1
|
Pezzuto F, Lunardi F, Vedovelli L, Fortarezza F, Urso L, Grosso F, Ceresoli GL, Kern I, Vlacic G, Faccioli E, Schiavon M, Gregori D, Rea F, Pasello G, Calabrese F. P14/ARF-Positive Malignant Pleural Mesothelioma: A Phenotype With Distinct Immune Microenvironment. Front Oncol 2021; 11:653497. [PMID: 33828993 PMCID: PMC8019896 DOI: 10.3389/fonc.2021.653497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The CDKN2A gene plays a central role in the pathogenesis of malignant pleural mesothelioma (MPM). The gene encodes for two tumor suppressor proteins, p16/INK4A and p14/ARF, frequently lost in MPM tumors. The exact role of p14/ARF in MPM and overall its correlation with the immune microenvironment is unknown. We aimed to determine whether there is a relationship between p14/ARF expression, tumor morphological features, and the inflammatory tumor microenvironment. METHODS Diagnostic biopsies from 76 chemo-naive MPMs were evaluated. Pathological assessments of histotype, necrosis, inflammation, grading, and mitosis were performed. We evaluated p14/ARF, PD-L1 (tumor proportion score, TPS), and Ki-67 (percentage) by immunohistochemistry. Inflammatory cell components (CD3+, CD4+, CD8+ T lymphocytes; CD20+ B-lymphocytes; CD68+ and CD163+ macrophages) were quantified as percentages of positive cells, distinguishing between intratumoral and peritumoral areas. The expression of p14/ARF was associated with several clinical and pathological characteristics. A random forest-based machine-learning algorithm (Boruta) was implemented to identify which variables were associated with p14/ARF expression. RESULTS p14/ARF was evaluated in 68 patients who had a sufficient number of tumor cells. Strong positivity was detected in 14 patients (21%) (11 epithelioid and 3 biphasic MPMs). At univariate analysis, p14/ARF-positive epithelioid mesotheliomas showed higher nuclear grade (G3) (p = 0.023) and higher PD-L1 expression (≥50%) (p = 0.042). The percentages of CD4 and CD163 in peritumoral areas were respectively higher and lower in p14/ARF positive tumors but did not reach statistical significance with our sample size (both p = 0.066). The Boruta algorithm confirmed the predictive value of PD-L1 percentage for p14/ARF expression in all histotypes. CONCLUSIONS p14/ARF-positive epithelioid mesotheliomas may mark a more aggressive pathological phenotype (higher nuclear grade and PD-L1 expression). Considering the results regarding the tumor immune microenvironment, p14/ARF-negative tumors seem to have an immune microenvironment less sensitive to immune checkpoint inhibitors, being associated with low PD-L1 and CD4 expression, and high CD163 percentage. The association between p14/ARF-positive MPMs and PD-L1 expression suggests a possible interaction of the two pathways. Confirmation of our preliminary results could be important for patient selection and recruitment in future clinical trials with anticancer immunotherapy.
Collapse
Affiliation(s)
- Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Loredana Urso
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Federica Grosso
- Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Mesothelioma and Rare Cancer Unit, Alessandria, Italy
| | | | - Izidor Kern
- Pathology Laboratory, University Clinic Golnik, Golnik, Slovenia
| | - Gregor Vlacic
- Pathology Laboratory, University Clinic Golnik, Golnik, Slovenia
| | - Eleonora Faccioli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Giulia Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Department of Oncology, Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
2
|
Zhu C, Hu H, Li J, Wang J, Wang K, Sun J. Identification of key differentially expressed genes and gene mutations in breast ductal carcinoma in situ using RNA-seq analysis. World J Surg Oncol 2020; 18:52. [PMID: 32156290 PMCID: PMC7063758 DOI: 10.1186/s12957-020-01820-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The aim of this study was to identify the key differentially expressed genes (DEGs) and high-risk gene mutations in breast ductal carcinoma in situ (DCIS). METHODS Raw data (GSE36863) were downloaded from the database of Gene Expression Omnibus (GEO), including three DCIS samples (DCIS cell lines MCF10.DCIS, Sum102, and Sum225) and one normal control sample (normal mammary epithelial cell line MCF10A). The DEGs were analyzed using NOIseq and annotated via DAVID. Motif scanning in the promoter region of DEGs was performed via SeqPos. Additionally, single nucleotide variations (SNVs) were identified via GenomeAnalysisTK and SNV risk was assessed via VarioWatch. Mutant genes with a high frequency and risk were validated by RT-PCR analyses. RESULTS Finally, 5391, 7073, and 7944 