1
|
Szmyd M, Zanib A, Behlow V, Hallman E, Pfiffner S, Yaldo R, Prudhomme N, Farrar K, Dinda S. Modulation of Estrogen Receptor Alpha (ERα) and Tumor Suppressor Gene BRCA1 in Breast Cancer Cells by Bazedoxifene Acetate (BZA). Cancers (Basel) 2024; 16:699. [PMID: 38398090 PMCID: PMC10886716 DOI: 10.3390/cancers16040699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Selective estrogen receptor modulators (SERMs) are steroid analogs with dual functionality, acting as partial estrogen receptor agonists to preserve postmenopausal bone density and as estrogen receptor antagonists in breast tissue. Bazedoxifene acetate (BZA) is an FDA-approved, third-generation SERM used in the treatment of osteoporosis in women. It demonstrates potential as a therapeutic option for breast cancer patients undergoing endocrine therapy. Our study aimed to assess BZA's effects on Estrogen Receptor Alpha (ERα) and tumor suppressor gene BRCA1 in T-47D and MCF-7 breast cancer cells, using Western blots, cellular viability, apoptosis assays, and RT-qPCR. Cells were cultured in 5% charcoal-stripped fetal bovine serum for six days to deplete endogenous steroids. Following a 24 h exposure to 2 µM BZA (optimal concentration determined from 1 nM-2 µM studies), Western blot analyses revealed reduced ERα and BRCA1 protein levels in both cell lines. ERα decreased by 48-63% and BRCA1 by 61-64%, indicating sensitivity to antiestrogens. Cytolocalization of ERα and BRCA1 remained unchanged after BZA and 17-β-estradiol (E2) treatment. ESR1 mRNA expression correlated with Western blot findings. Image cytometric analysis using the stain, propidium iodide, detected decreased cellular proliferation in T-47D and MCF-7 cells following a 6-day treatment ranging from 1 nM to 2 µM BZA. BZA treatment alone led to a tenfold reduction in cellular proliferation compared to estrogen-treated cells, suggesting antiproliferative effects. Understanding BZA's modulation of BRCA1 and ERα, along with their mechanistic interactions, is vital for comprehending its impact on breast cancer tumor suppressors and hormone receptors.
Collapse
Affiliation(s)
- Monica Szmyd
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Department of Foundational Medical Sciences, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Aisha Zanib
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Victoria Behlow
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Erin Hallman
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Samantha Pfiffner
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Raquel Yaldo
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Nina Prudhomme
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Katelyn Farrar
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Sumi Dinda
- Department of Clinical and Diagnostic Sciences, School of Health Sciences, Oakland University, Rochester, MI 48309, USA; (M.S.); (A.Z.); (V.B.); (E.H.); (S.P.); (R.Y.); (K.F.)
- Institute of Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI 48309, USA
- Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| |
Collapse
|
2
|
Desai N, Morris JS, Baladandayuthapani V. NetCellMatch: Multiscale Network-Based Matching of Cancer Cell Lines to Patients Using Graphical Wavelets. Chem Biodivers 2022; 19:e202200746. [PMID: 36279370 PMCID: PMC10066864 DOI: 10.1002/cbdv.202200746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/21/2022] [Indexed: 12/27/2022]
Abstract
Cancer cell lines serve as model in vitro systems for investigating therapeutic interventions. Recent advances in high-throughput genomic profiling have enabled the systematic comparison between cell lines and patient tumor samples. The highly interconnected nature of biological data, however, presents a challenge when mapping patient tumors to cell lines. Standard clustering methods can be particularly susceptible to the high level of noise present in these datasets and only output clusters at one unknown scale of the data. In light of these challenges, we present NetCellMatch, a robust framework for network-based matching of cell lines to patient tumors. NetCellMatch first constructs a global network across all cell line-patient samples using their genomic similarity. Then, a multi-scale community detection algorithm integrates information across topologically meaningful (clustering) scales to obtain Network-Based Matching Scores (NBMS). NBMS are measures of cluster robustness which map patient tumors to cell lines. We use NBMS to determine representative "avatar" cell lines for subgroups of patients. We apply NetCellMatch to reverse-phase protein array data obtained from The Cancer Genome Atlas for patients and the MD Anderson Cell Line Project for cell lines. Along with avatar cell line identification, we evaluate connectivity patterns for breast, lung, and colon cancer and explore the proteomic profiles of avatars and their corresponding top matching patients. Our results demonstrate our framework's ability to identify both patient-cell line matches and potential proteomic drivers of similarity. Our methods are general and can be easily adapted to other'omic datasets.
Collapse
Affiliation(s)
- Neel Desai
- Division of Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jeffrey S Morris
- Division of Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
3
|
Ravishankar K, Jiang X, Leddin EM, Morcos F, Cisneros GA. Computational compensatory mutation discovery approach: Predicting a PARP1 variant rescue mutation. Biophys J 2022; 121:3663-3673. [PMID: 35642254 PMCID: PMC9617126 DOI: 10.1016/j.bpj.2022.05.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/02/2022] Open
Abstract
The prediction of protein mutations that affect function may be exploited for multiple uses. In the context of disease variants, the prediction of compensatory mutations that reestablish functional phenotypes could aid in the development of genetic therapies. In this work, we present an integrated approach that combines coevolutionary analysis and molecular dynamics (MD) simulations to discover functional compensatory mutations. This approach is employed to investigate possible rescue mutations of a poly(ADP-ribose) polymerase 1 (PARP1) variant, PARP1 V762A, associated with lung cancer and follicular lymphoma. MD simulations show PARP1 V762A exhibits noticeable changes in structural and dynamical behavior compared with wild-type (WT) PARP1. Our integrated approach predicts A755E as a possible compensatory mutation based on coevolutionary information, and molecular simulations indicate that the PARP1 A755E/V762A double mutant exhibits similar structural and dynamical behavior to WT PARP1. Our methodology can be broadly applied to a large number of systems where single-nucleotide polymorphisms have been identified as connected to disease and can shed light on the biophysical effects of such changes as well as provide a way to discover potential mutants that could restore WT-like functionality. This can, in turn, be further utilized in the design of molecular therapeutics that aim to mimic such compensatory effect.
