1
|
Albert V, Bruss C, Tümen D, Piendl G, Weber F, Dahl E, Seitz S, Ortmann O, Wege AK, Brockhoff G. HER4 Affects Sensitivity to Tamoxifen and Abemaciclib in Luminal Breast Cancer Cells and Restricts Tumor Growth in MCF-7-Based Humanized Tumor Mice. Int J Mol Sci 2024; 25:7475. [PMID: 39000582 PMCID: PMC11242770 DOI: 10.3390/ijms25137475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
The impact of the HER4 receptor on the growth and treatment of estrogen receptor-positive breast cancer is widely uncertain. Using CRISPR/Cas9 technology, we generated stable HER4 knockout variants derived from the HER4-positive MCF-7, T-47D, and ZR-75-1 breast cancer cell lines. We investigated tumor cell proliferation as well as the cellular and molecular mechanisms of tamoxifen, abemaciclib, AMG232, and NRG1 treatments as a function of HER4 in vitro. HER4 differentially affects the cellular response to tamoxifen and abemaciclib treatment. Most conspicuous is the increased sensitivity of MCF-7 in vitro upon HER4 knockout and the inhibition of cell proliferation by NRG1. Additionally, we assessed tumor growth and immunological effects as responses to tamoxifen and abemaciclib therapy in humanized tumor mice (HTM) based on MCF-7 HER4-wildtype and the corresponding HER4-knockout cells. Without any treatment, the enhanced MCF-7 tumor growth in HTM upon HER4 knockout suggests a tumor-suppressive effect of HER4 under preclinical but human-like conditions. This phenomenon is associated with an increased HER2 expression in MCF-7 in vivo. Independent of HER4, abemaciclib and tamoxifen treatment considerably inhibited tumor growth in these mice. However, abemaciclib-treated hormone receptor-positive breast cancer patients with tumor-associated mdm2 gene copy gains or pronounced HER4 expression showed a reduced event-free survival. Evidently, the presence of HER4 affects the efficacy of tamoxifen and abemaciclib treatment in different estrogen receptor-positive breast cancer cells, even to different extents, and is associated with unfavorable outcomes in abemaciclib-treated patients.
Collapse
Affiliation(s)
- Veruschka Albert
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Christina Bruss
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Deniz Tümen
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gerhard Piendl
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Florian Weber
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
- Institute of Pathology, University of Regensburg, 93093 Regensburg, Germany
| | - Edgar Dahl
- Institute of Pathology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Anja K Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| |
Collapse
|
2
|
Bayat Z, Farhadi Z, Taherkhani A. Identification of potential biomarkers associated with poor prognosis in oral squamous cell carcinoma through integrated bioinformatics analysis: A pilot study. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
3
|
ZNF768 Expression Associates with High Proliferative Clinicopathological Features in Lung Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13164136. [PMID: 34439288 PMCID: PMC8391643 DOI: 10.3390/cancers13164136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Zinc-finger protein 768 (ZNF768) is a transcription factor that was recently shown to promote proliferation and repress senescence downstream of growth factor signaling in vitro. This protein was found to be overexpressed in a small cohort of lung adenocarcinoma (LUAD) compared to normal tissue, but its clinical value in this cancer remains unknown. The aim of this study was to determine whether ZNF768 associates with clinicopathological features in LUAD. We found that ZNF768 levels are often elevated in LUAD and that ZNF768 protein levels positively correlate with Ki-67 and other proliferative clinicopathological features in this cancer. Supporting a role for ZNF768 in promoting proliferation, ZNF768 depletion severely impairs proliferation in several lung cancer cell lines. Our study, that extends previous in vitro data, provides the first clinical observations supporting a possible role for ZNF768 in promoting cancer cell proliferation and tumor development in humans. Abstract Lung adenocarcinoma (LUAD) is the most common type of lung cancer and a leading cause of cancer-related deaths worldwide. Despite important recent advances, the prognosis for LUAD patients is still unfavourable, with a 5 year-survival rate close to 15%. Improving the characterization of lung tumors is important to develop alternative options for the diagnosis and the treatment of this disease. Zinc-finger protein 768 (ZNF768) is a transcription factor that was recently shown to promote proliferation and repress senescence downstream of growth factor signaling. Although ZNF768 protein levels were found to be elevated in LUAD compared to normal lung tissue, it is currently unknown whether ZNF768 expression associates with clinicopathological features in LUAD. Here, using tissue microarrays of clinical LUAD surgical specimens collected from 364 patients, we observed that high levels of ZNF768 is a common characteristic of LUAD. We show that ZNF768 protein levels correlate with high proliferative features in LUAD, including the mitotic score and Ki-67 expression. Supporting a role for ZNF768 in promoting proliferation, we report that ZNF768 depletion severely impairs proliferation in several lung cancer cell lines in vitro. A marked decrease in the expression of key proliferative genes was observed in cancer cell lines depleted from ZNF768. Altogether, our findings support a role for ZNF768 in promoting proliferation of LUAD.
Collapse
|
4
|
Duan S, Pagano M. Ubiquitin ligases in cancer: Functions and clinical potentials. Cell Chem Biol 2021; 28:918-933. [PMID: 33974914 PMCID: PMC8286310 DOI: 10.1016/j.chembiol.2021.04.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/23/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023]
Abstract
Ubiquitylation, a highly regulated post-translational modification, controls many cellular pathways that are critical to cell homeostasis. Ubiquitin ligases recruit substrates and promote ubiquitin transfer onto targets, inducing proteasomal degradation or non-degradative signaling. Accumulating evidence highlights the critical role of dysregulated ubiquitin ligases in processes associated with the initiation and progression of cancer. Depending on the substrate specificity and biological context, a ubiquitin ligase can act either as a tumor promoter or as a tumor suppressor. In this review, we focus on the regulatory roles of ubiquitin ligases and how perturbations of their functions contribute to cancer pathogenesis. We also briefly discuss current strategies for targeting or exploiting ubiquitin ligases for cancer therapy.
Collapse
Affiliation(s)
- Shanshan Duan
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA; Howard Hughes Medical Institute, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Shan P, Yang F, Qi H, Hu Y, Zhu S, Sun Z, Zhang Z, Wang C, Hou C, Yu J, Wang L, Zhou Z, Li P, Zhang H, Wang K. Alteration of MDM2 by the Small Molecule YF438 Exerts Antitumor Effects in Triple-Negative Breast Cancer. Cancer Res 2021; 81:4027-4040. [PMID: 33985974 DOI: 10.1158/0008-5472.can-20-0922] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/27/2020] [Accepted: 05/11/2021] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) exhibits a high mortality rate and is the most aggressive subtype of breast cancer. As previous studies have shown that histone deacetylases (HDAC) may represent molecular targets for TNBC treatment, we screened a small library of synthetic molecules and identified a potent HDAC inhibitor (HDACi), YF438, which exerts effective anti-TNBC activity both in vitro and in vivo. Proteomic and biochemical studies revealed that YF438 significantly downregulated mouse double minute 2 homolog (MDM2) expression. In parallel, loss of MDM2 expression or blocking MDM2 E3 ligase activity rendered TNBC cells less sensitive to YF438 treatment, revealing an essential role of MDM2 E3 ligase activity in YF438-induced inhibition of TNBC. Mechanistically, YF438 disturbed the interaction between HDAC1 and MDM2, induced the dissociation of MDM2-MDMX, and subsequently increased MDM2 self-ubiquitination to accelerate its degradation, which ultimately inhibited growth and metastasis of TNBC cells. In addition, analysis of clinical tissue samples demonstrated high expression levels of MDM2 in TNBC, and MDM2 protein levels closely correlated with TNBC progression and metastasis. Collectively, these findings show that MDM2 plays an essential role in TNBC progression and targeting the HDAC1-MDM2-MDMX signaling axis with YF438 may provide a promising therapeutic option for TNBC. Furthermore, this novel underlying mechanism of a hydroxamate-based HDACi in altering MDM2 highlights the need for further development of HDACi for TNBC treatment. SIGNIFICANCE: This study uncovers the essential role of MDM2 in TNBC progression and suggests that targeting the HDAC1-MDM2-MDMX axis with a hydroxamate-based HDACi could be a promising therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Peipei Shan
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China
| | - Feifei Yang
- School of Biological Science and Technology, University of Jinan, Jinan, P.R. China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China
| | - Yunjie Hu
- Weifang Medical University, Weifang, P.R. China
| | - Sujie Zhu
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China
| | - Zhenqing Sun
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Zhe Zhang
- Qingdao Municipal Hospital, Qingdao, Shandong, P.R. China
| | - Chuanxiao Wang
- Qingdao Municipal Hospital, Qingdao, Shandong, P.R. China
| | - Caixia Hou
- Qingdao Central Hospital, Qingdao, Shandong, P.R. China
| | - Jie Yu
- Qingdao Central Hospital, Qingdao, Shandong, P.R. China
| | - Lirong Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China
| | - Zhixia Zhou
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, P.R. China.
