1
|
Liu Y, Elmas A, Huang KL. Mutation impact on mRNA versus protein expression across human cancers. Gigascience 2025; 14:giae113. [PMID: 39775839 PMCID: PMC11702362 DOI: 10.1093/gigascience/giae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer mutations are often assumed to alter proteins, thus promoting tumorigenesis. However, how mutations affect protein expression-in addition to gene expression-has rarely been systematically investigated. This is significant as mRNA and protein levels frequently show only moderate correlation, driven by factors such as translation efficiency and protein degradation. Proteogenomic datasets from large tumor cohorts provide an opportunity to systematically analyze the effects of somatic mutations on mRNA and protein abundance and identify mutations with distinct impacts on these molecular levels. RESULTS We conduct a comprehensive analysis of mutation impacts on mRNA- and protein-level expressions of 953 cancer cases with paired genomics and global proteomic profiling across 6 cancer types. Protein-level impacts are validated for 47.2% of the somatic expression quantitative trait loci (seQTLs), including CDH1 and MSH3 truncations, as well as other mutations from likely "long-tail" driver genes. Devising a statistical pipeline for identifying somatic protein-specific QTLs (spsQTLs), we reveal several gene mutations, including NF1 and MAP2K4 truncations and TP53 missenses showing disproportional influence on protein abundance not readily explained by transcriptomics. Cross-validating with data from massively parallel assays of variant effects (MAVE), TP53 missenses associated with high tumor TP53 proteins are more likely to be experimentally confirmed as functional. CONCLUSION This study reveals that somatic mutations can exhibit distinct impacts on mRNA and protein levels, underscoring the necessity of integrating proteogenomic data to comprehensively identify functionally significant cancer mutations. These insights provide a framework for prioritizing mutations for further functional validation and therapeutic targeting.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abdulkadir Elmas
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
2
|
Leena Panigrahi L, Samal P, Ranjan Sahoo S, Sahoo B, Pradhan AK, Mahanta S, Rath SK, Arakha M. Nanoparticle-mediated diagnosis, treatment, and prevention of breast cancer. NANOSCALE ADVANCES 2024; 6:3699-3713. [PMID: 39050943 PMCID: PMC11265592 DOI: 10.1039/d3na00965c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 05/16/2024] [Indexed: 07/27/2024]
Abstract
By virtue of their advanced physicochemical properties, nanoparticles have attracted significant attention from researchers for application in diverse fields of medical science. Breast cancer, presenting a high risk of morbidity and mortality, frequently occurs in women and is considered a malignant tumor. Globally, breast cancer is considered the second leading cause of death. Accordingly, its poor prognosis, invasive metastasis, and relapse have motivated oncologists and nano-medical researchers to develop highly potent nanotherapies to cure this deadly disease. In this case, nanoparticles have emerged as responsive platforms for breast cancer management, providing new approaches to improve the diagnostic accuracy, deliver targeted therapies, and limit the progression of this disease. Recently, smart nano-carriers encapsulating drugs, ligands, and tracking probes have been developed for the specific therapy of breast cancers. Further, efforts have been devoted to developing various nano-systems with minimal toxicity. The aim of this review is to present a background on novel nanotheranostic methods that can be employed to diagnose and treat breast cancers and encourage readers to focus on the development of novel nanomedicine for breast cancers and other deadly diseases. In this context, we discuss different methods for the diagnosis, treatment, and prevention of breast cancers using different metal and metal oxide nanoparticles.
Collapse
Affiliation(s)
- Lipsa Leena Panigrahi
- Center For Biotechnology, Siksha O Anusandhan University Bhubaneswar Odisha 751003 India
| | - Pallavi Samal
- Center For Biotechnology, Siksha O Anusandhan University Bhubaneswar Odisha 751003 India
| | - Sameer Ranjan Sahoo
- Center For Biotechnology, Siksha O Anusandhan University Bhubaneswar Odisha 751003 India
| | - Banishree Sahoo
- Center For Biotechnology, Siksha O Anusandhan University Bhubaneswar Odisha 751003 India
| | - Arun Kumar Pradhan
- Center For Biotechnology, Siksha O Anusandhan University Bhubaneswar Odisha 751003 India
| | - Sailendra Mahanta
- School of Pharmacy, The Assam Kaziranga University Koraikhowa, NH-37 Jorhat Assam 785 006 India
| | - Sandip Kumar Rath
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine Atlanta Georgia USA
| | - Manoranjan Arakha
- Center For Biotechnology, Siksha O Anusandhan University Bhubaneswar Odisha 751003 India
| |
Collapse
|
3
|
Lin J, Chen ZF, Guo GD, Chen X. Impact of Alcian blue and periodic acid Schiff expression on the prognosis of gastric signet ring cell carcinoma. World J Gastrointest Oncol 2024; 16:687-698. [PMID: 38577442 PMCID: PMC10989384 DOI: 10.4251/wjgo.v16.i3.687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/27/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND The Alcian blue (AB) and periodic acid Schiff (PAS) stains are representative mucus markers in gastric signet ring cell carcinoma (SRCC). They are low-cost special staining methods used to detect acidic mucus and neutral mucus, respectively. However, the clinical importance of the special combined AB and PAS stain is unclear. AIM To investigate AB expression, PAS expression and the AB-to-PAS (A/P) ratio in gastric SRCC patients and to assess patient prognosis. METHODS Paraffin-embedded sections from 83 patients with gastric SRCC were stained with AB and PAS, and signet ring cell positivity was assessed quantitatively. Immunohistochemical staining for Ki67, protein 53 (P53) and human epidermal growth factor receptor 2 (HER2) was performed simultaneously. The cancer-specific survival (CSS) rate was estimated via Kaplan-Meier analysis. Cox proportional hazards models were used for univariate and multivariate survival analyses. RESULTS Kaplan-Meier survival analysis revealed that the 3-year CSS rate was significantly greater in the high-PAS-expression subgroup than in the low-PAS-expression subgroup (P < 0.001). The 3-year CSS rate in the A/P ≤ 0.5 group was significantly greater than that in the A/P > 0.5 group (P = 0.042). Univariate Cox regression analysis revealed that the factors affecting prognosis included tumor diameter, lymph node metastasis, vessel carcinoma embolus, tumor stage, the A/P ratio and the expression of Ki67, P53 and the PAS. Cox multivariate regression analysis confirmed that low PAS expression [hazard ratio (HR) = 3.809, 95% confidence interval (CI): 1.563-9.283, P = 0.003] and large tumor diameter (HR = 2.761, 95%CI: 1.086-7.020, P = 0.033) were independent risk factors for poor prognosis. CONCLUSION A/P > 0.5 is potentially a risk factor for prognosis, and low PAS expression is an independent risk factor in the prognosis of gastric SRCC. PAS expression and the A/P ratio could help in predicting the clinical prognosis of patients with SRCC.
Collapse
Affiliation(s)
- Juan Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Zhu-Feng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Internal Medicine, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Guo-Dong Guo
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, China
| | - Xin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Pathology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
4
|
Behl T, Kumar A, Vishakha, Sehgal A, Singh S, Sharma N, Yadav S, Rashid S, Ali N, Ahmed AS, Vargas-De-La-Cruz C, Bungau SG, Khan H. Understanding the mechanistic pathways and clinical aspects associated with protein and gene based biomarkers in breast cancer. Int J Biol Macromol 2023; 253:126595. [PMID: 37648139 DOI: 10.1016/j.ijbiomac.2023.126595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/22/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
Cancer is one of the most widespread and severe diseases with a huge mortality rate. In recent years, the second-leading mortality rate of any cancer globally has been breast cancer, which is one of the most common and deadly cancers found in women. Detecting breast cancer in its initial stages simplifies treatment, decreases death risk, and recovers survival rates for patients. The death rate for breast cancer has risen to 0.024 % in some regions. Sensitive and accurate technologies are required for the preclinical detection of BC at an initial stage. Biomarkers play a very crucial role in the early identification as well as diagnosis of women with breast cancer. Currently, a wide variety of cancer biomarkers have been discovered for the diagnosis of cancer. For the identification of these biomarkers from serum or other body fluids at physiological amounts, many detection methods have been developed. In the case of breast cancer, biomarkers are especially helpful in discovering those who are more likely to develop the disease, determining prognosis at the time of initial diagnosis and choosing the best systemic therapy. In this study we have compiled various clinical aspects and signaling pathways associated with protein-based biomarkers and gene-based biomarkers.
Collapse
Affiliation(s)
- Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India
| | - Ankush Kumar
- Institute of Pharmaceutical Sciences, IET Bhaddal Technical Campus, Ropar 140108, Punjab, India
| | - Vishakha
- Institute of Pharmaceutical Sciences, IET Bhaddal Technical Campus, Ropar 140108, Punjab, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, 141104 Ludhiana, Punjab, India
| | - Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana Ambala 133203, Haryana, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana Ambala 133203, Haryana, India
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow 226028, Uttar Pradesh, India
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia.
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadah 11451, Saudi Arabia
| | - Amira Saber Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza 12622, Egypt
| | - Celia Vargas-De-La-Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima 150001, Peru; E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410087, Romania; Doctoral School of Biomedical Sciences, University of Oradea, Oradea 410087, Romania
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| |
Collapse
|
5
|
Liu Y, Elmas A, Huang KL. Mutation Impact on mRNA Versus Protein Expression across Human Cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566942. [PMID: 38014015 PMCID: PMC10680725 DOI: 10.1101/2023.11.13.566942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cancer mutations are often assumed to alter proteins, thus promoting tumorigenesis. However, how mutations affect protein expression has rarely been systematically investigated. We conduct a comprehensive analysis of mutation impacts on mRNA- and protein-level expressions of 953 cancer cases with paired genomics and global proteomic profiling across six cancer types. Protein-level impacts are validated for 47.2% of the somatic expression quantitative trait loci (seQTLs), including mutations from likely "long-tail" driver genes. Devising a statistical pipeline for identifying somatic protein-specific QTLs (spsQTLs), we reveal several gene mutations, including NF1 and MAP2K4 truncations and TP53 missenses showing disproportional influence on protein abundance not readily explained by transcriptomics. Cross-validating with data from massively parallel assays of variant effects (MAVE), TP53 missenses associated with high tumor TP53 proteins were experimentally confirmed as functional. Our study demonstrates the importance of considering protein-level expression to validate mutation impacts and identify functional genes and mutations.
