1
|
Yi J, Tan S, Zeng Y, Zou L, Zeng J, Zhang C, Liu L, Fan P. Comprehensive Analysis of Prognostic and immune infiltrates for FOXPs Transcription Factors in Human Breast Cancer. Sci Rep 2022; 12:8896. [PMID: 35614183 PMCID: PMC9132954 DOI: 10.1038/s41598-022-12954-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/18/2022] [Indexed: 01/11/2023] Open
Abstract
Forkhead-box-P family include FOXP1/2/3/4 and its clinical significance still remains unclear in breast cancer (BRCA). We analysed the expressions of FOXPs in BRCA patients to determine diagnostic and prognostic values. Our results indicated that the transcriptional levels of FOXP3/4 were up-regulated in BRCA patients, but FOXP2 were down-regulated. No statistically significant correlation were found between the expression levels of FOXPs in Pathologic stage. FOXP2/3 had a significantly high AUC value in the detection of breast cancer, with 96.8% or 95.7% in accuracy respectively. Our study also suggested that BRCA patients with high transcription levels of FOXP1/2/4 were significantly associated with longer Overall Survival (OS). In contrast, BRCA patients with high transcription level of FOXP3 was not statistically related with OS. Our work revealed that FOXPs were closely related to the alteration of extensive immune checkpoints in breast invasive carcinoma. Additionally, FOXP3 has a significant positive correlation with PDCD1, CD274, CTLA4 and TMB in breast cancer, and FOXP3 expression showed a statistically significant correlation with infiltration of immune cells. Finally, we found that FOXP3 expression predicted the breast cancer cells response to anticancer drugs. Altogether, our work strongly suggested that FOXPs could serve as a biomarker for tumor detection, therapeutic design and prognosis.
Collapse
Affiliation(s)
- Jianing Yi
- Surgical Department of Breast and Thyroid Gland, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Siyi Tan
- Surgical Department of Medical Laboratory, Huazhi Biotechnology Co. Ltd, Changsha, Hunan, People's Republic of China
| | - Yuanjun Zeng
- Department of Pathology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Lianhong Zou
- Institute of Translational Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Jie Zeng
- Surgical Department of Breast and Thyroid Gland, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Chaojie Zhang
- Surgical Department of Breast and Thyroid Gland, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Luyao Liu
- Surgical Department of Breast and Thyroid Gland, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China.
| | - Peizhi Fan
- Surgical Department of Breast and Thyroid Gland, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Taouis K, Driouch K, Lidereau R, Lallemand F. Molecular Functions of WWOX Potentially Involved in Cancer Development. Cells 2021; 10:cells10051051. [PMID: 33946771 PMCID: PMC8145924 DOI: 10.3390/cells10051051] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 12/16/2022] Open
Abstract
The WW domain-containing oxidoreductase gene (WWOX) was cloned 21 years ago as a putative tumor suppressor gene mapping to chromosomal fragile site FRA16D. The localization of WWOX in a chromosomal region frequently altered in human cancers has initiated multiple current studies to establish its role in this disease. All of this work suggests that WWOX, due to its ability to interact with a large number of partners, exerts its tumor suppressive activity through a wide variety of molecular actions that are mostly cell specific.
Collapse
|
3
|
Migocka-Patrzałek M, Elias M. Muscle Glycogen Phosphorylase and Its Functional Partners in Health and Disease. Cells 2021; 10:cells10040883. [PMID: 33924466 PMCID: PMC8070155 DOI: 10.3390/cells10040883] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 02/07/2023] Open
Abstract
Glycogen phosphorylase (PG) is a key enzyme taking part in the first step of glycogenolysis. Muscle glycogen phosphorylase (PYGM) differs from other PG isoforms in expression pattern and biochemical properties. The main role of PYGM is providing sufficient energy for muscle contraction. However, it is expressed in tissues other than muscle, such as the brain, lymphoid tissues, and blood. PYGM is important not only in glycogen metabolism, but also in such diverse processes as the insulin and glucagon signaling pathway, insulin resistance, necroptosis, immune response, and phototransduction. PYGM is implicated in several pathological states, such as muscle glycogen phosphorylase deficiency (McArdle disease), schizophrenia, and cancer. Here we attempt to analyze the available data regarding the protein partners of PYGM to shed light on its possible interactions and functions. We also underline the potential for zebrafish to become a convenient and applicable model to study PYGM functions, especially because of its unique features that can complement data obtained from other approaches.
Collapse
|
4
|
Charan M, Verma AK, Hussain S, Misri S, Mishra S, Majumder S, Ramaswamy B, Ahirwar D, Ganju RK. Molecular and Cellular Factors Associated with Racial Disparity in Breast Cancer. Int J Mol Sci 2020; 21:ijms21165936. [PMID: 32824813 PMCID: PMC7460595 DOI: 10.3390/ijms21165936] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that racial differences can influence breast cancer incidence and survival rate. African American (AA) women are at two to three fold higher risk for breast cancer than other ethnic groups. AA women with aggressive breast cancers show worse prognoses and higher mortality rates relative to Caucasian (CA) women. Over the last few years, effective treatment strategies have reduced mortality from breast cancer. Unfortunately, the breast cancer mortality rate among AA women remains higher compared to their CA counterparts. The focus of this review is to underscore the racial differences and differential regulation/expression of genetic signatures in CA and AA women with breast cancer. Moreover, immune cell infiltration significantly affects the clinical outcome of breast cancer. Here, we have reviewed recent findings on immune cell recruitment in the tumor microenvironment (TME) and documented its association with breast cancer racial disparity. In addition, we have extensively discussed the role of cytokines, chemokines, and other cell signaling molecules among AA and CA breast cancer patients. Furthermore, we have also reviewed the distinct genetic and epigenetic changes in AA and CA patients. Overall, this review article encompasses various molecular and cellular factors associated with breast cancer disparity that affects mortality and clinical outcome.
Collapse
Affiliation(s)
- Manish Charan
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Ajeet K. Verma
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Shahid Hussain
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Swati Misri
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Sanjay Mishra
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Sarmila Majumder
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| | - Dinesh Ahirwar
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
- Correspondence: (D.A.); (R.K.G.)
| | - Ramesh K. Ganju
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
- Correspondence: (D.A.); (R.K.G.)
| |
Collapse
|
5
|
Xiao Z, Wei Z, Deng D, Zheng Z, Zhao Y, Jiang S, Zhang D, Zhang LJ, Fan M, Chen S, Wang S, Ding Y, Ye Y, Jiao H. Downregulation of Siah1 promotes colorectal cancer cell proliferation and migration by regulating AKT and YAP ubiquitylation and proteasome degradation. Cancer Cell Int 2020; 20:50. [PMID: 32082080 PMCID: PMC7020597 DOI: 10.1186/s12935-020-1124-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 01/25/2020] [Indexed: 12/11/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common malignant tumors in the world. Siah E3 ubiquitin protein ligase 1 (Siah1) has been identified as a tumor suppressor gene and plays an important role in the development of malignant tumors. However, the potential role and molecular mechanism of Siah1 in the development and progression of CRC is still unclear. Methods To explore the role and molecular mechanism of Siah1 in the development and progression of CRC, we examined the expression of Siah1 in CRC tissue samples and analyzed its association with progression and prognosis in CRC. In addition, overexpression and knockdown of Siah1 was used to investigate its activity in CRC cells. We also use bioinformatics to analyze and verify the significant roles of Siah1 in critical signaling pathways of CRC. Results We found that the expression of Siah1 was significantly downregulated in CRC tissues, and low expression of Siah1 was associated with aggressive TNM staging and poor survival of CRC patients. Moreover, we revealed that overexpression of Siah1 in CRC cells markedly inhibited CRC cell proliferation and invasion in vitro and in vivo, while knockdown of Siah1 enhanced CRC cell proliferation and invasion. Furthermore, we found that Siah1 prohibited cell proliferation and invasion in CRC partially through promoting AKT (the serine-threonine protein kinase) and YAP (yes associated protein) ubiquitylation and proteasome degradation to regulate the activity of MAPK(mitogen-activated protein kinase 1), PI3K-AKT (phosphatidylinositol 3-kinase-the serine-threonine protein kinase) and Hippo signaling pathways. Conclusions These findings suggested that Siah1 is a novel potential prognostic biomarker and plays a tumor suppressor role in the development and progression of CRC.
Collapse
Affiliation(s)
- Zhiyuan Xiao
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.,3Department of Pathology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, Guangdong China
| | - Zhigang Wei
- 4Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Danling Deng
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.,Department of Pathology, Shaoyang Central Hospital, Affiliated Shaoyang Hospital of University of South China, Shaoyang, Hunan China
| | - Zhe Zheng
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yali Zhao
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Shenglu Jiang
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Dan Zhang
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Ling-Jie Zhang
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Mingmei Fan
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Siqi Chen
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - ShuYang Wang
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yanqing Ding
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yaping Ye
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Hongli Jiao
- 1Department of Pathology, Nanfang Hospital and School of Basic Medical Science, Southern Medical University, Guangzhou, 510515 China.,2Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| |
Collapse
|
6
|
Kim JH, Hwang J, Jung JH, Lee HJ, Lee DY, Kim SH. Molecular networks of FOXP family: dual biologic functions, interplay with other molecules and clinical implications in cancer progression. Mol Cancer 2019; 18:180. [PMID: 31815635 PMCID: PMC6900861 DOI: 10.1186/s12943-019-1110-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023] Open
Abstract
Though Forkhead box P (FOXP) transcription factors comprising of FOXP1, FOXP2, FOXP3 and FOXP4 are involved in the embryonic development, immune disorders and cancer progression, the underlying function of FOXP3 targeting CD4 + CD25+ regulatory T (Treg) cells and the dual roles of FOXP proteins as an oncogene or a tumor suppressor are unclear and controversial in cancers to date. Thus, the present review highlighted research history, dual roles of FOXP proteins as a tumor suppressor or an oncogene, their molecular networks with other proteins and noncoding RNAs, cellular immunotherapy targeting FOXP3, and clinical implications in cancer progression.
