) and Transcriptional Co-activator with PDZ-binding Motif (TAZ) have both emerged as important drivers of cancer progression and metastasis. YAP and TAZ are often upregulated or nuclear localized in aggressive human cancers. There is abundant experimental evidence demonstrating that YAP or TAZ activation promotes cancer formation, tumor progression, and metastasis. In this review we summarize the evidence linking YAP/TAZ activation to metastasis, and discuss the roles of YAP and TAZ during each step of the metastatic cascade. Collectively, this evidence strongly suggests that inappropriate YAP or TAZ activity plays a causal role in cancer, and that targeting aberrant YAP/TAZ activation is a promising strategy for the treatment of metastatic disease. To this end, we also discuss several potential strategies for inhibiting YAP/TAZ activation in cancer and the challenges each strategy poses.
Collapse
Affiliation(s)
- Janine S A Warren
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
10
|
Barcus CE, Keely PJ, Eliceiri KW, Schuler LA. Prolactin signaling through focal adhesion complexes is amplified by stiff extracellular matrices in breast cancer cells. Oncotarget 2018; 7:48093-48106. [PMID: 27344177 PMCID: PMC5217003 DOI: 10.18632/oncotarget.10137] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/06/2016] [Indexed: 11/25/2022] Open
Abstract
Estrogen receptor α positive (ERα+) breast cancer accounts for most breast cancer deaths. Both prolactin (PRL) and extracellular matrix (ECM) stiffness/density have been implicated in metastatic progression of this disease. We previously demonstrated that these factors cooperate to fuel processes involved in cancer progression. Culture of ERα+ breast cancer cells in dense/stiff 3D collagen-I matrices shifts the repertoire of PRL signals, and increases crosstalk between PRL and estrogen to promote proliferation and invasion. However, previous work did not distinguish ECM stiffness and collagen density. In order to dissect the ECM features that control PRL signals, we cultured T47D and MCF-7 cells on polyacrylamide hydrogels of varying elastic moduli (stiffness) with varying collagen-I concentrations (ligand density). Increasing stiffness from physiological to pathological significantly augmented PRL-induced phosphorylation of ERK1/2 and the SFK target, FAK-Y925, with only modest effects on pSTAT5. In contrast, higher collagen-I ligand density lowered PRL-induced pSTAT5 with no effect on pERK1/2 or pFAK-Y925. Disrupting focal adhesion signaling decreased PRL signals and PRL/estrogen-induced proliferation more efficiently in stiff, compared to compliant, extracellular environments. These data indicate that matrix stiffness shifts the balance of PRL signals from physiological (JAK2/STAT5) to pathological (FAK/SFK/ERK1/2) by increasing PRL signals through focal adhesions. Together, our studies suggest that PRL signaling to FAK and SFKs may be useful targets in clinical aggressive ERα+ breast carcinomas.
Collapse
Affiliation(s)
- Craig E Barcus
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA.,Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Patricia J Keely
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA.,Laboratory for Cellular and Molecular Biology and Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA.,University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kevin W Eliceiri
- Laboratory for Cellular and Molecular Biology and Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706, USA.,University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA.,Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI 53706, USA.,University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
11
|
SRC Increases MYC mRNA Expression in Estrogen Receptor-Positive Breast Cancer via mRNA Stabilization and Inhibition of p53 Function. Mol Cell Biol 2018; 38:MCB.00463-17. [PMID: 29263157 DOI: 10.1128/mcb.00463-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/06/2017] [Indexed: 02/06/2023] Open
Abstract
The transcription factor gene MYC is important in breast cancer, and its mRNA is maintained at a high level even in the absence of gene amplification. The mechanism(s) underlying increased MYC mRNA expression is unknown. Here, we demonstrate that MYC mRNA was stabilized upon estrogen stimulation of estrogen receptor-positive breast cancer cells via SRC-dependent effects on a recently described RNA-binding protein, IMP1 with an N-terminal deletion (ΔN-IMP1). We also show that loss of the tumor suppressor p53 increased MYC mRNA levels even in the absence of estrogen stimulation. However, in cells with wild-type p53, SRC acted to overcome p53-mediated inhibition of estrogen-stimulated cell cycle entry and progression. SRC thus promotes cell proliferation in two ways: by stabilizing MYC mRNA and by inhibiting p53 function. Since estrogen receptor-positive breast cancers typically express wild-type p53, these studies establish a rationale for p53 status to be predictive for effective SRC inhibitor treatment in this subtype of breast cancer.
Collapse
|
12
|
Zarif JC, Miranti CK. The importance of non-nuclear AR signaling in prostate cancer progression and therapeutic resistance. Cell Signal 2016; 28:348-356. [PMID: 26829214 PMCID: PMC4788534 DOI: 10.1016/j.cellsig.2016.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
The androgen receptor (AR) remains the major oncogenic driver of prostate cancer, as evidenced by the efficacy of androgen deprivation therapy (ADT) in naïve patients, and the continued effectiveness of second generation ADTs in castration resistant disease. However, current ADTs are limited to interfering with AR ligand binding, either through suppression of androgen production or the use of competitive antagonists. Recent studies demonstrate 1) the expression of constitutively active AR splice variants that no longer depend on androgen, and 2) the ability of AR to signal in the cytoplasm independently of its transcriptional activity (non-genomic); thus highlighting the need to consider other ways to target AR. Herein, we review canonical AR signaling, but focus on AR non-genomic signaling, some of its downstream targets and how these effectors contribute to prostate cancer cell behavior. The goals of this review are to 1) re-highlight the continued importance of AR in prostate cancer as the primary driver, 2) discuss the limitations in continuing to use ligand binding as the sole targeting mechanism, 3) discuss the implications of AR non-genomic signaling in cancer progression and therapeutic resistance, and 4) address the need to consider non-genomic AR signaling mechanisms and pathways as a viable targeting strategy in combination with current therapies.
Collapse
Affiliation(s)
- Jelani C Zarif
- The James Buchanan Brady Urological Institute at The Johns Hopkins University School of Medicine Baltimore, MD 21287, United States
| | - Cindy K Miranti
- Lab of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, United States.
| |
Collapse
|
13
|
Chatterji T, Varkaris AS, Parikh NU, Song JH, Cheng CJ, Schweppe RE, Alexander S, Davis JW, Troncoso P, Friedl P, Kuang J, Lin SH, Gallick GE. Yes-mediated phosphorylation of focal adhesion kinase at tyrosine 861 increases metastatic potential of prostate cancer cells. Oncotarget 2016; 6:10175-94. [PMID: 25868388 PMCID: PMC4496348 DOI: 10.18632/oncotarget.3391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/16/2015] [Indexed: 01/15/2023] Open
Abstract
To study the role of FAK signaling complexes in promoting metastatic properties of prostate cancer (PCa) cells, we selected stable, highly migratory variants, termed PC3 Mig-3 and DU145 Mig-3, from two well-characterized PCa cell lines, PC3 and DU145. These variants were not only increased migration and invasion in vitro, but were also more metastatic to lymph nodes following intraprostatic injection into nude mice. Both PC3 Mig-3 and DU145 Mig-3 were specifically increased in phosphorylation of FAK Y861. We therefore examined potential alterations in Src family kinases responsible for FAK phosphorylation and determined only Yes expression was increased. Overexpression of Yes in PC3 parental cells and src-/-fyn-/-yes-/- fibroblasts selectively increased FAK Y861 phosphorylation, and increased migration. Knockdown of Yes in PC3 Mig-3 cells decreased migration and decreased lymph node metastasis following orthotopic implantation of into nude mice. In human specimens, Yes expression was increased in lymph node metastases relative to paired primary tumors from the same patient, and increased pFAK Y861 expression in lymph node metastases correlated with poor prognosis. These results demonstrate a unique role for Yes in phosphorylation of FAK and in promoting PCa metastasis. Therefore, phosphorylated FAK Y861 and increased Yes expression may be predictive markers for PCa metastasis.
Collapse
Affiliation(s)
- Tanushree Chatterji
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA
| | - Andreas S Varkaris
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nila U Parikh
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chien-Jui Cheng
- Department of Pathology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism, and Diabetes, and Department of Pathology, University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, Aurora, CO, USA
| | - Stephanie Alexander
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Cell Biology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - John W Davis
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Division of Endocrinology, Metabolism, and Diabetes, and Department of Pathology, University of Colorado Anschutz Medical Campus, University of Colorado Cancer Center, Aurora, CO, USA
| | - Jian Kuang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sue-Hwa Lin
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The David Koch Center for Applied Research in Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Programs in Cancer Biology and Cancer Metastasis, The University of Texas Graduate School of Biomedical Sciences at Houston, TX, USA
| |
Collapse
|
14
|
Wu X, Zahari MS, Renuse S, Nirujogi RS, Kim MS, Manda SS, Stearns V, Gabrielson E, Sukumar S, Pandey A. Phosphoproteomic Analysis Identifies Focal Adhesion Kinase 2 (FAK2) as a Potential Therapeutic Target for Tamoxifen Resistance in Breast Cancer. Mol Cell Proteomics 2015; 14:2887-900. [PMID: 26330541 DOI: 10.1074/mcp.m115.050484] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Indexed: 01/13/2023] Open
Abstract
Tamoxifen, an estrogen receptor-α (ER) antagonist, is an important agent for the treatment of breast cancer. However, this therapy is complicated by the fact that a substantial number of patients exhibit either de novo or acquired resistance. To characterize the signaling mechanisms underlying this resistance, we treated the MCF7 breast cancer cell line with tamoxifen for over six months and showed that this cell line acquired resistance to tamoxifen in vitro and in vivo. We performed SILAC-based quantitative phosphoproteomic profiling on the tamoxifen resistant and vehicle-treated sensitive cell lines to quantify the phosphorylation alterations associated with tamoxifen resistance. From >5600 unique phosphopeptides identified, 1529 peptides exhibited hyperphosphorylation and 409 peptides showed hypophosphorylation in the tamoxifen resistant cells. Gene set enrichment analysis revealed that focal adhesion pathway was one of the most enriched signaling pathways activated in tamoxifen resistant cells. Significantly, we showed that the focal adhesion kinase FAK2 was not only hyperphosphorylated but also transcriptionally up-regulated in tamoxifen resistant cells. FAK2 suppression by specific siRNA knockdown or a small molecule inhibitor repressed cellular proliferation in vitro and tumor formation in vivo. More importantly, our survival analysis revealed that high expression of FAK2 is significantly associated with shorter metastasis-free survival in estrogen receptor-positive breast cancer patients treated with tamoxifen. Our studies suggest that FAK2 is a potential therapeutic target for the management of hormone-refractory breast cancers.
