1
|
Gopinath P, Oviya RP, Gopisetty G. Oestrogen receptor-independent actions of oestrogen in cancer. Mol Biol Rep 2023; 50:9497-9509. [PMID: 37731028 DOI: 10.1007/s11033-023-08793-8] [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: 06/08/2023] [Accepted: 08/30/2023] [Indexed: 09/22/2023]
Abstract
Oestrogen, the primary female sex hormone, plays a significant role in tumourigenesis. The major pathway for oestrogen is via binding to its receptor [oestrogen receptor (ERα or β)], followed by nuclear translocation and transcriptional regulation of target genes. Almost 70% of breast tumours are ER + , and endocrine therapies with selective ER modulators (tamoxifen) have been successfully applied. As many as 25% of tamoxifen-treated patients experience disease relapse within 5 years upon completion of chemotherapy. In such cases, the ER-independent oestrogen actions provide a plausible explanation for the resistance, as well as expands the existing horizon of available drug targets. ER-independent oestrogen signalling occurs via one of the following pathways: signalling through membrane receptors, oxidative catabolism giving rise to genotoxic metabolites, effects on mitochondria and redox balance, and induction of inflammatory cytokines. The current review focuses on the non-classical oestrogen signalling, its role in cancer, and its clinical significance.
Collapse
Affiliation(s)
- Prarthana Gopinath
- Department of Molecular Oncology, Cancer Institute (WIA), Adyar, Chennai, 600020, India
| | - Revathi Paramasivam Oviya
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, 600119, India
| | - Gopal Gopisetty
- Department of Molecular Oncology, Cancer Institute (WIA), Adyar, Chennai, 600020, India.
| |
Collapse
|
2
|
Reis R, Dhawle R, Du Pasquier D, Tindall AJ, Frontistis Z, Mantzavinos D, de Witte P, Cabooter D. Electrochemical degradation of 17α-ethinylestradiol: Transformation products, degradation pathways and in vivo assessment of estrogenic activity. ENVIRONMENT INTERNATIONAL 2023; 176:107992. [PMID: 37244003 DOI: 10.1016/j.envint.2023.107992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Conventional water treatment methods are not efficient in eliminating endocrine disrupting compounds (EDCs) in wastewater. Electrochemical Advanced Oxidation Processes (eAOPs) offer a promising alternative, as they electro-generate highly reactive species that oxidize EDCs. However, these processes produce a wide spectrum of transformation products (TPs) with unknown chemical and biological properties. Therefore, a comprehensive chemical and biological evaluation of these remediation technologies is necessary before they can be safely applied in real-life situations. In this study, 17α-ethinylestradiol (EE2), a persistent estrogen, was electrochemically degraded using a boron doped diamond anode with sodium sulfate (Na2SO4) and sodium chloride (NaCl) as supporting electrolytes. Ultra-high performance liquid chromatography coupled to quadrupole time-of-flight mass spectrometry was used for the quantification of EE2 and the identification of TPs. Estrogenic activity was assessed using a transgenic medaka fish line. At optimal operating conditions, EE2 removal reached over 99.9% after 120 min and 2 min, using Na2SO4 and NaCl, respectively. The combined EE2 quantification and in vivo estrogenic assessment demonstrated the overall estrogenic activity was consistently reduced with the degradation of EE2, but not completely eradicated. The identification and time monitoring of TPs showed that the radical agents readily oxidized the phenolic A-ring of EE2, leading to the generation of hydroxylated and/or halogenated TPs and ring-opening products. eAOP revealed to be a promising technique for the removal of EE2 from water. However, caution should be exercised with respect to the generation of potentially toxic TPs.
Collapse
Affiliation(s)
- Rafael Reis
- Laboratory of Pharmaceutical Analysis, Department for Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, Leuven, Belgium
| | - Rebecca Dhawle
- Department of Chemical Engineering, University of Patras, 26500 Patras, Greece
| | - David Du Pasquier
- Laboratoire WatchFrog, Bâtiment Genavenir 3, 1 Rue Pierre Fontaine, 91000 Evry, France
| | - Andrew J Tindall
- Laboratoire WatchFrog, Bâtiment Genavenir 3, 1 Rue Pierre Fontaine, 91000 Evry, France
| | - Zacharias Frontistis
- Department of Chemical Engineering, University of Western Macedonia, GR-50132 Kozani, Greece; School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus
| | | | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, Leuven, Belgium
| | - Deirdre Cabooter
- Laboratory of Pharmaceutical Analysis, Department for Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, Leuven, Belgium.
| |
Collapse
|
3
|
Veisi P, Nikouei M, Cheraghi M, Shahgheibi S, Moradi Y. The association between the multiple birth and breast cancer incidence: an update of a systematic review and meta-analysis from 1983 to 2022. Arch Public Health 2023; 81:76. [PMID: 37106433 PMCID: PMC10142199 DOI: 10.1186/s13690-023-01089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND It has been assumed that perinatal factors such as multiple pregnancies may affect subsequent breast cancer risk in the mother. Considering the inconsistencies in the results of case-control and cohort studies published in the world, this meta-analysis was conducted in order to determine the exact association between multiple pregnancies (twins or more) and the breast cancer incidence. METHODS This study was performed as a meta-analysis based on PRISMA guidelines by searching the international databases of PubMed (Medline), Scopus, and Web of Science as well as by screening selected articles based on their subject, abstract and full text. The search time was from January 1983 to November 2022. Then the NOS checklist was used to evaluate the quality of the final selected articles. The indicators considered for the meta-analysis included the odds ratio (OR) and the risk ratio (RR) along with the confidence interval reported in the selected primary studies. The desired analyzes were performed with STATA software version 17 to be reported. RESULTS In this meta-analysis, 19 studies were finally selected for analysis, which fully met the inclusion criteria. Of these, 11 were case-control studies and 8 were cohort ones. Their sample size was 263,956 women (48,696 with breast cancer and 215,260 healthy) and 1,658,378 (63,328 twin or multiple pregnancies and 1,595,050 singleton pregnancies), respectively. After combining the results of cohort and case-control studies, the effect of multiple pregnancies on the breast cancer incidence was equal to 1.01 (95% CI: 0.89-1.14; I2: 44.88%, P: 0.06) and 0.89 (95% CI: 0.83-0.95; I2: 41.73%, P: 0.07), respectively. CONCLUSION The present meta-analysis results showed, in general, multiple pregnancies were one of the preventive factors of breast cancer.
Collapse
Affiliation(s)
- Pedram Veisi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maziar Nikouei
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mojtaba Cheraghi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Sholeh Shahgheibi
- Department of Obstetrics and Gynecology, School of Medicine, Besat Hospital, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Yousef Moradi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
- Department of Epidemiology and Biostatistics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
4
|
Selvaraj MK, Kaur J. Computational method for aromatase-related proteins using machine learning approach. PLoS One 2023; 18:e0283567. [PMID: 36989252 PMCID: PMC10057777 DOI: 10.1371/journal.pone.0283567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/12/2023] [Indexed: 03/30/2023] Open
Abstract
Human aromatase enzyme is a microsomal cytochrome P450 and catalyzes aromatization of androgens into estrogens during steroidogenesis. For breast cancer therapy, third-generation aromatase inhibitors (AIs) have proven to be effective; however patients acquire resistance to current AIs. Thus there is a need to predict aromatase-related proteins to develop efficacious AIs. A machine learning method was established to identify aromatase-related proteins using a five-fold cross validation technique. In this study, different SVM approach-based models were built using the following approaches like amino acid, dipeptide composition, hybrid and evolutionary profiles in the form of position-specific scoring matrix (PSSM); with maximum accuracy of 87.42%, 84.05%, 85.12%, and 92.02% respectively. Based on the primary sequence, the developed method is highly accurate to predict the aromatase-related proteins. Prediction scores graphs were developed using the known dataset to check the performance of the method. Based on the approach described above, a webserver for predicting aromatase-related proteins from primary sequence data was developed and implemented at https://bioinfo.imtech.res.in/servers/muthu/aromatase/home.html. We hope that the developed method will be useful for aromatase protein related research.