Collapse
Affiliation(s)
- Ju-Ha Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee university, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Jisung Hwang
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee university, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Ji Hoon Jung
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee university, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Hyo-Jung Lee
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee university, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, Rural Development Administration, National Institute of Horticultural and Herbal Science, Eumseong, 27709, Republic of Korea
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee university, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
7
|
Seachrist DD, Keri RA. The Activin Social Network: Activin, Inhibin, and Follistatin in Breast Development and Cancer. Endocrinology 2019; 160:1097-1110. [PMID: 30874767 PMCID: PMC6475112 DOI: 10.1210/en.2019-00015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022]
Abstract
Activins and inhibins are closely related protein heterodimers with a similar tissue distribution; however, these two complexes have opposing functions in development and disease. Both are secreted cytokine hormones, with activin the primary inducer of downstream signaling cascades and inhibin acting as a rheostat that exquisitely governs activin function. Adding to the complexity of activin signaling, follistatin, a highly glycosylated monomeric protein, binds activin with high affinity and restrains downstream pathway activation but through a mechanism distinct from that of inhibin. These three proteins were first identified as key ovarian hormones in the pituitary-gonadal axis that direct the synthesis and secretion of FSH from the pituitary, hence controlling folliculogenesis. Research during the past 30 years has expanded the roles of these proteins, first by discovering the ubiquitous expression of the trio and then by implicating them in a wide array of biological functions. In concert, these three hormones govern tissue development, homeostasis, and disease in multiple organ systems through diverse autocrine and paracrine mechanisms. In the present study, we have reviewed the actions of activin and its biological inhibitors, inhibin, and follistatin, in mammary gland morphogenesis and cancer.
Collapse
Affiliation(s)
- Darcie D Seachrist
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Ruth A Keri
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
- Division of General Medical Sciences–Oncology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
8
|
Role of Hedgehog Signaling in Breast Cancer: Pathogenesis and Therapeutics. Cells 2019; 8:cells8040375. [PMID: 31027259 PMCID: PMC6523618 DOI: 10.3390/cells8040375] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related mortality in women, only followed by lung cancer. Given the importance of BC in public health, it is essential to identify biomarkers to predict prognosis, predetermine drug resistance and provide treatment guidelines that include personalized targeted therapies. The Hedgehog (Hh) signaling pathway plays an essential role in embryonic development, tissue regeneration, and stem cell renewal. Several lines of evidence endorse the important role of canonical and non-canonical Hh signaling in BC. In this comprehensive review we discuss the role of Hh signaling in breast development and homeostasis and its contribution to tumorigenesis and progression of different subtypes of BC. We also examine the efficacy of agents targeting different components of the Hh pathway both in preclinical models and in clinical trials. The contribution of the Hh pathway in BC tumorigenesis and progression, its prognostic role, and its value as a therapeutic target vary according to the molecular, clinical, and histopathological characteristics of the BC patients. The evidence presented here highlights the relevance of the Hh signaling in BC, and suggest that this pathway is key for BC progression and metastasis.
Collapse
|
9
|
Loss of Heterozygosity (LOH) at 17p13 and 22q13 are Shared by Breast and Thyroid Carcinomas for Metastasis. Appl Immunohistochem Mol Morphol 2019; 27:e16-e19. [DOI: 10.1097/pai.0000000000000630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Fleming CA, McCarthy K, Ryan C, McCarthy A, O'Reilly S, O'Mahony D, Browne TJ, Redmond P, Corrigan MA. Evaluation of Discordance in Primary Tumor and Lymph Node Response After Neoadjuvant Therapy in Breast Cancer. Clin Breast Cancer 2017; 18:e255-e261. [PMID: 29246703 DOI: 10.1016/j.clbc.2017.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Neoadjuvant therapy (NAT) offers a unique opportunity to assess tumor response to systemic agents. However, a discrepancy may exist between the response of the primary tumor and involved nodes. We report on the frequency of response discordance after NAT in breast cancer. PATIENTS AND METHODS All consecutive node-positive patients receiving NAT in our department from 2009 to 2014 were identified. Patient demographics, and radiologic and pathologic features were tabulated. Tumor response was estimated by magnetic resonance imaging of the breast. Lymph node (LN) response was estimated from pathologic treatment response measurements. Statistical analysis was performed. RESULTS A total of 108 node-positive patients treated with NAT were eligible for inclusion. Median age was 51.73 years (range, 20-87 years). All patients underwent axillary clearance, and 62% underwent mastectomy. A 40% mean reduction in tumor size was observed. Statistically, a positive correlation between tumor and LN response after NAT was observed (Spearman correlation coefficient, r = 0.46, P < .001). Complete pathologic response was observed in 17 patients (15.7%). However, 21 patients experienced complete LN response, with only 81% of these patients (n = 17) experiencing a complete response in tumor also. A complete response was observed in tumor in 20 patients, and this predicted complete nodal response in 85% of cases (n = 17). Fifteen percent of primary tumors with complete pathologic response had persistently positive LNs. CONCLUSION A significant discordance exists between the primary tumor and LN response, representing a concern for the lack of response of occult regional or systemic metastases due to potential biologic heterogeneity.
Collapse
Affiliation(s)
| | - Karen McCarthy
- Breast Research Centre, Cork University Hospital, Cork, Ireland
| | - Ciara Ryan
- Department of Histopathology, Cork University Hospital, Cork, Ireland
| | - Aoife McCarthy
- Department of Histopathology, Cork University Hospital, Cork, Ireland
| | - Seamus O'Reilly
- Breast Research Centre, Cork University Hospital, Cork, Ireland
| | | | - Tara Jane Browne
- Department of Histopathology, Cork University Hospital, Cork, Ireland
| | - Paul Redmond
- Breast Research Centre, Cork University Hospital, Cork, Ireland
| | - Mark A Corrigan
- Breast Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|
11
|
Alexe G, Dalgin G, Ramaswamy R, Delisi C, Bhanot G. Data Perturbation Independent Diagnosis and Validation of Breast Cancer Subtypes Using Clustering and Patterns. Cancer Inform 2017. [DOI: 10.1177/117693510600200006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Molecular stratification of disease based on expression levels of sets of genes can help guide therapeutic decisions if such classifications can be shown to be stable against variations in sample source and data perturbation. Classifications inferred from one set of samples in one lab should be able to consistently stratify a different set of samples in another lab. We present a method for assessing such stability and apply it to the breast cancer (BCA) datasets of Sorlie et al. 2003 and Ma et al. 2003. We find that within the now commonly accepted BCA categories identified by Sorlie et al. Luminal A and Basal are robust, but Luminal B and ERBB2+ are not. In particular, 36% of the samples identified as Luminal B and 55% identified as ERBB2+ cannot be assigned an accurate category because the classification is sensitive to data perturbation. We identify a “core cluster” of samples for each category, and from these we determine “patterns” of gene expression that distinguish the core clusters from each other. We find that the best markers for Luminal A and Basal are (ESR1, LIV1, GATA-3) and (CCNE1, LAD1, KRT5), respectively. Pathways enriched in the patterns regulate apoptosis, tissue remodeling and the immune response. We use a different dataset (Ma et al. 2003) to test the accuracy with which samples can be allocated to the four disease subtypes. We find, as expected, that the classification of samples identified as Luminal A and Basal is robust but classification into the other two subtypes is not.
Collapse
Affiliation(s)
- G. Alexe
- Computational Biology Center, IBM Thomas J. Watson Research Center, Yorktown Heights, NY 10598, U.S.A
- The Simons Center for Systems Biology, Institute for Advanced Study, Princeton NJ 08540, U.S.A
| | - G.S. Dalgin
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, 2 Cummington Street, Boston, MA 02215, U.S.A
| | - R. Ramaswamy
- The Simons Center for Systems Biology, Institute for Advanced Study, Princeton NJ 08540, U.S.A
- School of Information Technology, Jawaharlal Nehru University, New Delhi 110 067, India
| | - C. Delisi
- Biomedical Engineering, Boston University, 44 Cummington Street, Boston, MA 02215, U.S.A
| | - G. Bhanot
- Computational Biology Center, IBM Thomas J. Watson Research Center, Yorktown Heights, NY 10598, U.S.A
- The Simons Center for Systems Biology, Institute for Advanced Study, Princeton NJ 08540, U.S.A
- Biomedical Engineering, Boston University, 44 Cummington Street, Boston, MA 02215, U.S.A
- Department of Biomedical Engineering and BioMaPS Institute, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
12
|
Yedjou CG, Tchounwou PB, Payton M, Miele L, Fonseca DD, Lowe L, Alo RA. Assessing the Racial and Ethnic Disparities in Breast Cancer Mortality in the United States. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:E486. [PMID: 28475137 PMCID: PMC5451937 DOI: 10.3390/ijerph14050486] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/22/2017] [Accepted: 04/26/2017] [Indexed: 01/01/2023]
Abstract
Breast cancer is the second leading cause of cancer related deaths among women aged 40-55 in the United States and currently affects more than one in ten women worldwide. It is also one of the most diagnosed cancers in women both in wealthy and poor countries. Fortunately, the mortality rate from breast cancer has decreased in recent years due to increased emphasis on early detection and more effective treatments in White population. Although the mortality rates have declined in some ethnic populations, the overall cancer incidence among African American and Hispanic populations has continued to grow. The goal of the present review article was to highlight similarities and differences in breast cancer morbidity and mortality rates primarily among African American women compared to White women in the United States. To reach our goal, we conducted a search of articles in journals with a primary focus on minority health, and authors who had published articles on racial/ethnic disparity related to breast cancer patients. A systematic search of original research was conducted using MEDLINE, PUBMED and Google Scholar databases. We found that racial/ethnic disparities in breast cancer may be attributed to a large number of clinical and non-clinical risk factors including lack of medical coverage, barriers to early detection and screening, more advanced stage of disease at diagnosis among minorities, and unequal access to improvements in cancer treatment. Many African American women have frequent unknown or unstaged breast cancers than White women. These risk factors may explain the differences in breast cancer treatment and survival rate between African American women and White women. New strategies and approaches are needed to promote breast cancer prevention, improve survival rate, reduce breast cancer mortality, and ultimately improve the health outcomes of racial/ethnic minorities.