Collapse
Affiliation(s)
- Xinyan Wu
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Muhammad Saddiq Zahari
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Santosh Renuse
- §Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | - Raja Sekhar Nirujogi
- §Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | - Min-Sik Kim
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Srikanth S Manda
- §Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | | | - Edward Gabrielson
- ‖Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | | | - Akhilesh Pandey
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; ¶Department of Oncology; ‖Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| |
Collapse
|
15
|
Acerbi I, Cassereau L, Dean I, Shi Q, Au A, Park C, Chen YY, Liphardt J, Hwang ES, Weaver VM. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol (Camb) 2015; 7:1120-34. [PMID: 25959051 DOI: 10.1039/c5ib00040h] [Citation(s) in RCA: 733] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumors are stiff and data suggest that the extracellular matrix stiffening that correlates with experimental mammary malignancy drives tumor invasion and metastasis. Nevertheless, the relationship between tissue and extracellular matrix stiffness and human breast cancer progression and aggression remains unclear. We undertook a biophysical and biochemical assessment of stromal-epithelial interactions in noninvasive, invasive and normal adjacent human breast tissue and in breast cancers of increasingly aggressive subtype. Our analysis revealed that human breast cancer transformation is accompanied by an incremental increase in collagen deposition and a progressive linearization and thickening of interstitial collagen. The linearization of collagen was visualized as an overall increase in tissue birefringence and was most striking at the invasive front of the tumor where the stiffness of the stroma and cellular mechanosignaling were the highest. Amongst breast cancer subtypes we found that the stroma at the invasive region of the more aggressive Basal-like and Her2 tumor subtypes was the most heterogeneous and the stiffest when compared to the less aggressive luminal A and B subtypes. Intriguingly, we quantified the greatest number of infiltrating macrophages and the highest level of TGF beta signaling within the cells at the invasive front. We also established that stroma stiffness and the level of cellular TGF beta signaling positively correlated with each other and with the number of infiltrating tumor-activated macrophages, which was highest in the more aggressive tumor subtypes. These findings indicate that human breast cancer progression and aggression, collagen linearization and stromal stiffening are linked and implicate tissue inflammation and TGF beta.
Collapse
Affiliation(s)
- I Acerbi
- Center for Bioengineering, Tissue Regeneration, Department of Surgery, UCSF, San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Blanckaert V, Kerviel V, Lépinay A, Joubert-Durigneux V, Hondermarck H, Chénais B. Docosahexaenoic acid inhibits the invasion of MDA-MB-231 breast cancer cells through upregulation of cytokeratin-1. Int J Oncol 2015; 46:2649-55. [PMID: 25825023 DOI: 10.3892/ijo.2015.2936] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/05/2015] [Indexed: 11/06/2022] Open
Abstract
Docosahexaenoic acid (DHA), the main member of the omega-3 essential fatty acid family, has been shown to reduce the invasion of the triple-negative breast cancer cell line MDA-MB-231, but the mechanism involved remains unclear. In the present study, a proteomic approach was used to define changes in protein expression induced by DHA. Proteins from crude membrane preparations of MDA-MB-231 cells treated with 100 µM DHA were separated by two-dimensional electrophoresis (2-DE) and differentially expressed proteins were identified using MALDI-TOF mass spectrometry. The main changes observed were the upregulation of Keratin, type Ⅱ cytoskeletal 1 (KRT1), catalase and lamin-A/C. Immunocytochemistry analyses confirmed the increase in KRT1 induced by DHA. Furthermore, in vitro invasion assays showed that siRNA against KRT1 was able to reverse the DHA-induced inhibition of breast cancer cell invasion. In conclusion, KRT1 is involved in the anti-invasive activity of DHA in breast cancer cells.
Collapse
Affiliation(s)
- Vincent Blanckaert
- Mer, Molécules, Santé (EA2160), IUML‑FR3473 CNRS, Université du Maine, F‑72085 Le Mans, France
| | - Vincent Kerviel
- Mer, Molécules, Santé (EA2160), IUML‑FR3473 CNRS, Université du Maine, F‑72085 Le Mans, France
| | - Alexandra Lépinay
- Mer, Molécules, Santé (EA2160), IUML‑FR3473 CNRS, Université du Maine, F‑72085 Le Mans, France
| | | | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Benoît Chénais
- Mer, Molécules, Santé (EA2160), IUML‑FR3473 CNRS, Université du Maine, F‑72085 Le Mans, France
| |
Collapse
|
17
|
Barcus CE, Holt EC, Keely PJ, Eliceiri KW, Schuler LA. Dense collagen-I matrices enhance pro-tumorigenic estrogen-prolactin crosstalk in MCF-7 and T47D breast cancer cells. PLoS One 2015; 10:e0116891. [PMID: 25607819 PMCID: PMC4301649 DOI: 10.1371/journal.pone.0116891] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/16/2014] [Indexed: 02/07/2023] Open
Abstract
Breast cancers that express estrogen receptor alpha (ERα+) constitute the majority of breast tumors. Estrogen is a major driver of their growth, and targeting ER-mediated signals is a largely successful primary therapeutic strategy. Nonetheless, ERα+ tumors also result in the most breast cancer mortalities. Other factors, including altered characteristics of the extracellular matrix such as density and orientation and consequences for estrogen crosstalk with other hormones such as prolactin (PRL), may contribute to these poor outcomes. Here we employed defined three dimensional low density/compliant and high density/stiff collagen-I matrices to investigate the effects on 17β-estradiol (E2) activity and PRL/E2 interactions in two well-characterized ERα+/PRLR+ luminal breast cancer cell lines in vitro. We demonstrate that matrix density modulated E2-induced transcripts, but did not alter the growth response. However, matrix density was a potent determinant of the behavioral outcomes of PRL/E2 crosstalk. High density/stiff matrices enhanced PRL/E2-induced growth mediated by increased activation of Src family kinases and insensitivity to the estrogen antagonist, 4-hydroxytamoxifen. It also permitted these hormones in combination to drive invasion and modify the alignment of collagen fibers. In contrast, low density/compliant matrices allowed modest if any cooperation between E2 and PRL to growth and did not permit hormone-induced invasion or collagen reorientation. Our studies demonstrate the power of matrix density to determine the outcomes of hormone actions and suggest that stiff matrices are potent collaborators of estrogen and PRL in progression of ERα+ breast cancer. Our evidence for bidirectional interactions between these hormones and the extracellular matrix provides novel insights into the regulation of the microenvironment of ERα+ breast cancer and suggests new therapeutic approaches.
Collapse
Affiliation(s)
- Craig E Barcus
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elizabeth C Holt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Patricia J Keely
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Laboratory for Cellular and Molecular Biology and Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kevin W Eliceiri
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Laboratory for Cellular and Molecular Biology and Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America; University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
18
|
Chang YL, Hsu YK, Wu TF, Huang CM, Liou LY, Chiu YW, Hsiao YH, Luo FJ, Yuan TC. Regulation of estrogen receptor α function in oral squamous cell carcinoma cells by FAK signaling. Endocr Relat Cancer 2014; 21:555-65. [PMID: 24825747 DOI: 10.1530/erc-14-0102] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Estrogen receptor α (ERA) is a DNA-binding transcription factor that plays an important role in the regulation of cell growth. Previous studies indicated that the expression of ERα in cell lines and tumors derived from oral squamous cell carcinoma (OSCC). The aim of this study was to examine the activity and function of ERα in OSCC cells and the mechanism underlying ERα activation. Immunochemical analyses in benign (n=11) and malignant (n=21) lesions of the oral cavity showed that ERα immunoreactivity was observed in 43% (9/21) of malignant lesions, whereas none of benign lesions showed ERα immunoreactivity. The ERα expression was also found in three OSCC cell lines and its transcriptional activity was correlated with cell growth. Addition of estradiol stimulated cell growth, whereas treatment of tamoxifen or knockdown of ERα expression caused reduced cell growth. Interestingly, the expression and activity of focal adhesion kinase (FAK) were associated with the phosphorylation of ERα at serine 118 in OSCC cells. Elevated expression of FAK in the slow-growing SCC25 cells caused increases in ERα phosphorylation, transcriptional activity, and cell growth rate, whereas knockdown of FAK expression in the rapid-growing OECM-1 cells led to reduced ERα phosphorylation and activity and retarded cell growth. Inhibition of the activity of protein kinase B (AKT), but not ERK, abolished FAK-promoted ERα phosphorylation. These results suggest that OSCC cells expressed functional ERα, whose activity can be enhanced by FAK/AKT signaling, and this was critical for promoting cell growth. Thus, FAK and ERα can serve as the therapeutic targets for the treatment of OSCC.