DEGs were identified in DCIS, Sum102, and Sum22 cell lines, respectively, when compared with MCF10A. VENN analysis of the three cell lines revealed 603 upregulated and 1043 downregulated DEGs, including 16 upregulated and 36 downregulated transcription factor (TF) genes. In addition, six TFs each (e.g., E2F1 and CREB1) were found to regulate the core up- and downregulated DEGs, respectively. Furthermore, SNV detection results revealed 1104 (MCF10.DCIS), 2833 (Sum102), and 1132 (Sum22) mutation sites. Four mutant genes (RWDD4, SDHC, SEPT7, and SFN) with high frequency and risk were identified. The results of RT-PCR analysis as well as bioinformatics analysis consistently demonstrated that the expression of RWDD4, SDHC, SEPT7, and SFN was downregulated in the tumor tissues as compared with that in adjacent non-tumor tissues. CONCLUSIONS The differentially expressed TFs, TFs regulating DEGs (e.g., E2F1 and CREB1), and high-frequency mutant genes (RWDD4, SDHC, SEPT7, and SFN) might play key roles in the pathogenesis of DCIS.
Collapse
Affiliation(s)
- Congyuan Zhu
- Department of General Surgery, the Affiliated Hospital of Jiangnan University (original area of Wuxi Third People's Hospital), No. 585 North Xingyuan Road, Liangxi District, Wuxi, 214002, Jiangsu, China.
| | - Hao Hu
- Department of General Surgery, the Affiliated Hospital of Jiangnan University (original area of Wuxi Third People's Hospital), No. 585 North Xingyuan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Jianping Li
- Department of General Surgery, the Affiliated Hospital of Jiangnan University (original area of Wuxi Third People's Hospital), No. 585 North Xingyuan Road, Liangxi District, Wuxi, 214002, Jiangsu, China.
| | - Jingli Wang
- Department of General Surgery, the Affiliated Hospital of Jiangnan University (original area of Wuxi Third People's Hospital), No. 585 North Xingyuan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Ke Wang
- Department of General Surgery, the Affiliated Hospital of Jiangnan University (original area of Wuxi Third People's Hospital), No. 585 North Xingyuan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Jingqiu Sun
- Department of General Surgery, the Affiliated Hospital of Jiangnan University (original area of Wuxi Third People's Hospital), No. 585 North Xingyuan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| |
Collapse
|
3
|
Aljabery F, Shabo I, Gimm O, Jahnson S, Olsson H. The expression profile of p14, p53 and p21 in tumour cells is associated with disease-specific survival and the outcome of postoperative chemotherapy treatment in muscle-invasive bladder cancer. Urol Oncol 2018; 36:530.e7-530.e18. [PMID: 30539751 DOI: 10.1016/j.urolonc.2018.05.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE We investigated the effects of alterations in the biological markers p14, p53, p21, and p16 in relation to tumour cell proliferation, T-category, N- category, lymphovascular invasion, and the ability to predict prognosis in patients with muscle-invasive bladder cancer (MIBC) treated with cystectomy and, if applicable, chemotherapy. MATERIALS AND METHODS We prospectively studied patients with urinary bladder cancer pathological stage pT1 to pT4 treated with cystectomy, pelvic lymph node dissection and postoperative chemotherapy. Tissue microarrays from paraffin-embedded cystectomy tumour samples were examined for expression of immunostaining of p14, p53, p21, p16 and Ki-67 in relation to other clinical and pathological factors as well as cancer-specific survival. RESULTS The median age of the 110 patients was 70 years (range 51-87 years), and 85 (77%) were male. Pathological staging was pT1 to pT2 (organ-confined) in 28 (25%) patients and pT3 to pT4 (non-organ-confined) in 82 (75%) patients. Lymph node metastases were found in 47 patients (43%). P14 expression was more common in tumours with higher T-stages (P = 0.05). The expression of p14 in p53 negative tumours was associated with a significantly shorter survival time (P=0.003). Independently of p53 expression, p14 expression was associated with an impaired response to chemotherapy (P=0.001). The expression of p21 in p53 negative tumours was associated with significantly decrease levels of tumour cell proliferation detected as Ki-67 expression (P=0.03). CONCLUSIONS The simultaneous expression of the senescence markers involved in the p53-pathway shows a more relevant correlation to the pathological outcome of MIBC than each protein separately. P14 expression in tumours with non-altered (p53-) tumours is associated with poor prognosis. P14 expression is associated with impaired response to chemotherapy. P21 expression is related to decreased tumour cell proliferation.
