51
|
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that are responsible for fine-tuning the spatial and temporal expression of a broad range of genes both during development and in adult tissues. This function is engrained in their ability to integrate a multitude of cellular and environmental signals and to act with remarkable fidelity. Several key members of the FOXA, FOXC, FOXM, FOXO and FOXP subfamilies are strongly implicated in cancer, driving initiation, maintenance, progression and drug resistance. The functional complexities of FOX proteins are coming to light and have established these transcription factors as possible therapeutic targets and putative biomarkers for specific cancers.
Collapse
Affiliation(s)
- Eric W-F Lam
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | | | | | | |
Collapse
|
52
|
Stumm L, Burkhardt L, Steurer S, Simon R, Adam M, Becker A, Sauter G, Minner S, Schlomm T, Sirma H, Michl U. Strong expression of the neuronal transcription factor FOXP2 is linked to an increased risk of early PSA recurrence in ERG fusion-negative cancers. J Clin Pathol 2013; 66:563-8. [PMID: 23559350 DOI: 10.1136/jclinpath-2012-201335] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Transcription factors of the forkhead box P (FOXP1-4) family have been implicated in various human cancer types before. The relevance and role of neuronal transcription factor FOXP2 in prostate cancer is unknown. METHODS A tissue microarray containing samples from more than 11 000 prostate cancers from radical prostatectomy specimens with clinical follow-up data was analysed for FOXP2 expression by immunohistochemistry. FOXP2 data were also compared with pre-existing ERG fusion (by fluorescence in situ hybridisation and immunohistochemistry) and cell proliferation (Ki67 labelling index) data. RESULTS There was a moderate to strong FOXP2 protein expression in basal and secretory cells of normal prostatic glands. As compared with normal cells, FOXP2 expression was lost or reduced in 25% of cancers. Strong FOXP2 expression was linked to advanced tumour stage, high Gleason score, presence of lymph node metastases and early tumour recurrence (p<0.0001; each) in ERG fusion-negative, but not in ERG fusion-positive cancers. High FOXP2 expression was linked to high Ki67 labelling index (p<0.0001) in all cancers irrespective of ERG fusion status. CONCLUSIONS These data demonstrate that similar high FOXP2 protein levels as in normal prostate epithelium exert a 'paradoxical' oncogenic role in 'non fusion-type' prostate cancer. It may be speculated that interaction of FOXP2 with members of pathways that are specifically activated in 'non fusion-type' cancers may be responsible for this phenomenon.
Collapse
Affiliation(s)
- Laura Stumm
- Institute of Pathology, University Medical Center, Hamburg-Eppendorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Abstract
Breast cancers are considered to be primarily regulated by estrogen signaling pathways because estrogen-dependent proliferation is observed in the majority of breast cancer cases. Thus, hormone therapy using antiestrogen drugs such as tamoxifen is effective for breast cancers expressing estrogen receptor α (ERα). However, acquired resistance during the endocrine therapy is a critical unresolved problem in breast cancer. Recently, a forkhead transcription factor FOXA1 has been reported to play an important role in the regulation of ERα-mediated transcription and proliferation of breast cancer. Interestingly, immunohistochemical analysis of breast cancer specimens has revealed that nuclear immunoreactivities of FOXP1 as well as those of FOXA1 are positively correlated with hormone receptor status, including ERα and progesterone receptor. In particular, the double-positive immunoreactivities of FOXP1 and FOXA1 are significantly associated with a favorable prognosis for survival of breast cancer patients receiving adjuvant tamoxifen therapy. The functions of FOXP1 and FOXA1 have been characterized in cultured cells; further, similar to FOXA1, FOXP1 is assumed to be a critical transcription factor for ERα signaling, and both forkhead transcription factors can serve as predictive factors for acquired endocrine resistance in breast cancer.
Collapse
|
54
|
Cancer genetics and genomics of human FOX family genes. Cancer Lett 2012; 328:198-206. [PMID: 23022474 DOI: 10.1016/j.canlet.2012.09.017] [Citation(s) in RCA: 290] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/20/2012] [Accepted: 09/21/2012] [Indexed: 12/16/2022]
Abstract
Forkhead-box (FOX) family proteins, involved in cell growth and differentiation as well as embryogenesis and longevity, are DNA-binding proteins regulating transcription and DNA repair. The focus of this review is on the mechanisms of FOX-related human carcinogenesis. FOXA1 is overexpressed as a result of gene amplification in lung cancer, esophageal cancer, ER-positive breast cancer and anaplastic thyroid cancer and is point-mutated in prostate cancer. FOXA1 overexpression in breast cancer and prostate cancer is associated with good or poor prognosis, respectively. Single nucleotide polymorphism (SNP) within the 5'-UTR of the FOXE1 (TTF2) gene is associated with thyroid cancer risk. FOXF1 overexpression in breast cancer is associated with epithelial-to-mesenchymal transition (EMT). FOXM1 is overexpressed owing to gene amplification in basal-type breast cancer and diffuse large B-cell lymphoma (DLBCL), and it is transcriptionally upregulated owing to Hedgehog-GLI, hypoxia-HIF1α or YAP-TEAD signaling activation. FOXM1 overexpression leads to malignant phenotypes by directly upregulating CCNB1, AURKB, MYC and SKP2 and indirectly upregulating ZEB1 and ZEB2 via miR-200b downregulation. Tumor suppressor functions of FOXO transcription factors are lost in cancer cells as a result of chromosomal translocation, deletion, miRNA-mediated repression, AKT-mediated cytoplasmic sequestration or ubiquitination-mediated proteasomal degradation. FOXP1 is upregulated as a result of gene fusion or amplification in DLBCL and MALT lymphoma and also repression of miRNAs, such as miR-1, miR-34a and miR-504. FOXP1 overexpression is associated with poor prognosis in DLBCL, gastric MALT lymphoma and hepatocellular carcinoma but with good prognosis in breast cancer. In neuroblastoma, the entire coding region of the FOXR1 (FOXN5) gene is fused to the MLL or the PAFAH1B gene owing to interstitial deletions. FOXR1 fusion genes function as oncogenes that repress transcription of FOXO target genes. Whole-genome sequencing data from tens of thousands of human cancers will uncover the mutational landscape of FOX family genes themselves as well as FOX-binding sites, which will be ultimately applied for cancer diagnostics, prognostics, and therapeutics.