Collapse
Affiliation(s)
- Clement G Yedjou
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Paul B Tchounwou
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Marinelle Payton
- Center of Excellence in Minority Health and Health Disparities, School of Public Health, Jackson State University, Jackson Medical Mall-Thad Cochran Center, 350 West Woodrow Wilson Avenue, Jackson, MS 39213, USA.
| | - Lucio Miele
- Department of Genetics, LSU Health Sciences Center, School of Medicine, 533 Bolivar Street, Room 657, New Orleans, LA 70112, USA.
| | - Duber D Fonseca
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Leroy Lowe
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4YW, UK.
| | - Richard A Alo
- Department of Civil and Environmental Engineering, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| |
Collapse
|
13
|
Zhang M, Wang B, Xu J, Wang X, Xie L, Zhang B, Li Y, Li J. CanProVar 2.0: An Updated Database of Human Cancer Proteome Variation. J Proteome Res 2017; 16:421-432. [PMID: 27977206 PMCID: PMC5515284 DOI: 10.1021/acs.jproteome.6b00505] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Identification and annotation of the mutations involved in oncogenesis and tumor progression are crucial for both cancer biology and clinical applications. Previously, we developed a public resource CanProVar, a human cancer proteome variation database for storing and querying single amino acid alterations in the human cancers. Since the publication of CanProVar, extensive cancer genomics efforts have revealed the enormous genomic complexity of various types of human cancers. Thus, there is an overwhelming need for comprehensive annotation of the genomic alterations at the protein level and making such knowledge easily accessible. Here, we describe CanProVar 2.0, a significantly expanded version of CanProVar, in which the amount of cancer-related variations and noncancer specific variations was increased by about 10-fold as compared to the previous version. To facilitate the interpretation of the variations, we added to the database functional data on potential impact of the cancer-related variations on 3D protein interaction and on the differential expression of the variant-bearing proteins between cancer and normal samples. The web interface allows for flexible queries based on gene or protein IDs, cancer types, chromosome locations, or pathways. An integrated protein sequence database containing variations that can be directly used for proteomics database searching can be downloaded.
Collapse
Affiliation(s)
- Menghuan Zhang
- Department of Bioinformatics & Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Bo Wang
- Department of Bioinformatics & Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jia Xu
- Department of Bioinformatics & Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaojing Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Suite NB100, Houston, TX 77030
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Suite NB100, Houston, TX 77030
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Bing Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Suite NB100, Houston, TX 77030
- Lester & Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Suite NB100, Houston, TX 77030
| | - Yixue Li
- Department of Bioinformatics & Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
- Key Lab of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Li
- Department of Bioinformatics & Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| |
Collapse
|
14
|
Mogilyansky E, Clark P, Quann K, Zhou H, Londin E, Jing Y, Rigoutsos I. Post-transcriptional Regulation of BRCA2 through Interactions with miR-19a and miR-19b. Front Genet 2016; 7:143. [PMID: 27630665 PMCID: PMC5005319 DOI: 10.3389/fgene.2016.00143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/21/2016] [Indexed: 12/31/2022] Open
Abstract
Breast cancer type 2, early onset susceptibility gene (BRCA2) is a major component of the homologous recombination DNA repair pathway. It acts as a tumor suppressor whose function is often lost in cancers. Patients with specific mutations in the BRCA2 gene often display discrete clinical, histopathological, and molecular features. However, a subset of sporadic cancers has wild type BRCA2 and display defects in the homology-directed repair pathway, which is the hallmark of ‘BRCAness.’ The mechanisms by which BRCAness arises are not well understood but post-transcriptional regulation of BRCA2 gene expression by microRNAs (miRNAs) may contribute to this phenotype. Here, we examine the post-transcriptional effects that some members of the six-miRNA cluster known as the miR-17/92 cluster have on the abundance of BRCA2’s messenger RNA (mRNA) and protein. We discuss two interactions involving the miR-19a and miR-19b members of the cluster and the 3′UTR of BRCA2’s mRNA. We investigated these miRNA:mRNA interactions in 15 cell lines derived from pancreatic, breast, colon, and kidney tissue. We show that over-expression of these two miRNAs results in a concomitant decrease of BRCA2’s mRNA and protein expression in a subset of the tested cell lines. Additionally, using luciferase reporter assays we identified direct interactions between miR-19a/miR-19b and a miRNA response element (MRE) in BRCA2’s 3′UTR. Our results suggest that BRCA2 is subject to a complex post-transcriptional regulatory program that has specific dependencies on the genetic and phenotypic background of cell types.
Collapse
Affiliation(s)
- Elena Mogilyansky
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Peter Clark
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia PA, USA
| | - Kevin Quann
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Honglei Zhou
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Eric Londin
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Yi Jing
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Isidore Rigoutsos
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| |
Collapse
|
15
|
Lehman HL, Stairs DB. Single and Multiple Gene Manipulations in Mouse Models of Human Cancer. CANCER GROWTH AND METASTASIS 2015; 8:1-15. [PMID: 26380553 PMCID: PMC4558888 DOI: 10.4137/cgm.s21217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/17/2015] [Accepted: 06/20/2015] [Indexed: 12/14/2022]
Abstract
Mouse models of human cancer play a critical role in understanding the molecular and cellular mechanisms of tumorigenesis. Advances continue to be made in modeling human disease in a mouse, though the relevance of a mouse model often relies on how closely it is able to mimic the histologic, molecular, and physiologic characteristics of the respective human cancer. A classic use of a genetically engineered mouse in studying cancer is through the overexpression or deletion of a gene. However, the manipulation of a single gene often falls short of mimicking all the characteristics of the carcinoma in humans; thus a multiple gene approach is needed. Here we review genetic mouse models of cancers and their abilities to recapitulate human carcinoma with single versus combinatorial approaches with genes commonly involved in cancer.
Collapse
Affiliation(s)
- Heather L Lehman
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Douglas B Stairs
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
16
|
Ackermann S, Kocak H, Hero B, Ehemann V, Kahlert Y, Oberthuer A, Roels F, Theißen J, Odenthal M, Berthold F, Fischer M. FOXP1 inhibits cell growth and attenuates tumorigenicity of neuroblastoma. BMC Cancer 2014; 14:840. [PMID: 25406647 PMCID: PMC4251948 DOI: 10.1186/1471-2407-14-840] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 10/30/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Segmental genomic copy number alterations, such as loss of 11q or 3p and gain of 17q, are well established markers of poor outcome in neuroblastoma, and have been suggested to comprise tumor suppressor genes or oncogenes, respectively. The gene forkhead box P1 (FOXP1) maps to chromosome 3p14.1, a tumor suppressor locus deleted in many human cancers including neuroblastoma. FoxP1 belongs to a family of winged-helix transcription factors that are involved in processes of cellular proliferation, differentiation and neoplastic transformation. METHODS Microarray expression profiles of 476 neuroblastoma specimens were generated and genes differentially expressed between favorable and unfavorable neuroblastoma were identified. FOXP1 expression was correlated to clinical markers and patient outcome. To determine whether hypermethylation is involved in silencing of FOXP1, methylation analysis of the 5' region of FOXP1 in 47 neuroblastomas was performed. Furthermore, FOXP1 was re-expressed in three neuroblastoma cell lines to study the effect of FOXP1 on growth characteristics of neuroblastoma cells. RESULTS Low expression of FOXP1 is associated with markers of unfavorable prognosis like stage 4, age >18 months and MYCN amplification and unfavorable gene expression-based classification (P < 0.001 each). Moreover, FOXP1 expression predicts patient outcome accurately and independently from well-established prognostic markers. Array-based CGH analysis of 159 neuroblastomas revealed that heterozygous loss of the FOXP1 locus was a rare event (n = 4), but if present, was associated with low FOXP1 expression. By contrast, DNA methylation analysis in 47 neuroblastomas indicated that hypermethylation is not regularly involved in FOXP1 gene silencing. Re-expression of FoxP1 significantly impaired cell proliferation, viability and colony formation in soft agar. Furthermore, induction of FOXP1 expression led to cell cycle arrest and apoptotic cell death of neuroblastoma cells. CONCLUSIONS Our results suggest that down-regulation of FOXP1 expression is a common event in high-risk neuroblastoma pathogenesis and may contribute to tumor progression and unfavorable patient outcome.
Collapse
Affiliation(s)
- Sandra Ackermann
- Department of Pediatric Oncology and Hematology and Center for Molecular Medicine Cologne (CMMC), Children's Hospital, University of Cologne, Kerpener Straße 62, Cologne 50924, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Karagöz Özen DS, Ozturk MA, Aydin Ö, Turna ZH, Ilvan S, Özgüroglu M. Receptor Expression Discrepancy between Primary and Metastatic Breast Cancer Lesions. Oncol Res Treat 2014; 37:622-6. [DOI: 10.1159/000368312] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/18/2014] [Indexed: 11/19/2022]
|
18
|
Roy D, Calaf GM. Allelic loss at chromosome 11q13 alters FGF3 gene expression in a human breast cancer progression model. Oncol Rep 2014; 32:2445-52. [PMID: 25333703 DOI: 10.3892/or.2014.3502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 08/26/2014] [Indexed: 11/06/2022] Open
Abstract
Identification of markers with the potential to predict tumorigenic behavior is important in breast cancer, due to the variability in clinical disease progression. Genetic alterations during neoplastic progression may appear as changes in total DNA content, single genes, or gene expression. Oncogenic alterations are thought to be prognostic indices for patients with breast cancer. Breast cancer deregulation can occur in the normal cellular process and can be measured by microsatellite instability (MSI)/loss of heterozygosity (LOH). Chromosome 11 is unique in this respect, as three regions of MSI/LOH have been identified (11p15-p15.5, 11q13-q13.3 and 11q23-q24). There are many important families of genes, such as FGF, CCND1, FADD, BAD and GAD2, that are located on chromosome 11 and these play a crucial role in breast cancer progression. Among them, different members of the fibroblast growth factor (FGF) family of genes are clustered around human chromosome 11q13 amplicon, which are constantly altering during breast cancer progression. Therefore, in this study, locus 11q13 and FGF3 gene (11q13) function were investigated in a radiation and estrogen breast cancer model induced by high-LET (α-particle) radiation and estrogen exposure. To assess the effect of ionizing radiation and estrogen at chromosome 11q13 loci and the subsequent role of FGF3 gene expression, various microsatellite markers were chosen in this region, and allelic loses (~20-45%) were identified by PCR-SSCP analysis. Results showed an increase in FGF3 protein expression and a 6- to 8-fold change in gene expression of FGF3 and associated genes. These deregulations could be utilized as an appropriate target for therapeutic intervention in breast cancer.