Collapse
Affiliation(s)
- Yi-Lin Chang
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Yu-Kan Hsu
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Tsung-Fan Wu
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Chieh-Ming Huang
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of ChinaDepartment of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Li-Yin Liou
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Ya-Wen Chiu
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Yu-Hsuan Hsiao
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Fuh-Jinn Luo
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| | - Ta-Chun Yuan
- Department of Life ScienceInstitute of Biotechnology, National Dong Hwa University, Hualien 97401, TaiwanDepartment of PathologyDepartment of DentistryMennonite Hospital, Hualien 970, Taiwan, Republic of China
| |
Collapse
|
19
|
Wang X, Qian H, Zhang S. Discovery of significant pathways in breast cancer metastasis via module extraction and comparison. IET Syst Biol 2014; 8:47-55. [PMID: 25014225 PMCID: PMC8687293 DOI: 10.1049/iet-syb.2013.0041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/03/2013] [Accepted: 12/30/2013] [Indexed: 09/29/2023] Open
Abstract
Discovering significant pathways rather than single genes or small gene sets involved in metastasis is becoming more and more important in the study of breast cancer. Many researches have shed light on this problem. However, most of the existing works are relying on some priori biological information, which may bring bias to the models. The authors propose a new method that detects metastasis-related pathways by identifying and comparing modules in metastasis and non-metastasis gene co-expression networks. The gene co-expression networks are built by Pearson correlation coefficients, and then the modules inferred in these two networks are compared. In metastasis and non-metastasis networks, 36 and 41 significant modules are identified. Also, 27.8% (metastasis) and 29.3% (non-metastasis) of the modules are enriched significantly for one or several pathways with p-value <0.05. Many breast cancer genes including RB1, CCND1 and TP53 are included in these identified pathways. Five significant pathways are discovered only in metastasis network: glycolysis pathway, cell adhesion molecules, focal adhesion, stathmin and breast cancer resistance to antimicrotubule agents, and cytosolic DNA-sensing pathway. The first three pathways have been proved to be closely associated with metastasis. The rest two can be taken as a guide for future research in breast cancer metastasis.
Collapse
Affiliation(s)
- Xiaochen Wang
- School of Mathematical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Huajie Qian
- School of Mathematical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Shuqin Zhang
- Center for Computational Systems Biology, School of Mathematical Sciences, Fudan University Shanghai, Shanghai 200433, People's Republic of China.
| |
Collapse
|
20
|
Elsberger B. Translational evidence on the role of Src kinase and activated Src kinase in invasive breast cancer. Crit Rev Oncol Hematol 2014; 89:343-51. [DOI: 10.1016/j.critrevonc.2013.12.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 12/04/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022] Open
|
21
|
Stebbing J, Lit LC, Zhang H, Darrington RS, Melaiu O, Rudraraju B, Giamas G. The regulatory roles of phosphatases in cancer. Oncogene 2014; 33:939-53. [PMID: 23503460 DOI: 10.1038/onc.2013.80] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/01/2013] [Indexed: 02/06/2023]
Abstract
The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3-kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies.
Collapse
Affiliation(s)
- J Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - L C Lit
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - H Zhang
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - R S Darrington
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - O Melaiu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - B Rudraraju
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - G Giamas
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
22
|
Liu X, Du L, Feng R. c-Src regulates cell cycle proteins expression through protein kinase B/glycogen synthase kinase 3 beta and extracellular signal-regulated kinases 1/2 pathways in MCF-7 cells. Acta Biochim Biophys Sin (Shanghai) 2013; 45:586-92. [PMID: 23615537 DOI: 10.1093/abbs/gmt042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
We have demonstrated that c-Src suppression inhibited the epithelial to mesenchymal transition in human breast cancer cells. Here, we investigated the role of c-Src on the cell cycle progression using siRNAs and small molecule inhibitor 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). Western blot analysis demonstrated the down-regulation of cyclin D1 and cyclin E and up-regulation of p27 Kip1 after c-Src suppression by PP2. Incubation of cells in the presence of PP2 significantly blocked the phosphorylation of extracellular signal-regulated kinases 1/2 (ERK1/2), protein kinase B (AKT), and glycogen synthase kinase 3 beta (GSK3β). Specific pharmacological inhibitors of MEK1/2/ERK1/2 and phosphatidylinositide 3-kinase/AKT pathways were used to demonstrate the relationship between the signal cascade and cell cycle proteins expression. The expression of cyclin D1 and cyclin E were decreased after inhibition of ERK1/2 or AKT activity, whereas the p27 Kip1 expression was increased. In addition, knockdown of c-Src by siRNAs reduced cell proliferation and phosphorylation of ERK1/2, AKT, and GSK3β. After c-Src depletion by siRNAs, we observed significant down-regulation of cyclin D1 and cyclin E, and up-regulation of p27 Kip1. These results suggest that c-Src suppression by PP2 or siRNAs may regulate the progression of cell cycle through AKT/GSK3β and ERK1/2 pathways.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | | | | |
Collapse
|
23
|
Somlo G, Atzori F, Strauss LC, Geese WJ, Specht JM, Gradishar WJ, Rybicki A, Sy O, Vahdat LT, Cortes J. Dasatinib plus capecitabine for advanced breast cancer: safety and efficacy in phase I study CA180004. Clin Cancer Res 2013; 19:1884-93. [PMID: 23403636 DOI: 10.1158/1078-0432.ccr-12-0652] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Dasatinib is an Src family kinase inhibitor with modest activity in advanced breast cancer. We aimed to assess toxicity and maximum tolerated dose (MTD) for dasatinib plus capecitabine, estimate efficacy, and explore effects on angiogenesis. EXPERIMENTAL DESIGN Dose levels (DL) were dasatinib 50 mg twice daily (DL1), 70 mg twice daily (DL2 and DL3), or 100 mg daily (DL3a); plus capecitabine on days 1 to 14 of a 21-day cycle, at 825 mg/m(2) twice daily (DL1 and DL2) or 1,000 mg/m(2) twice daily [DL3 and DL3a (MTD)]. DL3a was expanded to evaluate safety/efficacy. Plasma samples were collected for biomarker analysis. RESULTS Thirty-one and 21 patients were treated in the escalation and expansion phases. Sixty percent of tumors were hormone receptor-positive. Most common adverse events (AE) were any grade nausea (58%), hand-foot syndrome (44%), diarrhea (33%), fatigue (33%), vomiting (31%), and asthenia (31%). Most common grade 3/4 AEs were hand-foot syndrome (12%), diarrhea (8%), fatigue (8%), pleural effusion (8%), and vomiting (6%). The MTD was defined at DL3a (capecitabine 1,000 mg/m(2) twice daily and dasatinib 100 mg daily). Of 25 response-evaluable patients treated at DL3a, confirmed partial response was noted in 24% and stable disease in an additional 32%; median progression-free survival was 14.4 weeks. Significant decreases in plasma VEGF-A and increases in VEGFR-2 and collagen-IV were observed. CONCLUSIONS Dasatinib 100 mg once daily plus capecitabine 1,000 mg/m(2) twice daily were tolerable and were associated with clinical benefit in 56% of response-evaluable patients. Biomarker changes were consistent with an antiangiogenic effect.
Collapse
Affiliation(s)
- George Somlo
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Roop RP, Ma CX. Endocrine resistance in breast cancer: molecular pathways and rational development of targeted therapies. Future Oncol 2012; 8:273-92. [PMID: 22409464 DOI: 10.2217/fon.12.8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endocrine resistance presents a major challenge in the management of estrogen receptor (ER)-positive breast cancer and is an area under intense investigation. Although the underlying mechanism is still poorly understood, many studies point towards the 'cross-talk' between ER and growth factor receptor signaling pathways as the key in the development of estrogen-independent growth in breast cancer. This review aims to provide the reader our current understanding of various molecular pathways that mediate endocrine resistance and that are being evaluated as therapeutic targets for ER-positive breast cancer. While most of the agents that target these pathways have only been tested in Phase I or small Phase II trials, some have shown encouraging results. A critical issue that remains is the development of research strategies and clinical trials that take into account the molecular heterogeneity of ER-positive breast cancer.