Collapse
Affiliation(s)
- Yuqi Liu
- Center for Transformative Disease Modeling, Department of Genetics and Genomic Sciences, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Abdulkadir Elmas
- Center for Transformative Disease Modeling, Department of Genetics and Genomic Sciences, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Kuan-lin Huang
- Center for Transformative Disease Modeling, Department of Genetics and Genomic Sciences, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
6
|
Shomali N, Kamrani A, Heris JA, Shahabi P, Nasiri H, Sadeghvand S, Ghahremanzadeh K, Akbari M. Dysregulation of P53 in breast cancer: Causative factors and treatment strategies. Pathol Res Pract 2023; 247:154539. [PMID: 37257244 DOI: 10.1016/j.prp.2023.154539] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
One of the most prevalent cancers impacting women worldwide is breast cancer. Although there are several risk factors for breast cancer, the p53 gene's function has recently received much attention. The "gatekeeper" gene, or p53, is sometimes referred to as such since it is crucial in controlling cell proliferation and preventing the development of malignant cells. By identifying DNA damage and initiating cellular repair processes, p53 usually functions as a tumor-suppressor. But p53 gene alterations can result in a lack of function, allowing cells to divide out of control and perhaps triggering the onset of cancer. Various factors, such as mutation genes, signaling pathways, and hormones, can dysregulate P53 proteins and cause breast cancer. A promising strategy for individualized cancer treatment involves focusing on p53 mutations in breast cancer. While numerous techniques, including gene therapy and small compounds, have shown promise, further study is required to create safe and efficient treatments to target p53 mutations in breast cancer successfully.
Collapse
Affiliation(s)
- Navid Shomali
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Amin Kamrani
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Sadeghvand
- Pediatrics Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Abstract
Somatic stem cells are distinguished by their capacity to regenerate themselves and also to produce daughter cells that will differentiate. Self-renewal is achieved through the process of asymmetric cell division which helps to sustain tissue morphogenesis as well as maintain homeostasis. Asymmetric cell division results in the development of two daughter cells with different fates after a single mitosis. Only one daughter cell maintains "stemness" while the other differentiates and achieves a non-stem cell fate. Stem cells also have the capacity to undergo symmetric division of cells that results in the development of two daughter cells which are identical. Symmetric division results in the expansion of the stem cell population. Imbalances and deregulations in these processes can result in diseases such as cancer. Adult mammary stem cells (MaSCs) are a group of cells that play a critical role in the expansion of the mammary gland during puberty and any subsequent pregnancies. Furthermore, given the relatively long lifespans and their capability to undergo self-renewal, adult stem cells have been suggested as ideal candidates for transformation events that lead to the development of cancer. With the possibility that MaSCs can act as the source cells for distinct breast cancer types; understanding their regulation is an important field of research. In this review, we discuss asymmetric cell division in breast/mammary stem cells and implications on further research. We focus on the background history of asymmetric cell division, asymmetric cell division monitoring techniques, identified molecular mechanisms of asymmetric stem cell division, and the role asymmetric cell division may play in breast cancer.
Collapse
Affiliation(s)
| | - Brian W Booth
- Department of Bioengineering, Head-Cellular Engineering Laboratory, 401-1 Rhodes Engineering Research Center, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
8
|
Aldakheel FM, Abuderman AA, Alali BH, Mateen A, Alduraywish SA, jamil K, Alqahtani MS, Syed R. Smoking and P53 polymorphism association with chromosomal aberration in lung cancer. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2021; 33:101533. [DOI: 10.1016/j.jksus.2021.101533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Kuchur OA, Kuzmina DO, Dukhinova MS, Shtil AA. The p53 Protein Family in the Response of Tumor Cells to Ionizing Radiation: Problem Development. Acta Naturae 2021; 13:65-76. [PMID: 34707898 PMCID: PMC8526179 DOI: 10.32607/actanaturae.11247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/24/2020] [Indexed: 12/05/2022] Open
Abstract
Survival mechanisms are activated in tumor cells in response to therapeutic ionizing radiation. This reduces a treatment's effectiveness. The p53, p63, and p73 proteins belonging to the family of proteins that regulate the numerous pathways of intracellular signal transduction play a key role in the development of radioresistance. This review analyzes the p53-dependent and p53-independent mechanisms involved in overcoming the resistance of tumor cells to radiation exposure.
Collapse
Affiliation(s)
- O. A. Kuchur
- ITMO University, Saint-Petersburg, 191002 Russia
| | | | | | - A. A. Shtil
- ITMO University, Saint-Petersburg, 191002 Russia
- Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russia
| |
Collapse
|
10
|
Huang KL, Scott AD, Zhou DC, Wang LB, Weerasinghe A, Elmas A, Liu R, Wu Y, Wendl MC, Wyczalkowski MA, Baral J, Sengupta S, Lai CW, Ruggles K, Payne SH, Raphael B, Fenyö D, Chen K, Mills G, Ding L. Spatially interacting phosphorylation sites and mutations in cancer. Nat Commun 2021; 12:2313. [PMID: 33875650 PMCID: PMC8055881 DOI: 10.1038/s41467-021-22481-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 02/17/2021] [Indexed: 11/18/2022] Open
Abstract
Advances in mass-spectrometry have generated increasingly large-scale proteomics datasets containing tens of thousands of phosphorylation sites (phosphosites) that require prioritization. We develop a bioinformatics tool called HotPho and systematically discover 3D co-clustering of phosphosites and cancer mutations on protein structures. HotPho identifies 474 such hybrid clusters containing 1255 co-clustering phosphosites, including RET p.S904/Y928, the conserved HRAS/KRAS p.Y96, and IDH1 p.Y139/IDH2 p.Y179 that are adjacent to recurrent mutations on protein structures not found by linear proximity approaches. Hybrid clusters, enriched in histone and kinase domains, frequently include expression-associated mutations experimentally shown as activating and conferring genetic dependency. Approximately 300 co-clustering phosphosites are verified in patient samples of 5 cancer types or previously implicated in cancer, including CTNNB1 p.S29/Y30, EGFR p.S720, MAPK1 p.S142, and PTPN12 p.S275. In summary, systematic 3D clustering analysis highlights nearly 3,000 likely functional mutations and over 1000 cancer phosphosites for downstream investigation and evaluation of potential clinical relevance.
Collapse
Affiliation(s)
- Kuan-Lin Huang
- Department of Genetics and Genomics, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Adam D Scott
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel Cui Zhou
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Liang-Bo Wang
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Amila Weerasinghe
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Abdulkadir Elmas
- Department of Genetics and Genomics, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruiyang Liu
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Yige Wu
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael C Wendl
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessika Baral
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Sohini Sengupta
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Chin-Wen Lai
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Kelly Ruggles
- Center for Health Informatics and Bioinformatics, New York University School of Medicine, New York, NY, USA
| | - Samuel H Payne
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Benjamin Raphael
- Lewis-Sigler Institute, Princeton University, Princeton, NJ, USA
| | - David Fenyö
- Center for Health Informatics and Bioinformatics, New York University School of Medicine, New York, NY, USA
| | - Ken Chen
- Departments of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon Mills
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
11
|
Bocchi M, Sousa Pereira ND, Furuya RK, Motoori Fernandes CY, Losi-Guembarovski R, Vitiello GAF, Amarante MK, Watanabe MAE. Expression of Ki67 and p53 Proteins: Breast Cancer Aggressivity Markers in Brazilian Young Patients. J Adolesc Young Adult Oncol 2020; 10:379-388. [PMID: 32716670 DOI: 10.1089/jayao.2020.0037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: The increase in breast cancer (BC) cases in young women is of great importance since the tumor behavior in this group is generally more aggressive than in their older counterparts, and strategies for early diagnosis and prognostication are needed. Therefore, this work sought to investigate prognostic markers associated with young (<44 years old) BC patients. Methods: Two hundred thirty-six primary tumor tissues from 232 BC patients, of which 44 had less than 44 years at diagnosis were evaluated regarding the expression of estrogen and progesterone receptors (ER and PR), human epidermal growth factor receptor 2 (HER2), Ki67, and p53 (used as an indicator of p53 mutations) through immunohistochemistry. Also, data regarding tumor size, histopathological grade (HG), lymph node metastasis disease stage, and patients' survival status were collected. Results: Early age tumors had higher Ki67 expression and p53 mutations, and these markers were positively correlated with each other and associated worse prognosis parameters, such as negativity for ER and PR and positivity for HER2, and with higher HG, tumor size, and disease stage. In young patients, Ki67 correlated with ER, PR, and HG, whereas p53 correlated with HER2 and disease stage. Also, Ki67 associated with BC death independently of time from diagnosis, patients age, tumor size, and disease stage, and showed a trend toward a positive correlation with death in young patients, but not in the older group. Conclusion: Young BC patients were more likely to have intensely proliferative tumors with p53 mutations and these markers may hold prognostic relevance in BC, especially in this subgroup of patients.
Collapse
Affiliation(s)
- Mayara Bocchi
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, Brazil
| | - Nathalia de Sousa Pereira
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, Brazil
| | | | - Caroline Yukari Motoori Fernandes
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, Brazil
| | | | - Glauco Akelinghton Freire Vitiello
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, Brazil
| | - Marla Karine Amarante
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, Brazil
| | - Maria Angelica Ehara Watanabe
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, Brazil
| |
Collapse
|
12
|
Parker TM, Henriques V, Beltran A, Nakshatri H, Gogna R. Cell competition and tumor heterogeneity. Semin Cancer Biol 2020; 63:1-10. [DOI: 10.1016/j.semcancer.2019.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/24/2022]
|
13
|
Kopnin B. Genetic Events Responsible for Colorectal Tumorigenesis: Achievements and Challenges. TUMORI JOURNAL 2018; 79:235-43. [PMID: 8249174 DOI: 10.1177/030089169307900401] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal carcinogenesis is a multistep process that is accompanied by accumulation of changes in proto-oncogenes and tumor-suppressor genes. APC/MCC, RAS, DCC, p53 mutations and/or allelic losses, hyperexpression of c-MYC and RB genes, as well as other genomic alterations appear at characteristic stages of tumor development and are observed in most neoplasms. However, consideration of each of these abnormalities leaves many unanswered questions. The striking data on recurrent amplification of the RB tumor-suppressor gene as well as suppressive activities of protein kinase C and activated RAS genes, at least in some colon carcinoma cell lines, suggest the unusual effects of some signalling pathways in colonic epithelial cells. The results obtained to date indicate that distinct sets of genetic changes may underlie the development of colorectal tumors.