Collapse
Affiliation(s)
- Debasish Roy
- Department of Natural Sciences, Hostos College of the City University of New York, Bronx, NY, USA
| | - Gloria M Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica, Chile
| |
Collapse
|
19
|
Kapoor A, Vogel VG. Prognostic factors for breast cancer and their use in the clinical setting. Expert Rev Anticancer Ther 2014; 5:269-81. [PMID: 15877524 DOI: 10.1586/14737140.5.2.269] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Breast cancer is the second leading cause of cancer deaths in US women today. This year, approximately 216,000 US women will be diagnosed with invasive breast cancer and another 60,000 with in situ disease. Numerous factors can quantify individual risks for breast cancer, guide therapy and predict outcome. This review focuses on the clinical, pathologic, molecular and genetic prognostic tools available for use in patients with breast cancer, and their impact on clinical decisions and treatment selection.
Collapse
Affiliation(s)
- Avina Kapoor
- University of Pittsburgh School of Medicine, Department of Medicine, Magee-Womens Hospital, 300 Halket Street, Room 3524, Pittsburgh, PA 15213-3180, USA.
| | | |
Collapse
|
20
|
|
21
|
CORRÊA NATÁSSIAC, KUASNE HELLEN, FARIA JERUSAA, SEIXAS CIÇAC, SANTOS IRIAG, ABREU FRANCINEB, NONOGAKI SUELY, ROCHA RAFAELM, SILVA GERLUZAAPARECIDABORGES, GOBBI HELENICE, ROGATTO SILVIAR, GOES ALFREDOM, GOMES DAWIDSONA. Genomic and phenotypic profiles of two Brazilian breast cancer cell lines derived from primary human tumors. Oncol Rep 2013; 29:1299-307. [PMID: 23404580 PMCID: PMC3621816 DOI: 10.3892/or.2013.2284] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/14/2012] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most common type of cancer among women worldwide. Research using breast cancer cell lines derived from primary tumors may provide valuable additional knowledge regarding this type of cancer. Therefore, the aim of this study was to investigate the phenotypic profiles of MACL-1 and MGSO-3, the only Brazilian breast cancer cell lines available for comparative studies. We evaluated the presence of hormone receptors, proliferation, differentiation and stem cell markers, using immunohistochemical staining of the primary tumor, cultured cells and xenografts implanted in immunodeficient mice. We also investigated the ability of the cell lines to form colonies and copy number alterations by array comparative genomic hybridization. Histopathological analysis showed that the invasive primary tumor from which the MACL-1 cell line was derived, was a luminal A subtype carcinoma, while the ductal carcinoma in situ (DCIS) that gave rise to the MGSO-3 cell line was a HER2 subtype tumor, both showing different proliferation levels. The cell lines and the tumor xenografts in mice preserved their high proliferative potential, but did not maintain the expression of the other markers assessed. This shift in expression may be due to the selection of an 'establishment' phenotype in vitro. Whole-genome DNA evaluation showed a large amount of copy number alterations (CNAs) in the two cell lines. These findings render MACL-1 and MGSO-3 the first characterized Brazilian breast cancer cell lines to be potentially used for comparative research.
Collapse
Affiliation(s)
- NATÁSSIA C.R. CORRÊA
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte
| | - HELLEN KUASNE
- Department of Biological Sciences, State University of Londrina, Londrina
| | - JERUSA A.Q.A. FARIA
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte
| | - CIÇA C.S. SEIXAS
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte
| | - IRIA G.D. SANTOS
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte
| | | | - SUELY NONOGAKI
- Department of Anatomic Pathology, A.C. Camargo Hospital, São Paulo
| | - RAFAEL M. ROCHA
- Department of Anatomic Pathology, A.C. Camargo Hospital, São Paulo
| | | | - HELENICE GOBBI
- Department of Anatomic Pathology, Federal University of Minas Gerais, Belo Horizonte
| | - SILVIA R. ROGATTO
- NeoGene Laboratory, CIPE
- Department of Urology, School of Medicine, Paulista State University, Botucatu, Brazil
| | - ALFREDO M. GOES
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte
| | - DAWIDSON A. GOMES
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte
| |
Collapse
|
22
|
HER2 expression in breast cancer with nonamplified HER2 and gains of chromosome 17 centromere. Appl Immunohistochem Mol Morphol 2012; 20:367-74. [PMID: 22417858 DOI: 10.1097/pai.0b013e31823fc207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gains of chromosome 17 centromere (CEP17) may be accompanied by gains of chromosome 17q. To evaluate the effect of CEP17 gains (CEP17>3 copies per tumor nucleus) on the expression of the HER2 gene, which is located on chromosome 17q12-21.32, we analyzed HER2 amplification and expression in breast carcinomas with and without CEP17 gains. We isolated tumor nuclei from frozen tissues of 37 breast carcinomas for analysis of the HER2 gene and CEP17 by fluorescence in situ hybridization. HER2 expression was detected by immunohistochemistry (IHC) performed on formalin-fixed, paraffin-embedded sections of the corresponding tumors. Tumors with amplified HER2 as determined by both HER2 copy number and HER2/CEP17 ratio were detected in 29.7% (11/37). CEP17 gains were significantly associated with HER2 amplification (P=0.005) but not associated with estrogen receptor status, tumor grade, and lymph node status (P>0.05). In contrast, HER2 amplification was significantly associated with estrogen receptor negativity (P=0.020) but not with tumor grade and lymph node status (P>0.05). IHC analysis was performed in 7 HER2-amplified tumors and all of these were IHC 3+, which were used as positive controls. Among HER2-non-amplified tumors with CEP17 gains, only 1 tumor (1/8, 12.5%) was IHC 3+. However, none of the HER2-non-amplified tumors without CEP17 gains was IHC 3+. In HER2-non-amplified tumors, there was no significant association between HER2 protein expression as detected by IHC and CEP17 or HER2 copy number (P=0.999, P=0.785, respectively). These findings indicate that in the absence of HER2 amplification, CEP17 gains do not have a significant effect on HER2 protein expression.
Collapse
|
23
|
Quan L, Dittmar A, Zhou Y, Hutson A, Stassen APM, Demant P. Susceptibility loci affecting ERBB2/neu-induced mammary tumorigenesis in mice. Genes Chromosomes Cancer 2012; 51:631-43. [PMID: 22419448 DOI: 10.1002/gcc.21949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 01/19/2012] [Accepted: 01/25/2012] [Indexed: 02/05/2023] Open
Abstract
Twenty percent of breast cancers exhibit amplification or overexpression of ERBB2/neu and a poor prognosis. As the susceptibility genes controlling ERBB2 tumorgenesis are unknown, in a genetic mapping project we crossed transgenic mice expressing the neu oncogene under control of MMTV promoter with recombinant congenic (RC) strains, which provided a high mapping power. RC strains differed considerably in tumor latency (P = 0.0002), suggesting a strong genetic control of tumor development. Linkage analysis in neu-transgene carrying F2 hybrids between the most susceptible and most resistant RC strain revealed three mammary tumor susceptibility (Mts) loci with main effects, Mts1 (chr. 4), Mts2 (chr. 10), Mts3 (chr. 19), and two interacting loci Mts4 (chr.6) and Mts5 (chr. 8), significantly affecting mammary tumor latency. Suggestive significance levels indicated control of tumor numbers by Mts1 alone and in interaction with Mts5, and by two additional interacting loci on chromosomes 1 and 8. These loci combined explain to a large extent the tumor latency and number in individual F2 mouse. We also identified a suggestive locus on chromosome 17 controls metastasis to the lung. The loci Mts1, Mts1b, and Mts3 are located in the Naad4-4,5 and Naad19-2 LOH-regions of neu-induced mammary tumors, corresponding to the frequent human breast cancer LOH-regions 1p34/1p36, and 10q25, respectively. These results expand the knowledge of ERBB2 tumorigenesis and point to a combined control of specific tumor phenotypes by germ-line polymorphisms and somatic alterations.
Collapse
Affiliation(s)
- Lei Quan
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | |
Collapse
|
24
|
Mayer JA, Pham T, Wong KL, Scoggin J, Sales EV, Clarin T, Pircher TJ, Mikolajczyk SD, Cotter PD, Bischoff FZ. FISH-based determination of HER2 status in circulating tumor cells isolated with the microfluidic CEE™ platform. Cancer Genet 2012; 204:589-95. [PMID: 22200084 DOI: 10.1016/j.cancergen.2011.10.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/21/2011] [Accepted: 10/24/2011] [Indexed: 11/16/2022]
Abstract
Determination of HER2 status in breast cancer patients is considered standard practice for therapy selection. However, tumor biopsy in patients with recurrent and/or metastatic disease is not always feasible. Thus, circulating tumor cells (CTCs) are an alternative source of tumor cells for analysis of HER2. An antibody cocktail for recovery of variable, high- and low-, EpCAM-expressing tumor cells was developed based on FACS evaluation and then verified by CTC enumeration (based on CK and CD45 staining) with comparison to EpCAM-only and with CellSearch® (n=19). HER2 fluorescence in situ hybridization (FISH) on all (CK+ and CK-) captured cells was compared to HER2 status on the primary tumors (n=54) of patients with late stage metastatic/recurrent breast cancer. Capture of low EpCAM-expressing tumor cells increased from 27% to 76% when using the cocktail versus EpCAM alone, respectively. Overall, CTC detection with the OncoCEE™ platform was better compared to CellSearch® (68% vs. 89%, respectively), and a 93% concordance in HER2 status was observed. HER2 FISH analysis of CK+ and CK- CTCs is feasible using the CEE™ platform. Although larger clinical studies are warranted, the results demonstrate adequate sensitivity and specificity as needed for incorporation into laboratory testing.