Collapse
Affiliation(s)
| | - Xianli Jiang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emmett M Leddin
- Department of Chemistry, University of North Texas, Denton, Texas
| | - Faruck Morcos
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas; Department of Bioengineering, The University of Texas at Dallas, Richardson, Texas; Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas.
| | - G Andrés Cisneros
- Department of Chemistry, University of North Texas, Denton, Texas; Department of Physics, The University of Texas at Dallas, Richardson, Texas; Department of Chemistry, The University of Texas at Dallas, Richardson, Texas.
| |
Collapse
|
4
|
Sang L, He YJ, Kang J, Ye H, Bai W, Luo XD, Sun J. Mitochondrial Deoxyguanosine Kinase Regulates NAD + Biogenesis Independent of Mitochondria Complex I Activity. Front Oncol 2020; 10:570656. [PMID: 33392072 PMCID: PMC7775518 DOI: 10.3389/fonc.2020.570656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Overexpression of DGUOK promotes mitochondria oxidative phosphorylation and lung adenocarcinoma progression. However, the role and mechanism of DGUOK in regulation of mitochondria function and lung cancer progression still poorly understood. Here we demonstrated that DGUOK regulated NAD+ biogenesis. Depletion of the DGUOK significantly decreased NAD+ level. Furthermore, knockout of the DGUOK considerably reduced expression of the NMNAT2, a key molecule controlling NAD+ synthesis, at both mRNA and protein levels. Ectopic expression of the NMNAT2 abrogated the effect of knockdown of DGUOK on NAD+. Notably, this regulation is independent of DGUOK -mediated mitochondria complex I activity. We also showed that NMNAT2 was highly expressed in lung adenocarcinoma and negatively correlated with the patient overall survival. Our study suggested that DGUOK regulates NAD+ in a NMNAT2 dependent manner and DGUOK-NMNAT2-NAD+ axis could be a potential therapeutic target in lung adenocarcinoma.
Collapse
Affiliation(s)
- Lei Sang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Ying-Jie He
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Jiaxin Kang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Hongyi Ye
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Weiyu Bai
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xiao-Dong Luo
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Jianwei Sun
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| |
Collapse
|
5
|
Arif T, Anwar N. Promoter Hypermethylation and Expression Changes of BRCA1 Gene in a Cohort of Sporadic Breast Cancer Cases among Pakistani Population. Asian Pac J Cancer Prev 2020; 21:2395-2401. [PMID: 32856871 PMCID: PMC7771953 DOI: 10.31557/apjcp.2020.21.8.2395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Indexed: 11/25/2022] Open
Abstract
Objective: The purpose of our study was to determine the frequency of BRCA1 promoter hypermethylation and its association with expression changes of BRCA1 and main morphological features in sporadic breast cancer. Methods: A retrospective review of cases was performed to select those with specific morphological features suggestive of breast cancer. BRCA1 promoter hypermethylation and changes in protein expression were evaluated in 30 cancerous and 30 non-cancerous tissue samples. A tissue microarray containing samples from normal and tumor tissue was prepared and stained for BRCA1 protein expression using a commercially available monoclonal antibody against BRCA1 (Ab-1) clone MS110 (mAb). DNA was extracted using modified protocol of Qiagen minikit. DNA was modified using a Bisulfite conversion kit and BRCA1 hypermethylation was detected using a methylation specific PCR. Results: Promoter hypermethylation was negative in 30 non-cancerous samples with retained BRCA1 protein expression. Methylation was positive in 82.6% (n=19/23) of the sporadic cancer samples that had loss of BRCA1 expression and 50% (n=2/4) of the samples with equivocal protein expression. Methylation was negative in all the sporadic breast cancer samples (n=3/3) with retained protein expression. Chi-square analysis showed significant association of BRCA1 promoter methylation with decreased protein expression (P=0.016) and co-existence of loss of BRCA1 and Her2neu at chromosome 17 (P=0.026) respectively. There was no significant association of BRCA1 methylation with morphological features excluding necrosis (P=0.035). Promoter hypermethylation was found to be most common (68.75%) among Triple Negative Breast Cancer (TNBC) females less than 45 years old. Conclusion: Our study suggests that BRCA1 promoter hypermethylation has significant contribution in sporadic breast carcinogenesis. This was our preliminary study in Pakistan. Further studies aimed to determine the in-depth mechanisms of BRCA1 epigenetics in TNBC. BRCAness enriched phenotype in TNBC might be used as a biomarker for the exploitation of therapeutic and clinical implications.