Collapse
Affiliation(s)
- Ryan P Roop
- Washington University School of Medicine in Saint Louis, Department of Medicine, Divisions of Hematology & Oncology, St Louis, MO, USA
| | | |
Collapse
|
26
|
Anbalagan M, Moroz K, Ali A, Carrier L, Glodowski S, Rowan BG. Subcellular localization of total and activated Src kinase in African American and Caucasian breast cancer. PLoS One 2012; 7:e33017. [PMID: 22457730 PMCID: PMC3310861 DOI: 10.1371/journal.pone.0033017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 02/03/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Src, a non-receptor tyrosine kinase is elevated in cancer with expression and activity correlated with cell proliferation, adhesion, survival, motility, metastasis and angiogenesis. There is limited data on Src expression and subcellular localization in breast cancer and no information about expression in racial/ethnic groups. METHODOLOGY/PRINCIPAL FINDINGS The present study evaluated Src expression, activity, and subcellular localization in triple negative breast cancer (TNBC) and ERα positive breast cancer (ER+BC), cancer tissue and adjacent normal epithelial ducts, and Caucasian and African American cases. 79 paraffin embedded breast carcinoma cases were obtained from Tulane University Hospital between 2007-2009. 39 cases represented TNBC (33-African Americans, 4-Caucasians, 2-unknowns) and 40 cases represented ER+BC (21-African Americans, 16-Caucasians, 3-unknowns). Immunohistochemistry was used to measure staining distribution and intensity of total Src and activated phospho-SrcY416 (p-Y416Src) in carcinoma tissue and adjacent normal mammary ducts. In TNBC and ER+BC, total Src was significantly higher in cancer compared to adjacent normal ducts (P<0.0001) in both cell membrane and cytoplasm. In membranes, p-Y416Src was elevated in cancer compared to normal ducts. Total Src in the tumor cytoplasm was significantly higher in TNBC compared to ER+BC (P = 0.0028); conversely, p-Y416Src in the tumor cell membranes was higher in TNBC compared to ER+BC (P = 0.0106). Comparison between African American (n = 21) and Caucasian ER+BC (n = 16) revealed no significant difference in expression and localization of total Src and p-Y416Src. TNBC cases positive for lymph node metastasis showed elevated membrane p-Y416Src compared to lymph node negative TNBC (P = 0.027). CONCLUSION/SIGNIFICANCE Total Src and p-Y416Src were expressed higher in cancer compared to adjacent normal ducts. Cytoplasmic total Src and membrane p-Y416Src were significantly higher in TNBC compared to ER+BC. TNBC cases with lymph node metastasis showed elevated membrane p-Y416Src. Taken together, Src was elevated in the membrane and cytoplasm of more aggressive TNBC.
Collapse
Affiliation(s)
- Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Krzysztof Moroz
- Section of Surgical Pathology and Cytopathology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Alaa Ali
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Latonya Carrier
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Seth Glodowski
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
27
|
Luminal breast cancer metastasis is dependent on estrogen signaling. Clin Exp Metastasis 2012; 29:493-509. [PMID: 22427027 DOI: 10.1007/s10585-012-9466-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 01/10/2012] [Indexed: 12/29/2022]
Abstract
Luminal breast cancer is the most frequently encountered type of human breast cancer and accounts for half of all breast cancer deaths due to metastatic disease. We have developed new in vivo models of disseminated human luminal breast cancer that closely mimic the human disease. From initial lesions in the tibia, locoregional metastases develop predictably along the iliac and retroperitoneal lymph node chains. Tumors cells retain their epithelioid phenotype throughout the process of dissemination. In addition, systemically injected metastatic MCF-7 cells consistently give rise to metastases in the skeleton, floor of mouth, adrenal glands, as well as in the lungs, liver, brain and mammary fat pad. We show that growth of luminal breast cancer metastases is highly dependent on estrogen in a dose-dependent manner and that estrogen withdrawal induces rapid growth arrest of metastatic disease. On the other hand, even though micrometastases at secondary sites remain viable in the absence of estrogen, they are dormant and do not progress to macrometastases. Thus, homing to and seeding of secondary sites do not require estrogen. Moreover, in sharp contrast to basal-like breast cancer metastasis in which transforming growth factor-β signaling plays a key role, luminal breast cancer metastasis is independent of this cytokine. These findings have important implications for the development of targeted anti-metastatic therapy for luminal breast cancer.
Collapse
|
28
|
Elsberger B, Paravasthu DM, Tovey SM, Edwards J. Shorter disease-specific survival of ER-positive breast cancer patients with high cytoplasmic Src kinase expression after tamoxifen treatment. J Cancer Res Clin Oncol 2012; 138:327-32. [PMID: 22134837 DOI: 10.1007/s00432-011-1096-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/07/2011] [Indexed: 11/24/2022]
Abstract
BACKGROUND Src kinase, a non-receptor tyrosine kinase, is overexpressed and highly activated in a number of human cancers and appears to show a significant relationship with breast cancer progression. Recent in vitro studies have suggested that Src kinase may be involved in tamoxifen resistance. METHODS Immunohistochemistry was performed on 392 resected breast cancers using an antibody to c-Src. Expression was assessed using the weighted histoscore method. RESULTS Forty-five percentage of breast tumours exhibited nuclear, 46% cytoplasmic and 7% membrane expression. Lymph node positivity correlated with cytoplasmic c-Src tumour expression levels (P < 0.001). Nuclear c-Src correlated negatively with cytoplasmic and membrane c-Src expression (P < 0.001, P = 0.005). High expression levels of cytoplasmic c-Src was associated with worse disease-specific survival (P = 0.026) after completing 5 years of tamoxifen therapy. However, high expression of c-Src at any cellular location did not show any association with de novo relapse on tamoxifen (c-Src nuc P = 0.906, c-Src cyto P = 0.735 and c-Src memb P = 0.791). CONCLUSIONS No translational evidence was found in this study to support a role for Src kinase in developing de novo tamoxifen resistance. However, based on our findings on late clinical outcome, patients with high cytoplasmic c-Src may be selected for continuing endocrine therapy to prevent worsening prognosis.
Collapse
Affiliation(s)
- B Elsberger
- Institute of Cancer, College of Medical, Veterinary and Life Sciences, University of Glasgow, Western Infirmary, McGregor Building, Dumbarton Road, Glasgow, G11 6NT, UK
| | | | | | | |
Collapse
|
29
|
Mayer EL, Baurain JF, Sparano J, Strauss L, Campone M, Fumoleau P, Rugo H, Awada A, Sy O, Llombart-Cussac A. A phase 2 trial of dasatinib in patients with advanced HER2-positive and/or hormone receptor-positive breast cancer. Clin Cancer Res 2011; 17:6897-904. [PMID: 21903773 DOI: 10.1158/1078-0432.ccr-11-0070] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE SRC-family kinases (SFK) are involved in numerous oncogenic signaling pathways. A phase 2 trial of dasatinib, a potent oral tyrosine kinase inhibitor of SFKs, was carried out in patients with human epidermal growth factor receptor 2-positive (HER2+) and/or hormone receptor-positive (HR+) advanced breast cancer. EXPERIMENTAL DESIGN Patients with measurable tumors and progression after chemotherapy and HER2 and/or HR-targeted agents in adjuvant or metastatic settings (maximum of two prior metastatic setting regimens) received twice daily dasatinib. Primary endpoint was Response Evaluation Criteria in Solid Tumors-defined response rate. Secondary endpoints included toxicity and limited pharmacokinetics. RESULTS Seventy patients (55 years median age) were treated, 83% of HER2+ patients had received prior HER2-directed therapy, and 61% of HR+ patients had received prior endocrine therapy in the advanced setting. Dasatinib starting dose was reduced from 100 to 70 mg twice daily to limit toxicity. Median therapy duration was 1.8 months in both dose groups and most discontinuations were due to progression. Of 69 evaluable patients, three had confirmed partial responses and six had stable disease for 16 weeks or more (disease control rate = 13.0%); all nine of these tumors were HR+ (two were also HER2+). The most common drug-related toxicities were gastrointestinal complaints, headache, asthenia, and pleural effusion. Grade 3-4 toxicity occurred in 37% of patients and was comparable between doses; drug-related serious adverse events were less frequent with 70 mg twice daily than 100 mg twice daily. CONCLUSION Limited single-agent activity was observed with dasatinib in patients with advanced HR+ breast cancer.
Collapse
Affiliation(s)
- Erica L Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Role of Src in breast cancer cell migration and invasion in a breast cell/bone-derived cell microenvironment. Breast Cancer Res Treat 2011; 133:201-14. [PMID: 21894461 DOI: 10.1007/s10549-011-1753-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 08/18/2011] [Indexed: 10/17/2022]
Abstract
The preferential metastasis of breast cancer cells to bone comprises a complex set of events including homing and preferential growth, which may require unique factors produced by bone or other cells in the immediate microenvironment. In this study, an in vitro co-culture system composed of bone mesenchymal stem cells and breast cancer cell lines is used to examine the role of Src kinase on breast cancer cell migration and invasion in the presence of bone-derived cells. This research shows that Src kinase activity in breast cancer cell lines with either high or low levels of endogenous Src activity is increased by bone-derived cell-conditioned medium but not HS68 fibroblast-conditioned medium. Breast cancer cells exhibit enhanced migration in co-culture with bone-derived cells but not HS68 fibroblasts or no co-cultured cells. Inhibition of Src kinase activity using the inhibitors PP2 or saracatinib or using siRNA abrogates the preferential migration of the breast cancer cell lines in response to bone-derived cells. Inhibition of Src activity with saracatinib does not have any significant effect on breast cancer cell invasion in the presence of bone-derived cells. Factors are identified that are produced preferentially by bone-derived cells over HS68 cells that may impact breast cancer cell behavior. This research implicates Src kinase as an important effector of bone-derived cell signals on breast cancer cell migration.