Collapse
|
25
|
García Parra-Pérez FA, Zavala-Pompa A, Pacheco-Calleros J, Cortés-Gutiérrez EI, Cerda-Flores RM, Lara-Miranda S, Dávila-Rodríguez MI. Monosomy of chromosome 8 could be considered as a primary preneoplastic event in breast cancer: A preliminary study. Oncol Lett 2011; 3:445-449. [PMID: 22740929 DOI: 10.3892/ol.2011.484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 08/23/2011] [Indexed: 11/06/2022] Open
Abstract
This pilot study analyzed and compared the presence of chromosome 8 aneusomy in Mexican women with breast cancer and adjacent, intraductal, proliferative lesions. To determine the chromosome 8 copy number, we performed fluorescence in situ hybridization in nine patients (1800 cells) who underwent mastectomy. We selected two tissue samples from each patient, one corresponding to the invasive ductal carcinoma (IDC) and the other adjacent to the intraductal proliferative lesion (IPL). Breast tissue from 17 autopsy samples (1700 cells) was used as a control. The number of cells with monosomy, disomy and polysomy per subject and type of tissue were compared among the three groups of tissue with the RxC statistical software package using 50,000 total replicates. Chromosome 8 aneusomy was found in 66 and 67% of cells from the IDC and IPL samples, respectively. Monosomy was detected significantly more frequently in IPL compared with IDC samples (49.11 vs. 27.11%; p=0.0000), whereas polysomy was significantly more frequent in IDC compared with IPL samples (40.11 vs. 16.99%; p=0.00000). Control cells showed 92.3% disomy. These findings suggest that polysomy of chromosome 8 is more frequently observed in IDC and that monosomy is more frequent in tissue of IPL. Therefore, monosomy may be considered as a primary preneoplastic event. Future studies should be performed to increase the amount of breast tissue with ductal proliferative changes and with cancer, in order to support the results of this pilot study.
Collapse
Affiliation(s)
- Fabiola A García Parra-Pérez
- Department of Anatomical Pathology, High Specialty Medical Unit No.25, Mexican Institute of Social Security (IMSS)
| | | | | | | | | | | | | |
Collapse
|
26
|
Ke MW, Hsu JT, Jiang YN, Cheng WTK, Ju YT. Characterization of hTERT-immortalized caprine mammary epithelial cells. Reprod Domest Anim 2011; 47:553-61. [PMID: 22044690 DOI: 10.1111/j.1439-0531.2011.01916.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The aim of this article is to demonstrate and characterize caprine mammary epithelial cells (CMC) immortalized with human telomerase reverse transcriptase (hTERT) gene. Five immortalized CMCs were assigned to either myoepithelial or luminal epithelial groups based on their morphology and expression of cell lineage-specific intermediate filaments. Telomeric repeat amplification protocol revealed various telomerase activities in CMCs associated with their distinct proliferation potential. Karyotypic analysis showed three CMCs retained their modal Capra hircus chromosome number (2n = 60), whereas the remaining two CMCs were abnormal at 2n = 19 and 2n = 36. CMCs with abnormal karyotypes lost p53 protein after chemical-induced DNA damage and showed anchorage-independent growth in soft agar assay. In terms of functional differentiation, luminal CMCs organized into alveolus-like structures when grown in Matrigel. Furthermore, αs1- and β-casein gene was induced in luminal CMCs in response to lacto-hormones stimulation. Together these results showed that hTERT-immortalized CMCs retained major characteristics of mammary epithelial cells, and stability of the genome is required for maintaining normal mammary epithelium function. Application of CMCs can provide valuable models to study alveologenesis and lactogenesis of mammary epithelium and test the feasibility of recombinant constructs designed for the generation of transgenic livestock.
Collapse
Affiliation(s)
- M W Ke
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
27
|
Arslan C, Sari E, Aksoy S, Altundag K. Variation in hormone receptor and HER-2 status between primary and metastatic breast cancer: review of the literature. Expert Opin Ther Targets 2010; 15:21-30. [PMID: 21105765 DOI: 10.1517/14656566.2011.537260] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Hormone and human epidermal growth factor receptor 2 (HER-2) receptors are two important pharmaceutical targets that affect the survival of patients with metastatic breast cancer. Discordance of hormone and HER-2 receptors were reported in a series of studies. Receptor status was reported to change in both directions, yet alteration occurs mostly in the loss of positivity for both receptors. We do not know both the exact mechanism of this process or the contribution rate of technical mistakes; a number of mechanisms might be responsible. Factors suggested include: tumor heterogeneity, clonal selection of tumor cell subpopulations, genetic instability of tumor cells, local or systemic treatments, the time interval between primary tumor and metastasis, receptor status determination techniques, and the site of metastasis. AREAS COVERED IN THIS REVIEW Studies of estrogen, progesterone and HER-2 receptor discordance between primary and metastasis of breast cancer are summarized. Laboratory evaluation of estrogen, progesterone and HER-2 receptors, and possible causes of receptor discordance, are summarized. Literature data are reviewed; the major shortcoming of these studies is that they are mostly retrospective. WHAT THE READER WILL GAIN The reader will read a concise literature review about the studies on estrogen, progesterone and HER-2 receptor discordances between primary and metastasis of breast cancer. TAKE HOME MESSAGE We do not know whether the changes in receptor expression account for a true biological phenomenon or may result from inconsistent measurement. However, in light of current data, for the treatment plans that target the receptors, biopsy specimen from the metastatis of breast cancer must also be evaulated for alterations in the receptor status.
Collapse
Affiliation(s)
- Cagatay Arslan
- Hacettepe University Institute of Oncology, Ankara, Turkey
| | | | | | | |
Collapse
|
28
|
Shackleford TJ, Claret FX. JAB1/CSN5: a new player in cell cycle control and cancer. Cell Div 2010; 5:26. [PMID: 20955608 PMCID: PMC2976740 DOI: 10.1186/1747-1028-5-26] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 10/18/2010] [Indexed: 12/20/2022] Open
Abstract
c-Jun activation domain-binding protein-1 (Jab1) acts as a modulator of intracellular signaling and affects cellular proliferation and apoptosis, through its existence as a monomer or as the fifth component of the constitutive photomorphogenic-9 signalosome (CSN5). Jab1/CSN5 is involved in transcription factor specificity, deneddylation of NEDD8, and nuclear-to-cytoplasmic shuttling of key molecules. Jab1/CSN5 activities positively and negatively affect a number of pathways, including integrin signaling, cell cycle control, and apoptosis. Also, more recent studies have demonstrated the intriguing roles of Jab1/CSN5 in regulating genomic instability and DNA repair. The effects of Jab1/CSN5's multiple protein interactions are generally oncogenic in nature, and overexpression of Jab1/CSN5 in cancer provides evidence that it is involved in the tumorigenic process. In this review, we highlight our current knowledge of Jab1/CSN5 function and the recent discoveries in dissecting the Jab1 signaling pathway. Further, we also discuss the regulation of Jab1/CSN5 in cancers and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Terry J Shackleford
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA.
| | | |
Collapse
|
29
|
Van Dang C, McMahon SB. Emerging Concepts in the Analysis of Transcriptional Targets of the MYC Oncoprotein: Are the Targets Targetable? Genes Cancer 2010; 1:560-567. [PMID: 21533016 DOI: 10.1177/1947601910379011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Activation of the MYC oncoprotein is among the most ubiquitous events in human cancer. MYC functions in part as a sequence-specific regulator of transcription. Although early searches for direct downstream target genes that explain MYC's potent biological activity were met with enthusiasm, the postgenomic decade has brought the realization that MYC regulates the transcription of not just a manageably small handful of target genes but instead up to 15% of all active loci. As the dust has begun to settle, two important concepts have emerged that reignite hope that understanding MYC's downstream targets might still prove valuable for defining critical nodes for therapeutic intervention in cancer patients. First, it is now clear that MYC target genes are not a random sampling of the cellular transcriptome but instead fall into specific, critical biochemical pathways such as metabolism, chromatin structure, and protein translation. In retrospect, we should not have been surprised to discover that MYC rewires cell physiology in a manner designed to provide the tumor cell with greater biosynthetic properties. However, the specific details that have emerged from these studies are likely to guide the development of new clinical tools and strategies. This raises the second concept that instills renewed optimism regarding MYC target genes. It is now clear that not all MYC target genes are of equal functional relevance. Thus, it may be possible to discern, from among the thousands of potential MYC target genes, those whose inhibition will truly debilitate the tumor cell. In short, targeting the targets may ultimately be a realistic approach after all.
Collapse
Affiliation(s)
- Chi Van Dang
- Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | |
Collapse
|
30
|
Sari E, Guler G, Hayran M, Gullu I, Altundag K, Ozisik Y. Comparative study of the immunohistochemical detection of hormone receptor status and HER-2 expression in primary and paired recurrent/metastatic lesions of patients with breast cancer. Med Oncol 2010; 28:57-63. [PMID: 20099049 DOI: 10.1007/s12032-010-9418-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 01/06/2010] [Indexed: 10/19/2022]
Abstract
Recent studies have shown some degrees of discordance in ER, PR and HER-2 immunohistochemical expression between primary and recurrent/metastatic lesions (RML). Analysis was made on 78 patients with MBC whose ER, PR and/or HER-2 status were known both on the tissue samples of primary and RML. Among the RML sites, 29.5% were locoregional, 70.5% were distant metastatic sites. Among 75 patients with known ER expression on both primary and RML, 36% (n = 27) showed discordance on ER expression. Among 72 patients with known PR expression on both primary and RML, 54.2% (n = 39) showed discordance on PR expression. Among 61 patients with known HER-2 expression on both primary and RML, 14.7% (n = 9) showed discordance on HER-2 expression. No differences were observed when we compared patients who have discordant ER and HER-2 status with patients who have concordant results between the primary tumor and paired RML with respect to site of biopsy (locoregional vs distant metastasis) and prior therapies (chemotherapy and endocrine therapy). As these discordant results make changes in treatment decision, a biopsy of the metastatic lesion could be recommended in patients with MBC when feasible. Larger series are needed to identify the potential effect of prior therapies and site of metastasis on discordant results.
Collapse
Affiliation(s)
- Ebru Sari
- Department of Medical Oncology, Hacettepe University Institute of Oncology, Sihhiye, Ankara 06100, Turkey
| | | | | | | | | | | |
Collapse
|
31
|
Pénzváltó Z, Mihály Z, Gyorffy B. [Gene expression based multigene prognostic and predictive tests in breast cancer]. Magy Onkol 2010; 53:351-9. [PMID: 20071307 DOI: 10.1556/monkol.53.2009.4.4] [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]
Abstract
Patient tailored therapy will serve the fundamentals of future cancer treatment. For this it will be imperative to characterize the tumor and to acquire precise predictive and prognostic information. We can achieve this by using not only monogenic (like ER, PR, HER-2, Ki-67) but also multigene assays, which can provide answers to several diagnostic questions simultaneously. We present a summary of currently available RT-PCR and microarray based multigene tests including MammaPrint, Oncotype DX, BLN Assay, Theros Breast Cancer Index SM, MapQuant DX, ARUP Breast Bioclassifier, Celera Metastatic Score, eXagen BCtm, Invasive Gene Signature, Wound Response Indicator and Mammostrat. Two of these (Oncotype DX and MammaPrint) are already incorporated in several diagnostic protocols. However, multiple unsolved issues deteriorate the value of these tests: generally the validation is poor, the gene sets do not confirm each other, the associated costs are high and the necessary bioinformatics is highly complex.