Collapse
Affiliation(s)
- B Kopnin
- Institute of Carcinogenesis, Cancer Research Center, Moscow
| |
Collapse
|
14
|
TRRAP is essential for regulating the accumulation of mutant and wild-type p53 in lymphoma. Blood 2018; 131:2789-2802. [PMID: 29653964 DOI: 10.1182/blood-2017-09-806679] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 04/07/2018] [Indexed: 12/26/2022] Open
Abstract
Tumors accumulate high levels of mutant p53 (mutp53), which contributes to mutp53 gain-of-function properties. The mechanisms that underlie such excessive accumulation are not fully understood. To discover regulators of mutp53 protein accumulation, we performed a large-scale RNA interference screen in a Burkitt lymphoma cell line model. We identified transformation/transcription domain-associated protein (TRRAP), a constituent of several histone acetyltransferase complexes, as a critical positive regulator of both mutp53 and wild-type p53 levels. TRRAP silencing attenuated p53 accumulation in lymphoma and colon cancer models, whereas TRRAP overexpression increased mutp53 levels, suggesting a role for TRRAP across cancer entities and p53 mutations. Through clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 screening, we identified a 109-amino-acid region in the N-terminal HEAT repeat region of TRRAP that was crucial for mutp53 stabilization and cell proliferation. Mass spectrometric analysis of the mutp53 interactome indicated that TRRAP silencing caused degradation of mutp53 via the MDM2-proteasome axis. This suggests that TRRAP is vital for maintaining mutp53 levels by shielding it against the natural p53 degradation machinery. To identify drugs that alleviated p53 accumulation similarly to TRRAP silencing, we performed a small-molecule drug screen and found that inhibition of histone deacetylases (HDACs), specifically HDAC1/2/3, decreased p53 levels to a comparable extent. In summary, here we identify TRRAP as a key regulator of p53 levels and link acetylation-modifying complexes to p53 protein stability. Our findings may provide clues for therapeutic targeting of mutp53 in lymphoma and other cancers.
Collapse
|
15
|
Zhu Y, Aupperlee MD, Zhao Y, Tan YS, Kirk EL, Sun X, Troester MA, Schwartz RC, Haslam SZ. Pubertal and adult windows of susceptibility to a high animal fat diet in Trp53-null mammary tumorigenesis. Oncotarget 2018; 7:83409-83423. [PMID: 27825136 PMCID: PMC5347778 DOI: 10.18632/oncotarget.13112] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/19/2016] [Indexed: 11/25/2022] Open
Abstract
Premenopausal breast cancer is associated with increased animal fat consumption among normal weight, but not overweight women (Farvid et al., 2014). Our previous findings in obesity-resistant BALB/c mice similarly showed promotion of carcinogen-induced mammary tumorigenesis by a diet high in saturated animal fat (HFD). This effect was specific to pubertal versus adult HFD. This study identifies the effects of HFD during puberty versus adulthood in Trp53-null transplant BALB/c mice and investigates its mechanism of enhancing tumorigenesis. Either pubertal or adult HFD is sufficient to increase incidence of Trp53-null mammary tumors. Puberty-restricted HFD exposure promoted tumor cell proliferation, increased angiogenesis, and increased recruitment of total and M2 macrophages in epithelial tumors. Adult-restricted exposure to HFD similarly increased proliferation, angiogenesis, recruitment of total and M2 macrophages, and additionally reduced apoptosis. Adult HFD also increased incidence of spindle cell carcinomas resembling claudin-low breast cancer, and thus adult HFD in the Trp53-null transplantation system may be a useful model for human claudin low breast cancer. Importantly, these results on Trp53-null and our prior studies on DMBA-induced mammary tumorigenesis demonstrate a pubertal window of susceptibility to the promotional effects of HFD, indicating the potential of early life dietary intervention to reduce breast cancer risk.
Collapse
Affiliation(s)
- Yirong Zhu
- Cell and Molecular Biology Program and Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, USA
| | - Mark D Aupperlee
- Department of Physiology and Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, USA
| | - Yong Zhao
- Department of Physiology and Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, USA
| | - Ying Siow Tan
- Department of Physiology and Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, USA
| | - Erin L Kirk
- Department of Epidemiology, University of North Carolina at Chapel Hill, NC, USA
| | - Xuezheng Sun
- Department of Epidemiology, University of North Carolina at Chapel Hill, NC, USA
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Richard C Schwartz
- Department of Microbiology and Molecular Genetics and Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, USA
| | - Sandra Z Haslam
- Department of Physiology and Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
16
|
Soleimani A, Rahmani Y, Farshchian N, Delpisheh A, Khassi K, Shahmohammadi A, Amirifard N. The Evaluation of p53 Polymorphism at Codon 72 and Association With Breast Cancer in Iran: A Systematic Review and Meta-analysis. J Cancer Prev 2017; 21:288-293. [PMID: 28053964 PMCID: PMC5207614 DOI: 10.15430/jcp.2016.21.4.288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/05/2016] [Accepted: 12/05/2016] [Indexed: 11/21/2022] Open
Abstract
Background Breast cancer is the most common cancer among women in Iran and the world. Multiple environmental factors and genetic variations such as genetic polymorphisms are of its main causes. p53 gene plays an important role in conserving and sustaining the genome as a tumor suppressing gene. Change and polymorphism at codon 72 of p53 gene are correlated with increased risk of lung, mouth, endometrial, prostate, and colorectal cancers, and could be considered an indicator of susceptibility to breast cancer. Methods Twelve studies (1,190 cases and 1,145 control studies with evaluation of three types of Arg/Arg, Arg/Pro, and Pro/Pro genotypes) have been conducted using keywords, such as polymorphism at codon 72, gene p53 polymorphisms, and the relation between polymorphisms and breast cancer, from databases in Iran, including Magiran, Medlibe, Sid, and Iranmedex, as well as Latin databases such as PubMed, Google Scholar, Science Direct, and Scopus. Results The OR for Arg/Arg is 1.58 (95% CI: 2.45 to 1.01), the OR for Arg/Pro is 0.75 (95% CI: 1.10 to 0.51), and the OR for Pro/Pro is 0.62 (95% CI: 0.93 to 0.42). p53 gene polymorphism at codon 72 is statistically significant in Arg/Arg and Pro/Pro genotypes. Conclusions Arg/Arg genotype can be considered as a risk factor for breast cancer, and Pro/Pro genotype can be accounted for as a protective factor against breast cancer.
Collapse
Affiliation(s)
- Abozar Soleimani
- Department of Epidemiology, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
| | - Yousef Rahmani
- Clinical Research Development Center, Imam Ali and Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Negin Farshchian
- Cancer Research Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Delpisheh
- Department of Epidemiology, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
| | - Kivan Khassi
- Province Health Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Afshar Shahmohammadi
- Clinical Research Development Center, Imam Ali and Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasrin Amirifard
- Cancer Research Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
17
|
Guerra E, Cimadamore A, Simeone P, Vacca G, Lattanzio R, Botti G, Gatta V, D'Aurora M, Simionati B, Piantelli M, Alberti S. p53, cathepsin D, Bcl-2 are joint prognostic indicators of breast cancer metastatic spreading. BMC Cancer 2016; 16:649. [PMID: 27538498 PMCID: PMC4991058 DOI: 10.1186/s12885-016-2713-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/11/2016] [Indexed: 02/04/2023] Open
Abstract
Background Traditional prognostic indicators of breast cancer, i.e. lymph node diffusion, tumor size, grading and estrogen receptor expression, are inadequate predictors of metastatic relapse. Thus, additional prognostic parameters appear urgently needed. Individual oncogenic determinants have largely failed in this endeavour. Only a few individual tumor growth drivers, e.g. mutated p53, Her-2, E-cadherin, Trops, did reach some prognostic/predictive power in clinical settings. As multiple factors are required to drive solid tumor progression, clusters of such determinants were expected to become stronger indicators of tumor aggressiveness and malignant progression than individual parameters. To identify such prognostic clusters, we went on to coordinately analyse molecular and histopathological determinants of tumor progression of post-menopausal breast cancers in the framework of a multi-institutional case series/case-control study. Methods A multi-institutional series of 217 breast cancer cases was analyzed. Twenty six cases (12 %) showed disease relapse during follow-up. Relapsed cases were matched with a set of control patients by tumor diameter, pathological stage, tumor histotype, age, hormone receptors and grading. Histopathological and molecular determinants of tumor development and aggressiveness were then analyzed in relapsed versus non-relapsed cases. Stepwise analyses and model structure fitness assessments were carried out to identify clusters of molecular alterations with differential impact on metastatic relapse. Results p53, Bcl-2 and cathepsin D were shown to be coordinately associated with unique levels of relative risk for disease relapse. As many Ras downstream targets, among them matrix metalloproteases, are synergistically upregulated by mutated p53, whole-exon sequence analyses were performed for TP53, Ki-RAS and Ha-RAS, and findings were correlated with clinical phenotypes. Notably, TP53 insertion/deletion mutations were only detected in relapsed cases. Correspondingly, Ha-RAS missense oncogenic mutations were only found in a subgroup of relapsing tumors. Conclusions We have identified clusters of specific molecular alterations that greatly improve prognostic assessment with respect to singularly-analysed indicators. The combined analysis of these multiple tumor-relapse risk factors promises to become a powerful approach to identify patients subgroups with unfavourable disease outcome. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2713-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emanuela Guerra
- Unit of Cancer Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | | | - Pasquale Simeone
- Unit of Cancer Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Giovanna Vacca
- Unit of Cancer Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Rossano Lattanzio
- Unit of Cancer Pathology, CeSI-MeT, University of Chieti, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, University 'G. D'Annunzio', Chieti, Italy
| | - Gerardo Botti
- Department of Pathology "Foundation G.Pascale", National Cancer Institute, Naples, Italy
| | - Valentina Gatta
- Department of Psychological, Health ad Territorial Sciences, School of Medicine and Life Sciences, University 'G. D'Annunzio', Chieti, Italy
| | - Marco D'Aurora
- Department of Psychological, Health ad Territorial Sciences, School of Medicine and Life Sciences, University 'G. D'Annunzio', Chieti, Italy
| | | | - Mauro Piantelli
- Unit of Cancer Pathology, CeSI-MeT, University of Chieti, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, University 'G. D'Annunzio', Chieti, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI-MeT, University of Chieti, Chieti, Italy. .,Department of Neurosciences, Imaging and Clinical Sciences, University 'G. D'Annunzio', Chieti, Italy.