Collapse
Affiliation(s)
- Firas Aljabery
- Department of Urology, and Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, SE 581 85, Linköping, Sweden.
| | - Ivan Shabo
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, SE 171 77, Stockholm, Sweden. Department of Breast, Endocrine and Sarcoma Surgery, Karolinska University Hospital, SE 171 76, Stockholm, Sweden
| | - Oliver Gimm
- Department of Surgery and Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, SE 581 85, Linköping, Sweden
| | - Staffan Jahnson
- Department of Urology, and Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, SE 581 85, Linköping, Sweden
| | - Hans Olsson
- Department of Pathology and Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, SE 581 85, Linköping, Sweden
| |
Collapse
|
4
|
Pare R, Shin JS, Lee CS. Increased expression of senescence markers p14(ARF) and p16(INK4a) in breast cancer is associated with an increased risk of disease recurrence and poor survival outcome. Histopathology 2016; 69:479-91. [PMID: 26843058 DOI: 10.1111/his.12948] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/31/2016] [Indexed: 11/28/2022]
Abstract
AIMS Breast cancer is a hormonally driven disease. Cellular senescence is an age-related irreversible cell cycle arrest at the G1 phase upon induction. The aim of this study was to characterize the expression patterns of the senescence markers p14(ARF) , p16(INK4a) and p21(WAF1/Cip1) during breast cancer progression in a large patient cohort. METHODS AND RESULTS We conducted a retrospective study of 1080 patients with invasive ductal carcinoma, no special type, over an 11-year period. We performed immunohistochemical staining on tissue microarrays that included normal, benign hyperplasia, ductal carcinoma in situ and invasive ductal carcinoma tissue from each patient. Invasive ductal carcinomas showed higher expression of p14(ARF) and p16(INK4a) but lower expression of p21(WAF1/Cip1) than non-malignant tissues. There were significant correlations of normal, benign, preinvasive and malignant tissues with p14(ARF) , p16(INK4a) and p21(WAF1/Cip1) expression (P < 0.05). Univariate comparison showed a correlation between high p16(INK4a) expression and poor survival (P = 0.000) and an increased risk of relapse (P = 0.000), whereas high p14(ARF) expression correlated only with an increased risk of relapse (P = 0.038). Multivariate analysis showed p16(INK4a) to be an important prognostic factor for overall survival (P = 0.011) and disease-free survival (P = 0.004), with p14(ARF) also being a significant prognostic factor for disease-free survival (P = 0.043). Moreover, patients showing both high p16(INK4a) expression and and high p14(ARF) expression had an adjusted three-fold increased risk of disease recurrence (P < 0.05) and a two-fold increased risk of all-cause-related death (P < 0.05). CONCLUSIONS These finding suggest p16(INK4a) expression and p14(ARF) expression may play an important role in the progression of proliferative breast tissue to invasive cancer, and may be useful as prognostic factors.
Collapse
Affiliation(s)
- Rahmawati Pare
- Discipline of Pathology, School of Medicine, Western Sydney University, Liverpool, NSW, Australia.,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Joo-Shik Shin
- Discipline of Pathology, School of Medicine, Western Sydney University, Liverpool, NSW, Australia.,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia.,Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW, Australia
| | - Cheok Soon Lee
- Discipline of Pathology, School of Medicine, Western Sydney University, Liverpool, NSW, Australia.,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia.,Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW, Australia.,Cancer Pathology, Bosch Institute, University of Sydney, Sydney, NSW, Australia.,South Western Sydney Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
5
|
Urso L, Calabrese F, Favaretto A, Conte P, Pasello G. Critical review about MDM2 in cancer: Possible role in malignant mesothelioma and implications for treatment. Crit Rev Oncol Hematol 2015; 97:220-30. [PMID: 26358421 DOI: 10.1016/j.critrevonc.2015.08.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 07/02/2015] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
The tumor suppressor p53 regulates genes involved in DNA repair, metabolism, cell cycle arrest, apoptosis and senescence. p53 is mutated in about 50% of the human cancers, while in tumors with wild-type p53 gene, the protein function may be lost because of overexpression of Murine Double Minute 2 (MDM2). MDM2 targets p53 for ubiquitylation and proteasomal degradation. p53 reactivation through MDM2 inhibitors seems to be a promising strategy to sensitize p53 wild-type cancer cells to apoptosis. Moreover, additional p53-independent molecular functions of MDM2, such as neoangiogenesis promotion, have been suggested. Thus, MDM2 might be a target for anticancer treatment because of its antiapoptotic and proangiogenetic role. Malignant pleural mesothelioma (MPM) is an aggressive asbestos-related tumor where wild-type p53 might be present. The present review gives a complete landscape about the role of MDM2 in cancer pathogenesis, prognosis and treatment, with particular focus on Malignant Pleural Mesothelioma.