Collapse
Affiliation(s)
- Zsófia Pénzváltó
- Magyar Tudományos Akadémia és Semmelweis Egyetem Gyermekgyógyászati és Nefrológiai Kutatócsoportja I. Gyermekklinika 1083 Budapest Bókay u. 53/54.
| | | | | |
Collapse
|
32
|
Boettcher M, Fredebohm J, Gholami AM, Hachmo Y, Dotan I, Canaani D, Hoheisel JD. Decoding pooled RNAi screens by means of barcode tiling arrays. BMC Genomics 2010; 11:7. [PMID: 20051122 PMCID: PMC2824726 DOI: 10.1186/1471-2164-11-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 01/05/2010] [Indexed: 12/30/2022] Open
Abstract
Background RNAi screens via pooled short hairpin RNAs (shRNAs) have recently become a powerful tool for the identification of essential genes in mammalian cells. In the past years, several pooled large-scale shRNA screens have identified a variety of genes involved in cancer cell proliferation. All of those studies employed microarray analysis, utilizing either the shRNA's half hairpin sequence or an additional shRNA-associated 60 nt barcode sequence as a molecular tag. Here we describe a novel method to decode pooled RNAi screens, namely barcode tiling array analysis, and demonstrate how this approach can be used to precisely quantify the abundance of individual shRNAs from a pool. Results We synthesized DNA microarrays with six overlapping 25 nt long tiling probes complementary to each unique 60 nt molecular barcode sequence associated with every shRNA expression construct. By analyzing dilution series of expression constructs we show how our approach allows quantification of shRNA abundance from a pool and how it clearly outperforms the commonly used analysis via the shRNA's half hairpin sequences. We further demonstrate how barcode tiling arrays can be used to predict anti-proliferative effects of individual shRNAs from pooled negative selection screens. Out of a pool of 305 shRNAs, we identified 28 candidate shRNAs to fully or partially impair the viability of the breast carcinoma cell line MDA-MB-231. Individual validation of a subset of eleven shRNA expression constructs with potential inhibitory, as well as non-inhibitory, effects on the cell line proliferation provides further evidence for the accuracy of the barcode tiling approach. Conclusions In summary, we present an improved method for the rapid, quantitative and statistically robust analysis of pooled RNAi screens. Our experimental approach, coupled with commercially available lentiviral vector shRNA libraries, has the potential to greatly facilitate the discovery of putative targets for cancer therapy as well as sensitizers of drug toxicity.
Collapse
Affiliation(s)
- Michael Boettcher
- Department for Functional Genome Analysis, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
33
|
Fordyce C, Fessenden T, Pickering C, Jung J, Singla V, Berman H, Tlsty T. DNA damage drives an activin a-dependent induction of cyclooxygenase-2 in premalignant cells and lesions. Cancer Prev Res (Phila) 2009; 3:190-201. [PMID: 20028875 DOI: 10.1158/1940-6207.capr-09-0229] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cyclooxygenase-2 (COX-2) catalyzes the rate-limiting step in the synthesis of prostaglandins. Its overexpression induces numerous tumor-promoting phenotypes and is associated with cancer metastasis and poor clinical outcome. Although COX-2 inhibitors are promising chemotherapeutic and chemopreventative agents for cancer, the risk of significant cardiovascular and gastrointestinal complications currently outweighs their potential benefits. Systemic complications of COX-2 inhibition could be avoided by specifically decreasing COX-2 expression in epithelial cells. To that end, we have investigated the signal transduction pathway regulating the COX-2 expression in response to DNA damage in breast epithelial cells. In variant human mammary epithelial cells that have silenced p16 (vHMEC), double-strand DNA damage or telomere malfunction results in a p53- and activin A-dependent induction of COX-2 and continued proliferation. In contrast, telomere malfunction in HMEC with an intact p16/Rb pathway induces cell cycle arrest. Importantly, in ductal carcinoma in situ lesions, high COX-2 expression is associated with high gammaH2AX, TRF2, activin A, and telomere malfunction. These data show that DNA damage and telomere malfunction can have both cell-autonomous and cell-nonautonomous consequences and can provide a novel mechanism for the propagation of tumorigenesis.
Collapse
Affiliation(s)
- Colleen Fordyce
- Department of Pathology, University of California, San Francisco, 94143, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Fine B, Hodakoski C, Koujak S, Su T, Saal LH, Maurer M, Hopkins B, Keniry M, Sulis ML, Mense S, Hibshoosh H, Parsons R. Activation of the PI3K pathway in cancer through inhibition of PTEN by exchange factor P-REX2a. Science 2009; 325:1261-5. [PMID: 19729658 DOI: 10.1126/science.1173569] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
PTEN (phosphatase and tensin homolog on chromosome 10) is a tumor suppressor whose cellular regulation remains incompletely understood. We identified phosphatidylinositol 3,4,5-trisphosphate RAC exchanger 2a (P-REX2a) as a PTEN-interacting protein. P-REX2a mRNA was more abundant in human cancer cells and significantly increased in tumors with wild-type PTEN that expressed an activated mutant of PIK3CA encoding the p110 subunit of phosphoinositide 3-kinase subunit alpha (PI3Kalpha). P-REX2a inhibited PTEN lipid phosphatase activity and stimulated the PI3K pathway only in the presence of PTEN. P-REX2a stimulated cell growth and cooperated with a PIK3CA mutant to promote growth factor-independent proliferation and transformation. Depletion of P-REX2a reduced amounts of phosphorylated AKT and growth in human cell lines with intact PTEN. Thus, P-REX2a is a component of the PI3K pathway that can antagonize PTEN in cancer cells.
Collapse
Affiliation(s)
- Barry Fine
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kai K, Zhang Z, Yamashita H, Yamamoto Y, Miura Y, Iwase H. Loss of heterozygosity at the ATBF1-A locus located in the 16q22 minimal region in breast cancer. BMC Cancer 2008; 8:262. [PMID: 18796146 PMCID: PMC2564977 DOI: 10.1186/1471-2407-8-262] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Accepted: 09/16/2008] [Indexed: 01/07/2023] Open
Abstract
Background Loss of heterozygosity (LOH) on the long arm of chromosome 16 is one of the most frequent genetic events in solid tumors. Recently, the AT-motif binding factor 1 (ATBF1)-A gene, which has been assigned to chromosome 16q22.3-23.1, was identified as a plausible candidate for tumor suppression in solid tumors due to its functional inhibition of cell proliferation and high mutation rate in prostate cancer. We previously reported that a reduction in ATBF1-A mRNA levels correlated with a worse prognosis in breast cancer. However, the mechanisms regulating the reduction of ATBF1-A mRNA levels (such as mutation, methylation in the promoter region, or deletion spanning the coding region) have not been fully examined. In addition, few studies have analyzed LOH status at the ATBF1-A locus, located in the 16q22 minimal region. Methods Profiles of ATBF1-A mRNA levels that we previously reported for 127 cases were used. In this study, breast cancer specimens as well as autologous blood samples were screened for LOH using 6 polymorphic microsatellite markers spanning chromosome band 16q22. For mutational analysis, we selected 12 cases and analyzed selected spots in the ATBF1-A coding region at which mutations have been frequently reported in prostate cancer. Results Forty-three cases that yielded clear profiles of LOH status at both D16S3106 and D16S3018 microsatellites, nearest to the location of the ATBF1-A gene, were regarded as informative and were classified into two groups: LOH (22 cases) and retention of heterozygosity (21 cases). Comparative assessment of the ATBF1-A mRNA levels according to LOH status at the ATBF1-A locus demonstrated no relationship between them. In the 12 cases screened for mutational analysis, there were no somatic mutations with amino acid substitution or frameshift; however, two germ line alterations with possible polymorphisms were observed. Conclusion These findings imply that ATBF1-A mRNA levels are regulated at the transcriptional stage, but not by genetic mechanisms, deletions (LOH), or mutations.
Collapse
Affiliation(s)
- Kazuharu Kai
- Department of Breast and Endocrine Surgery, Faculty of Medical and Pharmaceutical Science, Kumamoto University, Kumamoto, Japan.
| | | | | | | | | | | |
Collapse
|
36
|
Fukamachi K, Imada T, Ohshima Y, Xu J, Tsuda H. Purple corn color suppresses Ras protein level and inhibits 7,12-dimethylbenz[a]anthracene-induced mammary carcinogenesis in the rat. Cancer Sci 2008; 99:1841-6. [PMID: 18616524 PMCID: PMC11159976 DOI: 10.1111/j.1349-7006.2008.00895.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Anthocyanins belong to the class of phenolic compounds collectively named flavonoids. Many anthocyanins are reported to have inhibitory effects on carcinogenesis. Purple corn color (PCC), an anthocyanin containing extract of purple corn seeds, is used as a food colorant. The major anthocyanin in PCC is cyanidin 3-O-beta-D-glucoside (C3-G). The present study was conducted to assess the influence of dietary PCC on 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary carcinogenesis in rats. PCC significantly inhibited DMBA-induced mammary carcinogenesis in human c-Ha-ras proto-oncogene transgenic (Hras128) rats and in their non-transgenic counterparts. PCC and C3-G also inhibited cell viability and induced apoptosis in mammary tumor cells derived from Hras128 rat mammary carcinomas. At the molecular level, PCC and C3-G treatment resulted in a preferential activation of caspase-3 and reduction of Ras protein levels in tumor cells. It is proposed that C3-G could act as a chemopreventive and possibly chemotherapeutic agent for cancers with mutations in ras. Secondly, the in vitro-in vivo system used in this study can be utilized for screening for cancer preventive compounds that act via Ras down-regulation.