Collapse
Affiliation(s)
- Taqdees Arif
- MPhil Molecular Pathology and Genomics, Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
| | - Natasha Anwar
- Consultant Molecular Pathologist, Agha Khan University Hospital Lahore Stat Lab, Lahore, Pakistan
| |
Collapse
|
6
|
Putri JF, Bhargava P, Dhanjal JK, Yaguchi T, Sundar D, Kaul SC, Wadhwa R. Mortaparib, a novel dual inhibitor of mortalin and PARP1, is a potential drug candidate for ovarian and cervical cancers. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:499. [PMID: 31856867 PMCID: PMC6923857 DOI: 10.1186/s13046-019-1500-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Background Mortalin is enriched in a large variety of cancers and has been shown to contribute to proliferation and migration of cancer cells in multiple ways. It has been shown to bind to p53 protein in cell cytoplasm and nucleus causing inactivation of its tumor suppressor activity in cancer cells. Several other activities of mortalin including mitochondrial biogenesis, ATP production, chaperoning, anti-apoptosis contribute to pro-proliferative and migration characteristics of cancer cells. Mortalin-compromised cancer cells have been shown to undergo apoptosis in in vitro and in vivo implying that it could be a potential target for cancer therapy. Methods We implemented a screening of a chemical library for compounds with potential to abrogate cancer cell specific mortalin-p53 interactions, and identified a new compound (named it as Mortaparib) that caused nuclear enrichment of p53 and shift in mortalin from perinuclear (typical of cancer cells) to pancytoplasmic (typical of normal cells). Biochemical and molecular assays were used to demonstrate the effect of Mortaparib on mortalin, p53 and PARP1 activities. Results Molecular homology search revealed that Mortaparib is a novel compound that showed strong cytotoxicity to ovarian, cervical and breast cancer cells. Bioinformatics analysis revealed that although Mortaparib could interact with mortalin, its binding with p53 interaction site was not stable. Instead, it caused transcriptional repression of mortalin leading to activation of p53 and growth arrest/apoptosis of cancer cells. By extensive computational and experimental analyses, we demonstrate that Mortaparib is a dual inhibitor of mortalin and PARP1. It targets mortalin, PARP1 and mortalin-PARP1 interactions leading to inactivation of PARP1 that triggers growth arrest/apoptosis signaling. Consistent with the role of mortalin and PARP1 in cancer cell migration, metastasis and angiogenesis, Mortaparib-treated cells showed inhibition of these phenotypes. In vivo tumor suppression assays showed that Mortaparib is a potent tumor suppressor small molecule and awaits clinical trials. Conclusion These findings report (i) the discovery of Mortaparib as a first dual inhibitor of mortalin and PARP1 (both frequently enriched in cancers), (ii) its molecular mechanism of action, and (iii) in vitro and in vivo tumor suppressor activity that emphasize its potential as an anticancer drug.
Collapse
Affiliation(s)
- Jayarani F Putri
- DBT-AIST International Laboratory for Advanced Biomedicine [DAILAB], DAICENTER, National Institute of Advanced Industrial Science and Technology [AIST], Central 5-41, Higashi 1-1-1, Tsukuba, Ibaraki, 305 8565, Japan
| | - Priyanshu Bhargava
- DBT-AIST International Laboratory for Advanced Biomedicine [DAILAB], DAICENTER, National Institute of Advanced Industrial Science and Technology [AIST], Central 5-41, Higashi 1-1-1, Tsukuba, Ibaraki, 305 8565, Japan
| | - Jaspreet Kaur Dhanjal
- DBT-AIST International Laboratory for Advanced Biomedicine [DAILAB], DAICENTER, National Institute of Advanced Industrial Science and Technology [AIST], Central 5-41, Higashi 1-1-1, Tsukuba, Ibaraki, 305 8565, Japan.,DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology [IIT] Delhi, New Delhi, Hauz Khas, 110 016, India
| | - Tomoko Yaguchi
- DBT-AIST International Laboratory for Advanced Biomedicine [DAILAB], DAICENTER, National Institute of Advanced Industrial Science and Technology [AIST], Central 5-41, Higashi 1-1-1, Tsukuba, Ibaraki, 305 8565, Japan
| | - Durai Sundar
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology [IIT] Delhi, New Delhi, Hauz Khas, 110 016, India
| | - Sunil C Kaul
- DBT-AIST International Laboratory for Advanced Biomedicine [DAILAB], DAICENTER, National Institute of Advanced Industrial Science and Technology [AIST], Central 5-41, Higashi 1-1-1, Tsukuba, Ibaraki, 305 8565, Japan.
| | - Renu Wadhwa
- DBT-AIST International Laboratory for Advanced Biomedicine [DAILAB], DAICENTER, National Institute of Advanced Industrial Science and Technology [AIST], Central 5-41, Higashi 1-1-1, Tsukuba, Ibaraki, 305 8565, Japan.