Collapse
Affiliation(s)
| | - Jasmeet Kaur
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
5
|
Firoozi Z, Mohammadisoleimani E, Dastsooz H, Daraei A, Dastgheib SA, Raoofat A, Mansoori H, Mansoori Y, Naghizadeh MM. Altered Expression of hsa_circ_0001445 and hsa_circ_0020397 in Breast Cancer Representing Associations with BMI and Reproductive Factors. ARCHIVES OF IRANIAN MEDICINE 2022; 25:817-827. [PMID: 37543909 PMCID: PMC10685847 DOI: 10.34172/aim.2022.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 11/21/2021] [Indexed: 08/08/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs), one of the recent subclasses of non-coding RNAs (ncRNAs), show pivotal functions in regulation of gene expression and have significant roles in malignancies including breast cancer (BC). This study was aimed to assess the hsa_circ_0001445 and hsa_circ_0020397 expression and role in BC, as well as the potential circRNA/miRNA/mRNA crosstalk in these contexts. METHODS The expression of hsa_circ_0001445 and hsa_circ_0020397 in 50 breast tumors and 50 normal tissues adjacent to the tumors was investigated using quantitative real-time polymerase chain reaction (qRT-PCR). Finally, bioinformatics analyses were used to uncover hsa_circ_0001445, hsa_circ_0020397-miRNA-mRNA potential regulatory networks. RESULTS The hsa_circ_0001445 expression was considerably downregulated in malignant tissues compared to their normal counterparts (P=0.020), while the hsa_circ_0020397 showed an upregulated pattern (P<0.001). Additionally, it was observed that the higher expression of hsa_circ_0001445 was associated with hair dye avoidance (P=0.034) and normal body mass index (BMI) (P=0.016) while hsa_circ_0020397 over-expression had an important association with a lack of vitamin D consumption (P=0.039). On the other hand, lower expression of hsa_circ_0001445 was significantly associated with age at menarche ˂14 years (P=0.027). Our study also revealed that the two circRNAs have potential ability to regulate key mRNAs and miRNAs in competing endogenous RNA (ceRNA) networks. CONCLUSION It is suggested that hsa_circ_0001445 and hsa_circ_0020397 with two opposite roles may be involved in BC development through sponging some miRNAs regulating ceRNA networks. However, their molecular interactions should be validated by further functional studies.
Collapse
Affiliation(s)
- Zahra Firoozi
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Hassan Dastsooz
- IIGM-Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo Cancer (IT), Torino, Italy
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina, Turin, Italy
| | - Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Raoofat
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hosein Mansoori
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | | |
Collapse
|
6
|
Aiyappa‐Maudsley R, Storr SJ, Rakha EA, Green AR, Ellis IO, Martin SG. CYP2S1 and CYP2W1 expression is associated with patient survival in breast cancer. J Pathol Clin Res 2022; 8:550-566. [PMID: 35902379 PMCID: PMC9535097 DOI: 10.1002/cjp2.291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/21/2022] [Accepted: 07/01/2022] [Indexed: 12/29/2022]
Abstract
The cytochrome P450 family of enzymes metabolise a wide range of compounds and play important roles in breast cancer pathogenesis due to their involvement in estrogen metabolism and the production of carcinogenic metabolites during this process. The orphan CYPs, CYP2S1, and CYP2W1 are reportedly upregulated in breast cancer. However, their expression and association with clinicopathological and survival parameters have not been previously assessed in a large cohort of breast cancers. Protein expression of CYP2S1 and CYP2W1 was assessed in early-stage invasive breast cancers (n = 1,426) using immunohistochemistry and correlated with various clinicopathological parameters and survival. mRNA expression of CYP2S1 and CYP2W1 was also assessed in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort. Low nuclear and cytoplasmic CYP2S1 was significantly associated with high-grade tumours (p ≤ 0.009), intermediate Nottingham prognostic index (NPI) group (p ≤ 0.025), high mitotic frequency (p ≤ 0.002), human epidermal growth factor receptor 2 (HER2)-negative disease (p ≤ 0.011), and ductal carcinoma (p ≤ 0.022). Cytoplasmic CYP2S1 was additionally associated with patients ≥50 years (p < 0.001), estrogen receptor (ER)-positive tumours (p = 0.011), and high nuclear pleomorphism (p = 0.003). Low cytoplasmic CYP2W1 was significantly associated with patients ≥50 years (p = 0.002), HER2-negative disease (p = 0.003), intermediate NPI (p = 0.013), and mitosis (p = 0.009). Low cytoplasmic CYP2S1 was significantly associated with adverse breast cancer specific survival (p = 0.034), which remained so in multivariate analysis (hazard ratio [HR]: 0.639; 95% confidence interval [CI]: 0.483-0.846; p = 0.002). Low nuclear CYP2W1 was significantly associated with adverse breast cancer specific survival (p = 0.012), with significance also maintained in multivariate analysis (HR: 0.677; 95% CI: 0.510-0.898; p = 0.007). No associations with survival were observed in the METABRIC cohort. CYP2S1 and CYP2W1 are associated with patient survival in breast cancer and may be important prognostic biomarkers.
Collapse
Affiliation(s)
- Radhika Aiyappa‐Maudsley
- Nottingham Breast Cancer Research Centre, School of Medicine, Biodiscovery InstituteUniversity of Nottingham, University ParkNottinghamUK,Present address:
Cancer Research Centre, Department of Molecular and Clinical Cancer MedicineUniversity of Liverpool, William Henry Duncan BuildingLiverpoolUK
| | - Sarah J Storr
- Nottingham Breast Cancer Research Centre, School of Medicine, Biodiscovery InstituteUniversity of Nottingham, University ParkNottinghamUK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, School of Medicine, Biodiscovery InstituteUniversity of Nottingham, University ParkNottinghamUK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, School of Medicine, Biodiscovery InstituteUniversity of Nottingham, University ParkNottinghamUK
| | - Ian O Ellis
- Nottingham Breast Cancer Research Centre, School of Medicine, Biodiscovery InstituteUniversity of Nottingham, University ParkNottinghamUK
| | - Stewart G Martin
- Nottingham Breast Cancer Research Centre, School of Medicine, Biodiscovery InstituteUniversity of Nottingham, University ParkNottinghamUK
| |
Collapse
|
7
|
R S, R C, Mohideen K, Adtani PN, Subramani V, Balasubramaniam M. Expression of Sex Hormones in Oral Squamous Cell Carcinoma: A Systematic Review on Immunohistochemical Studies. Cureus 2022; 14:e25384. [PMID: 35765387 PMCID: PMC9233754 DOI: 10.7759/cureus.25384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 12/16/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most widespread oral malignancy. In the western world, smoking and alcohol remain the most common risk factors, whereas smokeless tobacco and areca nut consumption are the prevalent etiological factors in South Asia. Numerous markers were studied till date to identify the disease. However, the role of sex hormones in OSCC is poorly defined. Hormonal receptors play a role in breast cancer, prostate cancer, and human papillomavirus (HPV) driven oropharyngeal cancers. It is proven that the expression of estrogen receptor-α improved the overall survival of the patient with HPV-positive oropharyngeal cancer. Thus, this review article aims to determine the expression of sex hormones by immunohistochemistry in OSCC. This systematic review was performed with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) Statement Criteria 2020. Relevant articles were collected from the electronic database in PubMed and Cochrane till 2021. Immunohistochemical studies on sex hormones and their role in OSCC published in English literature were included. We excluded case reports, case series, and abstract-only articles. The sample size of the selected studies varied, and these studies measured the parameters such as sex hormones and OSCC. Furthermore, all the studies used different sex hormones and their association with oral cancer through the immunohistochemical process. Thus, this review summarizes that sex hormones influence the biopathology of oral cancer and have a prognostic significance in the tumor subgroups.