Collapse
Affiliation(s)
- Loredana Urso
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Italy
| | - Adolfo Favaretto
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - PierFranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Giulia Pasello
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy.
| |
Collapse
|
6
|
Subcellular localization of Mdm2 expression and prognosis of breast cancer. Int J Clin Oncol 2013; 19:842-51. [PMID: 24292333 DOI: 10.1007/s10147-013-0639-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/01/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Mouse double minute 2 (Mdm2) is a negative regulator of the tumor suppressor p53. The p53-Mdm2 pathway may play a role in cancer development and prognosis, although the role of p53-Mdm2 in breast cancer remains unclear. METHODS p53 and Mdm2 expressions were determined by immunohistochemistry of tissue microarrays of 865 breast cancer patients who underwent surgery. Clinicopathological characteristics and survival data were analyzed. Mdm2 expression was categorized into four groups: negative, cytoplasm positive, nucleus positive, and concurrent nuclear and cytoplasm positive (N+&C+). RESULTS Negative, cytoplasm-positive, nucleus-positive, and N+&C+ expressions of Mdm2 were observed in 59.2, 10.9, 27.8, and 2.1 % of patients, respectively. The N+&C+ group was associated with larger tumor size, higher grade, negativity for estrogen and progesterone receptors, HER2 positivity, high Ki-67 index, p53 positivity, and triple negative breast cancer. p53-positive tumors showed poorer overall survival than p53-negative tumors. The nucleus-positive and N+&C+ groups showed poorer disease-free survival than the negative and cytoplasm-positive groups. In multivariate analysis, nuclear Mdm2 expression including the N+&C+ group was significantly related to poor prognosis. CONCLUSIONS Concurrent nuclear and cytoplasmic Mdm2 expression was an independent prognostic factor in patients with breast cancer. Subcellular localization of Mdm2 expression should be considered in the evaluation of Mdm2 in breast cancer.
Collapse
|
7
|
Down-regulation of c9orf86 in human breast cancer cells inhibits cell proliferation, invasion and tumor growth and correlates with survival of breast cancer patients. PLoS One 2013; 8:e71764. [PMID: 23977139 PMCID: PMC3743754 DOI: 10.1371/journal.pone.0071764] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Accepted: 07/06/2013] [Indexed: 11/29/2022] Open
Abstract
C9orf86 which is a novel subfamily within the Ras superfamily of GTPases, is overexpressed in the majority of primary breast tumors. Few functional studies have focused on the C9orf86 protein; therefore, in this study, we explored the role of C9orf86 in breast carcinogenesis. In our study, we found that silencing of C9orf86 by siRNA in MCF-7 and SK-BR-3 cells resulted in suppressed cell proliferation as well as in vitro cell invasion capabilities. Moreover, knockdown of C9orf86 inhibited tumor growth in nude mice. Cell cycle and apoptotic assays showed that the anti-proliferative effect of C9orf86-siRNA was mediated by arresting cells in the G1 phase and promoting apoptosis. In addition, we found that patients with high levels of C9orf86 expression showed a significant trend towards worse survival compared to patients with low C9orf86 expression (P = 0.002). These results provide evidence that C9orf86 represents a novel and clinically useful biomarker for BC patients and plays an important role during the progression of BC.