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong, P.R. China.
| |
Collapse
|
6
|
Diez-Calzadilla NA, Noguera Salvá R, Soriano Sarrió P, Martínez-Jabaloyas JM. Genetic profile and immunohistochemical study of clear cell renal carcinoma: Pathological-anatomical correlation and prognosis. Cancer Treat Res Commun 2021; 27:100374. [PMID: 33932757 DOI: 10.1016/j.ctarc.2021.100374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) accounts for 2-3% of all tumors being the most frequent solid lesion in the kidney. OBJECTIVE To determine what genetic alterations and immunohistochemical (IHC) of clear cell renal carcinoma (ccRCC) are associated with prognosis and tumor aggressiveness. PATIENTS AND METHODS Experimental analytical study with 57 patients who underwent radical and partial nephrectomy between 2005 and 2011, all with diagnosis of ccRCC and minimum post-operative follow-up of 36 months. The pathological study included IHC determination of biomarkers associated (CAIX, CAM 5.2, CD10, c-erbB-2, EGFR, HIF-1a, Ki67, MDM2, PAX-2 y 8, p53, survivin and VEGFR 1 and 2). Genetic analysis was carried out using multiplex ligation-dependent probe amplification (MLPA). Clinical data were collected and summarized using an access-type database, adding genetic analysis and IHC data of each patient's tumor sample. IHC statistical analysis included Chi-square, Kruskal-Wallis and multivariate analysis. The genetic analysis was performed using multivariate logistic regression (normal/deletion-duplication). Significance level p<0.05. RESULTS Pathologic stage was: pT1 (61.8%), pT2 (32.7%); pT3-T4 (5.4%); 16.3% were pN+ and 19.3% M1. 23.6% recurred being predominantly to distance in 83.3%. 27.3% of patients died (73.3% ccCCR). CAIX (Carbonic anhydrase IX) and tumor size were associated with worse Fuhrman grade (p = 0.035; p = 0.001 respectively). Deletion-duplication of genes increased the likelihood: of death (APC, Bcl-2 and CDKN2A by 11, 7 and 4 respectively and SMAD4 reduced the probability by 88%); tumor recurrence (CDKN2A by fifteen fold and VHL reduced the probability by 87%); pT greater than 2 (CCND2, MDM2 and WT1 multiplied by 6, 7 and 9); risk of N+ (CDK4 and EBF1 by 13); distant metastases (BRCA2 and DLEU1 by 5); Fuhrman grade ≥3 (BRCA1, BRCA2 and p53 by 40, 75 and 34 respectively, while that FHIT reduced by 96%). Deletion-duplication of CDK4 and DCC increased survival by a factor of 13 and 16, while that DLEU1 and RUNX1 decreased survival time by 80%. CONCLUSION CAIX and tumor size are associated with increased aggressiveness. The mutations to level 5q, 9p, 11p, 12, 13q, 17, 18q and 21q are associated with more aggressive tumors and with worse survival rate.
Collapse
Affiliation(s)
- N A Diez-Calzadilla
- Department of Urology. Hospital de Sagunto, Av. Ramón y Cajal, s/n, 46520. Sagunto, Valencia-España.
| | - R Noguera Salvá
- Department of Pathology. Faculty of Medicine. University of Valencia, Av. Blasco Ibáñez, 13. 46010. Valencia-España
| | - P Soriano Sarrió
- Department of Pathology. Faculty of Medicine. University of Valencia, Av. Blasco Ibáñez, 13. 46010. Valencia-España
| | - J M Martínez-Jabaloyas
- Department of Urology. Hospital Clínico Universitario de Valencia, Av. Blasco Ibáñez, 17, 46010. Valencia- España.
| |
Collapse
|
7
|
Xian J, Cheng Y, Qin X, Cao Y, Luo Y, Cao Y. Progress in the research of p53 tumour suppressor activity controlled by Numb in triple-negative breast cancer. J Cell Mol Med 2020; 24:7451-7459. [PMID: 32501652 PMCID: PMC7339219 DOI: 10.1111/jcmm.15366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/16/2019] [Accepted: 04/22/2020] [Indexed: 11/26/2022] Open
Abstract
Numb is known as a cell fate determinant as it identifies the direction of cell differentiation via asymmetrical partitioning during mitosis. It is considered as a tumour suppressor, and a frequent loss of Numb expression in breast cancer is noted. Numb forms a tri-complex with p53 and E3 ubiquitin ligase HDM2 (also known as MDM2), thereby preventing the ubiquitination and degradation of p53. In this study, we examined Numb expression in 125 patients with triple-negative breast cancer (TNBC). The results showed that 61 (48.8%) patients presented with a deficient or decreased Numb expression. The percentage of Ki67 > 14% in the retained Numb group was significantly lower than that in the decreased and deficient Numb groups (86.00% vs. 98.40%, P = .0171). This study aimed to detect the expression and migration of Numb, HDM2 and p53 in the membrane, cytoplasmic and nuclear fractions of normal mammary epithelial cell line MCF-10A and basal-like TNBC cell line MDA-MB-231. We obtained the cell fractions to identify changes in these three protein levels after the re-expression of NUMB in the MDA-MB-231 cells and the knocking down of NUMB in the MCF-10A cells. Results showed that Numb regulates p53 levels in the nucleus where the protein levels of Numb are positively correlated with p53 levels, regardless if it is re-expressed in the MDA-MB-231 cells or knocked down in the MCF-10A cells. Moreover, HDM2 was remarkably decreased only in the membrane fraction of NUMB knock-down cells; however, its mRNA levels were increased significantly. Our results reveal a previously unknown molecular mechanism that Numb can migrate into the nucleus and interact with HDM2 and p53.
Collapse
Affiliation(s)
- Jie Xian
- School of Basic Medical SciencesMedical University of ChongqingChongqingChina
| | - Yu Cheng
- School of Laboratory MedicineMedical University of ChongqingChongqingChina
| | - Xue Qin
- School of Basic Medical SciencesMedical University of ChongqingChongqingChina
| | - Yijia Cao
- Breast SurgeryChongqing Traditional Chinese Medicine HospitalChongqingChina
| | - Yetao Luo
- School of Public Health and ManagementMedical University of ChongqingChongqingChina
| | - Youde Cao
- School of Basic Medical SciencesMedical University of ChongqingChongqingChina
| |
Collapse
|
8
|
Gluck WL, Gounder MM, Frank R, Eskens F, Blay JY, Cassier PA, Soria JC, Chawla S, de Weger V, Wagner AJ, Siegel D, De Vos F, Rasmussen E, Henary HA. Phase 1 study of the MDM2 inhibitor AMG 232 in patients with advanced P53 wild-type solid tumors or multiple myeloma. Invest New Drugs 2019; 38:831-843. [PMID: 31359240 PMCID: PMC7211202 DOI: 10.1007/s10637-019-00840-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022]
Abstract
Background This open-label, first-in-human, phase 1 study evaluated AMG 232, an oral selective MDM2 inhibitor in patients with TP53 wild-type (P53WT), advanced solid tumors or multiple myeloma (MM). Methods In the dose escalation (n = 39), patients with P53WT refractory solid tumors enrolled to receive once-daily AMG 232 (15, 30, 60, 120, 240, 480, and 960 mg) for seven days every 3 weeks (Q3W). In the dose expansion (n = 68), patients with MDM2-amplified (well-differentiated and de-differentiated liposarcomas [WDLPS and DDLPS], glioblastoma multiforme [GBM], or other solid tumors [OST]), MDM2-overexpressing ER+ breast cancer (BC), or MM received AMG 232 at the maximum tolerated dose (MTD). Safety, pharmacokinetics, pharmacodynamics, and efficacy were assessed. Results AMG 232 had acceptable safety up to up to 240 mg. Three patients had dose-limiting toxicities of thrombocytopenia (n = 2) and neutropenia (n = 1). Due to these and other delayed cytopenias, AMG 232 240 mg Q3W was determined as the highest tolerable dose assessed in the dose expansion. Adverse events were typically mild/moderate and included diarrhea, nausea, vomiting, fatigue, decreased appetite, and anemia. AMG 232 plasma concentrations increased dose proportionally. Increases in serum macrophage inhibitor cytokine-1 from baseline were generally dose dependent, indicating p53 pathway activation. Per local review, there were no responses. Stable disease (durability in months) was observed in patients with WDLPS (3.9), OST (3.3), DDLPS (2.0), GBM (1.8), and BC (1.4–2.0). Conclusions In patients with P53WT advanced solid tumors or MM, AMG 232 showed acceptable safety and dose-proportional pharmacokinetics, and stable disease was observed.
Collapse
Affiliation(s)
- W Larry Gluck
- Prisma Health - Upstate, Institute for Translational Oncology Research, 900 W. Faris Rd., 3rd Floor, Greenville, SC, 29605, USA.
| | - Mrinal M Gounder
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | - Ferry Eskens
- Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jean Yves Blay
- Department of Medicine, Centre Léon Bérard, Lyon, France
| | | | - Jean-Charles Soria
- Department of Medicine, The Institute Gustave-Roussy, Paris, France.,Université Paris Sud, Orsay, France
| | - Sant Chawla
- Sarcoma Oncology Center, Cancer Center of Southern California, Santa Monica, CA, USA
| | - Vincent de Weger
- Department of Internal Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andrew J Wagner
- Center for Sarcoma and Bone Oncology and Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - David Siegel
- Multiple Myeloma Division, John Theurer Cancer Center at the Hackensack University Medical Center, Hackensack, NJ, USA
| | - Filip De Vos
- Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Erik Rasmussen
- Oncology Early Development, Amgen Inc., Thousand Oaks, CA, USA
| | - Haby A Henary
- Oncology Early Development, Amgen Inc., Thousand Oaks, CA, USA
| |
Collapse
|
9
|
Rubio-Patiño C, Trotta AP, Chipuk JE. MDM2 and mitochondrial function: One complex intersection. Biochem Pharmacol 2018; 162:14-20. [PMID: 30391206 DOI: 10.1016/j.bcp.2018.10.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022]
Abstract
Decades of research reveal that MDM2 participates in cellular processes ranging from macro-molecular metabolism to cancer signaling mechanisms. Two recent studies uncovered a new role for MDM2 in mitochondrial bioenergetics. Through the negative regulation of NDUFS1 (NADH:ubiquinone oxidoreductase 75 kDa Fe-S protein 1) and MT-ND6 (NADH dehydrogenase 6), MDM2 decreases the function and efficiency of Complex I (CI). These observations propose several important questions: (1) Where does MDM2 affect CI activity? (2) What are the cellular consequences of MDM2-mediated regulation of CI? (3) What are the physiological implications of these interactions? Here, we will address these questions and position these observations within the MDM2 literature.