Collapse
Affiliation(s)
- Katsumi Fukamachi
- Department of Molecular Toxicology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | |
Collapse
|
37
|
Hachana M, Trimeche M, Ziadi S, Amara K, Gaddas N, Mokni M, Korbi S. Prevalence and characteristics of the MMTV-like associated breast carcinomas in Tunisia. Cancer Lett 2008; 271:222-30. [PMID: 18639977 DOI: 10.1016/j.canlet.2008.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2008] [Revised: 03/02/2008] [Accepted: 06/09/2008] [Indexed: 12/16/2022]
Abstract
The involvement of a retrovirus homologous to the mouse mammary tumor virus (MMTV) in the pathogenesis of human breast cancer (BC) has long been assumed, but has never been proven. Previous studies have reported the detection of MMTV-like env sequences in variable proportions that did not exceed 40% of BC cases in several countries. However, these viral sequences have been found in higher proportion (74%) in Tunisian diagnosed with BC during the seventies. This study is an attempt to evaluate the current prevalence of MMTV-like env gene in BC in Tunisian women. We used semi-nested PCR that amplify a 190-bp MMTV-like env sequence, followed by direct sequencing to screen a series of 122 cases of BC randomly selected. The findings were correlated to clinicopathological data and immunohistochemical expression status of progesterone and oestrogen receptors, HER2, and P53. Specific MMTV-like env sequences were found in 17 (13.9%) cases of breast carcinomas, whereas the same sequences were not detected in matched normal breast tissues. The presence of the viral sequences correlates inversely with progesterone receptor expression (6.8% versus 20.3%; P=0.03) and HER2 overexpression (3.1% versus 17.7%; P=0.04). This present study confirms the presence of MMTV-like env sequences in BC in Tunisian women but describes an important decrease in the prevalence of the viral sequences compared with previous studies. This reduction may be due to some changes in the virological characteristics or exposure to the virus.
Collapse
Affiliation(s)
- M Hachana
- Department of Pathology, Farhat Hached Hospital, Sousse 4000, Tunisia
| | | | | | | | | | | | | |
Collapse
|
38
|
Urquidi V, Goodison S. Genomic signatures of breast cancer metastasis. Cytogenet Genome Res 2007; 118:116-29. [PMID: 18000362 DOI: 10.1159/000108292] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 09/28/2006] [Indexed: 01/04/2023] Open
Abstract
Despite significant advances in the treatment of primary cancer, the ability to predict the metastatic behavior of a patient's cancer, as well as to detect and eradicate such recurrences, remain major clinical challenges in oncology. While many potential molecular biomarkers have been identified and tested previously, none have greatly improved the accuracy of specimen evaluation over routine histopathological criteria and they predict individual outcomes poorly. However, the recent introduction of high-throughput microarray technology has opened new avenues in genomic investigation of cancer, and through application in tissue-based studies and appropriate animal models, has facilitated the identification of gene expression signatures that are associated with the lethal progression of breast cancer. The use of these approaches has the potential to greatly impact our knowledge of tumor biology, to provide efficient biomarkers, and enable development towards customized prognostication and therapies for the individual.
Collapse
Affiliation(s)
- V Urquidi
- Department of Medicine, University of Florida, Jacksonville, FL, USA
| | | |
Collapse
|
39
|
Expression of the forkhead transcription factor FOXP1 is associated with that of estrogen receptor-beta in primary invasive breast carcinomas. Breast Cancer Res Treat 2007; 111:453-9. [PMID: 18026833 DOI: 10.1007/s10549-007-9812-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Accepted: 10/31/2007] [Indexed: 12/26/2022]
Abstract
We previously identified a correlation between estrogen receptor alpha (ERalpha) and the candidate tumour suppressor gene Forkhead Box P1 (FOXP1), whose nuclear protein expression in breast tumours was associated with improved patient survival. However, the expression pattern of FOXP1 in normal breast tissue is more reminiscent of the second receptor, ERbeta, which has an emerging role as a tumour suppressor in breast cancer and critically may underlie the ability of some ERalpha-negative tumours to respond to tamoxifen. In a series of 283 breast cancers, in which ERalpha-positive tumours were treated with tamoxifen, the nuclear expression of ERbeta correlated significantly with ERalpha (p = 0.004), low-tumour grade (p = 0.008) and nuclear FOXP1 (p = 0.01). High-grade tumours exhibited significantly more cytoplasmic ERbeta than the low-grade tumours (p = 0.006). Regression analysis demonstrated that FOXP1 expression was most closely related to nuclear ERbeta (p = 0.021). Neither, nuclear or cytoplasmic ERbeta expression demonstrated prognostic significance. FOXP1 is not estrogen regulated and silencing FOXP1 expression, using siRNA, did not affect ERalpha, ERbeta or progesterone receptor expression, suggesting ER and FOXP1 co-expression may reflect a common regulatory mechanism.
Collapse
|
40
|
Lusa L, McShane LM, Reid JF, De Cecco L, Ambrogi F, Biganzoli E, Gariboldi M, Pierotti MA. Challenges in projecting clustering results across gene expression-profiling datasets. J Natl Cancer Inst 2007; 99:1715-23. [PMID: 18000217 DOI: 10.1093/jnci/djm216] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Gene expression microarray studies for several types of cancer have been reported to identify previously unknown subtypes of tumors. For breast cancer, a molecular classification consisting of five subtypes based on gene expression microarray data has been proposed. These subtypes have been reported to exist across several breast cancer microarray studies, and they have demonstrated some association with clinical outcome. A classification rule based on the method of centroids has been proposed for identifying the subtypes in new collections of breast cancer samples; the method is based on the similarity of the new profiles to the mean expression profile of the previously identified subtypes. METHODS Previously identified centroids of five breast cancer subtypes were used to assign 99 breast cancer samples, including a subset of 65 estrogen receptor-positive (ER+) samples, to five breast cancer subtypes based on microarray data for the samples. The effect of mean centering the genes (i.e., transforming the expression of each gene so that its mean expression is equal to 0) on subtype assignment by method of centroids was assessed. Further studies of the effect of mean centering and of class prevalence in the test set on the accuracy of method of centroids classifications of ER status were carried out using training and test sets for which ER status had been independently determined by ligand-binding assay and for which the proportion of ER+ and ER- samples were systematically varied. RESULTS When all 99 samples were considered, mean centering before application of the method of centroids appeared to be helpful for correctly assigning samples to subtypes, as evidenced by the expression of genes that had previously been used as markers to identify the subtypes. However, when only the 65 ER+ samples were considered for classification, many samples appeared to be misclassified, as evidenced by an unexpected distribution of ER+ samples among the resultant subtypes. When genes were mean centered before classification of samples for ER status, the accuracy of the ER subgroup assignments was highly dependent on the proportion of ER+ samples in the test set; this effect of subtype prevalence was not seen when gene expression data were not mean centered. CONCLUSIONS Simple corrections such as mean centering of genes aimed at microarray platform or batch effect correction can have undesirable consequences because patient population effects can easily be confused with these assay-related effects. Careful thought should be given to the comparability of the patient populations before attempting to force data comparability for purposes of assigning subtypes to independent subjects.
Collapse
Affiliation(s)
- Lara Lusa
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Györffy B. Resistance-associated signatures in breast cancer. Recent Results Cancer Res 2007; 176:37-50. [PMID: 17607915 DOI: 10.1007/978-3-540-46091-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A major obstacle in the treatment of breast cancer is the lack of adequate methods for predicting patient response to a particular chemotherapy regime. To date, single tumour markers have provided limited success. DNA array technologies identifying thousands of genes simultaneously can help to solve this problem. We investigated cancer cell lines sensitive and resistant to the topoisomerase inhibitors doxorubicin and mitoxantrone. These drugs are used in several different breast cancer treatment protocols. We have identified the top genes best associated with resistance against each cytostatic agent. We applied our gene expression signatures to a set of pre-characterised patients receiving doxorubicin monotherapy. The patients classified as sensitive to chemotherapy exhibited longer survival than the resistant ones. In summary, in our study we have successfully transferred experimental results to a clinical problem, and managed to perform a predictive test for a selected monotherapy protocol. However, many different studies have been performed using microarrays, each producing a different gene list for the same classification problem. It is likely that future diagnostic tools will include the results of several different laboratories, focus on genes validated on different technological platforms and use large cohorts of patients.
Collapse
Affiliation(s)
- Balázs Györffy
- Joint Research Laboratory, Semmelweis University Budapest and Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
42
|
Ottewell PD, Coleman RE, Holen I. From genetic abnormality to metastases: murine models of breast cancer and their use in the development of anticancer therapies. Breast Cancer Res Treat 2006; 96:101-13. [PMID: 16319986 DOI: 10.1007/s10549-005-9067-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous mouse models of mammary cancer have been developed that mimic selective aspects of human disease. The use of these models has enabled preclinical chemotherapeutic, chemoprevention, and genetic therapy studies in vivo, the testing of gene delivery systems, and the identification of tumour and metastasis suppressor and inducer genes. This review has discussed the most abundantly used murine models of mammary cancer including: spontaneous tumours, chemically induced tumours, orthotopic and syngeneic tumour transplantation, injected tumours, and genetically engineered mice with a predisposition to neoplasia. Each model has been discussed with regards to its merits and limitations for investigating the genetic and phenotypic alterations involved in the human disease as well as its potential usefulness for the development of new treatment strategies. To date no single mouse model is available with the ability to replicate the entire disease process, however, existing models continue to provide invaluable insights into breast cancer induction and progression that would be impossible to obtain using in vitro models alone.
Collapse
Affiliation(s)
- P D Ottewell
- Academic Unit of Clinical Oncology, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, UK.
| | | | | |
Collapse
|
43
|
Sun X, Zhou Y, Otto KB, Wang M, Chen C, Zhou W, Subramanian K, Vertino PM, Dong JT. Infrequent mutation of ATBF1 in human breast cancer. J Cancer Res Clin Oncol 2006; 133:103-5. [PMID: 16932943 DOI: 10.1007/s00432-006-0148-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 07/20/2006] [Accepted: 07/28/2006] [Indexed: 10/24/2022]
Abstract
Deletion at chromosome 16q is frequent in prostate and breast cancers, suggesting the existence of one or more tumor suppressor genes in 16q. Recently, the transcription factor ATBF1 at 16q22 was identified as a strong candidate tumor suppressor gene in prostate cancer, and loss of ATBF1 expression was associated with poorer prognosis in breast cancer. In the present study, we examined mutation, expression, and promoter methylation of ATBF1 in 32 breast cancer cell lines. Only 2 of the 32 cancer cell lines had mutations, although 18 nucleotide polymorphisms were detected. In addition, 24 of 32 (75%) cancer cell lines had reduced ATBF1 mRNA levels, yet promoter methylation was not involved in gene silencing. These findings suggest that ATBF1 plays a role in breast cancer through transcriptional downregulation rather than mutations.