| |
Collapse
|
7
|
Li SY, Wu HC, Mai HF, Zhen JX, Li GS, Chen SJ. Microarray-based analysis of whole-genome DNA methylation profiling in early detection of breast cancer. J Cell Biochem 2018; 120:658-670. [PMID: 30203578 DOI: 10.1002/jcb.27423] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022]
Abstract
Emerging evidence indicated that changes in DNA methylation early in breast cancer (BC) development might be clinically relevant for therapeutic decisions. Through analysis of whole-genome gene expression microarray and DNA methylation microarray, we explored genes with abnormal DNA methylation in BC for early detection. Firstly, human BC tissues and adjacent non-cancerous tissues were collected from nine BC patients. Gene expression microarray sequencing was conducted for identifying differentially expressed genes and DNA methylation microarray sequencing for differentially methylated genes in BC. Differentially expressed genes and methylated genes in BC were further explored using the Cancer Genome Atlas database. The correlation between DNA methylation and gene expression was illustrated by multiple comparisons. In other 60 clinical samples, methylation specific polymerase chain reaction (PCR) and reverse transcription quantitative PCR were applied for the methylation of HOXA4 and IGF1 genes in BC and adjacent non-cancerous tissues. In total, 1680 upregulated genes and 1249 downregulated genes were determined in BC. Chromosome 16 and 17 showed more differentially methylated genes, and DNA methylation level was increased in BC tissues in each gene region. Chromosome 19 showed more differentially methylated genes, and DNA methylation level was increased in BC tissues in the exoniensis 1, untranslated region-5 and transcriptional start site 200 gene regions. In other 60 clinical samples, HOXA4 and IGF1 in BC tissues presented increased DNA methylation and decreased gene expression in BC. MCF7 cells treated with RG108 showed decreased HOXA4 and IGF1 expressions. It was estimated that HOXA4 and IGF1 were identified with increased DNA methylation and decreased gene expression in BC, which may serve as biomarkers in early BC detection.
Collapse
Affiliation(s)
- Shao-Ying Li
- Department of Thyroid and Breast Surgery, Baoan Maternal and Child Health Hospital, Jinan University, Sanming Project of Medicine in Shenzhen (SZSM201606088), Shenzhen, China
| | - Hua-Cong Wu
- Department of Thyroid and Breast Surgery, Baoan Maternal and Child Health Hospital, Jinan University, Sanming Project of Medicine in Shenzhen (SZSM201606088), Shenzhen, China
| | - Hui-Fen Mai
- Department of Thyroid and Breast Surgery, Baoan Maternal and Child Health Hospital, Jinan University, Sanming Project of Medicine in Shenzhen (SZSM201606088), Shenzhen, China
| | - Jian-Xin Zhen
- Department of Thyroid and Breast Surgery, Baoan Maternal and Child Health Hospital, Jinan University, Sanming Project of Medicine in Shenzhen (SZSM201606088), Shenzhen, China
| | - Gui-Sen Li
- Department of Thyroid and Breast Surgery, Baoan Maternal and Child Health Hospital, Jinan University, Sanming Project of Medicine in Shenzhen (SZSM201606088), Shenzhen, China
| | - Shao-Jun Chen
- Department of Breast Surgery, Shenzhen Maternal and Child Health Hospital, Shenzhen, China
| |
Collapse
|
8
|
Hesari Z, Nourbakhsh M, Hosseinkhani S, Abdolvahabi Z, Alipour M, Tavakoli-Yaraki M, Ghorbanhosseini SS, Yousefi Z, Jafarzadeh M, Yarahmadi S. Down-regulation of NAMPT expression by mir-206 reduces cell survival of breast cancer cells. Gene 2018; 673:149-158. [PMID: 29886033 DOI: 10.1016/j.gene.2018.06.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 01/10/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD) is a critical coenzyme for all living cells. Nicotinamide phosphoribosyltransferase (NAMPT) functions as a key enzyme in the salvage pathway of NAD biosynthesis. Cancer cells have higher rate of NAD consumption and therefore NAMPT is essential for their survival. Thus, we investigated the effect of NAMPT inhibition by miR-206 on breast cancer cell survival. Breast cancer cells were transfected with miR-206 mimic, inhibitor and their negative controls. NAMPT levels were assessed by real-time PCR as well as western blotting. Cell survival assay and quantification of NAD level were performed by using colorimetric methods. Apoptosis assay was performed by labeling cells with Annexin V-FITC and propidium iodide followed by the flow cytometric analysis. Bioinformatics analysis was done to assess whether NAMPT 3'-UTR is a direct target of miR-206 and the results were confirmed by the luciferase reporter assay. NAMPT 3'-UTR was shown to be a direct target of miR-206. miR-206 reduced NAMPT expression at the protein level, leading to a significant decrease in the intracellular NAD level and subsequent decline in cell survival and induction of apoptosis. Targeting of NAMPT-mediated NAD salvage pathway by miR-206 might provide a new insight in the possible molecular mechanism of breast cancer cell growth regulation. This pathway might provide a new approach for breast cancer therapy.
Collapse
Affiliation(s)
- Zahra Hesari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zohreh Abdolvahabi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Alipour
- Department of Nano biotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran; Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran; Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences (JUMS), Jahrom, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Zeynab Yousefi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Meisam Jafarzadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Yarahmadi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Makvandi M, Pantel A, Schwartz L, Schubert E, Xu K, Hsieh CJ, Hou C, Kim H, Weng CC, Winters H, Doot R, Farwell MD, Pryma DA, Greenberg RA, Mankoff DA, Simpkins F, Mach RH, Lin LL. A PET imaging agent for evaluating PARP-1 expression in ovarian cancer. J Clin Invest 2018; 128:2116-2126. [PMID: 29509546 PMCID: PMC5919879 DOI: 10.1172/jci97992] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/28/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP) inhibitors are effective in a broad population of patients with ovarian cancer; however, resistance caused by low enzyme expression of the drug target PARP-1 remains to be clinically evaluated in this context. We hypothesize that PARP-1 expression is variable in ovarian cancer and can be quantified in primary and metastatic disease using a novel PET imaging agent. METHODS We used a translational approach to describe the significance of PET imaging of PARP-1 in ovarian cancer. First, we produced PARP1-KO ovarian cancer cell lines using CRISPR/Cas9 gene editing to test the loss of PARP-1 as a resistance mechanism to all clinically used PARP inhibitors. Next, we performed preclinical microPET imaging studies using ovarian cancer patient-derived xenografts in mouse models. Finally, in a phase I PET imaging clinical trial we explored PET imaging as a regional marker of PARP-1 expression in primary and metastatic disease through correlative tissue histology. RESULTS We found that deletion of PARP1 causes resistance to all PARP inhibitors in vitro, and microPET imaging provides proof of concept as an approach to quantify PARP-1 in vivo. Clinically, we observed a spectrum of standard uptake values (SUVs) ranging from 2-12 for PARP-1 in tumors. In addition, we found a positive correlation between PET SUVs and fluorescent immunohistochemistry for PARP-1 (r2 = 0.60). CONCLUSION This work confirms the translational potential of a PARP-1 PET imaging agent and supports future clinical trials to test PARP-1 expression as a method to stratify patients for PARP inhibitor therapy. TRIAL REGISTRATION Clinicaltrials.gov NCT02637934. FUNDING Research reported in this publication was supported by the Department of Defense OC160269, a Basser Center team science grant, NIH National Cancer Institute R01CA174904, a Department of Energy training grant DE-SC0012476, Abramson Cancer Center Radiation Oncology pilot grants, the Marsha Rivkin Foundation, Kaleidoscope of Hope Foundation, and Paul Calabresi K12 Career Development Award 5K12CA076931.