Collapse
|
55
|
Forkhead box P1 overexpression and its clinicopathologic significance in peripheral T-cell lymphoma, not otherwise specified. Hum Pathol 2012; 43:1322-7. [DOI: 10.1016/j.humpath.2011.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 10/24/2011] [Accepted: 10/26/2011] [Indexed: 11/18/2022]
|
56
|
Feng J, Zhang X, Zhu H, Wang X, Ni S, Huang J. High expression of FoxP1 is associated with improved survival in patients with non-small cell lung cancer. Am J Clin Pathol 2012; 138:230-5. [PMID: 22904134 DOI: 10.1309/ajcpdhqfnyjz01yg] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
FoxP1 has been reported to be expressed in several types of human malignant tumors, and has been associated with metastasis and patient prognosis. Quantitative real-time polymerase chain reaction (PCR) and immunohistochemical analysis with tissue microarray were used to characterize the expression of FoxP1 in non-small cell lung cancer (NSCLC). It was revealed that the expression of FoxP1 messenger RNA (mRNA) and protein was significantly higher in NSCLC tissue than in corresponding peritumoral tissue (P = .013 and P < .001, respectively). The expression of FoxP1 protein in NSCLC was related to gender, histologic type, and 5-year survival rate (all P < .05). Finally, we evaluated the prognostic significance of the expression of FoxP1 in a group of patients. Kaplan-Meier survival and Cox regression analyses showed that low expression of FoxP1 (P < .001) and later stage grouping by TNM (P = .022) were independent factors predicting poor prognosis for NSCLC.
Collapse
Affiliation(s)
- Jian Feng
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Xuesong Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Huijun Zhu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Xudong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Songshi Ni
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jianfei Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
57
|
Jiang W, Li L, Tang Y, Zhang WY, Liu WP, Li GD. Expression of FOXP1 in mucosa-associated lymphoid tissue lymphoma suggests a large tumor cell transformation and predicts a poorer prognosis in the positive thyroid patients. Med Oncol 2012; 29:3352-9. [PMID: 22736042 DOI: 10.1007/s12032-012-0288-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Accepted: 06/14/2012] [Indexed: 02/05/2023]
Abstract
The forkhead box protein P1 (FOXP1) expression resulted from chromosome translocation was found in MALT lymphoma, and its nuclear expression in diffuse large B cell lymphoma has been believed to be a poor prognostic factor. In our study, FOXP1 expression was investigated in its relationship to the occurrence of large tumor cells, clinical features, and prognosis in a series of 115 MALT lymphomas divided into two groups with or without the large tumor cells. All cases were morphologically reviewed, and FOXP1 expression was detected both in mRNA and protein levels by real-time PCR, immunochemical staining, and Western blot hybridization. All available clinical data were collected. In the MALT lymphoma with large cells, FOXP1 expression was higher at both mRNA (P = 0.008) and protein (P = 0.000) levels than that in group without large cells, and most large tumor cells showed FOXP1 positivity. It was also found that cases beyond Ann Arbor stage I have a higher FOXP1 expression rate than cases in stage I (P = 0.01), moreover, FOXP1-positive group has more plasmacytic differentiation (P = 0.025), deeper filtrating depth in digestive tract (P = 0.039), and a higher Ki67 proliferation index (P = 0.022). However, no statistical significance was identified in the involved anatomic sites and prognosis. Our data demonstrated the close relationship between FOXP1 nuclear expression and the occurrence of large tumor cells in MALT lymphoma, which suggested the possibility of large cell transformation of FOXP1-positive cases. And FOXP1 positivity was associated with enhanced invasion and proliferation ability of tumor cells. In the thyroid cases, the FOXP1 positivity showed a poorer prognosis (P = 0.043), but the significance was not found in the overall survival analysis (P = 0.1123).
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/analysis
- Blotting, Western
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Female
- Forkhead Transcription Factors/analysis
- Forkhead Transcription Factors/biosynthesis
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Lymphoma, B-Cell, Marginal Zone/metabolism
- Lymphoma, B-Cell, Marginal Zone/mortality
- Lymphoma, B-Cell, Marginal Zone/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Prognosis
- Real-Time Polymerase Chain Reaction
- Repressor Proteins/analysis
- Repressor Proteins/biosynthesis
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/mortality
- Thyroid Neoplasms/pathology
- Tissue Array Analysis
Collapse
Affiliation(s)
- Wei Jiang
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guoxuexiang Street, Chengdu, Sichuan, 610041, People's Republic of China
| | | | | | | | | | | |
Collapse
|
58
|
Association of double-positive FOXA1 and FOXP1 immunoreactivities with favorable prognosis of tamoxifen-treated breast cancer patients. Discov Oncol 2012; 3:147-59. [PMID: 22476979 DOI: 10.1007/s12672-012-0111-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 03/16/2012] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is primarily a hormone-dependent tumor that can be regulated by the status of the steroid hormones estrogen and progesterone. Forkhead box A1 (FOXA1) is a member of the forkhead box transcription factor family and functions as a pioneer factor of the estrogen receptor (ER) in breast cancer. In the present study, we demonstrate that FOXA1 mRNA was upregulated by estrogen and that estrogen receptor-α (ERα) recruitment to ER-binding sites in the vicinity of the FOXA1 gene was increased by estrogen in ERα-positive MCF-7 breast cancer cells. The estrogen-induced FOXA1 upregulation was repressed by 4-hydroxytamoxifen treatment. We also demonstrated that the proliferation and the migration of MCF-7 cells were decreased by FOXA1-specific small interfering RNA (siRNA; siFOXA1). Furthermore, siFOXA1 decreased the estrogen response element-driven transcription and the estrogen-dependent upregulation of ERα target genes in MCF-7 cells. Next, the immunohistochemical analyses of FOXA1 were performed using two groups of breast cancer specimens. The nuclear immunoreactivity of FOXA1 was detected in 80 (74%) of 108 human invasive breast cancers and was negatively correlated with tumor grade and positively correlated with hormone receptor status, including ERα and progesterone receptor, pathological tumor size, and immunoreactivity of FOXP1, another FOX family transcription factor. FOXA1 immunoreactivity was significantly elevated in the relapse-free breast cancer patients treated with tamoxifen. Notably, the double-positive immunoreactivities of FOXA1 and FOXP1 were significantly associated with a favorable prognosis for the relapse-free and overall survival of patients with tamoxifen-treated breast cancer, with lower P values compared with FOXA1 or FOXP1 immunoreactivity alone. These results suggest that FOXA1 plays an important role in the proliferation and migration of breast cancer cells by modulating estrogen signaling and that the double-positive immunoreactivities of FOXA1 and FOXP1 are associated with a favorable prognosis of tamoxifen-treated breast cancer.