| |
Collapse
|
18
|
Danforth DN. Genomic Changes in Normal Breast Tissue in Women at Normal Risk or at High Risk for Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:109-46. [PMID: 27559297 PMCID: PMC4990153 DOI: 10.4137/bcbcr.s39384] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/17/2016] [Accepted: 04/19/2016] [Indexed: 12/12/2022]
Abstract
Sporadic breast cancer develops through the accumulation of molecular abnormalities in normal breast tissue, resulting from exposure to estrogens and other carcinogens beginning at adolescence and continuing throughout life. These molecular changes may take a variety of forms, including numerical and structural chromosomal abnormalities, epigenetic changes, and gene expression alterations. To characterize these abnormalities, a review of the literature has been conducted to define the molecular changes in each of the above major genomic categories in normal breast tissue considered to be either at normal risk or at high risk for sporadic breast cancer. This review indicates that normal risk breast tissues (such as reduction mammoplasty) contain evidence of early breast carcinogenesis including loss of heterozygosity, DNA methylation of tumor suppressor and other genes, and telomere shortening. In normal tissues at high risk for breast cancer (such as normal breast tissue adjacent to breast cancer or the contralateral breast), these changes persist, and are increased and accompanied by aneuploidy, increased genomic instability, a wide range of gene expression differences, development of large cancerized fields, and increased proliferation. These changes are consistent with early and long-standing exposure to carcinogens, especially estrogens. A model for the breast carcinogenic pathway in normal risk and high-risk breast tissues is proposed. These findings should clarify our understanding of breast carcinogenesis in normal breast tissue and promote development of improved methods for risk assessment and breast cancer prevention in women.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Abstract
Inactivation of the p53 tumor suppressor gene is one of the commonest genetic changes identified in human breast cancer. In this review, the structure and function of the p53 gene and its protein products will be discussed, with particular reference to p53 alterations that contribute to carcinogenesis. The frequency and pattern of p53 alterations in breast cancer will be outlined, laboratory methods for their detection briefly summarized, and the potential use of p53 as a prognostic and predictive marker discussed.
Collapse
|
20
|
Platform Presentations. Toxicol Pathol 2016. [DOI: 10.1177/019262339302100612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
21
|
Thompson MA, Edmonds MD, Liang S, McClintock-Treep S, Wang X, Li S, Eischen CM. miR-31 and miR-17-5p levels change during transformation of follicular lymphoma. Hum Pathol 2015; 50:118-26. [PMID: 26997445 DOI: 10.1016/j.humpath.2015.11.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022]
Abstract
The 30% of patients whose indolent follicular lymphoma transforms to aggressive diffuse large B-cell lymphoma (DLBCL) have poor survival. Reliable predictors of follicular B-cell lymphoma transformation to DLBCL are lacking, and diagnosis of those that will progress is challenging. MicroRNA, which regulates gene expression, has critical functions in the growth and progression of many cancers and contributes to the pathogenesis of lymphoma. Using 5 paired samples from patients who presented with follicular lymphoma and progressed to DLBCL, we identified specific microRNA differentially expressed between the two. Specifically, miR-17-5p levels were low in follicular lymphoma and increased as the disease transformed. In contrast, miR-31 expression was high in follicular lymphoma and decreased as the lymphoma progressed. These results were confirmed in additional unpaired cases of low-grade follicular lymphoma (n = 13) and high-grade follicular lymphoma grade 3 or DLBCL (n = 17). Loss of miR-31 expression in DLBCL was not due to deletion of the locus. Changes in miR-17-5p and miR-31 were not correlated with immunophenotype, genetics, or status of the MYC oncogene. However, increased miR-17-5p expression did significantly correlate with increased expression of p53 protein, which is indicative of mutant TP53. Two pro-proliferative genes, E2F2 and PI3KC2A, were identified as direct messenger RNA targets of miR-31, suggesting that these may contribute to follicular lymphoma transformation. Our results indicate that changes in miR-31 and miR-17-5p reflect the transformation of follicular lymphoma to an aggressive large B-cell lymphoma and may, along with their targets, be viable markers for this process.
Collapse
MESH Headings
- 3' Untranslated Regions
- Adult
- Aged
- Aged, 80 and over
- Binding Sites
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Child
- Class I Phosphatidylinositol 3-Kinases
- Disease Progression
- E2F2 Transcription Factor/genetics
- E2F2 Transcription Factor/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Humans
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/metabolism
- Lymphoma, Follicular/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Neoplasm Grading
- Phenotype
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Transfection
Collapse
Affiliation(s)
- Mary Ann Thompson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Mick D Edmonds
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Shan Liang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Sara McClintock-Treep
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Xuan Wang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Shaoying Li
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Christine M Eischen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232.
| |
Collapse
|
22
|
Ono M, Tsuda H, Kobayashi T, Takeshita F, Takahashi RU, Tamura K, Akashi-Tanaka S, Moriya T, Yamasaki T, Kinoshita T, Yamamoto J, Fujiwara Y, Ochiya T. The expression and clinical significance of ribophorin II (RPN2) in human breast cancer. Pathol Int 2015; 65:301-8. [PMID: 25881688 DOI: 10.1111/pin.12297] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/19/2015] [Indexed: 01/08/2023]
Abstract
Ribophorin II (RPN2), part of the N-oligosaccharyltransferase complex, is highly expressed in breast cancer stem cells and is associated with tumor metastasis through interaction with mutant p53. The clinicopathological implication of RPN2 expression is undetermined. We examined immunohistochemically the expression levels of RPN2 and p53 in primary breast cancer tissues surgically resected from 218 patients. The correlations of RPN2 expression with the intrinsic subtype defined by hormone receptors (HRs) and HER2, clinicopathological parameters, p53 expression, and patients' clinical outcomes were examined. RPN2 was positive in 139 (64%), and the incidence of RPN2 expression was higher in the triple-negative breast cancer (TNBC) (HR-/HER2-) (65%) and HER2-enriched (HR-/HER2+) subtype (95%) than in the luminal A-like (HR+/HER2-) subtype (58%) (P = 0.0009). RPN2 expression was also correlated with p53 nuclear accumulation (P = 0.04). The RPN2-positive/p53-positive patient group showed significantly poorer prognosis than the RPN2-negative group for disease-free survival (P = 0.05) and for overall survival (P = 0.02). By multivariate analyses, the combination of RPN2 and p53 was not an independent prognostic factor. RPN2 expression was correlated with clinically aggressive features of breast cancer. These data support the further clinical application of anti-RPN2 therapy and the development of personalized medicine.
Collapse
Affiliation(s)
- Makiko Ono
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Hitoshi Tsuda
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan.,Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Takayuki Kobayashi
- Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Fumitaka Takeshita
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Ryou-U Takahashi
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenji Tamura
- Breast and Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Sadako Akashi-Tanaka
- Breast and Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Tomoyuki Moriya
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Tamio Yamasaki
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Takayuki Kinoshita
- Breast and Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Junji Yamamoto
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yasuhiro Fujiwara
- Breast and Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
23
|
Abstract
Locally advanced breast cancer (LABC) constitutes a heterogeneous entity that includes advanced-stage primary tumours, cancers with extensive nodal involvement and inflammatory breast carcinomas. Although the definition of LABC can be broadened to include some large operable breast tumours, we use this term to strictly refer to inoperable cancers that are included in the above-mentioned categories. The prognosis of such tumours is often unfavourable; despite aggressive treatment, many patients eventually develop distant metastases and die from the disease. Advances in systemic therapy, including radiation treatment, surgical techniques and the development of new targeted agents have significantly improved clinical outcomes for patients with this disease. Notwithstanding these advances, LABC remains an important clinical problem, particularly in developing countries and those without widely adapted breast cancer awareness programmes. The optimal management of LABC requires a multidisciplinary approach, a well-coordinated treatment schedule and close cooperation between medical, surgical and radiation oncologists. In this Review, we discuss the current state of the art and possible future treatment strategies for patients with LABC.
Collapse
|
24
|
Sun R, Wang X, Zhu H, Mei H, Wang W, Zhang S, Huang J. Prognostic value of LAMP3 and TP53 overexpression in benign and malignant gastrointestinal tissues. Oncotarget 2014; 5:12398-409. [PMID: 25362357 PMCID: PMC4322976 DOI: 10.18632/oncotarget.2643] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/28/2014] [Indexed: 02/06/2023] Open
Abstract
Lysosomal associated membrane protein 3 (LAMP3) is a newly identified tumor-specific protein. It is a downstream target gene of tumor suppressor TP53 and its expression has been associated with hypoxia-induced metastasis and poor overall survival in cervical and breast cancers. However, little is known of LAMP3 protein expression in gastrointestinal cancer and its prognostic value. We determined protein expression of LAMP3 and TP53 in both gastric (n=750) and colorectal (n=479) tissues by immunohistochemistry analysis on tissue microarray (TMA), their expression was correlated with patients' clinical parameters. LAMP3 and TP53 protein expression was significantly higher in cancerous tissues compared to normal and benign tissues. In both gastric and colorectal cancers, high LAMP3 protein expression (LAMP3+) was significantly associated with tumor stage (P=0.014 and P<0.001). No correlation between LAMP3 and TP53 expression was observed. Patients with high LAMP3 expression but not high TP53 expression had a poor overall survival (for gastric cancer P<0.001, CI: 1.762-4.567; for colorectal cancer P=0.036, CI: 1.062-5.980). Our data suggest that epithelial LAMP3 expression is an independent prognostic marker for gastrointestinal cancer.