Collapse
Affiliation(s)
- Camila Rubio-Patiño
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA
| | - Andrew Paul Trotta
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA; The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1130, New York, NY 10029, USA.
| |
Collapse
|
10
|
Mansour MA. Ubiquitination: Friend and foe in cancer. Int J Biochem Cell Biol 2018; 101:80-93. [PMID: 29864543 DOI: 10.1016/j.biocel.2018.06.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/05/2023]
Abstract
Dynamic modulation and posttranslational modification of proteins are tightly controlled biological processes that occur in response to physiological cues. One such dynamic modulation is ubiquitination, which marks proteins for degradation via the proteasome, altering their localization, affecting their activity, and promoting or interfering with protein interactions. Hence, ubiquitination is crucial for a plethora of physiological processes, including cell survival, differentiation and innate and adaptive immunity. Similar to kinases, components of the ubiquitination system are often deregulated, leading to a variety of diseases, such as cancer and neurodegenerative disorders. In a context-dependent manner, ubiquitination can regulate both tumor-suppressing and tumor-promoting pathways in cancer. This review outlines how components of the ubiquitination systems (e.g. E3 ligases and deubiquitinases) act as oncogenes or tumor suppressors according to the nature of their substrates. Furthermore, I interrogate how the current knowledge of the differential roles of ubiquitination in cancer lead to technical advances to inhibit or reactivate the components of the ubiquitination system accordingly.
Collapse
Affiliation(s)
- Mohammed A Mansour
- Institute of Cancer Sciences, University of Glasgow, United Kingdom; The CRUK Beatson Institute, Glasgow, Switchback Road, G61 1BD, United Kingdom; Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
11
|
Zhang R, Wu X, Xia X, Khanniche A, Song F, Zhang B, Wang Y, Ge H. OVA12 promotes tumor growth by regulating p53 expression in human cancer cells. Oncotarget 2017; 8:52854-52865. [PMID: 28881777 PMCID: PMC5581076 DOI: 10.18632/oncotarget.17501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 04/10/2017] [Indexed: 11/30/2022] Open
Abstract
Ovarian cancer-associated antigen 12 (OVA12) was first identified in an ovarian carcinoma complementary DNA (cDNA) expression library and has been shown to play an important role in tumor growth. Here, we found that overexpression of OVA12 accelerated tumor growth in different tumor cells, whereas OVA12 depletion was associated with the opposite effect. Moreover, knocking down OVA12 led to a significant increase in the protein levels of p53, and the overexpression of OVA12 significantly decreased endogenous p53 levels. In addition, OVA12 stimulated p53 polyubiquitination and degradation by the proteasome and promoted tumor growth at least partially through the p53 pathway. Taken together, these results indicate that OVA12 is a negative regulator of p53 and that inhibition of OVA12 expression might serve as a therapeutic target to restore tumor suppression.
Collapse
Affiliation(s)
- Renfeng Zhang
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xicai Wu
- Clinical Laboratory, People's Hospital of Rizhao, Rizhao, China
| | - Xiangfeng Xia
- Department of Radiology, The Third People's Hospital of Rizhao, Rizhao, China
| | - Asma Khanniche
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feifei Song
- Department of Pathology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Bingchang Zhang
- Department of Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Ying Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hailiang Ge
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Yang H, Yu S, Wang W, Li X, Hou Y, Liu Z, Shi Y, Mu K, Niu G, Xu J, Wang H, Zhu J, Zhuang T. SHARPIN Facilitates p53 Degradation in Breast Cancer Cells. Neoplasia 2017; 19:84-92. [PMID: 28063307 PMCID: PMC5219588 DOI: 10.1016/j.neo.2016.12.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/01/2016] [Accepted: 12/05/2016] [Indexed: 11/30/2022]
Abstract
The ubiquitin binding protein SHAPRIN is highly expressed in human breast cancer, one of the most frequent female malignancies worldwide. Here, we perform SHARPIN depletion in breast cancer cells together with RNA sequencing. The global expression profiling showed p53 signaling as a potential SHARPIN target. SHARPIN depletion decreased cell proliferation, which effect could be rescue by p53 knocking down. Depletion SHARPIN significantly increases p53 protein level and its target genes in multiple breast cancer cell lines. Further experiment revealed that SHARPIN could facilitate p53 poly-ubiquitination and degradation in MDM2 dependent manner. Immuno-precipitation assay showed that SHARPIN associated with MDM2 and prolonged MDM2 protein stability. Analysis of public available database showed SHARPIN correlated with poor prognosis specifically in p53 wild-type breast cancer patients. Together, our finding revealed a novel modifier for p53/MDM2 complex and suggested SHARPIN as a promising target to restore p53 function in breast cancer.
Collapse
Affiliation(s)
- Huijie Yang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Sifan Yu
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Department of Renal Cancer and Melanoma, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Weilong Wang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Xin Li
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Yingxiang Hou
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Zhenhua Liu
- College of Life Science and Technology, Synthetic Biology, Medical Institute, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Yuanyuan Shi
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China
| | - Kun Mu
- Department of Pathology, Shandong University School of Medicine, Jinan, PR China
| | - Gang Niu
- Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd., Shenzhen, PR China
| | - Juntao Xu
- Department of Cancer genomics, LemonData biotech (Shenzhen) Ltd., Shenzhen, PR China
| | - Hui Wang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China.
| | - Jian Zhu
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China.
| | - Ting Zhuang
- Research Center for Immunology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, PR China.
| |
Collapse
|
13
|
Abstract
Inactivation of the p53 tumor suppressor gene is one of the commonest genetic changes identified in human breast cancer. In this review, the structure and function of the p53 gene and its protein products will be discussed, with particular reference to p53 alterations that contribute to carcinogenesis. The frequency and pattern of p53 alterations in breast cancer will be outlined, laboratory methods for their detection briefly summarized, and the potential use of p53 as a prognostic and predictive marker discussed.
Collapse
|
14
|
Oliner JD, Saiki AY, Caenepeel S. The Role of MDM2 Amplification and Overexpression in Tumorigenesis. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a026336. [PMID: 27194168 DOI: 10.1101/cshperspect.a026336] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mouse double minute 2 (MDM2) is a critical negative regulator of the tumor suppressor p53, playing a key role in controlling its transcriptional activity, protein stability, and nuclear localization. MDM2 expression is up-regulated in numerous cancers, resulting in a loss of p53-dependent activities, such as apoptosis and cell-cycle arrest. Genetic amplification and inheritance of MDM2 promoter single-nucleotide polymorphisms (SNPs) are the two best-studied mechanisms for up-regulating MDM2 activity. This article provides an overview of these events in human cancer, highlighting the frequent occurrence of MDM2 amplification in sarcoma and the role of SNP309 and SNP285 in regulating MDM2 expression and cancer risk. The availability of large-scale genomic profiling datasets, like those from The Cancer Genome Atlas Research Network, have provided the opportunity to evaluate the consequences of MDM2 amplification and SNP inheritance across high-quality tumor samples from diverse cancer indications.
Collapse
Affiliation(s)
| | - Anne Y Saiki
- Oncology Research, Amgen, Thousand Oaks, California 91320
| | - Sean Caenepeel
- Oncology Research, Amgen, Thousand Oaks, California 91320
| |
Collapse
|
15
|
Swetzig WM, Wang J, Das GM. Estrogen receptor alpha (ERα/ESR1) mediates the p53-independent overexpression of MDM4/MDMX and MDM2 in human breast cancer. Oncotarget 2016; 7:16049-69. [PMID: 26909605 PMCID: PMC4941297 DOI: 10.18632/oncotarget.7533] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
MDM2 and MDM4 are heterodimeric, non-redundant oncoproteins that potently inhibit the p53 tumor suppressor protein. MDM2 and MDM4 also enhance the tumorigenicity of breast cancer cells in in vitro and in vivo models and are overexpressed in primary human breast cancers. Prior studies have characterized Estrogen Receptor Alpha (ERα/ESR1) as a regulator of MDM2 expression and an MDM2- and p53-interacting protein. However, similar crosstalk between ERα and MDM4 has not been investigated. Moreover, signaling pathways that mediate the overexpression of MDM4 in human breast cancer remain to be elucidated. Using the Cancer Genome Atlas (TCGA) breast invasive carcinoma patient cohort, we have analyzed correlations between ERα status and MDM4 and MDM2 expression in primary, treatment-naïve, invasive breast carcinoma samples. We report that the expression of MDM4 and MDM2 is elevated in primary human breast cancers of luminal A/B subtypes and associates with ERα-positive disease, independently of p53 mutation status. Furthermore, in cell culture models, ERα positively regulates MDM4 and MDM2 expression via p53-independent mechanisms, and these effects can be blocked by the clinically-relevant endocrine therapies fulvestrant and tamoxifen. Additionally, ERα also positively regulates p53 expression. Lastly, we report that endogenous MDM4 negatively regulates ERα expression and forms a protein complex with ERα in breast cancer cell lines and primary human breast tumor tissue. This suggests direct signaling crosstalk and negative feedback loops between ERα and MDM4 expression in breast cancer cells. Collectively, these novel findings implicate ERα as a central component of the p53-MDM2-MDM4 signaling axis in human breast cancer.
Collapse
Affiliation(s)
- Wendy M. Swetzig
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Molecular Pharmacology and Cancer Therapeutics, The University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Jianmin Wang
- Department of Bioinformatics and Biostatistics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Gokul M. Das
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Molecular Pharmacology and Cancer Therapeutics, The University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
16
|
Huang X, Dixit VM. Drugging the undruggables: exploring the ubiquitin system for drug development. Cell Res 2016; 26:484-98. [PMID: 27002218 PMCID: PMC4822129 DOI: 10.1038/cr.2016.31] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dynamic modulation of protein levels is tightly controlled in response to physiological cues. In mammalian cells, much of the protein degradation is carried out by the ubiquitin-proteasome system (UPS). Similar to kinases, components of the ubiquitin system are often dysregulated, leading to a variety of diseases, including cancer and neurodegeneration, making them attractive drug targets. However, so far there are only a handful of drugs targeting the ubiquitin system that have been approved by the FDA. Here, we review possible therapeutic intervention nodes in the ubiquitin system, analyze the challenges, and highlight the most promising strategies to target the UPS.