Collapse
|
59
|
Shigekawa T, Ijichi N, Ikeda K, Horie-Inoue K, Shimizu C, Saji S, Aogi K, Tsuda H, Osaki A, Saeki T, Inoue S. FOXP1, an Estrogen-Inducible Transcription Factor, Modulates Cell Proliferation in Breast Cancer Cells and 5-Year Recurrence-Free Survival of Patients with Tamoxifen-Treated Breast Cancer. Discov Oncol 2011. [DOI: 10.1007/s12672-011-0082-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
60
|
Chu YP, Chang CH, Shiu JH, Chang YT, Chen CY, Chuang WJ. Solution structure and backbone dynamics of the DNA-binding domain of FOXP1: insight into its domain swapping and DNA binding. Protein Sci 2011; 20:908-24. [PMID: 21416545 DOI: 10.1002/pro.626] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/15/2011] [Accepted: 03/02/2011] [Indexed: 11/09/2022]
Abstract
FOXP1 belongs to the P-subfamily of forkhead transcription factors and contains a conserved forkhead DNA-binding domain. According to size exclusion chromatography analysis, the forkhead domain of FOXP1 existed as a mixture of monomer and dimer. The dissociation constants of the forkhead domain of wild-type, C61S, and C61Y mutants of FOXP1 were 27.3, 28.8, and 332.0 μM, respectively. In contrast, FOXP1 A39P mutant formed only a monomer. NMR analysis also showed that FOXP1 C61S and C61Y mutants existed as a mixture. The solution structure of FOXP1 A39P/C61Y mutant was similar to the X-ray structure of the FOXP2 monomer. Comparison of backbone dynamics of FOXP1 A39P/C61Y and C61Y mutants showed that the residues preceding helix 3, the hinge region, exhibited the largest conformational exchange in FOXP1 monomer. The A39 residue of FOXP1 dimer has a lower order parameter with internal motion on the ps-ns timescale, suggesting that the dynamics of the hinge region of FOXP1 are important in the formation of the swapped dimer. The analysis also showed that the residues exhibiting the motions on the ps-ns and μs-ms timescales were located at the DNA-binding surface of FOXP1, suggesting the interactions between FOXP1 and DNA may be highly dynamic.
Collapse
Affiliation(s)
- Yuan-Ping Chu
- Department of Biochemistry and Molecular Biology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
61
|
Toma MI, Weber T, Meinhardt M, Zastrow S, Grimm MO, Füssel S, Wirth MP, Baretton GB. Expression of theForkheadTranscription Factor FOXP1 is Associated with Tumor Grade and Ki67 Expression in Clear Cell Renal Cell Carcinoma. Cancer Invest 2011; 29:123-9. [DOI: 10.3109/07357907.2010.535059] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
62
|
Newbury DF, Monaco AP. Genetic advances in the study of speech and language disorders. Neuron 2010; 68:309-20. [PMID: 20955937 PMCID: PMC2977079 DOI: 10.1016/j.neuron.2010.10.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2010] [Indexed: 11/29/2022]
Abstract
Developmental speech and language disorders cover a wide range of childhood conditions with overlapping but heterogeneous phenotypes and underlying etiologies. This characteristic heterogeneity hinders accurate diagnosis, can complicate treatment strategies, and causes difficulties in the identification of causal factors. Nonetheless, over the last decade, genetic variants have been identified that may predispose certain individuals to different aspects of speech and language difficulties. In this review, we summarize advances in the genetic investigation of stuttering, speech-sound disorder (SSD), specific language impairment (SLI), and developmental verbal dyspraxia (DVD). We discuss how the identification and study of specific genes and pathways, including FOXP2, CNTNAP2, ATP2C2, CMIP, and lysosomal enzymes, may advance our understanding of the etiology of speech and language disorders and enable us to better understand the relationships between the different forms of impairment across the spectrum.
Collapse
Affiliation(s)
- D F Newbury
- Wellcome Trust Centre for Human Genetics, Headington, Oxford, UK.
| | | |
Collapse
|
63
|
Zhang Y, Li S, Yuan L, Tian Y, Weidenfeld J, Yang J, Liu F, Chokas AL, Morrisey EE. Foxp1 coordinates cardiomyocyte proliferation through both cell-autonomous and nonautonomous mechanisms. Genes Dev 2010; 24:1746-57. [PMID: 20713518 DOI: 10.1101/gad.1929210] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiomyocyte proliferation is high in early development and decreases progressively with gestation, resulting in the lack of a robust cardiomyocyte proliferative response in the adult heart after injury. Little is understood about how both cell-autonomous and nonautonomous signals are integrated to regulate the balance of cardiomyocyte proliferation during development. In this study, we show that a single transcription factor, Foxp1, can control the balance of cardiomyocyte proliferation during development by targeting different pathways in the endocardium and myocardium. Endocardial loss of Foxp1 results in decreased Fgf3/Fgf16/Fgf17/Fgf20 expression in the heart, leading to reduced cardiomyocyte proliferation. This loss of myocardial proliferation can be rescued by exogenous Fgf20, and is mediated, in part, by Foxp1 repression of Sox17. In contrast, myocardial-specific loss of Foxp1 results in increased cardiomyocyte proliferation and decreased differentiation, leading to increased myocardial mass and neonatal demise. We show that Nkx2.5 is a direct target of Foxp1 repression, and Nkx2.5 expression is increased in Foxp1-deficient myocardium. Moreover, transgenic overexpression of Nkx2.5 leads to increased cardiomyocyte proliferation and increased ventricular mass, similar to the myocardial-specific loss of Foxp1. These data show that Foxp1 coordinates the balance of cardiomyocyte proliferation and differentiation through cell lineage-specific regulation of Fgf ligand and Nkx2.5 expression.