Collapse
Affiliation(s)
- Mehran Makvandi
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Austin Pantel
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lauren Schwartz
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin Schubert
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kuiying Xu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Chia-Ju Hsieh
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Catherine Hou
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hyoung Kim
- Department of OBGYN, Division of Gynecology and Oncology
| | - Chi-Chang Weng
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Robert Doot
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael D. Farwell
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Daniel A. Pryma
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - David A. Mankoff
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Fiona Simpkins
- Department of OBGYN, Division of Gynecology and Oncology
| | - Robert H. Mach
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lilie L. Lin
- Department of Radiation Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Zhou SJ, Bi TQ, Qin CX, Yang XQ, Pang K. Expression of NAMPT is associated with breast invasive ductal carcinoma development and prognosis. Oncol Lett 2018; 15:6648-6654. [PMID: 29725408 DOI: 10.3892/ol.2018.8164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 01/19/2018] [Indexed: 01/29/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) possesses various functions in human cells, and altered NAMPT expression is associated with human carcinogenesis. The present study detected the expression of NAMPT in normal and cancerous breast tissues from 83 patients using immunohistochemistry, and analyzed its association with the clinicopathological and survival data of the patients. NAMPT was significantly overexpressed in the breast invasive ductal carcinoma tissues compared with adjacent normal mammary gland tissues. Upregulated NAMPT expression was associated with a larger tumor size, lymph node metastasis, advanced clinical tumor-node-metastasis stages, and estrogen receptor and progesterone receptor expression. Furthermore, NAMPT expression was associated with poor overall and disease-free survival in patients with breast cancer. In conclusion, NAMPT increased protein expression in tumor cells may contribute to the development and progression of breast invasive ductal carcinoma. Thus, detection of NAMPT expression might be useful as a biomarker for the early detection and prognosis prediction of breast cancer.
Collapse
Affiliation(s)
- Shao-Jie Zhou
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Tie-Qiang Bi
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Chun-Xin Qin
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Xiao-Qing Yang
- Department of Thyroid and Breast Surgery, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Kai Pang
- Department of Statistics, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| |
Collapse
|
11
|
Hwang ES, Song SB. Nicotinamide is an inhibitor of SIRT1 in vitro, but can be a stimulator in cells. Cell Mol Life Sci 2017; 74:3347-3362. [PMID: 28417163 PMCID: PMC11107671 DOI: 10.1007/s00018-017-2527-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 03/24/2017] [Accepted: 04/12/2017] [Indexed: 01/15/2023]
Abstract
Nicotinamide (NAM), a form of vitamin B3, plays essential roles in cell physiology through facilitating NAD+ redox homeostasis and providing NAD+ as a substrate to a class of enzymes that catalyze non-redox reactions. These non-redox enzymes include the sirtuin family proteins which deacetylate target proteins while cleaving NAD+ to yield NAM. Since the finding that NAM exerts feedback inhibition to the sirtuin reactions, NAM has been widely used as an inhibitor in the studies where SIRT1, a key member of sirtuins, may have a role in certain cell physiology. However, once administered to cells, NAM is rapidly converted to NAD+ and, therefore, the cellular concentration of NAM decreases rapidly while that of NAD+ increases. The result would be an inhibition of SIRT1 for a limited duration, followed by an increase in the activity. This possibility raises a concern on the validity of the interpretation of the results in the studies that use NAM as a SIRT1 inhibitor. To understand better the effects of cellular administration of NAM, we reviewed published literature in which treatment with NAM was used to inhibit SIRT1 and found that the expected inhibitory effect of NAM was either unreliable or muted in many cases. In addition, studies demonstrated NAM administration stimulates SIRT1 activity and improves the functions of cells and organs. To determine if NAM administration can generate conditions in cells and tissues that are stimulatory to SIRT1, the changes in the cellular levels of NAM and NAD+ reported in the literature were examined and the factors that are involved in the availability of NAD+ to SIRT1 were evaluated. We conclude that NAM treatment can hypothetically be stimulatory to SIRT1.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemungu, 163 Seoulsiripdaero, Seoul, 02504, Republic of Korea.