Collapse
Affiliation(s)
- Saranya R
- Oral and Maxillofacial Pathology, Sathyabama Dental College and Hospital, Sathyabama Institute of Science and Technology, Chennai, IND
| | - Chandini R
- Oral and Maxillofacial Pathology, Sathyabama Dental College and Hospital, Sathyabama Institute of Science and Technology, Chennai, IND
| | - Khadijah Mohideen
- Oral and Maxillofacial Pathology, Sathyabama Dental College and Hospital, Sathyabama Institute of Science and Technology, Chennai, IND
| | - Pooja N Adtani
- Basic Medical and Dental Sciences, College of Dentistry, Gulf Medical University, Ajman, ARE
| | - Vijayanirmala Subramani
- Oral and Maxillofacial Pathology, Faculty of Dental Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Murali Balasubramaniam
- Oral and Maxillofacial Pathology, Sathyabama Dental College and Hospital, Sathyabama Institute of Science and Technology, Chennai, IND
| |
Collapse
|
8
|
The 3,4-Quinones of Estrone and Estradiol Are the Initiators of Cancer whereas Resveratrol and N-acetylcysteine Are the Preventers. Int J Mol Sci 2021; 22:ijms22158238. [PMID: 34361004 PMCID: PMC8347442 DOI: 10.3390/ijms22158238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 11/25/2022] Open
Abstract
This article reviews evidence suggesting that a common mechanism of initiation leads to the development of many prevalent types of cancer. Endogenous estrogens, in the form of catechol estrogen-3,4-quinones, play a central role in this pathway of cancer initiation. The catechol estrogen-3,4-quinones react with specific purine bases in DNA to form depurinating estrogen-DNA adducts that generate apurinic sites. The apurinic sites can then lead to cancer-causing mutations. The process of cancer initiation has been demonstrated using results from test tube reactions, cultured mammalian cells, and human subjects. Increased amounts of estrogen-DNA adducts are found not only in people with several different types of cancer but also in women at high risk for breast cancer, indicating that the formation of adducts is on the pathway to cancer initiation. Two compounds, resveratrol, and N-acetylcysteine, are particularly good at preventing the formation of estrogen-DNA adducts in humans and are, thus, potential cancer-prevention compounds.
Collapse
|
9
|
Effect of Interaction between 17β-Estradiol, 2-Methoxyestradiol and 16α-Hydroxyestrone with Chromium (VI) on Ovary Cancer Line SKOV-3: Preliminary Study. Molecules 2020; 25:molecules25215214. [PMID: 33182506 PMCID: PMC7665134 DOI: 10.3390/molecules25215214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer is the leading cause of death from gynecologic malignancies. Some estrogens, as well as xenoestrogens, such as chromium (VI) (Cr(VI)), are indicated as important pathogenic agents. The objective of this study was to evaluate the role of estradiol and some its metabolites upon exposure to the metalloestrogen Cr(VI) in an in vitro model. The changes in cell viability of malignant ovarian cancer cells (SKOV-3 resistant to cisplatin) exposed to 17β-estradiol (E2) and its two metabolites, 2-methoxyestradiol (2-MeOE2) and 16α-hydroxyestrone (16α-OHE1), upon exposure to potassium chromate (VI) and its interactions were examined. The single and mixed models of action, during short and long times of incubation with estrogens, were applied. The different effects (synergism and antagonism) of estrogens on cell viability in the presence of Cr(VI) was observed. E2 and 16α-OHE1 caused a synergistic effect after exposure to Cr(VI). 2-MeOE2 showed an antagonistic effect on Cr(VI). The examined estrogens could be ranked according to the most protective effect or least toxicity in the order: 2-MeOE2 > E2 > 16α-OHE1. Early pre-incubation (24 h or 7 days) of cells with estrogens caused mostly an antagonistic effect-protective against the toxic action of Cr(VI). The beneficial action of estrogens on the toxic effect of Cr(VI), in the context of the risk of ovarian cancer, seems to be important and further studies are needed.
Collapse
|
10
|
Ozkaya HM, Sayitoglu M, Comunoglu N, Sun E, Keskin FE, Ozata D, Hocaoglu RH, Khodzaev K, Firtina S, Tanriover N, Gazioglu N, Oz B, Kadioglu P. G-protein Coupled Estrogen Receptor Expression in Growth Hormone Secreting and Non-Functioning Adenomas. Exp Clin Endocrinol Diabetes 2020; 129:634-643. [PMID: 33091936 DOI: 10.1055/a-1274-1330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE To evaluate the expression of G-protein coupled estrogen receptor (GPER1), aromatase, estrogen receptor α (ERα), estrogen receptor β (ERβ), pituitary tumor transforming gene (PTTG), and fibroblast growth factor 2 (FGF2) in GH-secreting and non-functioning adenomas (NFA). METHODS Thirty patients with acromegaly and 27 patients with NFA were included. Gene expression was determined via quantitative reverse transcription polymerase chain reaction (QRT-PCR). Protein expression was determined via immunohistochemistry. RESULTS There was no difference, in terms of gene expression of aromatase, ERα, PTTG, and FGF2 between the two groups (p>0.05 for all). ERβ gene expression was higher and GPER1 gene expression was lower in GH-secreting adenomas than NFAs (p<0.05 for all). Aromatase and ERβ protein expression was higher in GH-secreting adenomas than NFAs (p=0.01). None of the tumors expressed ERα. GPER1 expression was detected in 62.2% of the GH-secreting adenomas and 45% of NFAs. There was no difference in terms of GPER1, PTTG, FGF2 H scores between the two groups (p>0.05 for all). GPER1 gene expression was positively correlated to ERα, ERβ, PTTG, and FGF2 gene expression (p<0.05 for all). There was a positive correlation between aromatase and GPER1 protein expression (r=0.31; p=0.04). CONCLUSIONS GPER1 is expressed at both gene and protein level in a substantial portion of GH-secreting adenomas and NFAs. The finding of a positive correlation between GPER1 and ERα, ERβ, PTTG, and FGF2 gene expression and aromatase and GPER1 protein expression suggests GPER1 along with aromatase and classical ERs might mediate the effects of estrogen through upregulation of PTTG and FGF2.
Collapse
Affiliation(s)
- Hande Mefkure Ozkaya
- Department of Endocrinology and Metabolism, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul
| | - Muge Sayitoglu
- Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul
| | - Nil Comunoglu
- Department of Pathology, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul
| | - Eda Sun
- Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul
| | - Fatma Ela Keskin
- Department of Endocrinology and Metabolism, Demiroglu Bilim University, Istanbul
| | - Duygu Ozata
- Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul
| | - Rabia Hacer Hocaoglu
- Department of Internal Medicine, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul
| | - Khusan Khodzaev
- Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul
| | - Sinem Firtina
- Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul
| | - Necmettin Tanriover
- Department of Neurosurgery, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul.,Pituitary Center, Istanbul University-Cerrahpasa, Istanbul
| | - Nurperi Gazioglu
- Department of Neurosurgery, Demiroglu Bilim University, Istanbul
| | - Buge Oz
- Department of Pathology, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul
| | - Pinar Kadioglu
- Department of Endocrinology and Metabolism, Cerrahpasa Medical School, Istanbul University-Cerrahpasa, Istanbul.,Pituitary Center, Istanbul University-Cerrahpasa, Istanbul
| |
Collapse
|
11
|
Al-Otaibi JS, Almuqrin AH, Mary YS, Mary YS. Comprehensive quantum mechanical studies on three bioactive anastrozole based triazole analogues and their SERS active graphene complex. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128388] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
12
|
|
13
|
Majhi PD, Sharma A, Roberts AL, Daniele E, Majewski AR, Chuong LM, Black AL, Vandenberg LN, Schneider SS, Dunphy KA, Jerry DJ. Effects of Benzophenone-3 and Propylparaben on Estrogen Receptor-Dependent R-Loops and DNA Damage in Breast Epithelial Cells and Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:17002. [PMID: 31939680 PMCID: PMC7015622 DOI: 10.1289/ehp5221] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
BACKGROUND Endocrine-disrupting chemicals have been shown to have broad effects on development, but their mutagenic actions that can lead to cancer have been less clearly demonstrated. Physiological levels of estrogen have been shown to stimulate DNA damage in breast epithelial cells through mechanisms mediated by estrogen-receptor alpha (ERα). Benzophenone-3 (BP-3) and propylparaben (PP) are xenoestrogens found in the urine of >96% of U.S. OBJECTIVES We investigated the effect of BP-3 and PP on estrogen receptor-dependent transactivation and DNA damage at concentrations relevant to exposures in humans. METHODS In human breast epithelial cells, DNA damage following treatment with 17β-estradiol (E2), BP-3, and PP was determined by immunostaining with antibodies against γ-H2AX and 53BP1. Estrogenic responses were determined using luciferase reporter assays and gene expression. Formation of R-loops was determined with DNA: RNA hybrid-specific S9.6 antibody. Short-term exposure to the chemicals was also studied in ovariectomized mice. Immunostaining of mouse mammary epithelium was performed to quantify R-loops and DNA damage in vivo. RESULTS Concentrations of 1μM and 5μM BP-3 or PP increased DNA damage similar to that of E2 treatment in a ERα-dependent manner. However, BP-3 and PP had limited transactivation of target genes at 1μM and 5μM concentrations. BP-3 and PP exposure caused R-loop formation in a normal human breast epithelial cell line when ERα was introduced. R-loops and DNA damage were also detected in mammary epithelial cells of mice treated with BP-3 and PP. CONCLUSIONS Acute exposure to xenoestrogens (PP and BP-3) in mice induce DNA damage mediated by formation of ERα-dependent R-loops at concentrations 10-fold lower than those required for transactivation. Exposure to these xenoestrogens may cause deleterious estrogenic responses, such as DNA damage, in susceptible individuals. https://doi.org/10.1289/EHP5221.