Collapse
Affiliation(s)
- Yuzhen Zhang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Campbell AJ, Lyne L, Brown PJ, Launchbury RJ, Bignone P, Chi J, Roncador G, Lawrie CH, Gatter KC, Kusec R, Banham AH. Aberrant expression of the neuronal transcription factorFOXP2in neoplastic plasma cells. Br J Haematol 2010; 149:221-30. [DOI: 10.1111/j.1365-2141.2009.08070.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
65
|
Rayoo M, Yan M, Takano EA, Bates GJ, Brown PJ, Banham AH, Fox SB. Expression of the forkhead box transcription factor FOXP1 is associated with oestrogen receptor alpha, oestrogen receptor beta and improved survival in familial breast cancers. J Clin Pathol 2009; 62:896-902. [DOI: 10.1136/jcp.2009.065169] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background:The role of FOXP1 in sporadic breast cancers has been widely studied but its role in familial breast cancers is yet unexplored.Aims:To investigate FOXP1 expression in different molecular subtypes of familial breast cancers and to correlate its expression with clinicopathological parameters, oestrogen receptors (ER) and survival.Methods:Immunohistochemical staining for FOXP1 was performed in 126 familial breast carcinomas comprising 35 BRCA1, 34 BRCA2 and 57 BRCAX.Results:Nuclear FOXP1 expression ranged from focal weak to widespread strong expression. Expression of FOXP1 was higher in familial breast cancers (54%) compared with sporadic cancers (46%) (p<0.001). There was a significant correlation between FOXP1 with ERα (p = 0.038) and ERβ (p = 0.007) in familial breast cancers. FOXP1 was more highly expressed in familial breast cancers compared with sporadic cancers for luminal (p = 0.021) and basal (p<0.001), but not HER2 and null phenotypes (both p>0.05). The absence of FOXP1 expression was associated with a shorter relapse-free (p = 0.025) and overall survival (p = 0.009) in familial breast cancer. Negativity for FOXP1 was associated with a significantly worse overall survival in BRCA2 cancers (p = 0.021) and there was a non-significant separation of the survival curves for BRCA1 cancers (p = 0.183). No differences in survival were seen for BRCAX cancers (p = 0.762).Conclusion:Results suggest that FOXP1 demonstrates different expression patterns in familial breast cancers than sporadic tumours, even in tumours showing similar phenotypes. They also suggest a different role of FOXP1 as a tumour suppressor in familial tumours, which is unrelated to ER expression and may impact on therapeutic options.
Collapse
|
66
|
Nowak D, Le Toriellec E, Stern MH, Kawamata N, Akagi T, Dyer MJ, Hofmann WK, Ogawa S, Koeffler HP. Molecular allelokaryotyping of T-cell prolymphocytic leukemia cells with high density single nucleotide polymorphism arrays identifies novel common genomic lesions and acquired uniparental disomy. Haematologica 2009; 94:518-27. [PMID: 19278963 DOI: 10.3324/haematol.2008.001347] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND T-cell prolymphocytic leukemia is a rare aggressive lymphoproliferative disease with a mature T-cell phenotype and characteristic genomic lesions such as inv(14)(q11q34), t(14;14)(q11;q32) or t(X;14)(q28;q11), mutation of the ATM gene on chromosome 11 and secondary alterations such as deletions of chromosome 8p and duplications of 8q. DESIGN AND METHODS We analyzed malignant cells from 18 patients with T-cell prolymphocytic leukemia using high density 250K single nucleotide polymorphism arrays and molecular allelokaryotyping to refine understanding of known alterations and identify new target genes. RESULTS Our analyses revealed that characteristic disruptions of chromosome 14 are frequently unbalanced. In the commonly deleted region on chromosome 11, we found recurrent microdeletions targeting the microRNA 34b/c and the transcription factors ETS1 and FLI1. On chromosome 8, we identified genes such as PLEKHA2, NBS1, NOV and MYST3 to be involved in breakpoints. New recurrent alterations were identified on chromosomes 5p, 12p, 13q, 17 and 22 with a common region of acquired uniparental disomy in four samples on chromosome 17q. Single nucleotide polymorphism array results were confirmed by direct sequencing and quantitative real-time polymerase chain reaction. CONCLUSIONS The first high density single nucleotide polymorphism array allelokaryotyping of T-cell prolymphocytic leukemia genomes added substantial new details about established alterations in this disease and moreover identified numerous new potential target genes in common breakpoints, deletions and regions of acquired uniparental disomy.
Collapse
Affiliation(s)
- Daniel Nowak
- Division of Hematology and Oncology, Cedars Sinai Medical Center, UCLA School of Medicine, 8700 Beverly Blvd, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Generali D, Buffa FM, Berruti A, Brizzi MP, Campo L, Bonardi S, Bersiga A, Allevi G, Milani M, Aguggini S, Papotti M, Dogliotti L, Bottini A, Harris AL, Fox SB. Phosphorylated ERα, HIF-1α, and MAPK Signaling As Predictors of Primary Endocrine Treatment Response and Resistance in Patients With Breast Cancer. J Clin Oncol 2009; 27:227-34. [DOI: 10.1200/jco.2007.13.7083] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose We aimed to identify signaling pathways involved in the response and resistance to aromatase inhibitor therapy in patients with breast cancer. Patients and Methods One hundred fourteen women with T2-4 N0-1, estrogen receptor (ER) α–positive tumors were randomly assigned to neoadjuvant letrozole or letrozole plus metronomic cyclophosphamide. Twenty-four tumor proteins involved in apoptosis, cell survival, hypoxia, angiogenesis, growth factor, and hormone signaling were assessed by immunohistochemistry in pretreatment samples (eg, caspase 3, phospho- mammalian target of rapamycin, hypoxia-inducible factor 1α [HIF-1α], vascular endothelial growth factor, mitogen-activated protein kinase [MAPK], phosphorylated epidermal growth factor receptor, phosphorylated ERα [pERα]). A multivariate generalized linear regression approach was applied using a penalized least-square minimization to perform variable selection and regularization. Ten-fold cross-validation and iterative leave-one-out were employed to validate and test the model, respectively. Tumor size, nodal status, age, tumor grade, histological type, and treatment were included in the analysis. Results Ninety-one patients (81%) attained a disease response, 48 achieved a complete clinical response (43%) whereas 22 did not respond (19%). Increased pERα and decreased p44/42 MAPK were significant factors for complete response to treatment in all leave-one-out iterations. Increased p44/42 MAPK and HIF-1α were significant factors for treatment resistance in all leave-one-out iterations. There was no significant interaction between these variables and treatment. Conclusion Activated ERα form was an independent factor for sensitivity to chemoendocrine treatment, whereas HIF-1α and p44/42 MAPK were independent factors for resistance. Although further confirmatory analyses are needed, these findings have clear potential implications for future strategies in the management of clinical trials with aromatase inhibitors in the breast cancer.