| | - Seon Beom Song
- Department of Life Science, University of Seoul, Dongdaemungu, 163 Seoulsiripdaero, Seoul, 02504, Republic of Korea
| |
Collapse
|
12
|
Chen Y, Li Z, Dong Z, Beebe J, Yang K, Fu L, Zhang JT. 14-3-3σ Contributes to Radioresistance By Regulating DNA Repair and Cell Cycle via PARP1 and CHK2. Mol Cancer Res 2017; 15:418-428. [PMID: 28087741 DOI: 10.1158/1541-7786.mcr-16-0366] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/23/2016] [Accepted: 12/11/2016] [Indexed: 01/05/2023]
Abstract
14-3-3σ has been implicated in the development of chemo and radiation resistance and in poor prognosis of multiple human cancers. While it has been postulated that 14-3-3σ contributes to these resistances via inhibiting apoptosis and arresting cells in G2-M phase of the cell cycle, the molecular basis of this regulation is currently unknown. In this study, we tested the hypothesis that 14-3-3σ causes resistance to DNA-damaging treatments by enhancing DNA repair in cells arrested in G2-M phase following DNA-damaging treatments. We showed that 14-3-3σ contributed to ionizing radiation (IR) resistance by arresting cancer cells in G2-M phase following IR and by increasing non-homologous end joining (NHEJ) repair of the IR-induced DNA double strand breaks (DSB). The increased NHEJ repair activity was due to 14-3-3σ-mediated upregulation of PARP1 expression that promoted the recruitment of DNA-PKcs to the DNA damage sites for repair of DSBs. On the other hand, the increased G2-M arrest following IR was due to 14-3-3σ-induced Chk2 expression.Implications: These findings reveal an important molecular basis of 14-3-3σ function in cancer cell resistance to chemo/radiation therapy and in poor prognosis of human cancers. Mol Cancer Res; 15(4); 418-28. ©2017 AACR.
Collapse
Affiliation(s)
- Yifan Chen
- Departments of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China.,Guangdong Esophageal Cancer Institute, Guangzhou, China
| | - Zhaomin Li
- Departments of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Zizheng Dong
- Departments of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jenny Beebe
- Departments of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ke Yang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Liwu Fu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China. .,Guangdong Esophageal Cancer Institute, Guangzhou, China
| | - Jian-Ting Zhang
- Departments of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
13
|
FASN regulates cellular response to genotoxic treatments by increasing PARP-1 expression and DNA repair activity via NF-κB and SP1. Proc Natl Acad Sci U S A 2016; 113:E6965-E6973. [PMID: 27791122 DOI: 10.1073/pnas.1609934113] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fatty acid synthase (FASN), the sole cytosolic mammalian enzyme for de novo lipid synthesis, is crucial for cancer cell survival and associates with poor prognosis. FASN overexpression has been found to cause resistance to genotoxic insults. Here we tested the hypothesis that FASN regulates DNA repair to facilitate survival against genotoxic insults and found that FASN suppresses NF-κB but increases specificity protein 1 (SP1) expression. NF-κB and SP1 bind to a composite element in the poly(ADP-ribose) polymerase 1 (PARP-1) promoter in a mutually exclusive manner and regulate PARP-1 expression. Up-regulation of PARP-1 by FASN in turn increases Ku protein recruitment and DNA repair. Furthermore, lipid deprivation suppresses SP1 expression, which is able to be rescued by palmitate supplementation. However, lipid deprivation or palmitate supplementation has no effect on NF-κB expression. Thus, FASN may regulate NF-κB and SP1 expression using different mechanisms. Altogether, we conclude that FASN regulates cellular response against genotoxic insults by up-regulating PARP-1 and DNA repair via NF-κB and SP1.
Collapse
|
14
|
Shen Y, Lee YH, Panneerselvam J, Zhang J, Loo LWM, Fei P. Mutated Fanconi anemia pathway in non-Fanconi anemia cancers. Oncotarget 2016; 6:20396-403. [PMID: 26015400 PMCID: PMC4653013 DOI: 10.18632/oncotarget.4056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/22/2015] [Indexed: 01/01/2023] Open
Abstract
An extremely high cancer incidence and the hypersensitivity to DNA crosslinking agents associated with Fanconi Anemia (FA) have marked it to be a unique genetic model system to study human cancer etiology and treatment, which has emerged an intense area of investigation in cancer research. However, there is limited information about the relationship between the mutated FA pathway and the cancer development or/and treatment in patients without FA. Here we analyzed the mutation rates of the seventeen FA genes in 68 DNA sequence datasets. We found that the FA pathway is frequently mutated across a variety of human cancers, with a rate mostly in the range of 15 to 35 % in human lung, brain, bladder, ovarian, breast cancers, or others. Furthermore, we found a statistically significant correlation (p < 0.05) between the mutated FA pathway and the development of human bladder cancer that we only further analyzed. Together, our study demonstrates a previously unknown fact that the mutated FA pathway frequently occurs during the development of non-FA human cancers, holding profound implications directly in advancing our understanding of human tumorigenesis as well as tumor sensitivity/resistance to crosslinking drug-relevant chemotherapy.