Collapse
Affiliation(s)
- Prabin Dhangada Majhi
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Botany, Ravenshaw University, Cuttack, Odisha, India
| | - Aman Sharma
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Amy L. Roberts
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Elizabeth Daniele
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Aliza R. Majewski
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Lynn M. Chuong
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Amye L. Black
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Laura N. Vandenberg
- Department of Environmental Health Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Sallie S. Schneider
- University of Massachusetts Medical School, Baystate Campus, Springfield, Massachusetts, USA
- Pioneer Valley Life Sciences Institute, Springfield, Massachusetts, USA
| | - Karen A. Dunphy
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - D. Joseph Jerry
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
- Pioneer Valley Life Sciences Institute, Springfield, Massachusetts, USA
| |
Collapse
|
14
|
An In Vitro Model of Triple-Negative Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:35-46. [PMID: 31576538 DOI: 10.1007/978-3-030-22254-3_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
We have characterized two highly tumorigenic and metastatic basal B TNBC cell lines, XtMCF and LmMCF, with the additional values of having the normal and early-stage counterparts of them. This model allows the study of the evolution of TNBC, and investigates molecular pathways at different stages of transformation and progression in a relatively constant genetic background. This constitutes an ideal model for developing targeted therapy in two important fields in cancer biology which are the epithelial mesenchymal transition (EMT) and cancer stem cells (CSC).
Collapse
|
15
|
Lanxiang W, Bin W, Ge X, Yutang H, Chunjie W, Honghao Z. Long-term exposure of 4-hydroxyestradiol induces the cancer cell characteristics via upregulating CYP1B1 in MCF-10A cells. Toxicol Mech Methods 2019; 29:686-692. [PMID: 31364906 DOI: 10.1080/15376516.2019.1650146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Life-long estrogen exposure is one of the major risk factors in the development and progression of breast cancer. However, little is known about the molecular mechanisms, by which chronic exposure to estrogen contributes to breast carcinogenesis. The aim of the present study was to investigate the effects of long-term exposure with 4-hydroxyestradiol (4-OHE2) on acquired cancer characteristics of human mammary epithelial MCF-10A cells. The possible regulators were further studied in chronic 4-OHE2-treated MCF-10A cells. We observed that MCF-10A cells long-term exposed to 4-OHE2 acquire the characteristics of cancer cells, such as enhanced cell growth, EMT properties, and increased migration and invasiveness. Moreover, the expression of CYP1B1 was significantly elevated in long-term 4-OHE2-treated MCF-10A cells. Block of CYP1B1 significantly reduced the cancer cell characteristics in long-term 4-OHE2-treated MCF-10A cells. Our results indicated that 4-OHE2 mediated enhanced cancer cell characteristics in mammary epithelial cells are an important key event for breast carcinogenesis process. CYP1B1 partially contributes to the 4-OHE2 induced cancer cell characteristics in MCF-10A cells. Targeting CYP1B1 might offer a new strategy for the treatment of estrogen-induced breast cancer.
Collapse
Affiliation(s)
- Wu Lanxiang
- Institute of Life Sciences, Chongqing Medical University , Chongqing , China
| | - Wang Bin
- Institute of Life Sciences, Chongqing Medical University , Chongqing , China
| | - Xu Ge
- Institute of Life Sciences, Chongqing Medical University , Chongqing , China
| | - Huang Yutang
- Institute of Life Sciences, Chongqing Medical University , Chongqing , China
| | - Wen Chunjie
- Institute of Life Sciences, Chongqing Medical University , Chongqing , China
| | - Zhou Honghao
- Institute of Life Sciences, Chongqing Medical University , Chongqing , China.,Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Central South University , Changsha , China
| |
Collapse
|
16
|
Daraei A, Izadi P, Khorasani G, Nafissi N, Naghizadeh MM, Younosi N, Meysamie A, Mansoori Y, Nariman‐Saleh‐Fam Z, Bastami M, Saadatian Z, Zendehbad Z, Tavakkoly‐Bazzaz J. Methylation of progesterone receptor isoform A promoter in normal breast tissue: An epigenetic link between early age at menarche and risk of breast cancer? J Cell Biochem 2019; 120:12393-12401. [DOI: 10.1002/jcb.28505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Abdolreza Daraei
- Department of Genetics, Faculty of Medicine Babol University of Medical Sciences Babol Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine Tehran University of Medical Sciences Tehran Iran
| | - Ghasemali Khorasani
- Division of Plastic and Reconstructive Surgery, Imam Khomeini Hospital Complex Tehran University of Medical Sciences Tehran Iran
| | - Nahid Nafissi
- Department of Surgery, School of Medicine Iran University of Medical Sciences Tehran Iran
| | | | - Nasim Younosi
- Department of Surgery, School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Alipasha Meysamie
- Department of Community and Preventive Medicine Department, Medical Faculty Tehran University of Medical Sciences Tehran Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center Fasa University of Medical Sciences Fasa Iran
| | - Ziba Nariman‐Saleh‐Fam
- Women's Reproductive Health Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Milad Bastami
- Immunology Research Center, Stem Cell and Regenerative Medicine Institute Tabriz University of Medical Sciences Tabriz Iran
| | - Zahra Saadatian
- Department of Medical Genetics, Faculty of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Zahra Zendehbad
- Department of Medical Genetics, School of Medicine Tehran University of Medical Sciences Tehran Iran
| | - Javad Tavakkoly‐Bazzaz
- Department of Medical Genetics, School of Medicine Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
17
|
Resende AD, Leal S, Batista‐Pinto C, Garcez F, Sá SI. Hepatic effects of long‐term tamoxifen administration to cycling female rats. J Biochem Mol Toxicol 2019; 33:e22293. [DOI: 10.1002/jbt.22293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Albina Dolores Resende
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS)Gandra Portugal
- Interdisciplinary Center of Marine and Environmental Research (CIIMAR)Matosinhos Portugal
| | - Sandra Leal
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS)Gandra Portugal
- Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of PortoPorto Portugal
| | - Carla Batista‐Pinto
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS)Gandra Portugal
- Interdisciplinary Center of Marine and Environmental Research (CIIMAR)Matosinhos Portugal
| | - Fernanda Garcez
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS)Gandra Portugal
| | - Susana Isabel Sá
- Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of PortoPorto Portugal
- Unit of Anatomy, Department of BiomedicineFaculty of Medicine of the University of PortoPorto Portugal
| |
Collapse
|
18
|
Cui X, Liew Z, Hansen J, Lee PC, Arah OA, Ritz B. Cancers Preceding Parkinson's Disease after Adjustment for Bias in a Danish Population-Based Case-Control Study. Neuroepidemiology 2019; 52:136-143. [PMID: 30661072 DOI: 10.1159/000494292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 10/04/2018] [Indexed: 01/20/2023] Open
Abstract
Cancer patients are at lower risk of developing Parkinson's disease (PD) compared with the general population. One explanation is the negative association between smoking and PD, but PD risk is also lower for cancers not related to smoking. Another explanation is survival bias where death from cancer may act as a competing risk. We conducted a large population-based case-control study in Denmark and investigated whether cancer diagnosis reduced the risk of developing PD even after adjusting for important risk factors including smoking, physical activity, and lifetime oestrogen status. Using probabilistic bias analysis we quantified the influence of survival bias. We estimated negative point estimates (ORs) between cancers and PD for all cancers except skin, female breast, and ill-defined and unspecified 0.85 (95% CI 0.59-1.21); smoking-related cancers 0.75 (95% CI 0.45-1.23); and cancers not related to smoking 0.82 (95% CI 0.49-1.38) that are very similar to those previously reported for a much larger Danish register only based study, even though our confidence intervals include the null. These effect estimates shifted towards the null after accounting for survival bias but most bias-adjusted ORs remained below 1 within the range of priors considered in simulations. Overall, cancer patients have a lower risk of developing PD even after controlling for cancer-related lifestyles factors and correcting for survival bias.