Collapse
Affiliation(s)
- Daniele Generali
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Francesca M. Buffa
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Alfredo Berruti
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Maria P. Brizzi
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Leticia Campo
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Simone Bonardi
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Alessandra Bersiga
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Giovanni Allevi
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Manuela Milani
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Sergio Aguggini
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Mauro Papotti
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Luigi Dogliotti
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Alberto Bottini
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Adrian L. Harris
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| | - Stephen B. Fox
- From the Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine; Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Unità di Patologia Mammaria –Breast Cancer Unit and Anatomia Patologica, Azienda Instituti Ospitalieri di Cremona, Cremona; Anatomia Patologica; Oncologia Medica, Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Azienda Ospedaliera San Luigi di Orbassano, Orbassano, Italy; Peter
| |
Collapse
|
68
|
Thomas R, Duke SE, Karlsson EK, Evans A, Ellis P, Lindblad-Toh K, Langford CF, Breen M. A genome assembly-integrated dog 1 Mb BAC microarray: a cytogenetic resource for canine cancer studies and comparative genomic analysis. Cytogenet Genome Res 2008; 122:110-21. [PMID: 19096206 PMCID: PMC2874680 DOI: 10.1159/000163088] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2008] [Indexed: 12/17/2022] Open
Abstract
Molecular cytogenetic studies have been instrumental in defining the nature of numerical and structural chromosome changes in human cancers, but their significance remains to be fully understood. The emergence of high quality genome assemblies for several model organisms provides exciting opportunities to develop novel genome-integrated molecular cytogenetic resources that now permit a comparative approach to evaluating the relevance of tumor-associated chromosome aberrations, both within and between species. We have used the dog genome sequence assembly to identify a framework panel of 2,097 bacterial artificial chromosome (BAC) clones, selected at intervals of approximately one megabase. Each clone has been evaluated by multicolor fluorescence in situ hybridization (FISH) to confirm its unique cytogenetic location in concordance with its reported position in the genome assembly, providing new information on the organization of the dog genome. This panel of BAC clones also represents a powerful cytogenetic resource with numerous potential applications. We have used the clone set to develop a genome-wide microarray for comparative genomic hybridization (aCGH) analysis, and demonstrate its application in detection of tumor-associated DNA copy number aberrations (CNAs) including single copy deletions and amplifications, regional aneuploidy and whole chromosome aneuploidy. We also show how individual clones selected from the BAC panel can be used as FISH probes in direct evaluation of tumor karyotypes, to verify and explore CNAs detected using aCGH analysis. This cytogenetically validated, genome integrated BAC clone panel has enormous potential for aiding gene discovery through a comparative approach to molecular oncology.
Collapse
Affiliation(s)
- R Thomas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, Raleigh, NC 27606, USA
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Loss of heterozygosity and copy number abnormality in clear cell renal cell carcinoma discovered by high-density affymetrix 10K single nucleotide polymorphism mapping array. Neoplasia 2008; 10:634-42. [PMID: 18592004 DOI: 10.1593/neo.08160] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 04/15/2008] [Accepted: 04/18/2008] [Indexed: 12/25/2022] Open
Abstract
Genetic aberrations are crucial in renal tumor progression. In this study, we describe loss of heterozygosity (LOH) and DNA-copy number abnormalities in clear cell renal cell carcinoma (cc-RCC) discovered by genome-wide single nucleotide polymorphism (SNP) arrays. Genomic DNA from tumor and normal tissue of 22 human cc-RCCs was analyzed on the Affymetrix GeneChip Human Mapping 10K Array. The array data were validated by quantitative polymerase chain reaction and immunohistochemistry. Reduced DNA copy numbers were detected on chromosomal arm 3p in 91%, on chromosome 9 in 32%, and on chromosomal arm 14q in 36% of the tumors. Gains were detected on chromosomal arm 5q in 45% and on chromosome 7 in 32% of the tumors. Copy number abnormalities were found not only in FHIT and VHL loci, known to be involved in renal carcinogenesis, but also in regions containing putative new tumor suppressor genes or oncogenes. In addition, microdeletions were detected on chromosomes 1 and 6 in genes with unknown impact on renal carcinogenesis. In validation experiments, abnormal protein expression of FOXP1 (on 3p) was found in 90% of tumors (concordance with SNP array data in 85%). As assessed by quantitative polymerase chain reaction, PARK2 and PACRG were down-regulated in 57% and 100%, respectively, and CSF1R was up-regulated in 69% of the cc-RCC cases (concordance with SNP array data in 57%, 33%, and 38%). Genome-wide SNP array analysis not only confirmed previously described large chromosomal aberrations but also detected novel microdeletions in genes potentially involved in tumor genesis of cc-RCC.
Collapse
|
70
|
Takayama K, Horie-Inoue K, Ikeda K, Urano T, Murakami K, Hayashizaki Y, Ouchi Y, Inoue S. FOXP1 is an androgen-responsive transcription factor that negatively regulates androgen receptor signaling in prostate cancer cells. Biochem Biophys Res Commun 2008; 374:388-93. [DOI: 10.1016/j.bbrc.2008.07.056] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 07/10/2008] [Indexed: 10/21/2022]
|
71
|
Brown PJ, Kagaya R, Banham AH. Characterization of human FOXP1 isoform 2, using monoclonal antibody 4E3-G11, and intron retention as a tissue-specific mechanism generating a novel FOXP1 isoform. Histopathology 2008; 52:632-7. [DOI: 10.1111/j.1365-2559.2008.02990.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
72
|
Lulu Fu, Girling JE, Rogers PAW. Expression of Fox Head Protein 1 in Human Eutopic Endometrium and Endometriosis. Reprod Sci 2008; 15:243-52. [DOI: 10.1177/1933719107312626] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Lulu Fu
- Centre for Women's Health Research, Monash University Department of Obstetrics and Gynaecology, and Monash Institute of Medical Research, Monash Medical Centre, Clayton, Victoria, Australia
| | - Jane E. Girling
- Centre for Women's Health Research, Monash University Department of Obstetrics and Gynaecology, and Monash Institute of Medical Research, Monash Medical Centre, Clayton, Victoria, Australia,
| | - Peter A. W. Rogers
- Centre for Women's Health Research, Monash University Department of Obstetrics and Gynaecology, and Monash Institute of Medical Research, Monash Medical Centre, Clayton, Victoria, Australia
| |
Collapse
|
73
|
Potentially oncogenic B-cell activation-induced smaller isoforms of FOXP1 are highly expressed in the activated B cell-like subtype of DLBCL. Blood 2007; 111:2816-24. [PMID: 18077790 DOI: 10.1182/blood-2007-09-115113] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The FOXP1 forkhead transcription factor is targeted by recurrent chromosome translocations in several subtypes of B-cell non-Hodgkin lymphomas, where high-level FOXP1 protein expression has been linked to a poor prognosis. Western blotting studies of diffuse large B-cell lymphoma (DLBCL) cell lines unexpectedly identified the atypical high-level expression of 2 smaller, 60 to 65 kDa, FOXP1 isoforms in all 5 of those with the activated B cell (ABC)-like DLBCL subtype and in a subgroup of primary DLBCL. The anti-FOXP1 (JC12) monoclonal antibody cannot distinguish FOXP1 isoforms by immunohistochemistry, a finding that may be clinically relevant as high-level expression of the full-length FOXP1 protein was observed in some germinal center-derived DLBCLs. ABC-like DLBCL-derived cell lines were observed to express 2 novel, alternatively spliced FOXP1 mRNA isoforms, encoding N-terminally truncated proteins. These transcripts and the smaller protein isoforms were induced as a consequence of normal B-cell activation, which thus represents an additional mechanism for up-regulating FOXP1 expression in lymphomas. The expression of potentially oncogenic smaller FOXP1 isoforms may resolve the previously contradictory findings that FOXP1 represents a favorable prognostic marker in breast cancer and an adverse risk factor in B-cell lymphomas.