Collapse
Affiliation(s)
- Xiaodong Huang
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
17
|
Interaction between p53 codon 72 and MDM2 309T>G polymorphisms and the risk of hepatocellular carcinoma. Tumour Biol 2015; 37:3863-70. [PMID: 26476535 DOI: 10.1007/s13277-015-4222-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/12/2015] [Indexed: 12/11/2022] Open
Abstract
The p53 tumor suppressor and its negative regulator, murine double minute 2 (MDM2), play critical roles in carcinogenesis. P53 codon 72 and MDM2 309T>G polymorphisms could influence p53 and MDM2 function, respectively, and might affect cancer susceptibility. We therefore investigated the association between these two SNPs, alone or in combination, and the risk of hepatocellular carcinoma (HCC) in Chinese. In this case-control study, we genotyped p53 codon 72 and MDM2 309T>G polymorphisms in 985 HCC cases and 992 cancer-free age- and sex-matched controls and evaluated their associations with the risk of HCC. Although no significant main effects were found for these two SNPs in the single-locus analysis and stratified analysis by age, sex, smoking, drinking, and hepatitis B virus (HBV) infection, we found that individuals carrying at least one G allele of the MDM2 309T>G polymorphism had statistically significant increased risk of HCC among those with the p53 Pro/Pro genotype (adjusted odds ratio (OR) = 2.23, 95 % confidence interval (95%CI) = 1.20-4.14 for TG genotype; adjusted OR = 2.67, 95%CI = 1.32-5.42 for GG genotype), and the interaction between p53 codon 72 and MDM2 309T>G was significant (P interaction = 0.017). Our findings suggest that the interaction of p53 codon 72 and MDM2 309T>G may play an important role in the etiology of HCC. More studies with well-designed and large sample sizes are required to validate these observations.
Collapse
|
18
|
DRAKULIC DANIJELA, VICENTIC JELENAMARJANOVIC, SCHWIRTLICH MARIJA, TOSIC JELENA, KRSTIC ALEKSANDAR, KLAJN ANDRIJANA, STEVANOVIC MILENA. The overexpression of SOX2 affects the migration of human teratocarcinoma cell line NT2/D1. ACTA ACUST UNITED AC 2015; 87:389-404. [DOI: 10.1590/0001-3765201520140352] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/14/2014] [Indexed: 12/15/2022]
Abstract
The altered expression of the SOX2 transcription factor is associated with oncogenic or tumor suppressor functions in human cancers. This factor regulates the migration and invasion of different cancer cells. In this study we investigated the effect of constitutive SOX2 overexpression on the migration and adhesion capacity of embryonal teratocarcinoma NT2/D1 cells derived from a metastasis of a human testicular germ cell tumor. We detected that increased SOX2 expression changed the speed, mode and path of cell migration, but not the adhesion ability of NT2/D1 cells. Additionally, we demonstrated that SOX2 overexpression increased the expression of the tumor suppressor protein p53 and the HDM2 oncogene. Our results contribute to the better understanding of the effect of SOX2 on the behavior of tumor cells originating from a human testicular germ cell tumor. Considering that NT2/D1 cells resemble cancer stem cells in many features, our results could contribute to the elucidation of the role of SOX2 in cancer stem cells behavior and the process of metastasis.
Collapse
Affiliation(s)
| | | | | | - JELENA TOSIC
- University of Belgrade, Serbia; University of Lausanne, Switzerland
| | | | | | | |
Collapse
|
19
|
Dolfi SC, Jäger AV, Medina DJ, Haffty BG, Yang JM, Hirshfield KM. Fulvestrant treatment alters MDM2 protein turnover and sensitivity of human breast carcinoma cells to chemotherapeutic drugs. Cancer Lett 2014; 350:52-60. [PMID: 24747123 PMCID: PMC5500211 DOI: 10.1016/j.canlet.2014.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/28/2014] [Accepted: 04/09/2014] [Indexed: 12/01/2022]
Abstract
The human homologue of mouse double minute 2 (MDM2) is overexpressed in tumors and contributes to tumorigenesis through inhibition of p53 activity. We investigated the effect of the anti-estrogen fulvestrant on MDM2 expression and sensitivity of estrogen receptor positive human breast cancer cell lines to chemotherapeutics. Fulvestrant down-regulated MDM2 through increased protein turnover. Fulvestrant blocked estrogen-dependent up-regulation of MDM2 and decreased basal expression of MDM2 in the absence of estradiol. As combinations of fulvestrant with doxorubicin, etoposide or paclitaxel were synergistic, altering cell cycle distribution and increasing cell death, this provides rationale for testing combinatorial chemotherapy with fulvestrant as a novel therapeutic strategy for patients with advanced breast cancer.
Collapse
Affiliation(s)
- Sonia C Dolfi
- Department of Medicine, Rutgers Cancer Institute of New Jersey, Rutgers The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, United States
| | - Adriana V Jäger
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniel J Medina
- Department of Medicine, Rutgers Cancer Institute of New Jersey, Rutgers The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, United States
| | - Bruce G Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, United States
| | - Jin-Ming Yang
- Department of Pharmacology, The Penn State Cancer Institute, Pennsylvania State University College of Medicine, and Milton S. Hershey Medical Center, 500 University Drive, Hershey, PA 17033, United States
| | - Kim M Hirshfield
- Department of Medicine, Rutgers Cancer Institute of New Jersey, Rutgers The State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, United States.
| |
Collapse
|
20
|
Yu Q, Li Y, Mu K, Li Z, Meng Q, Wu X, Wang Y, Li L. Amplification of Mdmx and overexpression of MDM2 contribute to mammary carcinogenesis by substituting for p53 mutations. Diagn Pathol 2014; 9:71. [PMID: 24667108 PMCID: PMC3986947 DOI: 10.1186/1746-1596-9-71] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/18/2014] [Indexed: 01/10/2023] Open
Abstract
Background The p53 tumor suppressor gene is mutated or deleted in nearly half of human cancers. The murine double minute 2 (Mdm2) and Mdmx represent two important cellular regulators of p53. The aim of this study was to evaluate the abnormalities of p53, Mdmx and Mdm2 genes in archived breast cancers. Methods We assessed the genetic instability at p53, Mdmx and Mdm2 using high resolution multi-color fluorescent in situ hybridization (FISH) protocol and detected the expression status of the tumor protein p53 (TP53), MDMx and MDM2 by immunohistochemistry in 115 archived samples of infiltrating ductal breast carcinomas with foci of ductal carcinoma in situ (DCIS) components. Results The presence of p53 allelic loss and/or TP53 overexpression was observed in 38% out of all patients, and was significantly more often in larger, high grade, ER negative and high ki67 tumors. Mdmx amplification with low-level increase of gene copy number is at high frequency while Mdm2 amplification is rare in primary breast cancer. Mdmx amplification was seen in more invasive carcinomas than preinvasive lesions. MDMx and MDM2 overexpression were detected in 65% and 38% of all cases respectively. Moreover it was showed that most tumors contained either p53 dysfunction or Mdm2 alteration, but not both. This distribution was significant (P < 0.05). Inverse correlation between Mdmx amplification/overexpression and p53 disfunction was also observed (P < 0.05). Conclusions Our results suggest the involvement of Mdm2 and Mdmx in p53-independent breast carcinogenesis and Mdmx may contribute to the regulation of p53 independently of Mdm2. Virtual slides The virtual slides for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1450529994118798.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Li Li
- Department of Pathology, Shandong University School of Medicine, 44#,Wenhua Xi Road, 250012 Jinan, Shandong, PR, China.
| |
Collapse
|
21
|
Sarajlić A, Filipović A, Janjić V, Coombes RC, Pržulj N. The role of genes co-amplified with nicastrin in breast invasive carcinoma. Breast Cancer Res Treat 2013; 143:393-401. [DOI: 10.1007/s10549-013-2805-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022]
|
22
|
Iyer G, Al-Ahmadie H, Schultz N, Hanrahan AJ, Ostrovnaya I, Balar AV, Kim PH, Lin O, Weinhold N, Sander C, Zabor EC, Janakiraman M, Garcia-Grossman IR, Heguy A, Viale A, Bochner BH, Reuter VE, Bajorin DF, Milowsky MI, Taylor BS, Solit DB. Prevalence and co-occurrence of actionable genomic alterations in high-grade bladder cancer. J Clin Oncol 2013; 31:3133-40. [PMID: 23897969 DOI: 10.1200/jco.2012.46.5740] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE We sought to define the prevalence and co-occurrence of actionable genomic alterations in patients with high-grade bladder cancer to serve as a platform for therapeutic drug discovery. PATIENTS AND METHODS An integrative analysis of 97 high-grade bladder tumors was conducted to identify actionable drug targets, which are defined as genomic alterations that have been clinically validated in another cancer type (eg, BRAF mutation) or alterations for which a selective inhibitor of the target or pathway is under clinical investigation. DNA copy number alterations (CNAs) were defined by using array comparative genomic hybridization. Mutation profiling was performed by using both mass spectroscopy-based genotyping and Sanger sequencing. RESULTS Sixty-one percent of tumors harbored potentially actionable genomic alterations. A core pathway analysis of the integrated data set revealed a nonoverlapping pattern of mutations in the RTK-RAS-RAF and phosphoinositide 3-kinase/AKT/mammalian target of rapamycin pathways and regulators of G1-S cell cycle progression. Unsupervised clustering of CNAs defined two distinct classes of bladder tumors that differed in the degree of their CNA burden. Integration of mutation and copy number analyses revealed that mutations in TP53 and RB1 were significantly more common in tumors with a high CNA burden (P < .001 and P < .003, respectively). CONCLUSION High-grade bladder cancer possesses substantial genomic heterogeneity. The majority of tumors harbor potentially tractable genomic alterations that may predict for response to target-selective agents. Given the genomic diversity of bladder cancers, optimal development of target-specific agents will require pretreatment genomic characterization.