Collapse
Affiliation(s)
- Xin Cui
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Zeyan Liew
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Johnni Hansen
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Pei-Chen Lee
- Department of Health Care Management, College of Healthcare Administration and Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Onyebuchi A Arah
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, California, USA.,Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Center for Health Policy Research, UCLA, Los Angeles, California, USA
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, California, USA, .,Department of Neurology, Geffen School of Medicine, UCLA, Los Angeles, California, USA,
| |
Collapse
|
19
|
Atwood CS, Ekstein SF. Human versus non-human sex steroid use in hormone replacement therapies part 1: Preclinical data. Mol Cell Endocrinol 2019; 480:12-35. [PMID: 30308266 DOI: 10.1016/j.mce.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 03/12/2018] [Accepted: 10/04/2018] [Indexed: 11/24/2022]
Abstract
Prior to 2002, hormone replacement therapy (HRT) was considered to be an important component of postmenopausal healthcare. This was based on a plethora of basic, epidemiological and clinical studies demonstrating the health benefits of supplementation with human sex steroids. However, adverse findings from the Women's Health Initiative (WHI) studies that examined the 2 major forms of HRT in use in the US at that time - Premarin (conjugated equine estrogens; CEE) and Prempro (CEE + medroxyprogesterone acetate; MPA), cast a shadow over the use of any form of HRT. Here we review the biochemical and physiological differences between the non-human WHI study hormones - CEE and MPA, and their respective human counterparts 17β-estradiol (E2) and progesterone (P4). Preclinical data from the last 30 years demonstrate clear differences between human and non-human sex steroids on numerous molecular, physiological and functional parameters in brain, heart and reproductive tissue. In contrast to CEE supplementation, which is not always detrimental although certainly not as optimal as E2 supplementation, MPA is clearly not equivalent to P4, having detrimental effects on cognitive, cardiac and reproductive function. Moreover, unlike P4, MPA is clearly antagonistic of the positive effects of E2 and CEE on tissue function. These data indicate that minor chemical changes to human sex steroids result in physiologically distinct actions that are not optimal for tissue health and functioning.
Collapse
Affiliation(s)
- Craig S Atwood
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, USA; Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, WI, 53705, USA; School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, 6027, WA, Australia.
| | - Samuel F Ekstein
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, USA
| |
Collapse
|
20
|
Renaud L, Agarwal N, Richards DJ, Falcinelli S, Hazard ES, Carnevali O, Hyde J, Hardiman G. Transcriptomic analysis of short-term 17α-ethynylestradiol exposure in two Californian sentinel fish species sardine (Sardinops sagax) and mackerel (Scomber japonicus). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 244:926-937. [PMID: 30469287 DOI: 10.1016/j.envpol.2018.10.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/24/2018] [Accepted: 10/11/2018] [Indexed: 06/09/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are substances which disrupt normal functioning of the endocrine system by interfering with hormone regulated physiological pathways. Aquatic environments provide the ultimate reservoir for many EDCs as they enter rivers and the ocean via effluent discharges and accumulate in sediments. One EDC widely dispersed in municipal wastewater effluent discharges is 17α-ethynylestradiol (EE2), which is one of the most widely prescribed medicines. EE2 is a bio-active estrogen employed in the majority of oral contraceptive pill formulations. As evidence of the health risks posed by EDCs mount, there is an urgent need to improve diagnostic tools for monitoring the effects of pollutants. As the cost of high throughput sequencing (HTS) diminishes, transcriptional profiling of an organism in response to EDC perturbation presents a cost-effective way of screening a wide range of endocrine responses. Coastal pelagic filter feeding fish species analyzed using HTS provide an excellent tool for EDC risk assessment in the marine environment. Unfortunately, there are limited genome sequence data and annotation for many of these species including Pacific sardine (Sardinops sagax) and chub mackerel (Scomber japonicus), which limits the utility of molecular tools such as HTS to interrogate the effects of endocrine disruption. In this study, we carried out RNA sequencing (RNAseq) of liver RNA harvested from wild sardine and mackerel exposed for 5 h under laboratory conditions to a concentration of 12.5 pM EE2 in the tank water. We developed an analytical framework for transcriptomic analyses of species with limited genomic information. EE2 exposure altered expression patterns of key genes involved in important metabolic and physiological processes. The systems approach presented here provides a powerful tool for obtaining a comprehensive picture of endocrine disruption in aquatic organisms.
Collapse
Affiliation(s)
- Ludivine Renaud
- Department of Medicine, Nephrology, Medical University of South Carolina, Charleston, SC, USA
| | - Nisha Agarwal
- Biomedical Informatics Research Center, San Diego State University, San Diego, CA, USA
| | | | - Silvia Falcinelli
- Dipartimento di Scienze della Vita e Dell'Ambiente, Università Politecnica della Marche, 60131, Ancona, Italy
| | - E Starr Hazard
- MUSC Bioinformatics, Center for Genomics Medicine, Medical University of South Carolina, Charleston, SC, USA; Academic Affairs Faculty & Computational Biology Resource Center, Medical University of South Carolina, Charleston, SC, USA
| | - Oliana Carnevali
- Dipartimento di Scienze della Vita e Dell'Ambiente, Università Politecnica della Marche, 60131, Ancona, Italy
| | - John Hyde
- NOAA Fisheries, Southwest Fisheries Science Center, La Jolla, CA, USA
| | - Gary Hardiman
- Department of Medicine, Nephrology, Medical University of South Carolina, Charleston, SC, USA; Biomedical Informatics Research Center, San Diego State University, San Diego, CA, USA; MUSC Bioinformatics, Center for Genomics Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA; Laboratory for Marine Systems Biology, Hollings Marine Laboratory, Charleston, SC, USA; School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Stranmillis Road, Belfast BT9 5AG, UK.
| |
Collapse
|
21
|
Park SA. Catechol Estrogen 4-Hydroxyestradiol is an Ultimate Carcinogen in Breast Cancer. ACTA ACUST UNITED AC 2018. [DOI: 10.15616/bsl.2018.24.3.143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Sin-Aye Park
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| |
Collapse
|
22
|
Liu X, Huang X, Zhang S, Niu F, Ouyang Y, Shou Z, Liu J. Correlations between CYP3A4 polymorphism and susceptibility to breast cancer in Chinese Han population. Int J Clin Oncol 2018; 24:179-188. [PMID: 30218411 DOI: 10.1007/s10147-018-1346-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND CYP3A4 is a major enzyme catalyzing the metabolism of endogenous steroids that play an important role in the etiology of carcinogenesis. This study was designed to investigate the contribution of CYP3A4 polymorphism to breast cancer in Chinese Han female population. METHODS To examine whether variants of CYP3A4 contribute to breast cancer, 5 single-nucleotide polymorphisms (SNPs) of CYP3A4 were genotyped by Sequenom MassARRAY in 267 breast cancer patients and 302 healthy controls. Odds ratio (OR) and 95% confidence intervals (CIs) were calculated by unconditional logistic regression adjusted for age. RESULTS We found that the TT genotype of CYP3A4*1G (rs2242480) polymorphism was associated with increased risk of breast cancer using the fixed effects model (recessive model: OR = 2.34, p = 0.018). Stratified according to age, CYP3A4*1G increased the risk of breast cancer especially in less than 50-year-old group (codominant model OR = 3.68, p = 0.041; recessive model: OR = 3.55, p = 0.012). Furthermore, TT genotype of rs2242480 was associated with Cerb-B2 positive (recessive model: OR = 2.47, p = 0.025) and stage I/II (recessive model: OR = 2.32, p = 0.041). However, no statistically significant associations in other polymorphisms and haploview analysis were observed. CONCLUSIONS This study provides an evidence for polymorphism of CYP3A4 gene associated with the development of breast cancer, also a new insight into etiology of breast cancer. However, the underlying mechanism of the CYP3A4 gene in breast cancer is necessary for further study.