Collapse
|
74
|
Expression of the forkhead transcription factor FOXP1 is associated with that of estrogen receptor-beta in primary invasive breast carcinomas. Breast Cancer Res Treat 2007; 111:453-9. [PMID: 18026833 DOI: 10.1007/s10549-007-9812-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Accepted: 10/31/2007] [Indexed: 12/26/2022]
Abstract
We previously identified a correlation between estrogen receptor alpha (ERalpha) and the candidate tumour suppressor gene Forkhead Box P1 (FOXP1), whose nuclear protein expression in breast tumours was associated with improved patient survival. However, the expression pattern of FOXP1 in normal breast tissue is more reminiscent of the second receptor, ERbeta, which has an emerging role as a tumour suppressor in breast cancer and critically may underlie the ability of some ERalpha-negative tumours to respond to tamoxifen. In a series of 283 breast cancers, in which ERalpha-positive tumours were treated with tamoxifen, the nuclear expression of ERbeta correlated significantly with ERalpha (p = 0.004), low-tumour grade (p = 0.008) and nuclear FOXP1 (p = 0.01). High-grade tumours exhibited significantly more cytoplasmic ERbeta than the low-grade tumours (p = 0.006). Regression analysis demonstrated that FOXP1 expression was most closely related to nuclear ERbeta (p = 0.021). Neither, nuclear or cytoplasmic ERbeta expression demonstrated prognostic significance. FOXP1 is not estrogen regulated and silencing FOXP1 expression, using siRNA, did not affect ERalpha, ERbeta or progesterone receptor expression, suggesting ER and FOXP1 co-expression may reflect a common regulatory mechanism.
Collapse
|
75
|
Koon HB, Ippolito GC, Banham AH, Tucker PW. FOXP1: a potential therapeutic target in cancer. Expert Opin Ther Targets 2007; 11:955-65. [PMID: 17614763 PMCID: PMC4282158 DOI: 10.1517/14728222.11.7.955] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Forkhead Box P1 (FOXP1) is a member of the FOX family of transcription factors which have a broad range of functions. Foxp1 is widely expressed and has been shown to have a role in cardiac, lung and lymphocyte development. FOXP1 is targeted by recurrent chromosome translocations and its overexpression confers a poor prognosis in a number of types of lymphomas, suggesting it may function as an oncogene. In contrast, FOXP1 localises to a tumour suppressor locus at 3p14.1 and loss of FOXP1 expression in breast cancer is associated with a worse outcome, suggesting FOXP1 may function as a tumour suppressor in other tissue types. These data suggest that FOXP1 may not only be useful in prognosis but also may be used to develop FOXP1-directed therapeutic strategies.
Collapse
Affiliation(s)
- Henry B Koon
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, MA, USA
| | - Gregory C Ippolito
- University of Texas, Department of Molecular Genetics and Microbiology, Institute of Cellular and Molecular Biology, 1 University Station A5000, Austin, Texas 78712-0162, USA
| | - Alison H Banham
- University of Oxford, Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, Headington, Oxfordshire, OX3 9DU, UK
| | - Philip W Tucker
- University of Texas, Department of Molecular Genetics and Microbiology, Institute of Cellular and Molecular Biology, 1 University Station A5000, Austin, Texas 78712-0162, USA
- Author for correspondence Tel: +1 512 475 7705; Fax: +1 512 475 7707;
| |
Collapse
|
76
|
Koon HB, Ippolito GC, Banham AH, Tucker PW. FOXP1: a potential therapeutic target in cancer. Expert Opin Ther Targets 2007. [PMID: 17614763 DOI: 10.1517/14728222.11.7.95] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Forkhead Box P1 (FOXP1) is a member of the FOX family of transcription factors which have a broad range of functions. Foxp1 is widely expressed and has been shown to have a role in cardiac, lung and lymphocyte development. FOXP1 is targeted by recurrent chromosome translocations and its overexpression confers a poor prognosis in a number of types of lymphomas, suggesting it may function as an oncogene. In contrast, FOXP1 localises to a tumour suppressor locus at 3p14.1 and loss of FOXP1 expression in breast cancer is associated with a worse outcome, suggesting FOXP1 may function as a tumour suppressor in other tissue types. These data suggest that FOXP1 may not only be useful in prognosis but also may be used to develop FOXP1-directed therapeutic strategies.
Collapse
Affiliation(s)
- Henry B Koon
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, MA, USA
| | | | | | | |
Collapse
|
77
|
Thomas R, Duke SE, Bloom SK, Breen TE, Young AC, Feiste E, Seiser EL, Tsai PC, Langford CF, Ellis P, Karlsson EK, Lindblad-Toh K, Breen M. A Cytogenetically Characterized, Genome-Anchored 10-Mb BAC Set and CGH Array for the Domestic Dog. J Hered 2007; 98:474-84. [PMID: 17702974 DOI: 10.1093/jhered/esm053] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The generation of a 7.5x dog genome assembly provides exciting new opportunities to interpret tumor-associated chromosome aberrations at the biological level. We present a genomic microarray for array comparative genomic hybridization (aCGH) analysis in the dog, comprising 275 bacterial artificial chromosome (BAC) clones spaced at intervals of approximately 10 Mb. Each clone has been positioned accurately within the genome assembly and assigned to a unique chromosome location by fluorescence in situ hybridization (FISH) analysis, both individually and as chromosome-specific BAC pools. The microarray also contains clones representing the dog orthologues of 31 genes implicated in human cancers. FISH analysis of the 10-Mb BAC clone set indicated excellent coverage of each dog chromosome by the genome assembly. The order of clones was consistent with the assembly, but the cytogenetic intervals between clones were variable. We demonstrate the application of the BAC array for aCGH analysis to identify both whole and partial chromosome imbalances using a canine histiocytic sarcoma case. Using BAC clones selected from the array as probes, multicolor FISH analysis was used to further characterize these imbalances, revealing numerous structural chromosome rearrangements. We outline the value of a combined aCGH/FISH approach, together with a well-annotated dog genome assembly, in canine and comparative cancer studies.