Collapse
Affiliation(s)
- Gopa Iyer
- Memorial Sloan-Kettering Cancer Center, Cornell University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Label-free impedimetric immunosensor for ultrasensitive detection of cancer marker Murine double minute 2 in brain tissue. Biosens Bioelectron 2013; 39:220-5. [DOI: 10.1016/j.bios.2012.07.049] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/22/2012] [Accepted: 07/23/2012] [Indexed: 02/06/2023]
|
24
|
Zyada MM. Increased expression of MDM2 and NM23 are associated with malignant transformation of pleomorphic adenoma. Interv Med Appl Sci 2012. [DOI: 10.1556/imas.4.2012.1.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract
This study was designed to correlate the expression of both MDM2 and NM23 in pleomorphic adenoma (PA) to clinical background, histological features, local recurrence, and metastatic potentiality in this tumor. Expression of MDM2 and NM23 was studied immunohistochemically in 23 cases of PA. Our results demonstrated MDM2 and NM23 overexpression in almost all cases of PA. There was a significant difference of the MDM2 and NM23 mean values between benign PA and both carcinoma-ex PA and metastasizing PA. The results of this study show that the overexpression of NM23 and MDM2 oncoproteins could be used as reliable predictors of malignant progression as well as metastatic potentiality of PA.
Collapse
Affiliation(s)
- Manal M. Zyada
- 1 Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| |
Collapse
|
25
|
Middleton LP. The Diagnosis of Pleomorphic Lobular Carcinoma In Situ Warrants Complete Excision with Negative Margins. CURRENT BREAST CANCER REPORTS 2012. [DOI: 10.1007/s12609-012-0072-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
26
|
Chargari C, Leteur C, Ferté C, Deberne M, Lahon B, Rivera C, Bourhis J, Deutsch É. Ciblage pharmacologique de Mdm2 : bases biologiques et perspectives de radiosensibilisation. Cancer Radiother 2011; 15:316-22. [DOI: 10.1016/j.canrad.2011.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Accepted: 02/02/2011] [Indexed: 02/04/2023]
|
27
|
Ahmed AA, Etemadmoghadam D, Temple J, Lynch AG, Riad M, Sharma R, Stewart C, Fereday S, Caldas C, Defazio A, Bowtell D, Brenton JD. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol 2010; 221:49-56. [PMID: 20229506 PMCID: PMC3262968 DOI: 10.1002/path.2696] [Citation(s) in RCA: 569] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Numerous studies have tested the association between TP53 mutations in ovarian cancer and prognosis but these have been consistently confounded by limitations in study design, methodology, and/or heterogeneity in the sample cohort. High-grade serous (HGS) carcinoma is the most clinically important histological subtype of ovarian cancer. As these tumours may arise from the ovary, Fallopian tube or peritoneum, they are collectively referred to as high-grade pelvic serous carcinoma (HGPSC). To identify the true prevalence of TP53 mutations in HGPSC, we sequenced exons 2-11 and intron-exon boundaries in tumour DNA from 145 patients. HGPSC cases were defined as having histological grade 2 or 3 and FIGO stage III or IV. Surprisingly, pathogenic TP53 mutations were identified in 96.7% (n = 119/123) of HGPSC cases. Molecular and pathological review of mutation-negative cases showed evidence of p53 dysfunction associated with copy number gain of MDM2 or MDM4, or indicated the exclusion of samples as being low-grade serous tumours or carcinoma of uncertain primary site. Overall, p53 dysfunction rate approached 100% of confirmed HGPSCs. No association between TP53 mutation and progression-free or overall survival was found. From this first comprehensive mapping of TP53 mutation rate in a homogeneous group of HGPSC patients, we conclude that mutant TP53 is a driver mutation in the pathogenesis of HGPSC cancers. Because TP53 mutation is almost invariably present in HGPSC, it is not of substantial prognostic or predictive significance.
Collapse
Affiliation(s)
- Ahmed Ashour Ahmed
- Functional Genomics of Ovarian Cancer Laboratory, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abdel-Fatah TM, Powe DG, Agboola J, Adamowicz-Brice M, Blamey RW, Lopez-Garcia MA, Green AR, Reis-Filho JS, Ellis IO. The biological, clinical and prognostic implications of p53 transcriptional pathways in breast cancers. J Pathol 2010; 220:419-34. [PMID: 20044801 DOI: 10.1002/path.2663] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We hypothesized that the functional status of p53 transcriptional pathways, rather than p53 protein expression alone, could accurately discriminate between low- and high-risk breast carcinoma (BC) and inform about individuals' tumour biological behaviour. To test this, we studied a well-characterized series of 990 BCs with long-term follow-up, immunohistochemically profiled for p53, its main regulators and downstream genes. Results were validated in an independent series of patients (n = 245) uniformly treated with adjuvant anthracycline-based chemotherapy. Eleven p53 transcriptional phenotypes were identified with just two main clinical outcomes. (a) Low risk/good prognosis group (active/partially inactive p53 pathways), defined as p53(+/-)/MDM4(+)/MDM2(+/-)/Bcl2(+/-)/p21(+/-), p53(-)/MDM4(-)/MDM2(+)/Bcl2(+)/p21(+/-) and p53(+/-)/MDM4(-)/MMD2(-)/Bcl2(+)/p21(+/-). These tumours had favourable clinicopathological characteristics, including ER(+) and long survival after systemic adjuvant-therapy (AT). (b) High risk/poor prognosis group (completely inactive p53 pathways), defined as p53(+/-)/MDM4(-) MDM2(-)/Bcl2(-)/p21(-), p53(-)/MDM4(-) MDM2(+)/Bcl2(-)/p21(-) and p53(+/-)/MDM4(-)/MDM2(-)/Bcl2(-)/p21(+). These tumours were characterized by aggressive clinicopathological characteristics and showed shortened survival when treated with AT. Completely inactive p53 pathways but intact p21 axis p53(+/-)/MDM4(-)/MDM2(-)/Bcl2(-)/p21(+) had the worst prognosis, particularly patients who received AT. Multivariate Cox regression models, including validated prognostic factors for both test and validation series, revealed that the functional status of p53 transcriptional pathways was an independent prognosticator for BC-specific survival (HR 2.64 and 4.5, p < 0.001, respectively) and disease-free survival (HR 1.93 and 2.5, p < 0.001, respectively). In conclusion, p53 functional status determined by assessment of p53 regulatory and downstream targets provides independent prognostic value and may help determine more adequate therapeutic regimens for specific subgroups of breast cancer patients.
Collapse
Affiliation(s)
- Tarek M Abdel-Fatah
- Division of Pathology, School of Molecular Medical Sciences, Nottingham University Hospitals NHS Trust, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ahmed AA, Etemadmoghadam D, Temple J, Lynch AG, Riad M, Sharma R, Stewart C, Fereday S, Caldas C, Defazio A, Bowtell D, Brenton JD. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol 2010. [PMID: 20229506 DOI: 10.1002/path.2696] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Numerous studies have tested the association between TP53 mutations in ovarian cancer and prognosis but these have been consistently confounded by limitations in study design, methodology, and/or heterogeneity in the sample cohort. High-grade serous (HGS) carcinoma is the most clinically important histological subtype of ovarian cancer. As these tumours may arise from the ovary, Fallopian tube or peritoneum, they are collectively referred to as high-grade pelvic serous carcinoma (HGPSC). To identify the true prevalence of TP53 mutations in HGPSC, we sequenced exons 2-11 and intron-exon boundaries in tumour DNA from 145 patients. HGPSC cases were defined as having histological grade 2 or 3 and FIGO stage III or IV. Surprisingly, pathogenic TP53 mutations were identified in 96.7% (n = 119/123) of HGPSC cases. Molecular and pathological review of mutation-negative cases showed evidence of p53 dysfunction associated with copy number gain of MDM2 or MDM4, or indicated the exclusion of samples as being low-grade serous tumours or carcinoma of uncertain primary site. Overall, p53 dysfunction rate approached 100% of confirmed HGPSCs. No association between TP53 mutation and progression-free or overall survival was found. From this first comprehensive mapping of TP53 mutation rate in a homogeneous group of HGPSC patients, we conclude that mutant TP53 is a driver mutation in the pathogenesis of HGPSC cancers. Because TP53 mutation is almost invariably present in HGPSC, it is not of substantial prognostic or predictive significance.
Collapse
Affiliation(s)
- Ahmed Ashour Ahmed
- Functional Genomics of Ovarian Cancer Laboratory, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ahmed AA, Etemadmoghadam D, Temple J, Lynch AG, Riad M, Sharma R, Stewart C, Fereday S, Caldas C, Defazio A, Bowtell D, Brenton JD. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol 2010. [PMID: 20229506 DOI: 10.1002/path.2696]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Numerous studies have tested the association between TP53 mutations in ovarian cancer and prognosis but these have been consistently confounded by limitations in study design, methodology, and/or heterogeneity in the sample cohort. High-grade serous (HGS) carcinoma is the most clinically important histological subtype of ovarian cancer. As these tumours may arise from the ovary, Fallopian tube or peritoneum, they are collectively referred to as high-grade pelvic serous carcinoma (HGPSC). To identify the true prevalence of TP53 mutations in HGPSC, we sequenced exons 2-11 and intron-exon boundaries in tumour DNA from 145 patients. HGPSC cases were defined as having histological grade 2 or 3 and FIGO stage III or IV. Surprisingly, pathogenic TP53 mutations were identified in 96.7% (n = 119/123) of HGPSC cases. Molecular and pathological review of mutation-negative cases showed evidence of p53 dysfunction associated with copy number gain of MDM2 or MDM4, or indicated the exclusion of samples as being low-grade serous tumours or carcinoma of uncertain primary site. Overall, p53 dysfunction rate approached 100% of confirmed HGPSCs. No association between TP53 mutation and progression-free or overall survival was found. From this first comprehensive mapping of TP53 mutation rate in a homogeneous group of HGPSC patients, we conclude that mutant TP53 is a driver mutation in the pathogenesis of HGPSC cancers. Because TP53 mutation is almost invariably present in HGPSC, it is not of substantial prognostic or predictive significance.