Collapse
|
31
|
Huang C, Park CC, Hilsenbeck SG, Ward R, Rimawi MF, Wang YC, Shou J, Bissell MJ, Osborne CK, Schiff R. β1 integrin mediates an alternative survival pathway in breast cancer cells resistant to lapatinib. Breast Cancer Res 2011; 13:R84. [PMID: 21884573 PMCID: PMC3236347 DOI: 10.1186/bcr2936] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 05/24/2011] [Accepted: 08/31/2011] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION The overexpression of human epidermal growth factor receptor (HER)-2 in 20% of human breast cancers and its association with aggressive growth has led to widespread use of HER2-targeted therapies, such as trastuzumab (T) and lapatinib (L). Despite the success of these drugs, their efficacy is limited in patients whose tumors demonstrate de novo or acquired resistance to treatment. The β1 integrin resides on the membrane of the breast cancer cell, activating several elements of breast tumor progression including proliferation and survival. METHODS We developed a panel of HER2-overexpressing cell lines resistant to L, T, and the potent LT combination through long-term exposure and validated these models in 3D culture. Parental and L/T/LT-resistant cells were subject to HER2 and β1 integrin inhibitors in 3D and monitored for 12 days, followed by quantification of colony number. Parallel experiments were conducted where cells were either stained for Ki-67 and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) or harvested for protein and analyzed by immunoblot. Results were subjected to statistical testing using analysis of variance and linear contrasts, followed by adjustment with the Sidak method. RESULTS Using multiple cell lines including BT474 and HCC1954, we reveal that in L and LT resistance, where phosphorylation of EGFR/HER1, HER2, and HER3 are strongly inhibited, kinases downstream of β1 integrin--including focal adhesion kinase (FAK) and Src--are up-regulated. Blockade of β1 by the antibody AIIB2 abrogates this up-regulation and functionally achieves significant growth inhibition of L and LT resistant cells in 3D, without dramatically affecting the parental cells. SiRNA against β1 as well as pharmacologic inhibition of FAK achieve the same growth inhibitory effect. In contrast, trastuzumab-resistant cells, which retain high levels of phosphorylated EGFR/HER1, HER2, and HER3, are only modestly growth-inhibited by AIIB2. CONCLUSIONS Our data suggest that HER2 activity, which is suppressed in resistance involving L but not T alone, dictates whether β1 mediates an alternative pathway driving resistance. Our findings justify clinical studies investigating the inhibition of β1 or its downstream signaling moieties as strategies to overcome acquired L and LT resistance.
Collapse
Affiliation(s)
- Catherine Huang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Catherine C Park
- Department of Radiation Oncology, University of California San Francisco, 1600 Divisadero Street, MZ Bldg R, San Francisco, CA 94143, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Robin Ward
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Mothaffar F Rimawi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Yen-chao Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Jiang Shou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Mina J Bissell
- Department of Cancer & DNA Damage Responses, Lawrence Berkeley National Laboratories, One Cyclotron Road, Berkeley, CA 94720, USA
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77054, USA
| |
Collapse
|
32
|
Boonyaratanakornkit V. Scaffolding proteins mediating membrane-initiated extra-nuclear actions of estrogen receptor. Steroids 2011; 76:877-84. [PMID: 21354435 DOI: 10.1016/j.steroids.2011.02.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 01/14/2011] [Accepted: 02/16/2011] [Indexed: 12/30/2022]
Abstract
Estrogen mediates biological effects on cell proliferation, differentiation, and homeostasis through estrogen receptor (ER). In addition to functioning as a ligand-activated nuclear transcription factor to directly regulate gene transcription, ER also mediates rapid activation of signaling pathways independent of its transcriptional activity. A subpopulation of ER localized to the cell membrane or cytoplasm has been proposed to mediate ER activation of signaling pathways. This review focuses on recent advances in our understanding of mechanisms responsible for ER cytoplasm/membrane localization, where rapid extra-nuclear signaling is initiated. These mechanisms include lipid modification of the receptor (palmitoylation) and interactions with membrane and cytoplasmic adaptor proteins including caveolins, striatin, p130Cas, Shc, HPIP, MTA-1s, and MNAR/PELP1. While it is clear that ER mediates rapid extra-nuclear signaling resulting in activation of signaling pathways such as Src/MAPK and PI-3 kinase/Akt, how ER extra-nuclear signaling influences overall ER/estrogen physiology is still not well understood. Future studies defining physiological roles of ER extra-nuclear actions and crosstalk with its nuclear counterparts will be important to our overall understanding of estrogen and ER biological functions.
Collapse
Affiliation(s)
- Viroj Boonyaratanakornkit
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, MS-130, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Zheng S, Huang J, Zhou K, Zhang C, Xiang Q, Tan Z, Wang T, Fu X. 17β-Estradiol enhances breast cancer cell motility and invasion via extra-nuclear activation of actin-binding protein ezrin. PLoS One 2011; 6:e22439. [PMID: 21818323 PMCID: PMC3144228 DOI: 10.1371/journal.pone.0022439] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 06/27/2011] [Indexed: 12/21/2022] Open
Abstract
Estrogen promotes breast cancer metastasis. However, the detailed mechanism remains largely unknown. The actin binding protein ezrin is a key component in tumor metastasis and its over-expression is positively correlated to the poor outcome of breast cancer. In this study, we investigate the effects of 17β-estradiol (E2) on the activation of ezrin and its role in estrogen-dependent breast cancer cell movement. In T47-D breast cancer cells, E2 rapidly enhances ezrin phosphorylation at Thr567 in a time- and concentration-dependent manner. The signalling cascade implicated in this action involves estrogen receptor (ER) interaction with the non-receptor tyrosine kinase c-Src, which activates the phosphatidylinositol-3 kinase/Akt pathway and the small GTPase RhoA/Rho-associated kinase (ROCK-2) complex. E2 enhances the horizontal cell migration and invasion of T47-D breast cancer cells in three-dimensional matrices, which is reversed by transfection of cells with specific ezrin siRNAs. In conclusion, E2 promotes breast cancer cell movement and invasion by the activation of ezrin. These results provide novel insights into the effects of estrogen on breast cancer progression and highlight potential targets to treat endocrine-sensitive breast cancers.
Collapse
Affiliation(s)
- Shuhui Zheng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinghe Huang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chengxi Zhang
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuling Xiang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tinghuai Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| | - Xiaodong Fu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| |
Collapse
|
34
|
Hiscox S, Nicholson RI. Src kinase: a therapeutic opportunity in endocrine-responsive and resistant breast cancer. Expert Rev Endocrinol Metab 2011; 6:423-435. [PMID: 30754115 DOI: 10.1586/eem.11.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The intracellular kinase, Src, interacts with a diverse array of signaling elements, including the estrogen receptor to regulate breast cancer progression. Recent evidence has also implicated Src in mediating the response of breast cancer to endocrine agents and in the acquisition of antihormone resistance, a significant limiting factor to the clinical effectiveness of systemic endocrine therapy. A number of pharmacological inhibitors of Src kinase have been developed that are effective at suppressing breast cancer growth and invasion in vitro and inhibiting disease spread in vivo. Significantly, there appears to be added benefit when these agents are given in combination with anti-estrogens in endocrine-sensitive and -resistant models. These new findings suggest that Src inhibitors might have therapeutic value in breast cancer patients to improve endocrine response and circumvent resistance.
Collapse
Affiliation(s)
- Stephen Hiscox
- a Breast Cancer (Molecular Pharmacology) Group, Welsh School of Pharmacy, Redwood Building, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
- b
| | - Robert I Nicholson
- a Breast Cancer (Molecular Pharmacology) Group, Welsh School of Pharmacy, Redwood Building, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| |
Collapse
|
35
|
Castoria G, D'Amato L, Ciociola A, Giovannelli P, Giraldi T, Sepe L, Paolella G, Barone MV, Migliaccio A, Auricchio F. Androgen-induced cell migration: role of androgen receptor/filamin A association. PLoS One 2011; 6:e17218. [PMID: 21359179 PMCID: PMC3040221 DOI: 10.1371/journal.pone.0017218] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 01/25/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Androgen receptor (AR) controls male morphogenesis, gametogenesis and prostate growth as well as development of prostate cancer. These findings support a role for AR in cell migration and invasiveness. However, the molecular mechanism involved in AR-mediated cell migration still remains elusive. METHODOLOGY/PRINCIPAL FINDINGS Mouse embryo NIH3T3 fibroblasts and highly metastatic human fibrosarcoma HT1080 cells harbor low levels of transcriptionally incompetent AR. We now report that, through extra nuclear action, AR triggers migration of both cell types upon stimulation with physiological concentrations of the androgen R1881. We analyzed the initial events leading to androgen-induced cell migration and observed that challenging NIH3T3 cells with 10 nM R1881 rapidly induces interaction of AR with filamin A (FlnA) at cytoskeleton. AR/FlnA complex recruits integrin beta 1, thus activating its dependent cascade. Silencing of AR, FlnA and integrin beta 1 shows that this ternary complex controls focal adhesion kinase (FAK), paxillin and Rac, thereby driving cell migration. FAK-null fibroblasts migrate poorly and Rac inhibition by EHT impairs motility of androgen-treated NIH3T3 cells. Interestingly, FAK and Rac activation by androgens are independent of each other. Findings in human fibrosarcoma HT1080 cells strengthen the role of Rac in androgen signaling. The Rac inhibitor significantly impairs androgen-induced migration in these cells. A mutant AR, deleted of the sequence interacting with FlnA, fails to mediate FAK activation and paxillin tyrosine phosphorylation in androgen-stimulated cells, further reinforcing the role of AR/FlnA interaction in androgen-mediated motility. CONCLUSIONS/SIGNIFICANCE The present report, for the first time, indicates that the extra nuclear AR/FlnA/integrin beta 1 complex is the key by which androgen activates signaling leading to cell migration. Assembly of this ternary complex may control organ development and prostate cancer metastasis.