Collapse
Affiliation(s)
- Rachael Thomas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 4700 Hillsborough Street, Raleigh, NC 27606, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Banham AH, Boddy J, Launchbury R, Han C, Turley H, Malone PR, Harris AL, Fox SB. Expression of the forkhead transcription factor FOXP1 is associated both with hypoxia inducible factors (HIFs) and the androgen receptor in prostate cancer but is not directly regulated by androgens or hypoxia. Prostate 2007; 67:1091-8. [PMID: 17477366 DOI: 10.1002/pros.20583] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND FOXP1 is a member of the winged helix or forkhead transcription factors. Recent studies have indicated possible roles for FOXP1 as a candidate tumor suppressor gene and a potential estrogen receptor (ER) co-regulator in the development of breast cancer. This study investigated whether FOXP1 has a similar relationship to the androgen receptor (AR) in prostate cancer and how these factors relate to the presence of hypoxia. METHODS FOXP1, the AR and various hypoxia-regulated proteins (HIF-1alpha, HIF-2alpha, and VEGF) were measured with immunohistochemistry using a tissue microarray constructed from 167 archival radical prostatectomies. Statistical analyses compared the co-expression of these factors both with each other and conventional parameters including patient age, pre-operative prostate specific antigen (PSA), post-operative Gleason score, capsular invasion, surgical margin status, tumor volume, and PSA recurrence. The influence of hypoxia, dihydrotestosterone, and the AR blocker Casodex was investigated in prostate cell lines VCaP and LNCaP in vitro. RESULTS Expression of nuclear FOXP1 was significantly positively correlated with AR (P = 0.0001), hypoxia inducible factor 1alpha (HIF-1alpha) (P = 0.01), HIF-2alpha (P = 0.0001), and vascular endothelial growth factor (VEGF) (P = 0.007) expression. A positive significant relationship was also identified with the post-operative Gleason score (P = 0.03) but not with the other variables, including PSA recurrence (P > 0.05). There was no significant change in expression in FOXP1 protein levels under conditions of hypoxia (0.1%), dihydrotestosterone stimulation (10 or 100 nM), or androgen blockade with Casodex (1, 10, or 50 microM). CONCLUSION These findings suggest that there may be a hormonal and hypoxia independent regulatory mechanism coordinating the expression of HIFs, the AR, and FOXP1 in prostate tumors.
Collapse
Affiliation(s)
- Alison H Banham
- Nuffield Department Clinical Laboratory Sciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Schwarzbraun T, Ofner L, Gillessen-Kaesbach G, Schaperdoth B, Preisegger KH, Windpassinger C, Wagner K, Petek E, Kroisel PM. A new 3p interstitial deletion including the entireMITF gene causes a variation of Tietz/Waardenburg type IIA syndromes. Am J Med Genet A 2007; 143A:619-24. [PMID: 17318840 DOI: 10.1002/ajmg.a.31627] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Thomas Schwarzbraun
- Institute of Medical Biology and Human Genetics, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol 2006; 24:5373-80. [PMID: 17135638 DOI: 10.1200/jco.2006.05.9584] [Citation(s) in RCA: 884] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To assess the clinical significance of tumor-infiltrating FOXP3-positive regulatory T cells (TR) in breast cancer patients with long-term follow-up. PATIENTS AND METHODS FOXP3-positive TR were detected by immunohistochemistry with our new, extensively characterized FOXP3 monoclonal antibody, 236A/E7. Numbers of FOXP3-positive lymphocytes in tissue microarray cores from pure ductal carcinoma in situ (DCIS; n = 62), invasive breast cancer (n = 237) or from comparable areas of normal terminal duct lobular breast tissue (n = 10) were determined. A median cutoff of > or = 15 defined patients with high numbers of TR. RESULTS TR numbers were significantly higher in in situ and invasive breast carcinomas than in normal breast; invasive tumors have significantly higher numbers than DCIS (P = .001). High numbers of FOXP3-positive TR identified patients with DCIS at increased risk of relapse (P = .04) and patients with invasive tumors with both shorter relapse-free (P = .004) and overall survival (P = .007). High TR numbers were present in high-grade tumors (P < or = .001), in patients with lymph node involvement (P = .01), and in estrogen receptor (ER) -negative tumors (P = .001). Importantly, high numbers of TR within ER-positive tumors identified high-risk patients (P = .005). Unlike conventional clinicopathologic factors, high numbers of FOXP3-positive TR can identify patients at risk of relapse after 5 years. CONCLUSION These findings indicate that quantification of FOXP3-positive TR in breast tumors is valuable for assessing disease prognosis and progression, and that TR are an important therapeutic target for breast cancer. FOXP3-positive TR represent a novel marker for identifying late-relapse patients who may benefit from aromatase therapy after standard tamoxifen treatment.
Collapse
Affiliation(s)
- Gaynor J Bates
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital; Cancer Research UK Molecular Oncology Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
81
|
Li B, Samanta A, Song X, Furuuchi K, Iacono KT, Kennedy S, Katsumata M, Saouaf SJ, Greene MI. FOXP3 ensembles in T-cell regulation. Immunol Rev 2006; 212:99-113. [PMID: 16903909 DOI: 10.1111/j.0105-2896.2006.00405.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our recent studies have identified dynamic protein ensembles containing forkhead box protein 3 (FOXP3) that provide insight into the molecular complexity of suppressor T-cell activities, and it is our goal to determine how these ensembles regulate FOXP3's transcriptional activity in vivo. In this review, we summarize our current understanding of how FOXP3 expression is induced and how FOXP3 functions in vivo as a transcriptional regulator by assembling a multisubunit complex involved in histone modification as well as chromatin remodeling.
Collapse
Affiliation(s)
- Bin Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Schön C, Wochnik A, Rössner A, Donow C, Knöchel W. The FoxP subclass in Xenopus laevis development. Dev Genes Evol 2006; 216:641-6. [PMID: 16609867 DOI: 10.1007/s00427-006-0073-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Accepted: 03/20/2006] [Indexed: 11/24/2022]
Abstract
We have investigated the sequences and the expression patterns of different members of the Xenopus laevis FoxP gene subfamily during embryogenesis. Low stringency hybridisation of a tadpole cDNA library with an xlFoxP2 fragment led to the isolation of several splice variants of xlFoxP1, xlFoxP2 and xlFoxP4. These variants do not only differ by utilisation of different leader exons, but also by alternative usage of coding exons thereby leading to functional alterations. For xlFoxP1b, we show that insertion of an additional exon disrupts binding to the co-repressor C-terminal binding protein1. Temporal and spatial expression patterns of xlFoxP2 and xlFoxP4 were analysed by RT-PCR and by whole mount in situ hybridisation. xlFoxP2 transcripts are detected from mid-gastrula to late tadpole stages and are found to be localised to pronephros, branchial arches and distinct structures of the hind-, mid- and forebrain, including the ciliary marginal zone of the retina. xlFoxP4 RNA is already present in early cleavage stage embryos and accumulates from midblastula until the end of embryogenesis. Localised expression is found within the anterior neural fold, in the mid- and hindbrain, in the branchial arches as well as in the pancreas.