Collapse
Affiliation(s)
- Ahmed Ashour Ahmed
- Functional Genomics of Ovarian Cancer Laboratory, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
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: 307] [Impact Index Per Article: 20.5] [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
|
32
|
Vargas AC, Lakhani SR, Simpson PT. Pleomorphic lobular carcinoma of the breast: molecular pathology and clinical impact. Future Oncol 2009; 5:233-43. [DOI: 10.2217/14796694.5.2.233] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pleomorphic lobular carcinoma of the breast is a recently described morphological variant of classic invasive lobular carcinoma with an aggressive behavior. Morphologically, pleomorphic lobular carcinoma elicits a similar pattern of infiltrative growth as invasive lobular carcinoma, yet neoplastic cells have marked nuclear atypia and pleomorphism. Pleomorphic lobular carcinoma and the in situ counterpart, pleomorphic lobular carcinoma in situ, are frequently hormone receptor (estrogen-, progesterone- and androgen-receptor)-positive and E-cadherin and β-catenin-negative, attaining to their lobular nature. Tumors can also be positive for HER2, p53, ki67 and GCDFP-15 and harbor frequent chromosomal alterations involving gains on 1q and 16p, losses on 11q and 16q, and genomic amplifications in the region of 8q24, 11q13, 12q13, 17q12 and 20q13. Recent gene-expression profiling classified pleomorphic lobular carcinoma as ‘molecular apocrine’ tumors reflecting the frequent apocrine differentiation of the tumors. In support for the aggressive biological features described for pleomorphic lobular carcinoma, accumulating clinical data demonstrate that it has an aggressive clinical course. It is now important to define the most appropriate management strategy for patients diagnosed with pleomorphic lobular carcinoma.
Collapse
Affiliation(s)
- Ana-Cristina Vargas
- Molecular & Cellular Pathology, The University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Sunil R Lakhani
- Molecular & Cellular Pathology, The University of Queensland Centre for Clinical Research, Brisbane, Australia; School of Medicine, The University of Queensland, Brisbane, Australia; Royal Brisbane & Women’s Hospital, Brisbane, Australia
| | - Peter T Simpson
- Molecular & Cellular Pathology, University of Queensland Centre for Clinical Research, The University of Queensland, Building 71(918), Royal Brisbane & Women’s Hospital, Herston, QLD 4029, Australia
| |
Collapse
|
33
|
Hamid S, Yang YH, Peng KNL, Ismail SM, Zain RB, Lim KP, Wan Mustafa WM, Abraham MT, Teo SH, Cheong SC. MDM2 SNP309 does not confer an increased risk to oral squamous cell carcinoma but may modulate the age of disease onset. Oral Oncol 2008; 45:496-500. [PMID: 18804411 DOI: 10.1016/j.oraloncology.2008.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 05/02/2008] [Accepted: 06/24/2008] [Indexed: 12/28/2022]
Abstract
The MDM2 SNP309 has been associated with increased expression of the protein which could suppress p53 function, and has been shown to modulate risk to cancer. We have previously shown that overexpression of MDM2 is a common event in oral cancers. In the present study, we determined the association between the MDM2 SNP309 polymorphism and oral cancer in 207 oral cancer patients and 116 normal subjects. We genotyped the MDM2 SNP309 by PCR-RFLP. Logistic regression was adapted to calculate odds ratios for MDM2 SNP309 polymorphism from univariate and multivariable adjusted models. Our results suggest that MDM2 SNP309 does not confer increased risk to oral cancer (OR=1.55, 95% CI=0.77-3.11). However, the GG/TG genotype was associated with later disease onset in women above 55 years of age. Collectively, our data suggests that MDM2 SNP309 may modulate the risk to oral cancer and is a modifier of the age at oral cancer onset in women above the age of 55 years.
Collapse
Affiliation(s)
- Sharifah Hamid
- Oral Cancer Research Team, Cancer Research Initiatives Foundation (CARIF), 2nd Floor Outpatient Centre, Subang Jaya Medical Centre, 47500 Subang Jaya, Selangor, Malaysia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Simpson PT, Reis-Filho JS, Lambros MBK, Jones C, Steele D, Mackay A, Iravani M, Fenwick K, Dexter T, Jones A, Reid L, Da Silva L, Shin SJ, Hardisson D, Ashworth A, Schmitt FC, Palacios J, Lakhani SR. Molecular profiling pleomorphic lobular carcinomas of the breast: evidence for a common molecular genetic pathway with classic lobular carcinomas. J Pathol 2008; 215:231-44. [DOI: 10.1002/path.2358] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Marques YMFS, de Lima MDDM, de Melo Alves SDM, Soares FA, de Araújo VC, Pinto DDS, Mantesso A. Mdm2, p53, p21 and pAKT protein pathways in benign neoplasms of the salivary gland. Oral Oncol 2008; 44:903-8. [PMID: 18485798 DOI: 10.1016/j.oraloncology.2007.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 11/22/2007] [Accepted: 11/23/2007] [Indexed: 12/28/2022]
Abstract
The p53 protein can be altered virtually in all human cancers. In the absence of p53 mutations, p53 inactivation is possible via complex formation with other proteins, such as Mdm2. Previous studies have shown an overexpression of Mdm2 and lack of p53 expression in pleomorphic adenomas. The pAkt protein is closely related to Mdm2, and has not been previously reported in salivary gland tumors. The aim of this study was to analyze the expression of Mdm2, p53, p21 and pAkt proteins in pleomorphic adenomas and myoepitheliomas by immunohistochemistry, Western blotting and immunofluorescence techniques. Overexpression of Mdm2 and pAkt was present in all the cell lines and tumors studied, whereas the expression of p53 and p21 proteins was considered absent. In conclusion, the signaling pathway in benign salivary gland neoplasm showed an important participation of Mdm2 overexpression protein in tumor formation, progression through inactivation of p53 action, or both, and of pAkt overexpression through increased translocation of Mdm2 protein to cellular nuclei.
Collapse
|
36
|
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: 19.0] [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
|
37
|
Lønning PE, Knappskog S, Staalesen V, Chrisanthar R, Lillehaug JR. Breast cancer prognostication and prediction in the postgenomic era. Ann Oncol 2007; 18:1293-306. [PMID: 17317675 DOI: 10.1093/annonc/mdm013] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Expanding knowledge, together with implementation of new techniques, has fuelled the area of translational medical research aiming at improving prognostication as well as prediction in cancer therapy. At the same time, new discoveries have revealed a biological complexity we were unaware of only a decade ago. Thus, we are faced with novel challenges with respect to how we may explore issues such as prognostication and predict drug resistance in vivo. While microarray analysis exploring expression of thousands of genes in concert represents a major methodological advancement, discoveries such as the finding of different mechanisms of epigenetic silencing, intronic mutations, that most gene transcripts in the human genome are subject to alternative splicing and that hypersplicing seems to be a tumour-related phenomenon, exemplifies a complex pathology that may not be explored with use of single analytical methods only. This paper discusses clinical settings for studying drug resistance in vivo together with a discussion of contemporary biology in this field. Notably, each individual parameter which has been found correlated to drug resistance in vivo so far represents either a direct drug target or a factor involved in DNA repair or apoptosis. On the basis of these findings, we suggest drug resistance may be explored on the basis of upfront biological hypotheses.
Collapse
Affiliation(s)
- P E Lønning
- Section of Oncology, Institute of Medicine, University of Bergen, Norway.
| | | | | | | | | |
Collapse
|
38
|
Yang JY, Zong CS, Xia W, Wei Y, Ali-Seyed M, Li Z, Broglio K, Berry DA, Hung MC. MDM2 promotes cell motility and invasiveness by regulating E-cadherin degradation. Mol Cell Biol 2006; 26:7269-82. [PMID: 16980628 PMCID: PMC1592879 DOI: 10.1128/mcb.00172-06] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gene amplification and protein overexpression of MDM2, which is often found in certain types of cancers, indicate that MDM2 plays an important role in tumorigenesis. Interestingly, several clinical reports have demonstrated that amplification of the MDM2 gene correlates with the metastatic stage. Using an antibody array assay, we identified E-cadherin as an MDM2-binding protein and confirmed that E-cadherin is a substrate for the MDM2 E3 ubiquitin ligase. We demonstrate that MDM2 interacts in vivo with E-cadherin, resulting in its ubiquitination and degradation. This regulation appears to be clinically relevant, as we found a significant correlation between high MDM2 and low E-cadherin protein levels in resected tumor specimens recovered from breast cancer patients with lymph node metastases. Ectopic expression of MDM2 in breast cancer cells was found to disrupt cell-cell contacts and enhance cell motility and invasive potential. We found that E-cadherin and MDM2 colocalized on the plasma membrane and in the early endosome, where ubiquitin moieties were attached to E-cadherin. Blocking endocytosis with dominant-negative mutants of dynamin abolished the association of MDM2 with E-cadherin, prevented E-cadherin degradation, and attenuated cell motility as observed by fluorescence microscopy. Thus, we provide evidence to support a novel role for MDM2 in regulating cell adhesions by a mechanism that involves degrading and down-regulating the expression of E-cadherin via an endosome pathway. This novel MDM2-regulated pathway is likely to play a biologically relevant role in cancer metastasis.
Collapse
Affiliation(s)
- Jer-Yen Yang
- Department of Molecular and Cellular Oncology, University of Texas M. D. Anderson Cancer Center, Unit 108, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
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: 3.8] [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
|
40
|
Ding Y, Lee JF, Lu H, Lee MH, Yan DH. Interferon-inducible protein IFIXalpha1 functions as a negative regulator of HDM2. Mol Cell Biol 2006; 26:1979-96. [PMID: 16479015 PMCID: PMC1430239 DOI: 10.1128/mcb.26.5.1979-1996.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The 200-amino-acid repeat (HIN-200) gene family with the hematopoietic interferon (IFN)-inducible nuclear protein encodes highly homologous proteins involved in cell growth, differentiation, autoimmunity, and tumor suppression. IFIX is the newest member of the human HIN-200 family and is often downregulated in breast tumors and breast cancer cell lines. The expression of the longest isoform of IFIX gene products, IFIXalpha1, is associated with growth inhibition, suppression of transformation, and tumorigenesis. However, the mechanism underlying the tumor suppression activity of IFIXalpha1 is not well understood. Here, we show that IFIXalpha1 downregulates HDM2, a principal negative regulator of p53, at the posttranslational level. IFIXalpha1 destabilizes HDM2 protein and promotes its ubiquitination. The E3 ligase activity of HDM2 appears to be required for this IFIXalpha1 effect. Importantly, HDM2 downregulation is required for the IFIXalpha1-mediated increase of p53 protein levels, transcriptional activity, and nuclear localization, suggesting that IFIXalpha1 positively regulates p53 by acting as a negative regulator of HDM2. We found that IFIXalpha1 interacts with HDM2. Interestingly, the signature motif of the HIN-200 gene family, i.e., the 200-amino-acid HIN domain of IFIXalpha1, is sufficient not only for binding HDM2 but also for downregulating it, leading to p53 activation. Finally, we show that IFIX mediates HDM2 downregulation in an IFN-inducible system. Together, these results suggest that IFIXalpha1 functions as a tumor suppressor by repressing HDM2 function.