Collapse
Affiliation(s)
- Yihang Shen
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Yuan-Hao Lee
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Jayabal Panneerselvam
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lenora W M Loo
- Program of Epidemiology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Peiwen Fei
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| |
Collapse
|
15
|
Chen H, Wang S, Zhang H, Nice EC, Huang C. Nicotinamide phosphoribosyltransferase (Nampt) in carcinogenesis: new clinical opportunities. Expert Rev Anticancer Ther 2016; 16:827-38. [PMID: 27186719 DOI: 10.1080/14737140.2016.1190649] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Nicotinamide phosphoribosyltransferase (Nampt) is the rate-limiting enzyme that catalyzes the first step in the mammalian nicotinamide adenine dinucleotide (NAD) salvage pathway. Aberrant NAD metabolism was associated with oncogenic signal transduction, suggesting the critical roles of Nampt in tumorigenesis and metastasis. Additionally, Nampt can be secreted out of the cell, and this extracellular form of Nampt (eNampt) was shown to induce inflammation and angiogenesis due to its cytokine activity, which may also be involved in carcinogenesis. AREAS COVERED This article reviews recent advances in the studies of Nampt in carcinogenesis, with a special highlight on Nampt inhibitors and future clinical application, including cancer diagnosis, prognosis and therapy. Expert commentary: Nampt not only maintains the balance of cellular metabolism, but also has a profound influence on multiple aspects of carcinogenesis. Therefore, elucidation of these mechanisms opens the door for future clinical applications targeting this protein. Additional studies are needed to address important questions including the relationship between extracellular Nampt and carcinogenesis.
Collapse
Affiliation(s)
- Hang Chen
- a Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education & Department of Neurology , The Affiliated Hospital of Hainan Medical College , Haikou , China
| | - Shiyu Wang
- a Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education & Department of Neurology , The Affiliated Hospital of Hainan Medical College , Haikou , China
| | - Haiyuan Zhang
- a Key Laboratory of Tropical Diseases and Translational Medicine of Ministry of Education & Department of Neurology , The Affiliated Hospital of Hainan Medical College , Haikou , China
| | - Edouard C Nice
- b Department of Biochemistry and Molecular Biology , Monash University , Clayton , Australia
| | - Canhua Huang
- c State Key Laboratory for Biotherapy and Cancer Center, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu , China
| |
Collapse
|
16
|
Li D, Wu QJ, Bi FF, Chen SL, Zhou YM, Zhao Y, Yang Q. Effect of the BRCA1-SIRT1-EGFR axis on cisplatin sensitivity in ovarian cancer. Am J Transl Res 2016; 8:1601-1608. [PMID: 27186285 PMCID: PMC4859644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/29/2016] [Indexed: 06/05/2023]
Abstract
There is accumulating evidence that breast cancer 1 (BRCA1), sirtuin 1 (SIRT1), and epidermal growth factor receptor (EGFR) help to modulate cisplatin cytotoxicity. The role of dynamic crosstalk among BRCA1, SIRT1, and EGFR in cisplatin sensitivity remains largely unknown. We found that BRCA1, SIRT1, and EGFR levels were increased in cisplatin-resistant ovarian cancers compared with those in cisplatin-sensitive ovarian cancers. Hypomethylation in the BRCA1 promoter was associated with BRCA1 activation, significantly elevated SIRT1 levels, decreased nicotinamide adenine dinucleotide (NAD)-mediated SIRT1 activity, and decreased EGFR levels. Treatment with 5 and 10 μg/ml cisplatin induced a gradual increase in BRCA1 and SIRT1 levels and a gradual decrease in NAD levels and NAD-mediated SIRT1 activity, whereas EGFR levels were increased or decreased by treatment with 5 or 10 μg/ml cisplatin, respectively. The overexpression of SIRT1 or the enhancement of SIRT1 activity synergistically enhanced the BRCA1-mediated effects on EGFR transcription. In contrast, the knockdown of SIRT1 or the inhibition of SIRT1 activity inhibited the BRCA1-mediated effects on EGFR transcription. BRCA1 regulates EGFR through a BRCA1-mediated balance between SIRT1 expression and activity. Those results improve our understanding of the basic molecular mechanism underlying BRCA1-related cisplatin resistance in ovarian cancer.
Collapse
Affiliation(s)
- Da Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Fang-Fang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Si-Lei Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Yi-Ming Zhou
- Department of Medicine, Brigham and Women’s Hospital, Harvard Institutes of Medicine, Harvard Medical SchoolBoston, MA 02115, USA
| | - Yue Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical UniversityShenyang 110001, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| |
Collapse
|
17
|
Mazzotta A, Partipilo G, De Summa S, Giotta F, Simone G, Mangia A. Nuclear PARP1 expression and its prognostic significance in breast cancer patients. Tumour Biol 2015; 37:6143-53. [PMID: 26614429 DOI: 10.1007/s13277-015-4465-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/17/2015] [Indexed: 01/08/2023] Open
Abstract
Poly(adenosine diphosphate [ADP]-ribose) polymerase 1 (PARP1) plays important roles in DNA damage response pathways and is often overexpressed in various human tumors. Currently, the use of PARP inhibitors for breast cancer (BC) therapy is the subject of debate, and there is an urgent need to understand much the expression and prognostic role of the PARP1 protein. The aim was to investigate the clinicopathological and prognostic significance of PARP1 in BC patients. The PARP1 and breast cancer susceptibility gene 1 (BRCA1) protein expressions were evaluated in 114 BCs by immunohistochemistry. Disease-free survival (DFS) and overall survival (OS) were determined based on the Kaplan-Meier method. Our results showed that nuclear PARP1 expression was significantly associated with peritumoral vascular invasion (P = 0.046), chemotherapeutic treatment (P = 0.026), oestrogen receptor (ER; P = 0.013), human epidermal growth factor receptor 2 (HER2; P = 0.003) and BRCA1 (P < 0.001) expression. Survival analyses showed a significant association with clinical outcome in the subgroup of ER-negative patients (P = 0.017 for DFS and P = 0.048 for OS) and in the subgroup of patients treated with chemotherapeutic agents (P = 0.042 for DFS and P = 0.046 for OS). A significant correlation was also found for DFS in patients characterized by tumors without peritumoral vascular invasion (P = 0.022). More importantly, multivariate analyses revealed that high nuclear PARP1 expression was associated with decreased DFS (P = 0.012) and OS (P = 0.026). In conclusion, PARP1 expression may be used as an independent prognostic factor in BC patients. In addition, this study demonstrated that high PARP1 expression may represent a marker of poorer prognosis both for patients with worse clinical outcome and in less aggressive clinical conditions.