Collapse
Affiliation(s)
- Rongwei Sun
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Xudong Wang
- Department of Laboratory Medicine, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Huijun Zhu
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Haijun Mei
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Wei Wang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Shu Zhang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Jianfei Huang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| |
Collapse
|
25
|
Milosevic Z, Tanic N, Bankovic J, Stankovic T, Buta M, Lavrnic D, Milovanovic Z, Pupic G, Stojkovic S, Milinkovic V, Ito Y, Dzodic R. Genetic alterations in quadruple malignancies of a patient with multiple sclerosis: their role in malignancy development and response to therapy. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1826-1833. [PMID: 24817989 PMCID: PMC4014273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/05/2014] [Indexed: 06/03/2023]
Abstract
Multiple cancers represent 2.42% of all human cancers and are mainly double or triple cancers. Many possible causes of multiple malignancies have been reported such as genetic alterations, exposure to anti-cancer chemotherapy, radiotherapy, immunosuppressive therapy and reduced immunologic response. We report a female patient with multiple sclerosis and quadruple cancers of different embryological origin. Patient was diagnosed with stage III (T3, N1a, MO) medullary thyroid carcinoma (MTC), multicentric micropapillary thyroid carcinoma, scapular and lumbar melanomas (Clark II, Breslow II), and lobular invasive breast carcinoma (T1a, NO, MO). All tumors present in our patient except micropapillary thyroid carcinomas were investigated for gene alterations known to have a key role in cancer promotion and progression. Tumor samples were screened for the p16 alterations (loss of heterozygosity and homozygous deletions), loss of heterozygosity of PTEN, p53 alterations (mutational status and loss of heterozygosity) and mutational status of RET, HRAS and KRAS. Each type of tumor investigated had specific pattern of analyzed genetic alterations. The most prominent genetic changes were mutual alterations in PTEN and p53 tumor suppressors present in breast cancer and two melanomas. These co-alterations could be crucial for promoting development of multiple malignancies. Moreover the insertion in 4(th) codon of HRAS gene was common for all tumor types investigated. It represents frameshift mutation introducing stop codon at position 5 which prevents synthesis of a full-length protein. Since the inactivated RAS enhances sensitivity to tamoxifen and radiotherapy this genetic alteration could be considered as a good prognostic factor for this patient.
Collapse
Affiliation(s)
- Zorica Milosevic
- Institute for Biological Research “Sinisa Stankovic”, University of BelgradeBulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Nikola Tanic
- Institute for Biological Research “Sinisa Stankovic”, University of BelgradeBulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Jasna Bankovic
- Institute for Biological Research “Sinisa Stankovic”, University of BelgradeBulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Tijana Stankovic
- Institute for Biological Research “Sinisa Stankovic”, University of BelgradeBulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Marko Buta
- Institute for Oncology and Radiology of Serbia, University of BelgradePasterova 14, 11000 Belgrade, Serbia
| | - Dragana Lavrnic
- Institute for Neurology, Clinical Center of Serbia, University of BelgradeDr Subotica 6, 11000 Belgrade, Serbia
- School of Medicine, University of BelgradeDr Subotica 8, 11000 Belgrade, Serbia
| | - Zorka Milovanovic
- Institute for Oncology and Radiology of Serbia, University of BelgradePasterova 14, 11000 Belgrade, Serbia
| | - Gordana Pupic
- Institute for Oncology and Radiology of Serbia, University of BelgradePasterova 14, 11000 Belgrade, Serbia
| | - Sonja Stojkovic
- Institute for Biological Research “Sinisa Stankovic”, University of BelgradeBulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Vedrana Milinkovic
- Institute for Biological Research “Sinisa Stankovic”, University of BelgradeBulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Yasuhiro Ito
- Department of Surgery, Kuma Hospital8-2-35, Shimoyamate-dori, Chuo-ku, Kobe, 650-0011, Japan
| | - Radan Dzodic
- Institute for Oncology and Radiology of Serbia, University of BelgradePasterova 14, 11000 Belgrade, Serbia
- School of Medicine, University of BelgradeDr Subotica 8, 11000 Belgrade, Serbia
| |
Collapse
|
26
|
Ma H, Lu Y, Malone KE, Marchbanks PA, Deapen DM, Spirtas R, Burkman RT, Strom BL, McDonald JA, Folger SG, Simon MS, Sullivan-Halley J, Press MF, Bernstein L. Mortality risk of black women and white women with invasive breast cancer by hormone receptors, HER2, and p53 status. BMC Cancer 2013; 13:225. [PMID: 23642215 PMCID: PMC3648503 DOI: 10.1186/1471-2407-13-225] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 05/01/2013] [Indexed: 12/26/2022] Open
Abstract
Background Black women are more likely than white women to have an aggressive subtype of breast cancer that is associated with higher mortality and this may contribute to the observed black-white difference in mortality. However, few studies have investigated the black-white disparity in mortality risk stratified by breast cancer subtype, defined by estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) status. Furthermore, it is not known whether additional consideration of p53 protein status influences black-white differences in mortality risk observed when considering subtypes defined by ER, PR and HER2 status. Methods Four biomarkers were assessed by immunohistochemistry in paraffin-embedded breast tumor tissue from 1,204 (523 black, 681 white) women with invasive breast cancer, aged 35–64 years at diagnosis, who accrued a median of 10 years’ follow-up. Multivariable Cox proportional hazards regression models were fit to assess subtype-specific black-white differences in mortality risk. Results No black-white differences in mortality risk were observed for women with triple negative (ER-negative [ER-], PR-, and HER2-) subtype. However, older (50–64 years) black women had greater overall mortality risk than older white women if they had been diagnosed with luminal A (ER-positive [ER+] or PR+ plus HER2-) breast cancer (all-cause hazard ratio, HR, 1.88; 95% confidence interval, CI, 1.18 to 2.99; breast cancer-specific HR, 1.51; 95% CI, 0.83 to 2.74). This black-white difference among older women was further confined to those with luminal A/p53- tumors (all-cause HR, 2.22; 95% CI, 1.30 to 3.79; breast cancer-specific HR, 1.89; 95% CI, 0.93 to 3.86). Tests for homogeneity of race-specific HRs comparing luminal A to triple negative subtype and luminal A/p53- to luminal A/p53+ subtype did not achieve statistical significance, although statistical power was limited. Conclusions Our findings suggest that the subtype-specific black-white difference in mortality risk occurs mainly among older women diagnosed with luminal A/p53- breast cancer, which is most likely treatable. These results further suggest that factors other than subtype may be relatively more important in explaining the increased mortality risk seen in older black women.
Collapse
Affiliation(s)
- Huiyan Ma
- Division of Cancer Etiology, Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Shi D, Gu W. Dual Roles of MDM2 in the Regulation of p53: Ubiquitination Dependent and Ubiquitination Independent Mechanisms of MDM2 Repression of p53 Activity. Genes Cancer 2012; 3:240-8. [PMID: 23150757 DOI: 10.1177/1947601912455199] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
MDM2 oncogenic protein is the principal cellular antagonist of the p53 tumor suppresser gene. p53 activity needs exquisite control to elicit appropriate responses to differential cellular stress conditions. p53 becomes stabilized and active upon various types of stresses. However, too much p53 is not beneficial to cells and causes lethality. At the steady state, p53 activity needs to be leashed for cell survival. Early studies suggested that the MDM2 oncoprotein negatively regulates p53 activity through the induction of p53 protein degradation. MDM2 serves as an E3 ubiquitin ligase of p53; it catalyzes polyubiquitination and subsequently induces proteasome degradation to downregulate p53 protein level. However, the mechanism by which MDM2 represses p53 is not a single mode. Emerging evidence reveals another cellular location of MDM2-p53 interaction. MDM2 is recruited to chromatin, specifically the p53 responsive promoter regions, in a p53 dependent manner. MDM2 is proposed to directly inhibit p53 transactivity at chromatin. This article provides an overview of the mechanism by which p53 is repressed by MDM2 in both ubiquitination dependent and ubiquitination independent pathways.
Collapse
Affiliation(s)
- Dingding Shi
- Institute for Cancer Genetics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | |
Collapse
|
28
|
Abstract
Tumor suppressor p53 is critical for suppressing all types of human cancers, including breast cancer. The p53 gene is somatically mutated in over half of all human cancers. The majority of the p53 mutations are missense mutations, leading to the expression of the full-length p53 mutants. Several hotspot mutations, including R175H, are frequently detected in human breast cancer. P53 cancer mutants not only lose tumor suppression activity but, more problematically, also gain new oncogenic activities. Despite correlation of the expression of p53 cancer mutants and the poor prognosis of human breast cancer patients, the roles of p53 cancer mutants in promoting breast cancer remain unclear. We used the humanized p53 cancer mutant knock-in (R175H) mice and mouse mammary tumor virus (MMTV)-Wnt-1 transgenic (mWnt-1) mice to specifically address the gain of function of R175H in promoting breast cancer. Although both R175H/R175HmWnt-1(R175HmWnt-1) and p53(-/-)mWnt-1 mice died from mammary tumor at the same kinetics, which was much earlier than mWnt-1 mice, most of the R175HmWnt-1 mice developed multiple mammary tumors per mouse, whereas p53(-/-)mWnt-1 and mWnt-1 mice mostly developed one tumor per mouse. The multiple mammary tumors arose in the same R175HmWnt-1 mouse exhibited different histological characters. Moreover, R175H gain-of-function mutant expands the mammary epithelial stem cells (MESCs) that give rise to the mammary tumors. As ATM suppresses the expansion of MESCs, the inactivation of ATM by R175H in mammary epithelial cells (MECs) could contribute to the expansion of MESCs in R175HmWnt-1 mice. These findings provide the basis for R175H to promote the initiation of breast cancer by expanding MESCs.
Collapse
|
29
|
Costarelli L, Campagna D, Mauri M, Fortunato L. Intraductal proliferative lesions of the breast-terminology and biology matter: premalignant lesions or preinvasive cancer? Int J Surg Oncol 2012; 2012:501904. [PMID: 22655184 PMCID: PMC3357964 DOI: 10.1155/2012/501904] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 02/21/2012] [Indexed: 11/17/2022] Open
Abstract
Morphological criteria for the diagnosis of intraductal proliferative lesions of the breast have been an object of research and much controversy, and its terminology is rather confusing. Knowledge of the molecular aspects of this disease probably necessitates further research to clarify if these entities can be identified as breast cancer precursors or as a malignant preinvasive disease. These issues are of great interest not only for their biological implications, but also to the clinician who must understand the disease and direct therapies. Molecular studies have shown that epitheliosis (usual ductal hyperplasia) is not monoclonal, while malignant lesions (atypical ductal hyperplasia, flat epithelial atypia, low-grade and high-grade intraductal carcinoma) constantly show these characteristics. These malignant lesions, classified with a DIN grading system (ductal intraepithelial neoplasia), are not obligate precursors of invasive ductal carcinoma and do not represent different evolving grades in a linear model of cancerogenesis. Breast cancerogenesis probably has different pathways with different morphologic precursors.