Collapse
Affiliation(s)
- Yi Ding
- Department of Molecular and Cellular Oncology, The University of Texas, M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Wilkening S, Bermejo JL, Burwinkel B, Klaes R, Bartram CR, Meindl A, Bugert P, Schmutzler RK, Wappenschmidt B, Untch M, Hemminki K, Försti A. The single nucleotide polymorphism IVS1+309 in mouse double minute 2 does not affect risk of familial breast cancer. Cancer Res 2006; 66:646-8. [PMID: 16423991 DOI: 10.1158/0008-5472.can-05-3168] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The mouse double minute 2 (MDM2) oncoprotein promotes cell survival and cell cycle progression by inhibiting the p53 tumor suppressor protein. Further, MDM2 overexpression can inhibit DNA double-strand break repair in a p53-independent manner. Recently, it was shown that a single nucleotide polymorphism (SNP) in the MDM2 promoter was associated with an accelerated tumor formation in individuals with a p53 mutation. The present case-control study investigated the association of this SNP (IVS1+309) with the risk and the age of onset of familial breast cancer in patients with unknown p53 mutation status. Data from 549 women affected by familial breast cancer and 1,065 healthy controls were analyzed. The cases did not carry BRCA1/2 mutations. Cases and controls showed a similar genotype distribution and the SNP did not seem to modify the age of onset of familial breast cancer. The data were also examined taking into account the presence of any additional cancer after breast cancer and the family history of cases; however, no association was found. These results suggest that the SNP IVS1+309 alone affects neither the risk nor the age of onset of heritable breast cancer.
Collapse
Affiliation(s)
- Stefan Wilkening
- Department of Molecular Genetic Epidemiology, German Cancer Research Center (Deutsches Krebsforschungszentrum), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Turbin DA, Cheang MCU, Bajdik CD, Gelmon KA, Yorida E, De Luca A, Nielsen TO, Huntsman DG, Gilks CB. MDM2 protein expression is a negative prognostic marker in breast carcinoma. Mod Pathol 2006; 19:69-74. [PMID: 16258514 DOI: 10.1038/modpathol.3800484] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The protein encoded by the MDM2 oncogene inhibits the function of p53, leading to increased cell growth, avoidance of apoptosis, tolerance of genetic instability, and resistance to chemotherapy. The present study was performed to evaluate the relationship between MDM2 protein expression and survival in breast carcinoma. Two series of cases were used in this study: the first to identify the cutoff to be used in the interpretation of MDM2 immunostaining and perform preliminary survival analysis, and a second, independent series, to validate the findings from the first series and to perform multivariate analysis. For both series, archival sections of tissue microarrays were stained with anti-MDM2 antibody (NeoMarkers, Fremont, CA, USA) and MDM2 staining intensity was scored semiquantitatively. In the first series, 49 of 362 (14%) interpretable cases were positive for MDM2 expression, with 35 (10%) showing weak positivity and 14 (4%) strong positivity. Patients with MDM2-positive tumours had a significantly worse disease-specific survival than patients with MDM2-negative tumours (P=0.0022, 10-year DSS 61% (95% CI: 45-73) vs 73% (95% CI: 67-77)). No significant difference in survival was observed between patients with strongly and weakly MDM2-positive tumours (P=0.3). Accordingly, in the independent validation series weak and strong MDM2 positivity were combined and considered to be MDM2 positive. MDM2 expression was seen in 230/1747 (13%) interpretable cases in this series, with a significant difference (P<0.0001) in DSS between MDM2-negative and MDM2-positive cases (10 year DSS 58% (95% CI: 51-64) vs 73% (95% CI: 70-75)). MDM2 was an independent prognostic marker (HR=1.35, P=0.02) in a Cox regression model including MDM2 expression, tumour grade, nodal status, ER status and tumour size. Immunohistochemical studies of MDM2 in more than 2000 breast carcinomas show that MDM2 is an independent negative prognostic marker.
Collapse
Affiliation(s)
- Dmitry A Turbin
- Genetic Pathology Evaluation Centre of the Department of Pathology, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Murray SA, Yang S, Demicco E, Ying H, Sherr DH, Hafer LJ, Rogers AE, Sonenshein GE, Xiao ZXJ. Increased expression of MDM2, cyclin D1, and p27Kip1 in carcinogen-induced rat mammary tumors. J Cell Biochem 2005; 95:875-84. [PMID: 15844214 DOI: 10.1002/jcb.20414] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
It is thought that environmental pollutants, such as polycyclic aromatic hydrocarbons (PAH), contribute to human breast tumorigenesis, yet their roles remain incompletely elucidated. The prototypical PAH 7,12-dimethylbenz(alpha)anthracene (DMBA) specifically and effectively induces mammary tumor formation in rodent models. In an attempt to explore the molecular mechanisms by which PAH initiates and promotes mammary tumorigenesis, we examined the expression of several cell cycle regulators in rat mammary tumors induced by DMBA. Expression of cyclin D1, murine double minute-2 (MDM2), and Akt was up-regulated in tumors in comparison to normal mammary glands, as indicated by RT-PCR, Western blot analysis, and immunohistochemical staining. Expression of p27Kip1 protein was also elevated in the tumors with increased cytoplasmic localization. However, RB protein remained hyperphosphorylated. To directly test the effects of DMBA, the MCF-7 human breast cancer cells were treated. DMBA induced MDM2 expression in a dose- and time-dependent fashion in the MCF-7 cells, and this activation appeared to be p53 dependent. These data suggest that activation of cyclin D1, MDM2, and AKT as well as increased expression and cytoplasmic localization of p27Kip1 may play a role in this model of environmental pollutant-induced mammary tumorigenesis.
Collapse
Affiliation(s)
- Stephen A Murray
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Genetic and biochemical evidence have demonstrated a direct link between Mdm2 and cancer development. Elevated expression of Mdm2 is observed in a significant proportion of different types of cancer. The major contribution of Mdm2 to the development of cancer is through a tight inhibition of the activities and stability of the tumor suppressor p53. However, extensive studies over the past few years have identified p53-independent functions of Mdm2, in the regulation of several important cellular processes and multiple signaling pathways. The promotion of cell cycle progression by Mdm2 is mediated via p53 inhibition, and by regulating the pRb/E2F complex. Mdm2 is an important mediator of growth and survival signaling in the PI3K/Akt pathway, an activator of certain steroid hormone receptors, and an inhibitor of the TGF-beta growth restrictive pathway. Thus, the impact on these pathways by deregulated Mdm2, as often observed in cancer, can be oncogenic in a permissible environment. This renders Mdm2 as an important target for the development of anti-cancer drugs.
Collapse
Affiliation(s)
- Yaara Levav-Cohen
- The Hebrew University Hadassah Medical School, Lautenberg Center for General and Tumor Immunology, Jerusalem, Israel
| | | | | |
Collapse
|
45
|
Alkhalaf M, El-Mowafy AM, Abou-Zeid LA. Progesterone inhibition of MDM2 p90 protein in MCF-7 human breast cancer cell line is dependent on p53 levels. J Mol Genet Med 2005; 1:33-7. [PMID: 19565011 PMCID: PMC2702060 DOI: 10.4172/1747-0862.1000008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 06/16/2005] [Accepted: 06/20/2005] [Indexed: 11/11/2022] Open
Abstract
The mdm2 gene encodes several protein isoforms with different molecular weights (p90, p80, p76 and p57). MDM2 p90 (usually considered to be the major MDM2 protein) binds to and inactivates P53. We have recently shown that growth inhibition of MCF-7 human breast cancer cells by progesterone is associated with P53 down-regulation. In this work, we analyzed the expression pattern of MDM2 proteins in three human breast cancer cell lines by western blotting with anti-MDM2 antibodies. We found a prominent expression of MDM2 p57 protein in cell lines which have non-functional P53 protein (T47D and MDA-MB-231) as compared to the p90, p80 isoforms, whereas p90 was the major protein isoform in MCF-7 cells that contain functional P53 protein. When MCF-7 cells were treated with 100 nM of progesterone, MDM2 p90 was inhibited but the highly expressed MDM2 p57 isoform was not. The inhibition of MDM2 p90 protein by progesterone was abrogated in MCF-7 cells transfected with a P53 expressing vector. To our knowledge, this is the first report linking progesterone-induced growth inhibition with down-regulation of the MDM2 protein. We present evidence that reestablishing of P53 expression by transient transfection of P53 cDNA in these cells enhances the expression level of MDM2 p90 isoform. The data indicate that expression of MDM2 p90 is regulated through a P53-dependent pathway in response to progesterone.