Collapse
Affiliation(s)
- Gabriella Castoria
- Dipartimento di Patologia Generale, II Università di Napoli, Napoli, Italy
| | - Loredana D'Amato
- Dipartimento di Patologia Generale, II Università di Napoli, Napoli, Italy
| | | | - Pia Giovannelli
- Dipartimento di Patologia Generale, II Università di Napoli, Napoli, Italy
| | - Tiziana Giraldi
- Dipartimento di Patologia Generale, II Università di Napoli, Napoli, Italy
| | - Leandra Sepe
- Dipartimento di Biochimica e Biotecnologie Mediche, Università ‘Federico II’, Napoli, Italy
| | - Giovanni Paolella
- Dipartimento di Biochimica e Biotecnologie Mediche, Università ‘Federico II’, Napoli, Italy
| | - Maria Vittoria Barone
- European Laboratory for the Investigation of Food Induced Disease, Dipartimento di Pediatria, Università ‘Federico II’, Napoli, Italy
| | - Antimo Migliaccio
- Dipartimento di Patologia Generale, II Università di Napoli, Napoli, Italy
| | | |
Collapse
|
36
|
Abstract
Tyrosine kinases play significant roles in tumor progression and therapy resistance. Inhibitors of tyrosine kinases are on the forefront of targeted therapy. For prostate cancer, tyrosine kinases play an additional role in the development of castration-resistant disease state, the most troubling aspect of prostate cancinogenesis which presently defies any effective treatment. Among the 30 or so tyrosine kinases expressed in a typical prostate cancer cell, nearly one third of them have been implicated in prostate carcinogenesis. Interestingly, most of them channel signals through a trio of non-receptor tyrosine kinases, Src/Etk/FAK, referred here as Src tyrosine kinase complex. This complex has been shown to play a significant role in the aberrant activation of androgen receptor (AR) mediated by growth factors (e.g., epidermal growth factor (EGF)), cytokines (interleukin (IL)-6), chemokines (IL-8), and neurokines (gastrin-releasing peptide). These factors are induced and released from the prostate cancer to the stromal cells upon androgen withdrawal. The Src kinase complex has the ability to phosphorylate androgen receptor, resulting in the nuclear translocation and stabilization of un-liganded androgen receptor. Indeed, tyrosine kinase inhibitors targeting Src can inhibit androgen-independent growth of prostate cancer cells in vitro and in preclinical xenograft model. While effective in inducing growth arrest and inhibiting metastasis of castration-resistant tumors, Src inhibitors rarely induce a significant level of apoptosis. This is also reflected by the general ineffectiveness of tyrosine kinase inhibitors as monotherapy in clinical trials. One of the underlying causes of apoptosis resistance is "autophagy," which is induced by tyrosine kinase inhibitors and by androgen withdrawal. Autophagy is a self-digesting process to regenerate energy by removal of long-lived proteins and retired organelles to provide a survival mechanism to cells encountering stresses. Excessive autophagy, sometimes, could lead to type II programmed cell death. We demonstrated that autophagy blockade sensitizes prostate cancer cells toward Src tyrosine kinase inhibitor. Thus, a combination therapy based on Src tyrosine kinase inhibitor and autophagy modulator deserves further attention as a potential treatment for relapsed prostate cancer.
Collapse
Affiliation(s)
- Hsing-Jien Kung
- UC Davis Cancer Center, UCDMC, Res III, Rm. 2400, 4645 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
37
|
Fu XD, Russo E, Zullino S, Genazzani AR, Simoncini T. Sex steroids and breast cancer metastasis. Horm Mol Biol Clin Investig 2010; 3:383-9. [PMID: 25961210 DOI: 10.1515/hmbci.2010.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 10/19/2010] [Indexed: 11/15/2022]
Abstract
Sex steroids, particularly estrogen and progesterone, promote normal breast tissue growth and differentiation. Prolonged exposure of estrogen and/or progesterone is considered a risk factor for breast cancer carcinogenesis, and the effects of sex steroids on breast cancer metastasis are controversial. Emerging evidence indicates that sex steroids regulate breast cancer metastatic processes via nongenomic and genomic mechanisms. Through the regulation of actin-binding proteins estrogen and progesterone rapidly provoke actin cytoskeleton reorganization in breast cancer cells, leading to formation of membrane structures facilitating breast cancer cell migration and invasion. In addition, steroid receptors interact and trans-activate receptor tyrosine kinases (including epidermal growth factor receptor and insulin-like growth factor receptor), resulting in growth factor-like effects that promote cancer cell invasive behavior. Moreover, sex steroids regulate the expression of metastasis-associated molecules, such as E-cadherin, matrix metalloproteinases, growth factors, chemokines and their receptors, leading to epithelial-to-mesenchymal-like transition. However, there is also evidence that sex steroids and their receptors protect against breast cancer cell invasiveness through distinct mechanisms. Here, we present an overview of the currently identified actions of sex steroids on breast cancer metastasis and their potential clinical implications.
Collapse
|
38
|
Anaganti S, Fernández-Cuesta L, Langerød A, Hainaut P, Olivier M. p53-Dependent repression of focal adhesion kinase in response to estradiol in breast cancer cell-lines. Cancer Lett 2010; 300:215-24. [PMID: 21071137 DOI: 10.1016/j.canlet.2010.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 10/08/2010] [Accepted: 10/11/2010] [Indexed: 12/13/2022]
Abstract
Mutations in the TP53 suppressor gene are frequent in breast cancers. These mutations are associated with poor prognosis, thought to be due to proliferative advantage and poor response to chemotherapy associated with loss of p53 function. The focal adhesion kinase (FAK/PTK2), a tyrosine kinase, is over-expressed in a variety of human tumors including breast cancers. FAK is a critical regulator of adhesion and motility and its over-expression is associated with increased metastatic potential. Recently, FAK promoter has been shown to contain p53 responsive elements and to be down-regulated by DNA-damage in a p53-dependent manner. Here, we have used five estrogen-dependent breast cancer cells lines with different p53 status, including an isogenic model, to show that FAK expression was regulated in a p53-dependent manner in response to estradiol. FAK protein and mRNA expression were down-regulated by estradiol in wild-type but not mutant p53 cells. Moreover, silencing wild-type p53 increased FAK expression, while over expressing p53 repressed FAK expression. ChIP experiment showed that p53 bound to FAK promoter in the presence of estradiol in p53 wild-type but not in mutant p53 cells, suggesting a direct role of p53 in down regulating FAK mRNA expression. FAK mRNA expression was also found to correlate with TP53 mutation status in a series of breast tumors. Finally, loss of FAK down-regulation in p53 mutant cells was correlated with increased proliferation and invasion upon estradiol stimulation, while FAK silencing reduced invasion. These results suggest that p53 is an important down regulator of FAK and that loss of p53 function in breast cancer may contribute to the metastatic potential of estrogen-responsive tumors through uncontrolled FAK expression upon estrogens stimulation.
Collapse
|
39
|
Sanchez AM, Flamini MI, Baldacci C, Goglia L, Genazzani AR, Simoncini T. Estrogen receptor-alpha promotes breast cancer cell motility and invasion via focal adhesion kinase and N-WASP. Mol Endocrinol 2010; 24:2114-25. [PMID: 20880986 DOI: 10.1210/me.2010-0252] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ability of cancer cells to move and invade the surrounding environment is the basis of local and distant metastasis. Cancer cell movement requires dynamic remodeling of the cytoskeleton and cell membrane and is controlled by multiprotein complexes including focal adhesion kinase (FAK) or the Neural Wiskott-Aldrich Syndrome Protein (N-WASP). We show that 17β-estradiol induces phosphorylation of FAK and its translocation toward membrane sites where focal adhesion complexes are assembled. This process is triggered via a Gα/Gβ protein-dependent, rapid extranuclear signaling of estrogen receptor α interacts in a multiprotein complex with c-Src, phosphatidylinositol 3-OH kinase, and FAK. Within this complex FAK autophosphorylation ensues, and activated FAK recruits the small GTPase cdc42, which, in turn, triggers N-WASP phosphorylation. This results in the translocation of Arp2/3 complexes at sites where membrane structures related to cell movement are formed. Recruitment of FAK and N-WASP is necessary for cell migration and invasion induced by 17β-estradiol in breast cancer cells. Our findings identify an original mechanism through which estrogen promotes breast cancer cell motility and invasion. This information helps to understand the effects of estrogen on breast cancer metastasis and may provide new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Angel Matias Sanchez
- Molecular and Cellular Gynecological Endocrinology Laboratory, Department of Reproductive Medicine and Child Development, Division of Obstetrics and Gynecology, University of Pisa, Via Roma, 57, 56100, Pisa, Italy
| | | | | | | | | | | |
Collapse
|
40
|
Lee JH, Pyon JK, Kim DW, Lee SH, Nam HS, Kim CH, Kang SG, Lee YJ, Park MY, Jeong DJ, Cho MK. Elevated c-Src and c-Yes expression in malignant skin cancers. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:116. [PMID: 20796316 PMCID: PMC2936336 DOI: 10.1186/1756-9966-29-116] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Accepted: 08/27/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Src family kinases (SFKs) play an important role in cancer proliferation, survival, motility, invasiveness, metastasis, and angiogenesis. Among the SFKs, c-Src and c-Yes are particularly over-expressed or hyper-activated in many human epithelial cancers. However, only a few studies have attempted to define the expression and role of c-Src and c-Yes in cutaneous carcinomas. OBJECTIVES To investigate the expression of c-Src and c-Yes in cutaneous carcinomas to include malignant melanoma (MM), squamous cell carcinoma (SCC) and basal cell carcinoma (BCC). METHODS We examined 6 normal skin tissues and 18 malignant skin tumor tissues using western blotting for the expression of c-Src and c-Yes. In another set, 16 specimens of MM, 16 SCCs and 16 BCCs were analyzed for the expression of c-Src and c-Yes using immunohistochemical staining. RESULTS Western blotting showed that c-Src was expressed in all malignant skin tumors, but not in normal skin, while c-Yes was expressed in MM and SCC, but not in BCC and normal skin. Immunohistochemical staining results of c-Src and c-Yes in MM, SCC, and BCC mirrored those of the western blot analysis. CONCLUSIONS c-Src, rather than c-Yes, plays a key role in the proliferation and progression of malignant skin cancers.