Collapse
Affiliation(s)
| | - Domenico Campagna
- Department of Pathology, San Giovanni-Addolorata Hospital, Rome, Italy
| | - Maria Mauri
- Department of Medical Oncology, San Giovanni-Addolorata Hospital, Rome, Italy
| | - Lucio Fortunato
- Department of Surgery, San Giovanni-Addolorata Hospital, Rome, Italy
| |
Collapse
|
30
|
Yahya RS, Fouda MI, El-Baz HA, Mosa TE, ElMaksoud MDA. Serum Survivin and TP53 Gene Expression in Children with Acute Lymphoblastic Leukemia. IRANIAN JOURNAL OF PUBLIC HEALTH 2012; 41:37-44. [PMID: 23113120 PMCID: PMC3481665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 12/22/2011] [Indexed: 11/26/2022]
Abstract
BACKGROUND The aim of this study was to detect the prognostic significance of survivin level and the expression of total p53 in acute lymphoblastic leukemia (ALL) and its correlation to patients' outcome. METHODS Sixty two children newly diagnosed with acute lymphoblastic leukemia were treated with chemotherapy and followed up for 2 years or until death. Twenty apparently healthy volunteers with matched age and sex were taken as control. Survivin protein was measured by quantitative sandwich enzyme immunoassay and total human p53 was measured by Flow cytometry in peripheral blood at diagnosis and at complete remission. RESULTS A highly significant elevation (P<0.0001) was found in survivin protein and total p53 levels in acute lymphoblastic leukemia children patients at diagnosis compared to controls. At complete remission a significant decrease of the two indices were found in ALL patients compared to those at diagnosis (P<0.0001). Survivin protein and total p53 was significantly higher in non-survived compared to survived group (P<0.0001 & P=0.016, respectively). A positive correlation was found between survivin level and total human p53 level in children with ALL (r=0.501 & P<0.0001). CCONCLUSION: survivin protein is related to anti-apoptotic proteins and its high expression lead to unsuccessful treatment of ALL. Survivin and TP53 are new prognostic tools in ALL, independent of age and sex.
Collapse
Affiliation(s)
- Raida S Yahya
- Children Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt,Corresponding Author: Tel: 20 0122 49 79 953, E-mail address:
| | - Manal I Fouda
- Dept. of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hatim A El-Baz
- Dept. of Biochemistry, National Research Center, Cairo, Egypt
| | - Tamer E Mosa
- Dept. of Biochemistry, National Research Center, Cairo, Egypt
| | | |
Collapse
|
31
|
Moro J, Tinucci-Costa M, Silveira A, Gerardi D, Alessi A. Reactivity of p53 protein in canine transmissible venereal tumor. ARQ BRAS MED VET ZOO 2010. [DOI: 10.1590/s0102-09352010000200011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The expression of p53 protein was evaluated in canine transmissible venereal tumor (CTVT), as following: natural occurrence (n=8); resistant to chemotherapy (n=4); and allogeneic transplanted in progression (n=8), stable (n=8), and regression (n=8)stages. The collected specimens were submitted to GM1 immunohistochemical reaction. Results showed a mean percentage of immunomarked cells around 18.6% in CTVT of natural occurrence, 23.8% in CTVT resistant to chemotherapy, 22.9% in allogeneic transplanted CTVT in both progression and stable stages, and 35.8% in transplanted CTVT in regression stage. The results suggest that there is a functional abnormality in p53 gene and its products in the studied tumors; although, it is not possible to correlate the percentage of cells marked by p53 and a prognosis.
Collapse
|
32
|
Lee EJ, Kim TJ, Kim DS, Choi CH, Lee JW, Lee JH, Bae DS, Kim BG. p53 alteration independently predicts poor outcomes in patients with endometrial cancer: A clinicopathologic study of 131 cases and literature review. Gynecol Oncol 2010; 116:533-8. [DOI: 10.1016/j.ygyno.2009.11.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 11/11/2009] [Accepted: 11/13/2009] [Indexed: 01/10/2023]
|
33
|
Ma H, Wang Y, Sullivan-Halley J, Weiss L, Marchbanks PA, Spirtas R, Ursin G, Burkman RT, Simon MS, Malone KE, Strom BL, McDonald JA, Press MF, Bernstein L. Use of four biomarkers to evaluate the risk of breast cancer subtypes in the women's contraceptive and reproductive experiences study. Cancer Res 2010; 70:575-87. [PMID: 20068186 DOI: 10.1158/0008-5472.can-09-3460] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Epidemiologic studies suggest that some hormone-related risk factors in breast cancer differentially influence risk for disease subtypes classified by the status of the estrogen and progesterone receptors (ER/PR). However, it remains unclear whether human epidermal growth factor receptor 2 (HER2) or p53 expression status further differentiates these exposure-risk group associations. We evaluated the associations of oral contraceptive (OC) use and reproductive factors with incident invasive breast cancer subtypes among 1,197 population-based cases and 2,015 controls from the Los Angeles County or Detroit components of the Women's Contraceptive and Reproductive Experiences Study. Case-control comparisons by ER/PR/HER2/p53 status were conducted by multivariable polychotomous unconditional logistic regression methods. We found that OC use was not associated with any breast cancer subtype as defined by ER/PR/HER2/p53 status, except for a 2.9-fold increased risk of so-called triple-negative tumors (ER(-)/PR(-)/HER2(-)) among women of 45 to 64 years of age who started OC use before age 18. Parity was associated with a decreased risk of luminal A (ER(+) or PR(+), HER2(-)), luminal B (ER(+) or PR(+)/HER2(+)), and ER(-)/PR(-)/HER2(+) tumors. Age at first full-term pregnancy was positively associated with luminal A tumors among older women. Neither of these reproductive factors was associated with triple-negative tumors. Long duration of breast-feeding lowered the risk of triple-negative and luminal A tumors. p53 status did not define further differential risk patterns. Our findings offer evidence of differences in the hormone-related risk factors between triple-negative cancers and other ER/PR/HER2-defined subtypes of breast cancer.
Collapse
Affiliation(s)
- Huiyan Ma
- Division of Cancer Etiology, Department of Population Sciences, City of Hope Medical Center, Duarte, California 91010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kulić A, Sirotković-Skerlev M, Jelisavac-Cosić S, Herceg D, Kovac Z, Vrbanec D. Anti-p53 antibodies in serum: relationship to tumor biology and prognosis of breast cancer patients. Med Oncol 2009; 27:887-93. [PMID: 19763913 DOI: 10.1007/s12032-009-9301-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 08/25/2009] [Indexed: 02/06/2023]
Abstract
The aim of this study was to analyze the concentration of anti-p53 antibodies in the serum of breast cancer patients and to correlate these results with various clinical, pathological and biochemical parameters. We also wanted to assess the prognostic significance of these antibodies in our patients. Sera from 61 patients with breast cancer and 20 individuals without malignancies were analyzed using enzyme-linked immunoadsorbent assay. High levels of anti-p53 antibodies were detected in twenty-one (35%) breast cancer patients and one control (5%). The difference was statistically significant. We observed an inverse relationship between the anti-p53 antibodies and the age of the patients. We found significant association of anti-p53 antibodies with tumor size, histological grade of the tumors and the number of axillary lymph nodes involved. The levels of anti-p53 antibodies were higher in patients with negative estrogen and progesterone receptors in comparison with patients with positive steroid receptors, but the difference was not statistically significant. No relation was observed between anti-p53 antibodies neither with the Cathepsin D levels in the cytosol nor with the HER-2/neu extracellular domain in the serum. Patients with primary tumors and higher levels of anti-p53 antibodies had shorter 5-year survival than patients with lower levels of anti-p53 antibodies. Our results support the role of anti-p53 antibodies as a biomarker of less favorable phenotype as well as a prognostic factor for patients with breast cancer.
Collapse
Affiliation(s)
- A Kulić
- Department of Pathophysiology, University Hospital Center Zagreb, Kispatićeva 12, Zagreb, 10000, Croatia.
| | | | | | | | | | | |
Collapse
|
35
|
Immunohistochemical expression of dogTERT in canine testicular tumours in relation to PCNA, ki67 and p53 expression. Vet Res Commun 2009; 33:905-19. [DOI: 10.1007/s11259-009-9308-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2009] [Indexed: 11/27/2022]
|
36
|
Cheng H, Liu P, Wang ZC, Zou L, Santiago S, Garbitt V, Gjoerup OV, Iglehart JD, Miron A, Richardson AL, Hahn WC, Zhao JJ. SIK1 couples LKB1 to p53-dependent anoikis and suppresses metastasis. Sci Signal 2009; 2:ra35. [PMID: 19622832 PMCID: PMC2752275 DOI: 10.1126/scisignal.2000369] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resistance to anoikis, the subtype of apoptosis triggered by lack of adhesion, contributes to malignant transformation and the development of metastasis. Although several lines of evidence suggest that p53 plays a critical role in anoikis, the pathway(s) that connect cell detachment to p53 remain undefined. Here, through the use of a kinome-wide loss-of-function screen, we identify the serine-threonine kinase SIK1 (salt-inducible kinase 1) as a regulator of p53-dependent anoikis. Inactivation of SIK1 compromised p53 function in anoikis and allowed cells to grow in an anchorage-independent manner. In vivo, SIK1 loss facilitated metastatic spread and survival of disseminated cells as micrometastases in lungs. The presence of functional SIK1 was required for the activity of the kinase LKB1 in promoting p53-dependent anoikis and suppressing anchorage-independent growth, Matrigel invasion, and metastatic potential. In human cancers, decreased expression of the gene encoding SIK1 closely correlated with development of distal metastases in breast cancers from three independent cohorts. Together, these findings indicate that SIK1 links LKB1 to p53-dependent anoikis and suppresses metastasis.
Collapse
Affiliation(s)
- Hailing Cheng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Duan W, Gao L, Wu X, Hade EM, Gao JX, Ding H, Barsky SH, Otterson GA, Villalona-Calero MA. Expression of a mutant p53 results in an age-related demographic shift in spontaneous lung tumor formation in transgenic mice. PLoS One 2009; 4:e5563. [PMID: 19440353 PMCID: PMC2680060 DOI: 10.1371/journal.pone.0005563] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 04/18/2009] [Indexed: 11/30/2022] Open
Abstract
Background Mutations in the P53 gene are among the most common genetic abnormalities in human lung cancer. Codon 273 in the sequence-specific DNA binding domain is one of the most frequently mutated sites. Methodology To investigate the role of mutant p53 in lung tumorigenesis, a lung specific p53(273H) transgenic mouse model was developed. Rates of lung cancer formation in the transgenic animals and their littermates were evaluated by necropsy studies performed in progressive age cohorts ranging from 4 to 24 months. In order to establish the influence of other common genetic abnormalities in lung tumor formation in the animals, K-Ras gene mutation and p16INK4a (p16) promoter methylation were evaluated in a total of 281 transgenic mice and 189 non-transgenic littermates. Principal Findings At the age extremes of 4–12 and 22–24 months no differences were observed, with very low prevalence of tumors in animals younger than 12 months, and a relatively high prevalence at age 22 months or older. However, the transgenic mice had a significant higher lung tumor rate than their non-transgenic counterparts during the age of 13–21 months, suggesting an age-related shift in lung tumor formation induced by the lung-specific expression of the human mutant p53. Histopathology suggested a more aggressive nature for the transgenic tumors. Older mice (>13 months) had a significantly higher rate of p16 promoter methylation (17% v 82%). In addition, an age related effect was observed for K-Ras codons 12 or 13 mutations, but not for codon 61 mutations. Conclusions/Significance These results would suggest that the mutant p53(273H) contributes to an acceleration in the development of spontaneous lung tumors in these mice. Combination with other genetic and epigenetic alterations occurring after the age of 13 months is intimately linked to its oncogenic potential.