Collapse
Affiliation(s)
- Xu Liu
- Department of Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, #1120, Lianhua Road, Futian District, Shenzhen, 518036, Guangdong, China
| | - Xi Huang
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China
| | - Shanshan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, 710069, Shaanxi, China
| | - Fanglin Niu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, 710069, Shaanxi, China
| | - Yongri Ouyang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, 710069, Shaanxi, China
| | - Zhexing Shou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1277, Jiefang Avenue, Jianghan District, Wuhan, 430022, Hubei, China.
| | - Jikui Liu
- Department of Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, #1120, Lianhua Road, Futian District, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
23
|
Katuwal S, Martinsen JI, Kjaerheim K, Sparen P, Tryggvadottir L, Lynge E, Weiderpass E, Pukkala E. Occupational variation in the risk of female breast cancer in the Nordic countries. Cancer Causes Control 2018; 29:1027-1038. [DOI: 10.1007/s10552-018-1076-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/23/2018] [Indexed: 12/31/2022]
|
24
|
Farhood B, Goradel NH, Mortezaee K, Khanlarkhani N, Salehi E, Nashtaei MS, Mirtavoos-Mahyari H, Motevaseli E, Shabeeb D, Musa AE, Najafi M. Melatonin as an adjuvant in radiotherapy for radioprotection and radiosensitization. Clin Transl Oncol 2018; 21:268-279. [PMID: 30136132 DOI: 10.1007/s12094-018-1934-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
Abstract
It is estimated that more than half of cancer patients undergo radiotherapy during the course of their treatment. Despite its beneficial therapeutic effects on tumor cells, exposure to high doses of ionizing radiation (IR) is associated with several side effects. Although improvements in radiotherapy techniques and instruments could reduce these side effects, there are still important concerns for cancer patients. For several years, scientists have been trying to modulate tumor and normal tissue responses to IR, leading to an increase in therapeutic ratio. So far, several types of radioprotectors and radiosensitizers have been investigated in experimental studies. However, high toxicity of chemical sensitizers or possible tumor protection by radioprotectors creates a doubt for their clinical applications. On the other hand, the protective effects of these radioprotectors or sensitizer effects of radiosensitizers may limit some type of cancers. Hence, the development of some radioprotectors without any protective effect on tumor cells or low toxic radiosensitizers can help improve therapeutic ratio with less side effects. Melatonin as a natural body hormone is a potent antioxidant and anti-inflammatory agent that shows some anti-cancer properties. It is able to neutralize different types of free radicals produced by IR or pro-oxidant enzymes which are activated following exposure to IR and plays a key role in the protection of normal tissues. In addition, melatonin has shown the ability to inhibit long-term changes in inflammatory responses at different levels, thereby ameliorating late side effects of radiotherapy. Fortunately, in contrast to classic antioxidants, some in vitro studies have revealed that melatonin has a potent anti-tumor activity when used alongside irradiation. However, the mechanisms of its radiosensitive effect remain to be elucidated. Studies suggested that the activation of pro-apoptosis gene, such as p53, changes in the metabolism of tumor cells, suppression of DNA repair responses as well as changes in biosynthesis of estrogen in breast cancer cells are involved in this process. In this review, we describe the molecular mechanisms for radioprotection and radiosensitizer effects of melatonin. Furthermore, some other proposed mechanisms that may be involved are presented.
Collapse
Affiliation(s)
- B Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - N H Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - K Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - N Khanlarkhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - E Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - M S Nashtaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Infertility Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - H Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - E Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - D Shabeeb
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran.,Department of Physiology, College of Medicine, University of Misan, Amarah, Iraq
| | - A E Musa
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran.,Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - M Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
25
|
Shoombuatong W, Schaduangrat N, Nantasenamat C. Towards understanding aromatase inhibitory activity via QSAR modeling. EXCLI JOURNAL 2018; 17:688-708. [PMID: 30190660 PMCID: PMC6123608 DOI: 10.17179/excli2018-1417] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/10/2018] [Indexed: 12/14/2022]
Abstract
Aromatase is a rate-limiting enzyme for estrogen biosynthesis that is overproduced in breast cancer tissue. To block the growth of breast tumors, aromatase inhibitors (AIs) are employed to bind and inhibit aromatase in order to lower the amount of estrogen produced in the body. Although a number of synthetic aromatase inhibitors have been released for clinical use in the treatment of hormone-receptor positive breast cancer, these inhibitors may lead to undesirable side effects (e.g. increased rash, diarrhea and vomiting; effects on the bone, brain and heart) and therefore, the search for novel AIs continues. Over the past decades, there has been an intense effort in employing medicinal chemistry and quantitative structure-activity relationship (QSAR) to shed light on the mechanistic basis of aromatase inhibition. To the best of our knowledge, this article constitutes the first comprehensive review of all QSAR studies of both steroidal and non-steroidal AIs that have been published in the field. Herein, we summarize the experimental setup of these studies as well as summarizing the key features that are pertinent for robust aromatase inhibition.
Collapse
Affiliation(s)
- Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
26
|
van de Ven M, Liu X, van der Burg E, Klarenbeek S, Alexi X, Zwart W, Dijcks F, Bouwman P, Jonkers J. BRCA1-associated mammary tumorigenesis is dependent on estrogen rather than progesterone signaling. J Pathol 2018; 246:41-53. [PMID: 29877575 DOI: 10.1002/path.5105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/01/2018] [Accepted: 05/25/2018] [Indexed: 12/16/2022]
Abstract
Hereditary breast cancers in BRCA1 mutation carriers are mostly estrogen receptor α (ERα)-negative and progesterone receptor (PR)-negative; however, hormone depletion via bilateral oophorectomy does result in a marked reduction in breast cancer risk, suggesting that BRCA1-associated breast tumorigenesis is dependent on hormone signaling. We used geneticaly engineered mouse models to determine the individual influences of ERα and PR signaling on the development of BRCA1-deficient breast cancer. In line with the human data, BRCA1-deficient mouse mammary tumors are ERα-negative, and bilateral ovariectomy leads to abrogation of mammary tumor development. Hormonal replacement experiments in ovariectomized mice showed that BRCA1-deficient mammary tumor formation is promoted by estrogen but not by progesterone. In line with these data, mammary tumorigenesis was significantly delayed by the selective ERα downregulator fulvestrant, but not by the selective PR antagonist Org33628. Together, our results illustrate that BRCA1-associated tumorigenesis is dependent on estrogen signaling rather than on progesterone signaling, and call into question the utility of PR antagonists as a tumor prevention strategy for BRCA1 mutation carriers. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Marieke van de Ven
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Xiaoling Liu
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Xanthippi Alexi
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Fred Dijcks
- Synthon Biopharmaceuticals B.V, Nijmegen, The Netherlands
| | - Peter Bouwman
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Mondal B, Chen H, Wen W, Cavalieri EL, Rogan EG, Zahid M. Modulation of Cellular Response to Arsenic Trioxide Toxicity by Resveratrol. ACS OMEGA 2018; 3:5511-5515. [PMID: 29876539 PMCID: PMC5981766 DOI: 10.1021/acsomega.7b01727] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 05/10/2018] [Indexed: 05/07/2023]
Abstract
Arsenic trioxide (As2O3) is an environmental carcinogen and a putative endocrine disruptor. Resveratrol has been shown to reverse As2O3-induced oxidative damage. In immortalized but nontransformed estrogen receptor α-negative human breast cells (MCF10A), we observed that 25 μM resveratrol ameliorated As2O3-induced cytotoxicity. As2O3, in the presence or absence of 25 μM resveratrol, induced quinone reductase (NAD(P)H quinone dehydrogenase 1), via the induction of NFE2-related factor 2. As2O3 caused a repression of cytochrome P450 (CYP)1B1, but the addition of 25 μM resveratrol rescued the expression of cytochrome P450 1B1 and kept it at a constant level. Therefore, 25 μM resveratrol can modulate the effects of As2O3 on enzymes involved in estrogen metabolism.