Collapse
Affiliation(s)
- Christian Schön
- Abteilung Biochemie, Universität Ulm, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | | | | | | | | |
Collapse
|
83
|
Piva R, Penolazzi L, Lambertini E, Giordano S, Gambari R. Induction of apoptosis of human primary osteoclasts treated with a transcription factor decoy mimicking a promoter region of estrogen receptor alpha. Apoptosis 2006; 10:1079-94. [PMID: 16151641 DOI: 10.1007/s10495-005-0618-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this paper we investigated how the increase of human estrogen receptor alfa (ERalpha) gene expression may affect breast, osteoblast and osteoclast cells. Increase of ERalpha expression was obtained by interfering with the activity of a negative transcription factor and by removing it with a short and powerful decoy oligonucleotide (RA4-3') mimicking a region of distal promoter C of ERalpha gene. We provide evidence that this decoy was able to induce apoptosis in osteoclasts, but not in osteoblasts and in breast cancer cells, in an estrogen dependent manner. This effect was associated with increase of the levels of Caspase 3 and Fas receptor. Since ERalpha is important in the transcription of different genes and is involved in several pathological processes, including neoplastic and osteopenic diseases, our findings may be of relevance for a possible new therapeutical approach of such diseases.
Collapse
Affiliation(s)
- R Piva
- Department of Biochemistry and Molecular Biology, Ferrara University, Via Fossato di Mortara n.74, 44100 Ferrara, Italy
| | | | | | | | | |
Collapse
|
84
|
Giatromanolaki A, Koukourakis MI, Sivridis E, Gatter KC, Harris AL, Banham AH. Loss of expression and nuclear/cytoplasmic localization of the FOXP1 forkhead transcription factor are common events in early endometrial cancer: relationship with estrogen receptors and HIF-1alpha expression. Mod Pathol 2006; 19:9-16. [PMID: 16258506 DOI: 10.1038/modpathol.3800494] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The FOXP1 gene has been identified as a new member of the winged helix family of transcription factors that have important roles in cellular transformation, differentiation and proliferation. In this study, we examined the expression of FOXP1 in the normal and malignant endometrium (stage I endometrioid adenocarcinoma cases), showing a frequent deregulation of its expression in cancer. Proliferative endometrium showed predominantly nuclear localization of FOXP1, while exclusively weak cytoplasmic staining was present in the secretory phase. Loss of nuclear expression was the most striking event in endometrial adenocarcinoma. Nuclear expression ranged from 0 to 20% (median 0%). Cytoplasmic expression was noted more frequently, ranging from 0 to 90% of cancer cells (median 30%). Overall, 24/82 cases (29.3%) were observed to lack both nuclear and cytoplasmic FOXP1 expression. Tumors with exclusively cytoplasmic expression of FOXP1 were linked with deep myometrial invasion and hypoxia-inducible factors 1alpha (HIF-1alpha) expression. On the other hand, the presence of nuclear FOXP1 expression was significantly linked with ER-alpha reactivity. Survival analysis did not reveal significant differences among patients grouped by FOXP1 expression, presumably due to the high curability of stage I disease. This study provides evidence on pathways to be investigated to elucidate the interplay between FOXP1, ER-alpha and HIF-1alpha in hormone dependent cancers.
Collapse
|
85
|
Scharff C, Haesler S. An evolutionary perspective on FoxP2: strictly for the birds? Curr Opin Neurobiol 2005; 15:694-703. [PMID: 16266802 DOI: 10.1016/j.conb.2005.10.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2005] [Accepted: 10/21/2005] [Indexed: 10/25/2022]
Abstract
FoxP2 mutations in humans are associated with a disorder that affects both the comprehension of language and its production, speech. This discovery provided the first opportunity to analyze the genetics of language with molecular and neurobiological tools. The amino acid sequence and the neural expression pattern of FoxP2 are extremely conserved, from reptile to man. This suggests an important role for FoxP2 in vertebrate brains, regardless of whether they support imitative vocal learning or not. Its expression pattern pinpoints neural circuits that might have been crucial for the evolution of speech and language, including the basal ganglia and the cerebellum. Recent studies in songbirds show that during times of song plasticity FoxP2 is upregulated in a striatal region essential for song learning. This suggests that FoxP2 plays important roles both in the development of neural circuits and in the postnatal behaviors they mediate.
Collapse
Affiliation(s)
- Constance Scharff
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195 Berlin, Germany; Freie Universität Berlin, Department of Animal Behavior, Grunewaldstrasse 34, 12165 Berlin, Germany.
| | | |
Collapse
|
86
|
Lambertini E, Penolazzi L, Magaldi S, Giordano S, Senno LD, Piva R. Transcription factor decoy against promoter C of estrogen receptor α gene induces a functional ERα protein in breast cancer cells. Breast Cancer Res Treat 2005; 92:125-32. [PMID: 15986121 DOI: 10.1007/s10549-005-2413-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study addresses the hypothesis that transfection of oligonucleotide mimicking a negative regulatory sequence of promoter C of estrogen receptor alpha (ER alpha) gene is sufficient for its re-expression in ER-negative human cancer cell lines. Even if the negative transcription regulator subtracted by the transcription factor decoy is not yet been identified, we demonstrated that after this decoy treatment, the cells produced a functional ER alpha protein able to respond to 17-beta-estradiol and to transactivate a transfected estrogen response element (ERE)-regulated reporter gene. The effects of reactivated ER alpha protein and its estrogen dependence on endogenous target gene expression level, such as ER beta, have been also assessed. The proliferation of the cells transfected with low levels of decoy was significantly increased by estrogen and not by tamoxifen, suggesting that the levels of reactivated ER alpha in these decoy conditions confers a certain hormone sensitivity. On the contrary, high-level expression of ER alpha obtained at high doses of transfected decoy molecule produced a progressive decrease of cell proliferation. Since ER alpha is important in the transcription of different genes and its loss is involved in several pathological processes including neoplastic and chronic diseases, our findings may be of relevance for a possible new therapeutical approach of such diseases.
Collapse
Affiliation(s)
- Elisabetta Lambertini
- Department of Biochemistry and Molecular Biology, Ferrara University, 44100 Ferrara, Italy
| | | | | | | | | | | |
Collapse
|