Collapse
Affiliation(s)
- Jang Hyun Lee
- Molecular Cancer Research Center, College of Medicine, Soonchunhyang University, Chunan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bolós V, Gasent JM, López-Tarruella S, Grande E. The dual kinase complex FAK-Src as a promising therapeutic target in cancer. Onco Targets Ther 2010; 3:83-97. [PMID: 20616959 PMCID: PMC2895777 DOI: 10.2147/ott.s6909] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Indexed: 12/11/2022] Open
Abstract
Focal adhesion kinase (FAK) and steroid receptor coactivator (Src) are intracellular (nonreceptor) tyrosine kinases that physically and functionally interact to promote a variety of cellular responses. Plenty of reports have already suggested an additional central role for this complex in cancer through its ability to promote proliferation and anoikis resistance in tumor cells. An important role for the FAK/Src complex in tumor angiogenesis has also been established. Furthermore, FAK and Src have been associated with solid tumor metastasis through their ability to promote the epithelial mesenchymal transition. In fact, a strong correlation between increased FAK/Src expression/phosphorylation and the invasive phenotype in human tumors has been found. Additionally, an association for FAK/Src with resistances to the current anticancer therapies has already been established. Currently, novel anticancer agents that target FAK or Src are under development in a broad variety of solid tumors. In this article we will review the normal cellular functions of the FAK/Src complex as an effector of integrin and/or tyrosine kinase receptor signaling. We will also collect data about their role in cancer and we will summarize the most recent data from the FAK and Src inhibitors under clinical and preclinical development. Furthermore, the association of both these proteins with chemotherapy and hormonal therapy resistances, as a rationale for new combined therapeutic approaches with these novel agents, to abrogate treatment associated resistances, will also be reviewed.
Collapse
|
42
|
Vanderlaag KE, Hudak S, Bald L, Fayadat-Dilman L, Sathe M, Grein J, Janatpour MJ. Anterior gradient-2 plays a critical role in breast cancer cell growth and survival by modulating cyclin D1, estrogen receptor-alpha and survivin. Breast Cancer Res 2010; 12:R32. [PMID: 20525379 PMCID: PMC2917027 DOI: 10.1186/bcr2586] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 05/08/2010] [Accepted: 06/04/2010] [Indexed: 11/23/2022] Open
Abstract
Introduction Anterior-gradient 2 (AGR2) is an estrogen-responsive secreted protein. Its upregulation has been well documented in a number of cancers, particularly breast cancer, for which mixed data exist on the prognostic implications of AGR2 expression. Although emerging evidence indicates that AGR2 is associated with poor prognosis, its function and impact on cancer-relevant pathways have not been elucidated in breast cancer. Methods To investigate the biologic role of AGR2 in breast cancer, AGR2 was transiently knocked down, by using siRNA, in T47 D and ZR-75-1 (estrogen receptor-α (ER)-positive) and MDA-MB-231 and SK-BR-3 (ER-negative) human breast cancer cell lines. The impact of silencing AGR2 was evaluated in both anchorage-dependent and anchorage-independent growth (soft agar, spheroid) assays. Cell-cycle profiles in ER-positive cell lines were determined with BrdU incorporation, and cell death was measured with Annexin V, JC-1, and F7-26 staining. After transiently silencing AGR2 or stimulating with recombinant AGR2, modulation of key regulators of growth and survival pathways was assessed with Western blot. Combination studies of AGR2 knockdown with the antiestrogens tamoxifen and fulvestrant were carried out and assessed at the level of anchorage-dependent growth inhibition and target modulation (cyclin D1, ER). Results AGR2 knockdown inhibited growth in anchorage-dependent and anchorage-independent assays, with a more-pronounced effect in ER-positive cell lines. Cyclin D1 levels and BrdU incorporation were reduced with AGR2 knockdown. Conversely, cyclin D1 was induced with recombinant AGR2. AGR2 knockdown induced cell death in ZR-75-1 and T47 D cells, and also downregulated survivin and c-Myc. Evidence of AGR2-ER crosstalk was demonstrated by a reduction of ER at the protein level after transiently silencing AGR2. AGR2 knockdown in combination with fulvestrant or tamoxifen did not preclude the efficacy of the antiestrogens, but enhanced it. In addition, p-Src, implicated in tamoxifen resistance, was downregulated with AGR2 knockdown. Conclusions Transiently silencing AGR2 in ER-positive breast cancer cell lines inhibited cell growth and cell-cycle progression and induced cell death. Breast cancer drivers (ER and cyclin D1) as well as cancer-signaling nodes (pSrc, c-Myc, and survivin) were demonstrated to be downstream of AGR2. Collectively, the data presented support the utility of anti-AGR2 therapy in ER-positive breast cancers because of its impact on cancer-relevant pathways.
Collapse
Affiliation(s)
- Kathryn E Vanderlaag
- Schering-Plough Biopharma, 901 South California Avenue, Palo Alto, CA 94304, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Glondu-Lassis M, Dromard M, Lacroix-Triki M, Nirdé P, Puech C, Knani D, Chalbos D, Freiss G. PTPL1/PTPN13 regulates breast cancer cell aggressiveness through direct inactivation of Src kinase. Cancer Res 2010; 70:5116-26. [PMID: 20501847 DOI: 10.1158/0008-5472.can-09-4368] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The protein tyrosine phosphatase PTPL1/PTPN13, the activity of which is decreased through allelic loss, promoter methylation, or somatic mutations in some tumors, has been proposed as a tumor suppressor gene. Moreover, our recent clinical study identified PTPL1 expression level as an independent prognostic indicator of a favorable outcome for patients with breast cancer. However, how PTPL1 can affect tumor aggressiveness has not been characterized. Here, we first show that PTPL1 expression, assessed by immunohistochemistry, is decreased in breast cancer and metastasis specimens compared with nonmalignant tissues. Second, to evaluate whether PTPL1 plays a critical role in breast cancer progression, RNA interference experiments were performed in poorly tumorigenic MCF-7 breast cancer cells. PTPL1 inhibition drastically increased tumor growth in athymic mice and also enhanced several parameters associated with tumor progression, including cell proliferation on extracellular matrix components and cell invasion. Furthermore, the inhibition of Src kinase expression drastically blocked the effects of PTPL1 silencing on cell growth. In PTPL1 knockdown cells, the phosphorylation of Src on tyrosine 419 is increased, leading to the activation of its downstream substrates Fak and p130cas. Finally, substrate-trapping experiments revealed that Src tyrosine 419 is a direct target of the phosphatase. Thus, by identification of PTPL1 as the first phosphatase able to inhibit Src through direct dephosphorylation in intact cells, we presently describe a new mechanism by which PTPL1 inhibits breast tumor aggressiveness.
Collapse
|
44
|
Combining Src inhibitors and aromatase inhibitors: a novel strategy for overcoming endocrine resistance and bone loss. Eur J Cancer 2010; 46:2187-95. [PMID: 20471823 DOI: 10.1016/j.ejca.2010.04.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 03/18/2010] [Accepted: 04/14/2010] [Indexed: 11/21/2022]
Abstract
Aromatase inhibitors have largely replaced tamoxifen as the first-line treatment for postmenopausal women with metastatic, hormone receptor-positive (HR+) breast cancer. However, many patients develop clinical resistance with prolonged treatment, and oestrogen deprivation following aromatase inhibition can result in loss of bone mineral density. Furthermore, most patients with metastatic breast cancer develop bone metastases, and the resulting adverse skeletal-related events are a significant cause of patient morbidity. Src, a non-receptor tyrosine kinase, is a component of signalling pathways that regulate breast cancer cell proliferation, invasion and metastasis as well as osteoclast-mediated bone turnover. Preclinical evidence also suggests a role for Src in acquired endocrine resistance. As such, Src inhibition represents a logical strategy for the treatment of metastatic breast cancer. In vitro, combination therapy with Src inhibitors and endocrine agents, including aromatase inhibitors, has been shown to inhibit the proliferation and metastasis of both endocrine-responsive and endocrine-resistant breast cancer cell lines more effectively than either of the therapy alone. Src inhibition has also been shown to suppress osteoclast formation and activity. Combination therapy with aromatase inhibitors and Src inhibitors therefore represents a novel approach through which the development of both acquired resistance and bone pathology could be delayed. Data from clinical trials utilising such combinations will reveal if this strategy has the potential to improve patient outcomes.