Collapse
Affiliation(s)
- Bodhisattwa Mondal
- Department
of Environmental, Agricultural, and Occupational Health,
College of Public Health, and the Eppley Institute for Research in Cancer
and Allied Diseases, University of Nebraska
Medical Center, Omaha, Nebraska 68198, United
States
| | - Hongxia Chen
- Department
of Environmental, Agricultural, and Occupational Health,
College of Public Health, and the Eppley Institute for Research in Cancer
and Allied Diseases, University of Nebraska
Medical Center, Omaha, Nebraska 68198, United
States
| | - Weihua Wen
- Department
of Environmental, Agricultural, and Occupational Health,
College of Public Health, and the Eppley Institute for Research in Cancer
and Allied Diseases, University of Nebraska
Medical Center, Omaha, Nebraska 68198, United
States
| | - Ercole L. Cavalieri
- Department
of Environmental, Agricultural, and Occupational Health,
College of Public Health, and the Eppley Institute for Research in Cancer
and Allied Diseases, University of Nebraska
Medical Center, Omaha, Nebraska 68198, United
States
| | - Eleanor G. Rogan
- Department
of Environmental, Agricultural, and Occupational Health,
College of Public Health, and the Eppley Institute for Research in Cancer
and Allied Diseases, University of Nebraska
Medical Center, Omaha, Nebraska 68198, United
States
| | - Muhammad Zahid
- Department
of Environmental, Agricultural, and Occupational Health,
College of Public Health, and the Eppley Institute for Research in Cancer
and Allied Diseases, University of Nebraska
Medical Center, Omaha, Nebraska 68198, United
States
- E-mail: . Phone: 1-402-559-8912. Fax: 1-402-559-7259
| |
Collapse
|
28
|
Mansoori Y, Tabei MB, Askari A, Izadi P, Daraei A, Naghizadeh MM, Zendehbad Z, Bastami M, Nariman-Saleh-Fam Z, Mansoori H, Tavakkoly-Bazzaz J. A link between expression level of long-non-coding RNA ZFAS1 in breast tissue of healthy women and obesity. Int J Biol Markers 2018; 33:500-506. [PMID: 29690801 DOI: 10.1177/1724600818762258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Epidemiological and experimental literature indicates that the risk of breast cancer incidence is strongly linked to hormone-dependent factors, including reproductive history and obesity. However, the molecular mechanisms underlying the association between these factors and breast cancer risk are poorly understood. The aim of this study, therefore, was to determine whether obesity and reproductive history are associated with expression levels of two breast cancer-related long non-coding RNAs (lncRNAs), namely ZFAS1 and SRA1 in cancer-free breast tissues of women. METHODS In the current research, 145 healthy women were recruited, and the quantitative expression levels of the two lncRNAs were determined through qPCR assay after gathering the mammoplasty breast tissue samples. RESULTS It was found that women with body mass index (BMI)≥30 kg/m2 and BMI 25-29 kg/m2 show a low expression of ZFAS1 compared to the BMI<25 kg/m2 (P=0.031 and P=0.027, respectively). Then, the correlation analysis disclosed a negative correlation of ZFAS1 low expression with increasing BMI (r=-0.194, P=0.019). Interestingly, this analysis demonstrated a negative correlation between low expression of the ZFAS1 and high BMI in women with menarche age below 14 (r=-221; P=0.028). Lastly, it was also revealed that there was a negative association of the low expression level of ZFAS1 with increasing BMI in women through regression models (B=-0.048, P=0.019). CONCLUSIONS These findings suggest interesting clues about the links between high BMI and the expression levels of ZFAS1 in non-diseased breasts that may help us better understand the underlying mechanisms through which obesity contributes to breast carcinogenesis. However, such results need more validations in future research.
Collapse
Affiliation(s)
- Yaser Mansoori
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Bagher Tabei
- Department of Medical Genetics, School of Medical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Askari
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.,Bone and Joint Reconstraction Research Center, Shafa Orthopedic Haspital, Iran University of Medical Sciences, Tehran, IR Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Daraei
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Zahra Zendehbad
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Bastami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hosein Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.,Department of Medical Genetics, School of Medical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Bai X, Xie J, Sun S, Zhang X, Jiang Y, Pang D. The associations of genetic polymorphisms in CYP1A2 and CYP3A4 with clinical outcomes of breast cancer patients in northern China. Oncotarget 2018; 8:38367-38377. [PMID: 28418906 PMCID: PMC5503538 DOI: 10.18632/oncotarget.16359] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/27/2017] [Indexed: 01/23/2023] Open
Abstract
Background Cytochrome P450 (CYP) 1A2 and CYP3A4 may play a role in the differentiation of clinical outcomes among breast cancer women. This study aimed to analyze the association of genetic polymorphisms in the CYP1A2 and CYP3A4 genes with clinicopathological features, protein expression and prognosis of breast cancer in the northern Chinese population. Results Firstly, SNP rs11636419, rs17861162 and rs2470890 in the CYP1A2 were significantly associated with age and menstruation status. And SNP rs11636419 and rs17861162 were associated with the P53 status. Secondly, SNP rs2470890 was correlated with CYP1A2 protein expression under the co-dominant and dominant model (P = 0.017, P = 0.006, respectively). Thirdly, for SNP rs2470890, the Kaplan–Meier 5 year survival curves showed that patients carrying genotypes CT or TT had a worse OS compared with the genotype CC carriers under both codominant and dominant model (P < 0.001, P < 0.001, respectively). Materials and Methods Four single nucleotide polymorphisms (SNPs) were successfully genotyped in 459 breast cancer patients using the SNaPshot method. The associations of four polymorphisms with protein expression and clinicopathological characteristics were evaluated by Pearson's chi-square test. The Cox hazard regression analysis and Kaplan–Meier survival analysis were performed to evaluate the relationship between the SNPs and overall survival (OS) of breast cancer. Conclusions CYP1A2 rs2470890 was significantly associated with the prognosis of patients with breast cancer and could serve as an independent impact factor of prognosis of breast carcinoma.
Collapse
Affiliation(s)
- Xianan Bai
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin150040, China
| | - Jingjing Xie
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin150040, China
| | - Shanshan Sun
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin150040, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin150040, China
| | - Yongdong Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin150040, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin150040, China
| |
Collapse
|
30
|
Ashtekar SS, Bhatia NM, Bhatia MS. Development of leads targeting ER-α in breast cancer: An in silico exploration from natural domain. Steroids 2018; 131:14-22. [PMID: 29307843 DOI: 10.1016/j.steroids.2017.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/26/2017] [Accepted: 12/28/2017] [Indexed: 11/25/2022]
Abstract
The steroid, estrogen has been recognized as being important for stimulating the growth of breast cancers primarily mediated via the steroidal estrogen receptor-α (ER-α). Inhibition of estrogen activity by small molecules with increased target specificity has proven to be an effective treatment for breast cancer. After the success stories of SERMs and fulvestrant, there is a need for the development of new small molecule modulating ER-α is due to developing resistance and side effects to current breast cancer therapy. In this pursuit, we virtually screened 227 chemically diverse bioactive natural products to get the best hits having an ER-α binding affinity. The docking scores and protein-ligand interactions of the obtained hits were emulated with the clinically used selective estrogen modulators and ER-antagonists. The results revealed 18 potential hits, which were putatively classified as hits belonging to ER agonists, modulators, and antagonists. Furthermore, as most of the hits were found to comprise the chromene nucleus, the 2D and 3D QSAR studies were performed using a set of natural products and synthesized compounds containing this scaffold, to understand the structural requirements for improving activity against breast cancer. Additionally, a pharmacophore model was generated to investigate the pharmacophoric features of the explored scaffolds for an optimal anticancer activity. The results signify that these compounds with structural modification could serve as potential leads in the drug discovery process for the treatment of breast cancer.