Collapse
Affiliation(s)
- Annalisa Mazzotta
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy
| | - Giulia Partipilo
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy
| | - Simona De Summa
- Molecular Genetic Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy
| | - Francesco Giotta
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy
| | - Giovanni Simone
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy
| | - Anita Mangia
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco 65, 70124, Bari, Italy.
| |
Collapse
|
18
|
Granato T, Porpora MG, Longo F, Angeloni A, Manganaro L, Anastasi E. HE4 in the differential diagnosis of ovarian masses. Clin Chim Acta 2015; 446:147-55. [PMID: 25892674 DOI: 10.1016/j.cca.2015.03.047] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/25/2015] [Accepted: 03/09/2015] [Indexed: 12/12/2022]
Abstract
Ovarian masses, a common finding among pre- and post-menopausal women, can be benign or malignant. Ovarian cancer is the leading cause of death from gynecologic malignancy among women living in industrialized countries. According to the current guidelines, measurement of CA125 tumor marker remains the gold standard in the management of ovarian cancer. Recently, HE4 has been proposed as emerging biomarker in the differential diagnosis of adnexal masses and in the early diagnosis of ovarian cancer. Discrimination of benign and malignant ovarian tumors is very important for correct patient referral to institutions specialized in care and management of ovarian cancer. Tumor markers CA125 and HE4 are currently incorporated into the "Risk of Ovarian Malignancy Algorithm" (ROMA) with menopausal status for discerning malignant from benign pelvic masses. The availability of a good biomarker such as HE4, closely associated with the differential and early diagnosis of ovarian cancer, could reduce medical costs related to more expensive diagnostic procedures. Finally, it is important to note that HE4 identifies platinum non-responders thus enabling a switch to second line chemotherapy and improved survival.
Collapse
Affiliation(s)
- Teresa Granato
- CNR-IBPM, National Research Council, Viale Regina Elena 324, 00161 Rome, Italy
| | - Maria Grazia Porpora
- Department of Gynaecology, Obstetrics and Urology, "Sapienza" University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy
| | - Flavia Longo
- Department of Molecular Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy
| | - Antonio Angeloni
- Department of Molecular Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy
| | - Lucia Manganaro
- Department of Radiology, "Sapienza", University of Rome, Viale Regina Elena 324, 00161 Roma, Italy
| | - Emanuela Anastasi
- Department of Molecular Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
19
|
Wang XX, Li D. Linking BRCA1 to NAD World. Cell Cycle 2015; 14:941-2. [PMID: 25713994 DOI: 10.1080/15384101.2015.1006542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Xiu-Xia Wang
- a Department of Obstetrics and Gynecology ; Shengjing Hospital of China Medical University ; Shenyang , China
| | | |
Collapse
|
20
|
Fang YY, Bi FF, Zhou YM, Sun WP, Li CY, Liu Q, Zhao Y, Li D. Nicotinamide adenine dinucleotide (NAD) may affect DNA methyltransferase 1 through regulation of BRCA1 in ovarian cancer. Am J Cancer Res 2015; 5:1199-1206. [PMID: 26045998 PMCID: PMC4449447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/01/2015] [Indexed: 06/04/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a crucial molecule of energy production and signal transduction processes that have been linked to ovarian cancer development. Notably, emerging evidence has led to considerable interest in the role of DNA methyltransferase 1 (DNMT1) in the initiation and progression of ovarian cancer. However, dynamic crosstalk between NAD and DNMT1 is poorly understood. Here, we show that DNMT1 levels are upregulated, along with increased NAD levels in non-BRCA1-mutated ovarian cancer cells. In contrast, DNMT1 levels are not affected by increasing NAD levels in BRCA1-mutated ovarian cancer cells. Mechanistically, BRCA1 inactivity-mediated loss of H3K9ac enrichment around the core promoter inhibits DNMT1 transcription. Consistent with this, BRCA1 levels correlate with DNMT1 levels (R = 0.534, R < 0.001) in human ovarian cancer specimens. Therefore, these results highlight a novel regulatory effect of NAD on DNMT1, and further correlate the physiological properties of NAD metabolism with DNMT1-mediated biological processes. All of this may improve our understanding of the basic molecular mechanism underlying NAD- and DNMT1-related ovarian cancer progression.
Collapse
Affiliation(s)
- Yuan-Yuan Fang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Fang-Fang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Yi-Ming Zhou
- Department of Medicine, Brigham and Women’s Hospital, Harvard Institutes of Medicine, Harvard Medical SchoolBoston, MA 02115, USA
| | - Wu-Ping Sun
- Division of Cell Signaling, National Institute for Physiological SciencesOkazaki 444-8787, Japan
| | - Chun-Yan Li
- Department of Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijing 100005, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| | - Yue Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical UniversityShenyang 110001, China
| | - Da Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical UniversityShenyang 110004, China
| |
Collapse
|