Collapse
|
45
|
Hiscox S, Barnfather P, Hayes E, Bramble P, Christensen J, Nicholson RI, Barrett-Lee P. Inhibition of focal adhesion kinase suppresses the adverse phenotype of endocrine-resistant breast cancer cells and improves endocrine response in endocrine-sensitive cells. Breast Cancer Res Treat 2010; 125:659-69. [PMID: 20354780 DOI: 10.1007/s10549-010-0857-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 03/16/2010] [Indexed: 01/25/2023]
Abstract
Acquired resistance to endocrine therapy in breast cancer is a major clinical problem. Previous reports have demonstrated that cell models of acquired endocrine resistance have altered cell-matrix adhesion and a highly migratory phenotype, features which may impact on tumour spread in vivo. Focal adhesion kinase (FAK) is an intracellular kinase that regulates signalling pathways central to cell adhesion, migration and survival and its expression is frequently deregulated in breast cancer. In this study, we have used the novel FAK inhibitor PF573228 to address the role of FAK in the development of endocrine resistance. Whilst total-FAK expression was similar between endocrine-sensitive and endocrine-resistant MCF7 cells, FAK phosphorylation status (Y397 or Y861) was altered in resistance. PF573228 promoted a dose-dependent inhibition of FAK phosphorylation at Y397 but did not affect other FAK activation sites (pY407, pY576 and pY861). Endocrine-resistant cells were more sensitive to these inhibitory effects versus MCF7 (mean IC(50) for FAK pY397 inhibition: 0.43 μM, 0.05 μM and 0.13 μM for MCF7, TamR and FasR cells, respectively). Inhibition of FAK pY397 was associated with a reduction in TamR and FasR adhesion to, and migration over, matrix components. PF573228 as a single agent (0-1 μM) did not affect the growth of MCF7 cells or their endocrine-resistant counterparts. However, treatment of endocrine-sensitive cells with PF573228 and tamoxifen combined resulted in greater suppression of proliferation versus single agent treatment. Together these data suggest the importance of FAK in the process of endocrine resistance, particularly in the development of an aggressive, migratory cell phenotype and demonstrate the potential to improve endocrine response through combination treatment.
Collapse
Affiliation(s)
- Stephen Hiscox
- Welsh School of Pharmacy, Cardiff University, Redwood Building, Kind Edward VII Avenue, Cathays Park, Cardiff, CF10 3NB, UK.
| | | | | | | | | | | | | |
Collapse
|
46
|
Beierle EA, Ma X, Trujillo A, Kurenova EV, Cance WG, Golubovskaya VM. Inhibition of focal adhesion kinase and src increases detachment and apoptosis in human neuroblastoma cell lines. Mol Carcinog 2010; 49:224-34. [PMID: 19885861 DOI: 10.1002/mc.20592] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood. Focal adhesion kinase (FAK) is an intracellular kinase that is overexpressed in a number of human tumors including neuroblastoma, and regulates both cellular adhesion and survival. We have studied the effects of FAK inhibition upon neuroblastoma using adenovirus-containing FAK-CD (AdFAK-CD). Utilizing an isogenic MYCN+/MYCN- neuroblastoma cell line, we found that the MYCN+ cells are more sensitive to FAK inhibition with AdFAK-CD than their MYCN negative counterparts. In addition, we have shown that phosphorylation of Src is increased in the untreated isogenic MYCN- neuroblastoma cells, and that the decreased sensitivity of the MYCN- neuroblastoma cells to FAK inhibition with AdFAK-CD is abrogated by the addition of the Src family kinase inhibitor, PP2. The results of the current study suggest that both FAK and Src play a role in protecting neuroblastoma cells from apoptosis, and that dual inhibition of these kinases may be important when designing therapeutic interventions for this tumor.
Collapse
Affiliation(s)
- Elizabeth A Beierle
- Department of Surgery, University of Alabama, Birmingham, Alabama 35233, USA
| | | | | | | | | | | |
Collapse
|
47
|
Deng B, Yang X, Liu J, He F, Zhu Z, Zhang C. Focal adhesion kinase mediates TGF-β1-induced renal tubular epithelial-to-mesenchymal transition in vitro. Mol Cell Biochem 2010; 340:21-9. [DOI: 10.1007/s11010-010-0396-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Accepted: 02/03/2010] [Indexed: 11/29/2022]
|
48
|
Koshman YE, Piano MR, Russell B, Schwertz DW. Signaling responses after exposure to 5 alpha-dihydrotestosterone or 17 beta-estradiol in norepinephrine-induced hypertrophy of neonatal rat ventricular myocytes. J Appl Physiol (1985) 2009; 108:686-96. [PMID: 20044473 DOI: 10.1152/japplphysiol.00994.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Androgens appear to enhance, whereas estrogens mitigate, cardiac hypertrophy. However, signaling pathways in cells for short (3 min) and longer term (48 h) treatment with 17beta-estradiol (E2) or 5 alpha-dihydrotestosterone (DHT) are understudied. We compared the effect of adrenergic stimulation by norepinephrine (NE; 1 microM) alone or in combination with DHT (10 nM) or E2 (10 nM) treatment in neonatal rat ventricular myocytes (NRVMs) by cell area, protein synthesis, sarcomeric structure, gene expression, phosphorylation of extracellular signal-regulated (ERK), and focal adhesion kinases (FAK), and phospho-FAK nuclear localization. NE alone elicited the expected hypertrophy and strong sarcomeric organization, and DHT alone gave a similar but more modest response, whereas E2 did not alter cell size. Effects of NE dominated when used with either E2 or DHT with all combinations. Both sex hormones alone rapidly activated FAK but not ERK. Long-term or brief exposure to E2 attenuated NE-induced FAK phosphorylation, whereas DHT had no effect. Neither hormone altered NE-elicited ERK activation. Longer term exposure to E2 alone reduced FAK phosphorylation and reduced nuclear phospho-FAK, whereas its elevation was seen in the presence of NE with both sex hormones. The mitigating effects of E2 on the NE-elicited increase in cell size and the hypertrophic effect of DHT in NRVMs are in accordance with results observed in whole animal models. This is the first report of rapid, nongenomic sex hormone signaling via FAK activation and altered FAK trafficking to the nucleus in heart cells.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- Department of Physiology and Biophysics, University of Illinois at Chicago, MC 901, 835 South Wolcott Ave., Chicago, IL 60612-7342, USA
| | | | | | | |
Collapse
|
49
|
Saad F, Lipton A. SRC kinase inhibition: targeting bone metastases and tumor growth in prostate and breast cancer. Cancer Treat Rev 2009; 36:177-84. [PMID: 20015594 DOI: 10.1016/j.ctrv.2009.11.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 11/12/2009] [Accepted: 11/16/2009] [Indexed: 11/25/2022]
Abstract
Prostate and breast cancer cells preferentially metastasize to bone, whereupon a complex interaction between metastatic tumor cells, osteoclasts, and osteoblasts results in the development of bone lesions that cause significant pain and patient morbidity. For patients with bone lesions, the goals of treatment are to decrease tumor growth, prevent further metastases, and inhibit tumor-associated bone pathology. Preclinical data suggest that SRC, a nonreceptor tyrosine kinase, is an important signaling molecule during the processes of osteoclast-mediated bone resorption, tumor growth, and metastasis, and that SRC has a role in hormone receptor signaling and resistance. As such, SRC represents a logical target for the treatment of advanced metastatic prostate or breast cancer. SRC-targeting agents, including dasatinib, saracatinib, and bosutinib, are currently in clinical development for patients with solid tumors. Preliminary data from phase 1/2 trials, including tumor responses and bone-specific activity in patients with prostate or breast cancer, demonstrate that SRC inhibitors have potential in the clinical setting. Data arising from ongoing and future clinical trials will confirm whether SRC inhibitors provide clinical benefits for patients with advanced disease.
Collapse
Affiliation(s)
- Fred Saad
- University of Montreal, CHU Montreal, 1560 Sherbrooke East, Montreal, Quebec, Canada.
| | | |
Collapse
|
50
|
Abstract
Metastasis may arise years after removal of a primary tumor. The mechanisms allowing latent disseminated cancer cells to survive are unknown. We report that a gene expression signature of Src activation is associated with late-onset bone metastasis in breast cancer. This link is independent of hormone receptor status or breast cancer subtype. In breast cancer cells, Src is dispensable for homing to the bones or lungs but is critical for the survival and outgrowth of these cells in the bone marrow. Src mediates AKT regulation and cancer cell survival responses to CXCL12 and TNF-related apoptosis-inducing ligand (TRAIL), factors that are distinctively expressed in the bone metastasis microenvironment. Breast cancer cells that lodge in the bone marrow succumb in this environment when deprived of Src activity.
Collapse
|