Collapse
Affiliation(s)
- Wenrui Duan
- Comprehensive Cancer Center, The Ohio State University College of Medicine and Public Health, Columbus, Ohio, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Breast cancer is the field of medicine with the greatest presence of nanotechnological therapeutic agents in the clinic. A pegylated form of liposomally encapsulated doxorubicin is routinely used for treatment against metastatic cancer, and albumin nanoparticulate chaperones of paclitaxel were approved for locally recurrent and metastatic disease in 2005. These drugs have yielded substantial clinical benefit, and are steadily gathering greater beneficial impact. Clinical trials currently employing these drugs in combination with chemo and biological therapeutics exceed 150 worldwide. Despite these advancements, breast cancer morbidity and mortality is unacceptably high. Nanotechnology offers potential solutions to the historical challenge that has rendered breast cancer so difficult to contain and eradicate: the extreme biological diversity of the disease presentation in the patient population and in the evolutionary changes of any individual disease, the multiple pathways that drive disease progression, the onset of 'resistance' to established therapeutic cocktails, and the gravity of the side effects to treatment, which result from generally very poor distribution of the injected therapeutic agents in the body. A fundamental requirement for success in the development of new therapeutic strategies is that breast cancer specialists-in the clinic, the pharmaceutical and the basic biological laboratory-and nanotechnologists-engineers, physicists, chemists and mathematicians-optimize their ability to work in close collaboration. This further requires a mutual openness across cultural and language barriers, academic reward systems, and many other 'environmental' divides. This paper is respectfully submitted to the community to help foster the mutual interactions of the breast cancer world with micro- and nano-technology, and in particular to encourage the latter community to direct ever increasing attention to breast cancer, where an extraordinary beneficial impact may result. The paper initiates with an introductory overview of breast cancer, its current treatment modalities, and the current role of nanotechnology in the clinic. Our perspectives are then presented on what the greatest opportunities for nanotechnology are; this follows from an analysis of the role of biological barriers that adversely determine the biological distribution of intravascularly injected therapeutic agents. Different generations of nanotechnology tools for drug delivery are reviewed, and our current strategy for addressing the sequential bio-barriers is also presented, and is accompanied by an encouragement to the community to develop even more effective ones.
Collapse
|
39
|
Liu GY, Luo Q, Xiong B, Pan C, Yin P, Liao HF, Zhuang WC, Gao HZ. Tissue array for Tp53, C-myc, CCND1 gene over-expression in different tumors. World J Gastroenterol 2008; 14:7199-207. [PMID: 19084934 PMCID: PMC2776877 DOI: 10.3748/wjg.14.7199] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To rapidly detect molecular alterations in different malignancies and investigate the possible role of Tp53, C-myc, and CCND1 genes in development of tumors in human organs and their adjacent normal tissues, as well as the possible relation between well- and poorly-differentiated tumors.
METHODS: A tissue array consisting of seven different tumors was generated. The tissue array included 120 points of esophagus, 120 points of stomach, 80 points of rectum, 60 points of thyroid gland, 100 points of mammary gland, 80 points of liver, and 80 points of colon. Expressions of Tp53, C-myc, and CCND1 were determined by RNA in situ hybridization. 3’ terminal digoxin-labeled anti-sense single stranded oligonucleotide and locked nucleic acid modifying probe were used.
RESULTS: The expression level of Tp53 gene was higher in six different carcinoma tissue samples than in paracancerous tissue samples with the exception in colon carcinoma tissue samples (P < 0.05). The expression level of CCND1 gene was significantly different in different carcinoma tissue samples with the exception in esophagus and colon carcinoma tissue samples. The expression level of C-myc gene was different in esophagus carcinoma tissue samples (χ2 = 18.495, P = 0.000), stomach carcinoma tissue samples (χ2 = 23.750, P = 0.000), and thyroid gland tissue samples (χ2 = 10.999, P = 0.004). The intensity of signals was also different in different carcinoma tissue samples and paracancerous tissue samples.
CONCLUSION: Over-expression of the Tp53, CCND1, and C-myc genes appears to play a role in development of human cancer by regulating the expression of mRNA. Tp53, CCND1 and C-myc genes are significantly correlated with the development of different carcinomas.
Collapse
|
40
|
Ohta M, Sugimoto T, Seto M, Mohri D, Asaoka Y, Tada M, Tanaka Y, Yamaji Y, Kanai F, Kawabe T, Omata M. Genetic alterations in colorectal cancers with demethylation of insulin-like growth factor II. Hum Pathol 2008; 39:1301-1308. [PMID: 18619647 DOI: 10.1016/j.humpath.2008.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 02/05/2008] [Accepted: 02/19/2008] [Indexed: 10/21/2022]
Abstract
Loss of genomic imprinting is an epigenetic alteration of some cancers involving the absence of parental origin-specific expression of imprinted genes. Loss of genomic imprinting of insulin-like growth factor II is often detected in colorectal cancer. However, the genetic alterations accompanied by colorectal cancer with insulin-like growth factor II loss of genomic imprinting have not been fully determined. Genomic DNA samples were collected from 52 colorectal cancer tissues and analyzed. The loss of insulin-like growth factor II genomic imprinting status was determined by assessing the demethylation of the insulin-like growth factor II differentially methylated region using bisulfite sequencing. The molecular signatures were also examined: genetic mutations of KRAS, BRAF, and PIK3CA; the expression of CTNNB1 and TP53; and microsatellite instability status. Several cases of colorectal cancer with normal insulin-like growth factor II imprinting were located in the distal colon; in contrast, colorectal cancer with loss of genomic imprinting tended to be found in the proximal colon (22.7 versus 56.6%). The PIK3CA gene mutation was highly detected in normal imprinting tumors compared to colorectal cancers with insulin-like growth factor II loss of genomic imprinting (27.3% versus 6.7%). In multivariate analysis of these clinicopathologic and molecular factors of tumors, statistically significant relationships were observed among the proximal location of the tumor (odds ratio, 12.9; 95% confidence interval, 1.52-110.13), PIK3CA genetic mutation (odds ratio, 0.082; 95% confidence interval, 0.01-0.73), and insulin-like growth factor II genomic imprinting status. Our findings indicate that colorectal cancers with demethylation of the insulin-like growth factor II gene are distinct from normal imprinting tumors, both in clinical and genetic features.
Collapse
Affiliation(s)
- Miki Ohta
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Inflammatory breast cancer (IBC) is a rare, but aggressive form of breast cancer. Despite the progress related to the introduction of primary combination chemotherapy (CT) to the multimodality treatment regimen, the prognosis of IBC remains poor with long-term survival inferior to 50%. Until recently, IBC remained understudied at the molecular level. In the past 10 years, advances have been made in the molecular characterization of the disease. Recently, the use of experimental models and new high-throughput molecular profiling technologies have led to the identification of genes or pathways potentially involved in disease development, which might represent new clinically relevant targets. The aim of this review is to present and discuss what is known about the biology of this particularly aggressive form of breast cancer and to discuss how this knowledge could improve its management.
Collapse
Affiliation(s)
- Emmanuelle Charafe-Jauffret
- Département d'Oncologie Moléculaire, Institut Paoli-Calmettes and UMR599 INSERM, IFR137, 232 Boulevard Sainte-Marguerite, Marseille Cedex 09, France
| | | | | | | |
Collapse
|
42
|
Lu H, Goodell V, Disis ML. Humoral immunity directed against tumor-associated antigens as potential biomarkers for the early diagnosis of cancer. J Proteome Res 2008; 7:1388-94. [PMID: 18311901 DOI: 10.1021/pr700818f] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Over the past decade, it has been demonstrated that cancer is immunogenic, and multiple tumor antigens have been identified in cancer patients. It is now possible to potentially harness the immune response elicited by cancer growth as a potential diagnostic tool. Humoral immunity, or the development of autoantibodies against tumor-associated proteins, may be used as a marker for cancer exposure. Unlike circulating proteins that are shed by bulky tumors, serum autoantibodies are detectable even when antigen expression is minimal. This paper will review the methods used for tumor antigen discovery and overview what is known about autoantibodies targeting common cancer antigens with a focus on breast cancer. Data will be presented modeling the use of tumor antigen associated autoantibodies as a breast cancer diagnostic. The endogenous humoral immune response present in cancer patients may allow the identification of individuals exposed to the malignant transformation of somatic cells.