Collapse
|
8
|
Functional interplay between the DNA-damage-response kinase ATM and ARF tumour suppressor protein in human cancer. Nat Cell Biol 2013; 15:967-77. [DOI: 10.1038/ncb2795] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 05/24/2013] [Indexed: 12/25/2022]
|
9
|
Pare R, Yang T, Shin JS, Lee CS. The significance of the senescence pathway in breast cancer progression. J Clin Pathol 2013; 66:491-5. [PMID: 23539738 DOI: 10.1136/jclinpath-2012-201081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Invasive breast cancer develops through prolonged accumulation of multiple genetic changes. The progression to a malignant phenotype requires overriding of growth inhibition. It is evident that some breast cancers have an inherited basis, and both hereditary and sporadic cancers appear to involve molecular mechanisms that are linked to the cell cycle. Frequently, changes in the molecular pathways with gene deletions, point mutations and/or overexpression of growth factors can be seen in these cancers. Recent evidence also implicates the senescence pathway in breast carcinogenesis. It has a barrier effect towards excessive cellular growth, acting as the regulator of tumour initiation and progression. Later in carcinogenesis, acquisition of the senescence associated secretory phenotype may instead promote tumour progression by stimulating growth and transformation in adjacent cells. This two-edge role of senescence in cancer directs more investigations into the effects of the senescence pathway in the development of malignancy. This review presents the current evidence on the roles of senescence molecular pathways in breast cancer and its progression.
Collapse
Affiliation(s)
- Rahmawati Pare
- Discipline of Pathology, School of Medicine, University of Western Sydney, Liverpool, New South Wales, Australia
| | | | | | | |
Collapse
|
10
|
Bandyopadhyay K, Li P, Gjerset RA. The p14ARF alternate reading frame protein enhances DNA binding of topoisomerase I by interacting with the serine 506-phosphorylated core domain. PLoS One 2013; 8:e58835. [PMID: 23555599 PMCID: PMC3608632 DOI: 10.1371/journal.pone.0058835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/07/2013] [Indexed: 12/27/2022] Open
Abstract
In addition to its well-characterized function as a tumor suppressor, p14ARF (ARF) is a positive regulator of topoisomerase I (topo I), a central enzyme in DNA metabolism and a target for cancer therapy. We previously showed that topo I hyperphosphorylation, a cancer-associated event mediated by elevated levels of the protein kinase CK2, increases topo I activity and the cellular sensitivity to topo I-targeted drugs. Topo I hyperphosphorylation also increases its interaction with ARF. Because the ARF−topo I interaction could be highly relevant to DNA metabolism and cancer treatment, we identified the regions of topo I involved in ARF binding and characterized the effects of ARF binding on topo I function. Using a series of topo I deletion constructs, we found that ARF interacted with the topo I core domain, which encompasses most of the catalytic and DNA-interacting residues. ARF binding increased the DNA relaxation activity of hyperphosphorylated topo I by enhancing its association with DNA, but did not affect the topo I catalytic rate. In cells, ARF promoted the chromatin association of hyperphosphorylated, but not basal phosphorylated, topo I, and increased topo I-mediated DNA nicking under conditions of oxidative stress. The aberrant nicking was found to correlate with increased formation of DNA double-strand breaks, which are precursors of many genome destabilizing events. The results suggest that the convergent actions of oxidative stress and elevated CK2 and ARF levels, which are common features of cancer cells, lead to a dysregulation of topo I that may contribute both to the cellular response to topo I-targeted drugs and to genome instability.