Collapse
Affiliation(s)
- Moussa Alkhalaf
- Department of Biochemistry, Faculty of Medicine, and Department of Applied Therapeutics, Faculty of Pharmacy, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| | | | | |
Collapse
|
46
|
Pinto AE, André S, Laranjeira C, Soares J. Correlations of cell cycle regulators (p53, p21, pRb and mdm2) and c-erbB-2 with biological markers of proliferation and overall survival in breast cancer. Pathology 2005; 37:45-50. [PMID: 15875733 DOI: 10.1080/00313020400011250] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AIM The biological impact of cell cycle regulatory proteins on breast cancer progression is widely recognised, although mostly unclear. The aim of this preliminary study was to investigate the correlations of several cell cycle modulators (p53, p21, pRb, and mdm2) and c-erbB-2 expression with cell proliferation markers (S-phase fraction [SPF] and Ki-67) and overall survival in breast cancer. METHODS The series comprised 50 women with stage I-II invasive ductal breast carcinoma (median follow-up 87 months), who were selected for their tumour proliferative characteristics (15 low, 15 high, and 20 intermediate proliferative tumours). Tumour differentiation was assessed following the Nottingham grading criteria. Cell cycle regulators, oestrogen receptor status, and Ki-67 index were analysed by immunohistochemistry on paraffin embedded material (cut-offs 10%). c-erbB-2 was evaluated according to a standardised immunohistochemical assay and borderline cases were confirmed by FISH analysis. Ploidy and SPF were determined by DNA flow cytometry on frozen samples. Chi-square test and Fisher's exact test were applied to analyse the statistical significance of data. RESULTS Positive immunostaining was observed in nine (18%) p53+, 30 (60%) p21+, 13 (26%) pRb+, and one (2%) mdm2+ cases. c-erbB-2 expression was considered positive in 11 (22%) cases. In the subset of patients dead of the disease, a high incidence of c-erbB-2 over-expression (7/10, 70%) was verified. In general, no significant correlations among cell cycle regulators or between the latter and histopathological or proliferative characteristics were found. Only the p53-/p21+ phenotype significantly correlated with low SPF (p=0.048), and p21 positivity showed a trend to be associated with low SPF (p=0.083). No statistically significant correlations between cell cycle inhibitors and clinical outcome were found. On the contrary, c-erbB-2 over-expression showed significant correlations with DNA aneuploidy (p<0.001), high SPF (p<0.001), high tumour grading (p=0.008), lack of oestrogen receptors (p=0.036), and poor overall survival (p<0.001). CONCLUSIONS The results seem to indicate the lack of correlations of cell cycle regulatory proteins with cell proliferation markers and overall survival in breast cancer, in contrast to c-erbB-2 over-expression which was found to be associated with increased proliferation rate and worse prognosis.
Collapse
Affiliation(s)
- António E Pinto
- Departamento de Patologia Morfológica, Instituto Português de Oncologia de Francisco Gentil, Centro Regional Oncológico de Lisboa, SA, Portugal.
| | | | | | | |
Collapse
|
47
|
Zhou R, Frum R, Deb S, Deb SP. The growth arrest function of the human oncoprotein mouse double minute-2 is disabled by downstream mutation in cancer cells. Cancer Res 2005; 65:1839-48. [PMID: 15753382 DOI: 10.1158/0008-5472.can-03-3755] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have reported earlier that ectopic expression of mouse double minute-2 (MDM2) induces G1 arrest in normal cells. To explain occasional overexpression of MDM2 in cancer cells, we searched for deletion or substitution mutation in the growth suppressor domains of MDM2 in several breast cancer cell lines that overexpress the oncoprotein. Our results suggest the absence of alteration (deletion or substitution) in the open reading frame of MDM2 transcripts in such cells. Because the breast cancer cell line MCF-7 overexpresses MDM2, we isolated the full-length MDM2 transcript from this cell line. The MDM2 cDNA synthesized from transcripts isolated from MCF-7 cells induced inhibition of G1 to S phase transition in normal human diploid cells such as WI38, suggesting that the genetic alterations in breast cancer cells that overexpress MDM2 disable the growth arrest function of the oncoprotein. Consistently, overexpression of full-length MDM2 in MCF-7 cells over its high endogenous level did not inhibit G1-S transition efficiently. Although MDM2 overexpression was accompanied by CDK4 overexpression or absence of cdk4 inhibitor p16 in most breast cancer cells, we found remarkably high levels of cyclin A rather than cyclin E in these cells. Ectopic expression of cyclin A released MDM2-mediated inhibition of G1-S transition in normal human diploid WI38 cells. We propose that cancer cells expressing high levels of cyclin A escape MDM2-mediated G1 arrest, which may account for a selective growth advantage over normal cells.
Collapse
Affiliation(s)
- Ruizhe Zhou
- Department of Biochemistry and the Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298-0614, USA
| | | | | | | |
Collapse
|
48
|
Al-Kuraya K, Schraml P, Torhorst J, Tapia C, Zaharieva B, Novotny H, Spichtin H, Maurer R, Mirlacher M, Köchli O, Zuber M, Dieterich H, Mross F, Wilber K, Simon R, Sauter G. Prognostic relevance of gene amplifications and coamplifications in breast cancer. Cancer Res 2005; 64:8534-40. [PMID: 15574759 DOI: 10.1158/0008-5472.can-04-1945] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple different oncogenes have been described previously to be amplified in breast cancer including HER2, EGFR, MYC, CCND1, and MDM2. Gene amplification results in oncogene overexpression but may also serve as an indicator of genomic instability. As such, presence of one or several gene amplifications may have prognostic significance. To assess the prognostic importance of amplifications and coamplifications of HER2, EGFR, MYC, CCND1, and MDM2 in breast cancer, we analyzed a breast cancer tissue microarray containing samples from 2197 cancers with follow-up information. Fluorescence in situ hybridizations revealed amplifications of CCND1 in 20.1%, HER2 in 17.3%, MDM2 in 5.7%, MYC in 5.3%, and EGFR in 0.8% of the tumors. All gene amplifications were significantly associated with high grade. HER2 (P < 0.001) and MYC amplification (P < 0.001) were also linked to shortened survival. In case of HER2, this was independent of grade, pT, and pN categories. MYC amplification was almost 3 times more frequent in medullary cancer (15.9%), than in the histologic subtype with the second highest frequency (ductal; 5.6%; P = 0.0046). HER2 and MYC amplification were associated with estrogen receptor/progesterone receptor negativity (P < 0.001) whereas CCND1 amplification was linked to estrogen receptor/progesterone receptor positivity (P < 0.001). Coamplifications were more prevalent than expected based on the individual frequencies. Coamplifications of one or several other oncogenes occurred in 29.6% of CCND1, 43% of HER2, 55.7% of MDM2, 65% of MYC, and 72.8% of EGFR-amplified cancers. HER2/MYC-coamplified cancers had a worse prognosis than tumors with only one of these amplifications. Furthermore, a gradual decrease of survival was observed with increasing number of amplifications. In conclusion, these data support a major prognostic impact of genomic instability as determined by a broad gene amplification survey in breast cancer.
Collapse
Affiliation(s)
- Khawla Al-Kuraya
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Dornan D, Bheddah S, Newton K, Ince W, Frantz GD, Dowd P, Koeppen H, Dixit VM, French DM. COP1, the negative regulator of p53, is overexpressed in breast and ovarian adenocarcinomas. Cancer Res 2004; 64:7226-30. [PMID: 15492238 DOI: 10.1158/0008-5472.can-04-2601] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumor suppressor protein p53 plays a central role in protecting normal cells from undergoing transformation. Thus, it is fitting that cancer cells selectively dampen the p53 response to gain a selective growth advantage. In fact, the p53 gene is the most commonly mutated tumor suppressor gene in human cancers, and if the gene is not mutated, then other components of the p53 pathways are skewed to dampen the p53 response to stress. We recently identified COP1 as a novel and critical negative regulator of p53. COP1 is a RING finger-containing protein that targets p53 for degradation to the proteasome and is necessary for p53 turnover in normal and cancer cells. However, the association between COP1 and cancer remains to be determined. We performed expression analysis of COP1 in ovarian and breast cancer tissue microarrays. COP1 is significantly overexpressed in 81% (25 of 32) of breast and 44% (76 of 171) of ovarian adenocarcinoma as assessed by in situ hybridization and immunohistochemistry. Overexpression of COP1 correlated with a striking decrease in steady state p53 protein levels and attenuation of the downstream target gene, p21, in cancers that retain a wild-type p53 gene status. Overall, these results suggest that overexpression of COP1 contributes to the accelerated degradation of p53 protein in cancers and attenuates the tumor suppressor function of p53.
Collapse
Affiliation(s)
- David Dornan
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang Z, Wang H, Prasad G, Li M, Yu D, Bonner JA, Agrawal S, Zhang R. Radiosensitization by antisense anti-MDM2 mixed-backbone oligonucleotide in in vitro and in vivo human cancer models. Clin Cancer Res 2004; 10:1263-73. [PMID: 14977824 DOI: 10.1158/1078-0432.ccr-0245-03] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE The MDM2 oncogene, amplified or overexpressed in many human cancers, has been suggested to be a novel target for cancer therapy. We have demonstrated a second-generation antisense antihuman-MDM2 oligonucleotide to have antitumor activity when administered alone or in combination with cancer chemotherapeutic agents. In the present study, we investigated the effect of the antisense oligonucleotide on radiation therapy. EXPERIMENTAL DESIGN The in vitro radiosensitization activity was determined in cell lines of human cancers of prostate (LNCaP and PC3), breast (MCF-7 and MDA-MB-468), pancreas (PANC-1), and glioma (U87-MG and A172) and its in vivo radiosensitization activity in xenograft models of LNCaP, PC3, MCF-7, MDA-MB-468, and PANC-1. RESULTS In cells containing at least one functional p53 allele (LNCaP, U87-MG, and A172), after specific inhibition of MDM2 expression, p53 and p21 levels were elevated. In LNCaP cells, the Bax level was increased, and Bcl-2 and E2F1 levels were decreased. In PC3 cells that are p53 null, after inhibition of MDM2 expression, Bax and p21 levels were elevated, and E2F1 levels were decreased. On the basis of in vitro clonogenic assay, the antisense oligonucleotide, in a sequence-specific manner, significantly increased radiation-induced antiproliferation effects. It also increased radiation-induced inhibitory effects on tumor growth in SCID or nude mice bearing LNCaP, PC3, MCF-7, MDA-MB-468, and PANC-1 xenografts. CONCLUSIONS These results suggest that MDM2 has a role in radiation therapy of human cancers, regardless of p53 status, providing a basis for future development of MDM2 inhibitors, such as antisense oligonucleotides, as radiosensitizers.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | | | | | | | | | | | |
Collapse
|