Collapse
Affiliation(s)
- Xiaodong Sun
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kaminski R, Zagozdzon R, Fu Y, Mroz P, Fu W, Seng S, Avraham S, Avraham HK. Role of SRC kinases in Neu-induced tumorigenesis: challenging the paradigm using Csk homologous kinase transgenic mice. Cancer Res 2006; 66:5757-62. [PMID: 16740714 DOI: 10.1158/0008-5472.can-05-3536] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Amplification of the HER-2/neu (ErbB2) gene is observed in approximately 30% of human breast cancers, correlating with a poor clinical prognosis. Src kinases are also involved in the etiology of breast cancer, and their activation was suggested to be necessary for Neu-induced oncogenesis. To address whether Src activity is essential for Neu-mediated tumorigenesis, we used a physiologic inhibitor of Src kinase activity, the Csk homologous kinase (CHK), expressed as a mammary tissue-specific transgene. Our data, using a physiologic inhibitor of Src activity (CHK), showed that blocking of Neu-induced Src activity without altering Src expression levels had no significant effects on Neu-mediated mammary tumorigenesis in vivo. This contradicts the current paradigm that activation of Src kinases is essential for Neu-induced oncogenesis. This study is the first to distinguish between the kinase-dependent and kinase-independent actions of Src and shows that its kinase-dependent properties are not requisite for Neu-induced tumorigenesis.
Collapse
Affiliation(s)
- Rafal Kaminski
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center and Wellman Laboratory of Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Numerous genes are controlled by complex regulatory networks and involved in the development and progression of breast cancer, and these genes are the key factors determining each characteristic of the tumor. Gene expression profiling, a large scale analysis of gene expression, has created new possibilities for the molecular characterization of cancer. Systematic analysis of expression patterns of thousands of genes in tumor cells using DNA microarrays and correlation of these patterns to specific features of phenotypic variation may provide the basis for an improved taxonomy of cancer. These profiles have the potential to explain the genetic heterogeneity of breast cancer and allow treatment strategies to be planned in accordance with their probability of success in individual patients.
Collapse
Affiliation(s)
- Koichi Nagasaki
- Genome Center, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan.
| | | |
Collapse
|
46
|
Horie-Inoue K, Inoue S. Epigenetic and proteolytic inactivation of 14-3-3sigma in breast and prostate cancers. Semin Cancer Biol 2006; 16:235-9. [PMID: 16682214 DOI: 10.1016/j.semcancer.2006.03.006] [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: 12/27/2022]
Abstract
14-3-3sigma is an epithelial marker whose expression is induced by DNA damage through a p53-dependent pathway. 14-3-3sigma functions sequesters cyclin B1-CDC2 complexes outside the nucleus and thereby contributes to a G2 arrest. Down-regulation or lack of 14-3-3sigma is a frequent event in breast and prostate cancers. Epigenetic silencing by CpG methylation, p53 inactivation, and proteasome-dependent proteolysis leads to loss of 14-3-3sigma. Hypermethylation of the 14-3-3sigma gene is often observed in precancerous lesions and likely to be causally linked to the onset of cancer. Proteolytic inactivation of 14-3-3sigma has been recently found in breast and prostate cancers. In breast cancer, the estrogen-responsive E3 ubiquitin ligase Efp specifically targets 14-3-3sigma for degradation. The E2 ubiquitin conjugating enzyme UBC8 and Efp also mediates ISG15 modification of 14-3-3sigma. Detection of 14-3-3sigma inactivation on the protein or DNA methylation level may be used for cancer prognosis. Furthermore, 14-3-3sigma may be a potential therapeutic target in breast and prostate cancer.
Collapse
Affiliation(s)
- Kuniko Horie-Inoue
- Research Center for Genomic Medicine and Department of Molecular Biology, Saitama Medical School, Hidaka-shi, Japan
| | | |
Collapse
|
47
|
Yu Q, Sicinska E, Geng Y, Ahnström M, Zagozdzon A, Kong Y, Gardner H, Kiyokawa H, Harris LN, Stål O, Sicinski P. Requirement for CDK4 kinase function in breast cancer. Cancer Cell 2006; 9:23-32. [PMID: 16413469 DOI: 10.1016/j.ccr.2005.12.012] [Citation(s) in RCA: 301] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Revised: 11/18/2005] [Accepted: 12/09/2005] [Indexed: 02/08/2023]
Abstract
Cyclin D1 is overexpressed in the majority of human breast cancers. We previously found that mice lacking cyclin D1 are resistant to mammary carcinomas triggered by the ErbB-2 oncogene. In this study, we investigated which function of cyclin D1 is required for ErbB-2-driven mammary oncogenesis. We report that the ability of cyclin D1 to activate cyclin-dependent kinase CDK4 underlies the critical role for cyclin D1 in breast cancer formation. We also found that the continued presence of CDK4-associated kinase activity is required to maintain breast tumorigenesis. We analyzed primary human breast cancers and found high cyclin D1 levels in a subset (approximately 25%) of ErbB-2-overexpressing tumors. We propose that this subset of breast cancer patients might benefit from inhibiting CDK4 kinase.
Collapse
Affiliation(s)
- Qunyan Yu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Prostate cancer is one of the most commonly diagnosed and potentially devastating cancers in men, throughout the world. However, the clinical manifestation of this disease varies greatly, from indolent tumours, requiring little or no treatment, to those aggressive cancers which require radical therapies. Prostate cancer, like all other cancers, develops and progresses as a consequence of an accumulation of genetic changes. While several putative genes have been isolated for the development of breast, ovarian and colon cancer, the aetiology and pathogenesis of prostate cancer remains poorly understood. In this review, we discuss important genetic markers in early, metastatic and hormone refractory prostate cancer which may, in the future, be used as markers for diagnosis and prognosis, as well as targets for therapeutic intervention.
Collapse
Affiliation(s)
- S R J Bott
- Prostate Cancer Research Centre, Institute of Urology, University College London, 24 St Nicholas Place, Loughton, Essex IG1O 1BF, UK
| | | | | | | |
Collapse
|
49
|
Méndez O, Fernández Y, Peinado MA, Moreno V, Sierra A. Anti-apoptotic proteins induce non-random genetic alterations that result in selecting breast cancer metastatic cells. Clin Exp Metastasis 2005; 22:297-307. [PMID: 16170666 DOI: 10.1007/s10585-005-8751-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Accepted: 06/15/2005] [Indexed: 02/03/2023]
Abstract
To shed light on the relationships between over-expression of anti-apoptotic proteins, genomic instability, and the metastatic ability of breast cancer cells, we analyzed genetic changes in tumors and metastases by orthotopically injecting MDA-MB 435 cells transfected with anti-apoptotic genes Bcl-xL or Bcl-2 into nude mice. Tumors and metastasis variants were extracted by primary culture from breast, bone, lung, and lymph node from mice with 435/Bcl-xL, 435/Bcl-2, and 435/Neo tumors. Using the Arbitrarily Primed Polymerase Chain Reaction (AP-PCR), which permits the detection of allelic imbalances, we generated four different fingerprints utilizing four primers. We found that the genetic damage fraction (GDF) increased in 435/Bcl-2 (GDF=0.55) and 435/Bcl-xL cells (GDF=0.34), in regard to 435/Neo control cells (GDF=0.29), indicating that non-random genetic alterations occurred in cells secondary to Bcl-2 or Bcl-xL over-expression. Anti-apoptotic proteins render breast cancer cells susceptible to the in vivo acquisition of highly tumorigenic (Kruskal-Wallis, P=0.019) and metastatic (Kruskal-Wallis, P=0.004) activity. We therefore propose that genetic instability is a molecular mechanism favored by anti-apoptotic proteins involved in the selection of highly metastatic cells during tumorigenesis, a pathogenic event favoring the expansion of metastasis.
Collapse
Affiliation(s)
- Olga Méndez
- Centre d' Oncologia Molecular, Institut de Recerca Oncológica-IDIBELL, Barcelona, Spain
| | | | | | | | | |
Collapse
|
50
|
Fernandez SV, Russo IH, Russo J. Estradiol and its metabolites 4-hydroxyestradiol and 2-hydroxyestradiol induce mutations in human breast epithelial cells. Int J Cancer 2005; 118:1862-8. [PMID: 16287077 DOI: 10.1002/ijc.21590] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An elevated incidence of breast cancer in women has been associated with prolonged exposure to high levels of estrogens. Our laboratory demonstrated that treatment of the immortalized human breast epithelial cells MCF-10F with 17beta-estradiol (E2), 4-hydroxyestradiol (4-OHE2) or 2-hydroxyestradiol (2-OHE2) induces phenotypical changes indicative of neoplastic transformation. MCF-10F cells treated with E2, 4-OHE2 or 2-OHE2 formed colonies in agar methocel and lost their ductulogenic capacity in collagen, expressing phenotypes similar to those induced by the carcinogen benzo[a]pyrene. To investigate whether the transformation phenotypes were associated with genomic changes, cells treated with E2, 4-OHE2 or 2-OHE2 at different doses were analyzed using microsatellite markers. Since microsatellite instability (MSI) and loss of heterozygosity (LOH) in chromosomes 13 and 17 have been reported in human breast carcinomas, we tested these parameters in MCF-10F cells treated with E2, 2-OHE2, or 4-OHE2 alone or in combination with the antiestrogen ICI182780. MCF-10F cells treated with E2 or 4-OHE2, either alone or in combination with ICI182780, exhibited LOH in the region 13q12.3 with the marker D13S893 located at approximately 0.8 cM telomeric to BRCA2. Cells treated with E2 or 4-OHE2 at doses of 0.007 and 70 nM and 2-OHE2 only at a higher dose (3.6 microM) showed a complete loss of 1 allele with D13S893. For chromosome 17, differences were found using the marker TP53-Dint located in exon 4 of p53. Cells treated with E2 or 4-OHE2 at doses of 0.007 nM and 70 nM and 2-OHE2 only at a higher dose (3.6 microM) exhibited a 5 bp deletion in p53 exon 4. Our results show that E2 and its catechol estrogen metabolites are mutagenic in human breast epithelial cells. ICI182780 did not prevent these mutations, indicating that the carcinogenic effect of E2 is mainly through its reactive metabolites 4-OHE2 and 2-OHE2, with 4-OHE2 and E2 being mutagenic at lower doses than 2-OHE2.
Collapse
Affiliation(s)
- Sandra V Fernandez
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|