Collapse
Affiliation(s)
- Keya Bandyopadhyay
- Torrey Pines Institute for Molecular Studies San Diego, California, United States of America
| | - Pingchuan Li
- Torrey Pines Institute for Molecular Studies San Diego, California, United States of America
| | - Ruth A. Gjerset
- Torrey Pines Institute for Molecular Studies San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
11
|
Andrique L, Fauvin D, El Maassarani M, Colasson H, Vannier B, Séité P. ErbB380kDa, a nuclear variant of the ErbB3 receptor, binds to the Cyclin D1 promoter to activate cell proliferation but is negatively controlled by p14ARF. Cell Signal 2012; 24:1074-85. [DOI: 10.1016/j.cellsig.2012.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/16/2011] [Accepted: 01/04/2012] [Indexed: 01/11/2023]
|
12
|
Westhoff B, Colaluca IN, D'Ario G, Donzelli M, Tosoni D, Volorio S, Pelosi G, Spaggiari L, Mazzarol G, Viale G, Pece S, Di Fiore PP. Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci U S A 2009; 106:22293-8. [PMID: 20007775 PMCID: PMC2799768 DOI: 10.1073/pnas.0907781106] [Citation(s) in RCA: 315] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Indexed: 12/21/2022] Open
Abstract
Notch signaling regulates cell specification and homeostasis of stem cell compartments, and it is counteracted by the cell fate determinant Numb. Both Numb and Notch have been implicated in human tumors. Here, we show that Notch signaling is altered in approximately one third of non-small-cell lung carcinomas (NSCLCs), which are the leading cause of cancer-related deaths: in approximately 30% of NSCLCs, loss of Numb expression leads to increased Notch activity, while in a smaller fraction of cases (around 10%), gain-of-function mutations of the NOTCH-1 gene are present. Activation of Notch correlates with poor clinical outcomes in NSCLC patients without TP53 mutations. Finally, primary epithelial cell cultures, derived from NSCLC harboring constitutive activation of the Notch pathway, are selectively killed by inhibitors of Notch (gamma-secretase inhibitors), showing that the proliferative advantage of these tumors is dependent upon Notch signaling. Our results show that the deregulation of the Notch pathway is a relatively frequent event in NSCLCs and suggest that it might represent a possible target for molecular therapies in these tumors.
Collapse
MESH Headings
- Aged
- Base Sequence
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- DNA Mutational Analysis
- DNA, Neoplasm/genetics
- Female
- Gene Expression
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Middle Aged
- Mutation
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Transcription Factor HES-1
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Britta Westhoff
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
| | - Ivan N. Colaluca
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
| | - Giovanni D'Ario
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Maddalena Donzelli
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
| | - Daniela Tosoni
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
| | - Sara Volorio
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
| | - Giuseppe Pelosi
- European Institute of Oncology, Milan, Italy; and
- Dipartimento di Medicina, Chirurgia ed Odontoiatria, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Spaggiari
- European Institute of Oncology, Milan, Italy; and
- Dipartimento di Medicina, Chirurgia ed Odontoiatria, Università degli Studi di Milano, Milan, Italy
| | - Giovanni Mazzarol
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
| | - Giuseppe Viale
- European Institute of Oncology, Milan, Italy; and
- Dipartimento di Medicina, Chirurgia ed Odontoiatria, Università degli Studi di Milano, Milan, Italy
| | - Salvatore Pece
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
- Dipartimento di Medicina, Chirurgia ed Odontoiatria, Università degli Studi di Milano, Milan, Italy
| | - Pier Paolo Di Fiore
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
- European Institute of Oncology, Milan, Italy; and
- Dipartimento di Medicina, Chirurgia ed Odontoiatria, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
13
|
Colaluca IN, Tosoni D, Nuciforo P, Senic-Matuglia F, Galimberti V, Viale G, Pece S, Di Fiore PP. NUMB controls p53 tumour suppressor activity. Nature 2008; 451:76-80. [PMID: 18172499 DOI: 10.1038/nature06412] [Citation(s) in RCA: 304] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 10/23/2007] [Indexed: 01/04/2023]
Abstract
NUMB is a cell fate determinant, which, by asymmetrically partitioning at mitosis, controls cell fate choices by antagonising the activity of the plasma membrane receptor of the NOTCH family. NUMB is also an endocytic protein, and the NOTCH-NUMB counteraction has been linked to this function. There might be, however, additional functions of NUMB, as witnessed by its proposed role as a tumour suppressor in breast cancer. Here we describe a previously unknown function for human NUMB as a regulator of tumour protein p53 (also known as TP53). NUMB enters in a tricomplex with p53 and the E3 ubiquitin ligase HDM2 (also known as MDM2), thereby preventing ubiquitination and degradation of p53. This results in increased p53 protein levels and activity, and in regulation of p53-dependent phenotypes. In breast cancers there is frequent loss of NUMB expression. We show that, in primary breast tumour cells, this event causes decreased p53 levels and increased chemoresistance. In breast cancers, loss of NUMB expression causes increased activity of the receptor NOTCH. Thus, in these cancers, a single event-loss of NUMB expression-determines activation of an oncogene (NOTCH) and attenuation of the p53 tumour suppressor pathway. Biologically, this results in an aggressive tumour phenotype, as witnessed by findings that NUMB-defective breast tumours display poor prognosis. Our results uncover a previously unknown tumour suppressor circuitry.