Collapse
Affiliation(s)
- Hailing Lu
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington 98109, USA.
| | | | | |
Collapse
|
43
|
Triple negative breast cancer: current understanding of biology and treatment options. Curr Opin Obstet Gynecol 2008; 20:40-6. [DOI: 10.1097/gco.0b013e3282f40de9] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Graesslin O, Chantot-Bastaraud S, Lorenzato M, Birembaut P, Quéreux C, Daraï E. Fluorescence in situ Hybridization and Immunohistochemical Analysis of p53 Expression in Endometrial Cancer: Prognostic Value and Relation to Ploidy. Ann Surg Oncol 2007; 15:484-92. [DOI: 10.1245/s10434-007-9712-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2007] [Revised: 10/19/2007] [Accepted: 10/23/2007] [Indexed: 01/09/2023]
|
45
|
Ihemelandu CU, Leffall LD, Dewitty RL, Naab TJ, Mezghebe HM, Makambi KH, Adams-Campbell L, Frederick WA. Molecular Breast Cancer Subtypes in Premenopausal and Postmenopausal African-American Women: Age-Specific Prevalence and Survival. J Surg Res 2007; 143:109-18. [DOI: 10.1016/j.jss.2007.03.085] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 03/13/2007] [Accepted: 03/29/2007] [Indexed: 12/17/2022]
|
46
|
Ihemelandu CU, Leffall LD, Dewitty RL, Naab TJ, Mezghebe HM, Makambi KH, Adams-Campbell L, Frederick WA. Molecular Breast Cancer Subtypes in Premenopausal African-American Women, Tumor Biologic Factors and Clinical Outcome. Ann Surg Oncol 2007; 14:2994-3003. [PMID: 17647064 DOI: 10.1245/s10434-007-9477-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 05/14/2007] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Breast cancer is currently viewed as a heterogeneous disease made up of various subtypes, with distinct differences in prognosis. Our goal was to study the distribution and to characterize the clinical and biological factors that influence the behavior and clinical management of the different molecular breast cancer subtypes in premenopausal African-American women. METHODS A retrospective analysis of Howard University Hospital tumor registry, for all premenopausal African-American women aged less than 50 years, diagnosed with breast cancer from 1998-2005, was performed. RESULTS The luminal A subtype was the most prevalent (50.0%), vs basal-cell-like (23.2%), luminal B (14.1%), and HER-2/neu (12.7%). However when stratified by age groups, results showed that in the age group <35 years the basal-cell-like subtype was the most prevalent (55.6%), vs 25.9%, 14.8%, and 5.6% for luminal A, luminal B, and HER-2/neu subtypes, respectively (P < .000). P53 mutation was more prevalent in the basal-cell-like subtype compared to luminal A (48.0% vs 18.6%, P < .01). The expression of the Bcl-2 gene differed by subtype, with the luminal A and luminal B subtypes more likely to overexpress the Bcl-2 gene (89.1% luminal A, 80.0% luminal B vs 47.6% basal-cell-like and 40.0% HER-2/neu, P < .000). Though not statistically significant, HER-2/neu and basal-cell-like subtypes had the shortest survival time (P < .31). CONCLUSION The high prevalence of the basal-cell-like subtype in young premenopausal African-American women aged <35 years may contribute to the poorer prognosis observed in this cohort of African-American women.
Collapse
MESH Headings
- Adult
- Black or African American/genetics
- Biomarkers, Tumor/genetics
- Breast Neoplasms/ethnology
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/surgery
- Carcinoma, Basal Cell/ethnology
- Carcinoma, Basal Cell/genetics
- DNA Mutational Analysis
- Disease-Free Survival
- District of Columbia
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/physiology
- Genes, bcl-2/genetics
- Genes, erbB-2/genetics
- Genes, p53/genetics
- Hospitals, University
- Humans
- Middle Aged
- Neoplasms, Hormone-Dependent/ethnology
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/mortality
- Oligonucleotide Array Sequence Analysis
- Premenopause
- Prognosis
- Receptors, Estrogen/genetics
- Receptors, Progesterone/genetics
- Registries
- Retrospective Studies
- SEER Program
Collapse
Affiliation(s)
- Chukwuemeka U Ihemelandu
- Department of Surgery, Howard University Hospital, 2041 Georgia Avenue, N.W., Washington, D.C 20060, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lu X, Lu X, Wang ZC, Iglehart JD, Zhang X, Richardson AL. Predicting features of breast cancer with gene expression patterns. Breast Cancer Res Treat 2007; 108:191-201. [PMID: 18297396 DOI: 10.1007/s10549-007-9596-6] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2006] [Accepted: 04/05/2007] [Indexed: 01/17/2023]
Abstract
Data from gene expression arrays hold an enormous amount of biological information. We sought to determine if global gene expression in primary breast cancers contained information about biologic, histologic, and anatomic features of the disease in individual patients. Microarray data from the tumors of 129 patients were analyzed for the ability to predict biomarkers [estrogen receptor (ER) and HER2], histologic features [grade and lymphatic-vascular invasion (LVI)], and stage parameters (tumor size and lymph node metastasis). Multiple statistical predictors were used and the prediction accuracy was determined by cross-validation error rate; multidimensional scaling (MDS) allowed visualization of the predicted states under study. Models built from gene expression data accurately predict ER and HER2 status, and divide tumor grade into high-grade and low-grade clusters; intermediate-grade tumors are not a unique group. In contrast, gene expression data is inaccurate at predicting tumor size, lymph node status or LVI. The best model for prediction of nodal status included tumor size, LVI status and pathologically defined tumor subtype (based on combinations of ER, HER2, and grade); the addition of microarray-based prediction to this model failed to improve the prediction accuracy. Global gene expression supports a binary division of ER, HER2, and grade, clearly separating tumors into two categories; intermediate values for these bio-indicators do not define intermediate tumor subsets. Results are consistent with a model of regional metastasis that depends on inherent biologic differences in metastatic propensity between breast cancer subtypes, upon which time and chance then operate.
Collapse
Affiliation(s)
- Xuesong Lu
- Bioinformatics Division, TNLIST and Department of Automation, Tsinghua University, Beijing 100084, China.
| | | | | | | | | | | |
Collapse
|
48
|
Zhang P, Zhang J, Sheng H, Russo JJ, Osborne B, Buetow K. Gene functional similarity search tool (GFSST). BMC Bioinformatics 2006; 7:135. [PMID: 16536867 PMCID: PMC1421445 DOI: 10.1186/1471-2105-7-135] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Accepted: 03/14/2006] [Indexed: 11/21/2022] Open
Abstract
Background With the completion of the genome sequences of human, mouse, and other species and the advent of high throughput functional genomic research technologies such as biomicroarray chips, more and more genes and their products have been discovered and their functions have begun to be understood. Increasing amounts of data about genes, gene products and their functions have been stored in databases. To facilitate selection of candidate genes for gene-disease research, genetic association studies, biomarker and drug target selection, and animal models of human diseases, it is essential to have search engines that can retrieve genes by their functions from proteome databases. In recent years, the development of Gene Ontology (GO) has established structured, controlled vocabularies describing gene functions, which makes it possible to develop novel tools to search genes by functional similarity. Results By using a statistical model to measure the functional similarity of genes based on the Gene Ontology directed acyclic graph, we developed a novel Gene Functional Similarity Search Tool (GFSST) to identify genes with related functions from annotated proteome databases. This search engine lets users design their search targets by gene functions. Conclusion An implementation of GFSST which works on the UniProt (Universal Protein Resource) for the human and mouse proteomes is available at GFSST Web Server. GFSST provides functions not only for similar gene retrieval but also for gene search by one or more GO terms. This represents a powerful new approach for selecting similar genes and gene products from proteome databases according to their functions.
Collapse
Affiliation(s)
- Peisen Zhang
- Laboratory of Population Genetics, National Cancer Institute, NIH, Bethesda, USA
| | - Jinghui Zhang
- Laboratory of Population Genetics, National Cancer Institute, NIH, Bethesda, USA
| | - Huitao Sheng
- Columbia Genome Center, Columbia University, New York, USA
| | - James J Russo
- Columbia Genome Center, Columbia University, New York, USA
| | | | - Kenneth Buetow
- Laboratory of Population Genetics, National Cancer Institute, NIH, Bethesda, USA
| |
Collapse
|
49
|
Shimizu K, Ueda Y, Yamagishi H. Titration of serum p53 antibodies in patients with gastric cancer: a single-institute study of 40 patients. Gastric Cancer 2006; 8:214-9. [PMID: 16328595 DOI: 10.1007/s10120-005-0337-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Accepted: 05/19/2005] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alterations of the p53 tumor suppressor gene are the most commonly observed genetic abnormalities in many different types of human malignancies. The accumulation of mutant p53 often leads to the production of p53 antibody (p53-Ab) in the sera of patients with various cancers. To evaluate the clinical implications of serum p53-Abs in patients with gastric cancer, we compared p53-Abs with conventional tumor markers such as carcinoembryonic antigen (CEA) and carbohydrate antigen (CA)19-9. METHODS Serum samples were obtained preoperatively from 40 patients with histologically confirmed gastric adenocarcinoma, including 28 (70%) patients in stage Ia. The serum p53-Abs were assessed by enzyme-linked immunosorbent assay, using a new version of a highly specific, quantitative p53-Abs Kit (MESACUP Kit II). RESULTS p53-Abs were detected in 6 (15%) of 40 patients with gastric cancer, including 3 patients with early gastric cancer. Seven (17.5%) of the 40 patients were positive for CEA in serum. However, none of 7 patients with high CEA levels were positive for p53-Abs. No significant correlation of p53-Abs with patient age, sex, pathological parameters, tumor markers such as CEA and CA19-9, or poor survival (P = 0.116) was observed. CONCLUSION Although we employed the latest version of the p53-Abs Kit, the sensitivity of serum p53-Ab in gastric cancer patients was relatively low. No correlation was found between the presence of p53-Ab and the staging of cancer or survival. However, serum p53-Ab was detectable in patients with gastric cancer even in the early stages of disease. In addition, it may be independent of CEA and CA19-9.
Collapse
Affiliation(s)
- Keiji Shimizu
- Department of Surgery, Division of Digestive Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-dori Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | | | | |
Collapse
|
50
|
Brown NM, Stenzel TT, Friedman PN, Henslee J, Huper G, Marks JR. Evaluation of expression based markers for the detection of breast cancer cells. Breast Cancer Res Treat 2005; 97:41-7. [PMID: 16319979 DOI: 10.1007/s10549-005-9085-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Accepted: 09/23/2005] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Genes that are expressed in a highly tissue- or disease-specific manner provide possible targets for therapeutics, early detection of cancer, and monitoring of disease burden during and after treatment. Further, genes of this type that code for secreted or shed proteins may allow for serum detection of the product facilitating our ability to specifically detect the cancer in all circumstances. To this end, we are working towards identification and characterization of such genes that are specifically expressed in breast epithelium. In the current study, we have measured the expression of two markers that emerged from a screen of the Incyte LifeSeq Database and were subsequently shown to be highly restricted to breast epithelium termed BU101 (also called Lipophilin B) and BS106 (small mucin-like protein). These two novel markers were compared with two other candidate markers, Mammaglobin and Cytokeratin 19 (CK19). METHODS Utilizing quantitative real-time PCR, we compared the expression of these four genes in a series of 95 primary breast cancers, 9 lymph nodes from breast cancer patients, 13 lymph nodes from non-cancer patients and 10 normal breast tissues. RESULTS Cytokeratin was shown to be highly sensitive in detecting all breast cancers, while BU101, BS106 and Mammaglobin were more restricted. CONCLUSION While no one of the these markers efficiently detects all breast cancers, a combination of two or more could achieve a very high sensitivity in assaying for circulating or occult breast cancer cells.
Collapse
Affiliation(s)
- Nicholas M Brown
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|