Collapse
Affiliation(s)
- Ivan N Colaluca
- IFOM, the FIRC Institute for Molecular Oncology Foundation, Via Adamello 16, 20139, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Agrawal A, Yang J, Murphy RF, Agrawal DK. Regulation of the p14ARF-Mdm2-p53 pathway: an overview in breast cancer. Exp Mol Pathol 2006; 81:115-22. [PMID: 16919268 DOI: 10.1016/j.yexmp.2006.07.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Accepted: 07/07/2006] [Indexed: 12/22/2022]
Abstract
Knowledge of the roles of proteins that are abnormally suppressed or activated due to mutation in the DNA sequences of the common tumor suppressor genes, p14ARF and p53, is critical to the understanding the pathogenesis of breast cancer. Mdm2 is a mediator for the function of both p14ARF and p53. In this review article factors including Pokemon, Geminin, Twist, and Apigenin, which control the action of individual proteins in the p14ARF-Mdm2-p53 pathway in breast cancer as well the consequences of mutation 7 of p53 are discussed. The complexity of interaction of components of the pathway and the underlying development of cancer is emphasized. Opportunities for future therapeutic innovations are indicated.
Collapse
Affiliation(s)
- Anshu Agrawal
- Department of Biomedical Sciences, Medicine, and Medical Microbiology and Immunology, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE 68178, USA
| | | | | | | |
Collapse
|
15
|
Validation of transgenic models of breast cancer: ductal carcinoma in situ (DCIS) and Brca1-mutation-related breast cancer. ACTA ACUST UNITED AC 2005; 8. [PMID: 24371431 DOI: 10.1017/s1470903105003135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Available mouse models of ductal carcinoma in situ (DCIS) and BRCA1-mutation-related breast cancer are reviewed. The best validated mouse models of human DCIS are the conditional estrogen receptor α in mammary tissue (CERM) model initiated by deregulated estrogen receptor α and the serial explant mouse model initiated by p53 deficiency. At present the most useful and best validated mouse model of BRCA1-mutation-related breast cancer uses the cre-lox system to make a conditional Brca1 deletion targeted to mammary epithelial cells. The major shortcoming of the non-conditional Brca1 models is the high incidence of non-mammary tumor development. The use of mammary gland transplants or explants from these mice into nude hosts is one approach that could be used to circumvent this deficiency. Development and validation of a Brca1-mutation-related mouse model of basal cell breast cancer is an important next step.
Collapse
|
16
|
Vestey SB, Sen C, Calder CJ, Perks CM, Pignatelli M, Winters ZE. Activated Akt expression in breast cancer: Correlation with p53, Hdm2 and patient outcome. Eur J Cancer 2005; 41:1017-25. [PMID: 15862750 DOI: 10.1016/j.ejca.2005.02.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2004] [Revised: 01/20/2005] [Accepted: 02/03/2005] [Indexed: 10/25/2022]
Abstract
Activation of protein kinase-B/Akt (pAkt) is mediated by oestrogen and involves HER-2 in vitro, to phosphorylate Hdm2 and influence p53 cytoplasmic localisation and degradation. Expression of all active Akt isoforms (pAkt) were examined, together with p53/Hdm2 subcellular expression in invasive ductal breast cancers (IDCs), to evaluate whether in vitro findings were related to clinical data and determine the effect on outcome. Immunohistochemical expression of serine 473 specific phosphorylated Akt (pAkt) isoforms (Akt-1,2,3) was evaluated in 97 patients, together with subcellular expression of p53/Hdm2. The results show that pAkt was evaluable in 95 patients with cytoplasmic expression in 81% and more likely to be associated with larger tumours (P=0.007), with no correlation with HER-2 expression. pAkt correlated with increasing levels of cytoplasmic p53 (P=0.025) and was associated with a reduced disease-free survival (P=0.04; univariate). In conclusion, pAkt has implications in breast cancer growth through mechanisms inactivating p53 with an association with immunohistochemical p53 expression, which is preferentially cytoplasmic. Despite in vitro associations, pAkt appears to be a variable marker of HER-2 expression.
Collapse
Affiliation(s)
- S B Vestey
- University of Bristol, Department of Clinical Sciences at South Bristol--Surgery, Level 7, Bristol Royal Infirmary, Marlborough Street, Bristol BS2 8HW, UK
| | | | | | | | | | | |
Collapse
|