Collapse
Affiliation(s)
- Snehal S Ashtekar
- Department of Quality Assurance, Bharati Vidyapeeth College of Pharmacy, Kolhapur 416013, Maharashtra, India.
| | - Neela M Bhatia
- Department of Quality Assurance, Bharati Vidyapeeth College of Pharmacy, Kolhapur 416013, Maharashtra, India
| | - Manish S Bhatia
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur 416013, Maharashtra, India
| |
Collapse
|
31
|
Baicalein has protective effects on the 17β-estradiol-induced transformation of breast epithelial cells. Oncotarget 2018; 8:10470-10484. [PMID: 28060756 PMCID: PMC5354673 DOI: 10.18632/oncotarget.14433] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/13/2016] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic and systematic studies have indicated that flavonoid consumption is associated with a lower incidence of breast cancer. Baicalein is the primary flavonoid derived from the roots of Scutellaria baicalensis Georgi. In the current study, the long-term exposure of breast epithelial cells to 17β-estradiol (E2) was used to investigate the chemopreventive potential of baicalein on neoplastic transformation. The results demonstrated that baicalein significantly inhibited E2-induced cell growth, motility, and invasiveness, and suppressed E2-induced misshapen acini formation in 3D cultures. Furthermore, it inhibited the ability of E2-induced cells to form clones in agarose and tumors in NOD/SCID immunodeficient mice. Docking studies using Sybyl-X 1.2 software showed that baicalein could bind to both estrogen receptor-α (ERa) and G-protein coupled estrogen receptor 30 (GPR30), which are two critical E2-mediated pathways. Baicalein prevented the E2-induced ERa-mediated activation of nuclear transcriptional signaling by interfering with the trafficking of ERa into the nucleus and subsequent binding to estrogen response elements, thereby decreasing the mRNA levels of ERa target genes. It also inhibited E2-induced GPR30-mediated signal transduction, as well as the transcription of GPR30-regulated genes. Therefore, these results suggest that baicalein is a potential drug for reducing the risk of estrogen-dependent breast cancer.
Collapse
|
32
|
Lone SH, Bhat MA, Lone RA, Jameel S, Lone JA, Bhat KA. Hemisynthesis, computational and molecular docking studies of novel nitrogen containing steroidal aromatase inhibitors: testolactam and testololactam. NEW J CHEM 2018. [DOI: 10.1039/c8nj00063h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Combined DFT and molecular docking studies of synthesized steroidal lactams reveal their potential as aromatase inhibitors.
Collapse
Affiliation(s)
- Shabir H. Lone
- Department of Chemistry
- Govt Degree College Khanabal
- Anantnag
- India
| | - Muzzaffar A. Bhat
- Department of Chemistry
- Islamic University of Science and Technology
- Awantipora
- India
| | - Rayees A. Lone
- Bioorganic Chemistry Division Indian Institute of Integrative Medicine
- Srinagar
- India
| | - Salman Jameel
- Bioorganic Chemistry Division Indian Institute of Integrative Medicine
- Srinagar
- India
| | - Javeed A. Lone
- Bioorganic Chemistry Division Indian Institute of Integrative Medicine
- Srinagar
- India
| | - Khursheed A. Bhat
- Bioorganic Chemistry Division Indian Institute of Integrative Medicine
- Srinagar
- India
| |
Collapse
|
33
|
Kim JY, Choi HG, Lee HM, Lee GA, Hwang KA, Choi KC. Effects of bisphenol compounds on the growth and epithelial mesenchymal transition of MCF-7 CV human breast cancer cells. J Biomed Res 2017; 31:358-369. [PMID: 28808208 PMCID: PMC5548997 DOI: 10.7555/jbr.31.20160162] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bisphenol-A (BPA) has been considered as an endocrine disrupting chemical (EDC) because it can exert estrogenic properties. For bisphenol-S (BPS) and bisphenol-F (BPF) that are BPA analogs and substitutes, their risk to estrogen-dependent cancer has been reported rarely compared with the numerous cases of BPA. In this study, we examined whether BPA, BPS, and BPF can lead to the proliferation, migration, and epithelial mesenchymal transition (EMT) of MCF-7 clonal variant (MCF-7 CV) breast cancer cells expressing estrogen receptors (ERs). In a cell viability assay, BPA, BPS, and BPF significantly increased proliferation of MCF-7 CV cells compared to control (DMSO) as did 17β-estradiol (E2). In Western blotting assay, BPA, BPS, and BPF enhanced the protein expression of cell cycle progression genes such as cyclin D1 and E1. In addition, MCF-7 CV cells lost cell to cell contacts and acquired fibroblast-like morphology by the treatment of BPA, BPS, or BPF for 24 hours. In cell migration assay, BPA, BPS, and BPF accelerated the migration capability of MCF-7 CV cells as did E2. In relation with the EMT process, BPA, BPS, and BPF increased the protein expression ofN-cadherin, while they decreased the protein expression of E-cadherin. When BPA, BPS, and BPF were co-treated with ICI 182,780, an ER antagonist, proliferation effects were reversed, the expression of cyclin D1 and cyclin E1 was downregulated, and the altered cell migration and expression ofN-cadherin and E-cadherin by BPA, BPS, and BPF were restored to the control level. Thus, these results imply that BPS and BPF also have the risk of breast cancer progression as much as BPA in the induction of proliferation and migration of MCF-7 CV cells by regulating the protein expression of cell cycle-related genes and EMT markersvia the ER-dependent pathway.
Collapse
Affiliation(s)
- Ji-Youn Kim
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644 Republic of Korea
| | - Ho-Gyu Choi
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644 Republic of Korea
| | - Hae-Miru Lee
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644 Republic of Korea
| | - Geum-A Lee
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644 Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644 Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644 Republic of Korea
| |
Collapse
|
34
|
Daraei A, Izadi P, Khorasani G, Nafissi N, Naghizadeh MM, Younosi N, Meysamie A, Mansoori Y, Bastami M, Tavakkoly-Bazzaz J. Epigenetic Changes of the ESR1 Gene in Breast Tissue of Healthy Women: A Missing Link with Breast Cancer Risk Factors? Genet Test Mol Biomarkers 2017; 21:464-470. [DOI: 10.1089/gtmb.2017.0028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasemali Khorasani
- Division of Plastic and Reconstructive Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Nafissi
- Surgical Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Nasim Younosi
- Surgical Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alipasha Meysamie
- Community and Preventive Medicine Department, Medical Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Milad Bastami
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Wen C, Wu L, Fu L, Wang B, Zhou H. Unifying mechanism in the initiation of breast cancer by metabolism of estrogen (Review). Mol Med Rep 2017. [PMID: 28627646 DOI: 10.3892/mmr.2017.6738] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Excessive exposure to estrogen is associated with increased risk of breast cancer. The mechanisms of carcinogenesis in the breast caused by estrogen metabolism include formation of depurinating adducts which are released from DNA to generate apurinic sites, and production of reactive oxygen species (ROS). Excess ROS not only exerts genotoxicity by indirectly increasing genomic instability, but also stimulates progression of mammary carcinogenicity by inducing a redox‑associated signaling pathway. Estrogen metabolism enzymes serve an important role in estrogen metabolism. Alterations in the expression and activity of estrogen metabolism enzymes may influence estrogen metabolism homeostasis. The present review discusses the process of estrogen metabolism, the role of estrogen metabolites and ROS in breast carcinogenesis, and the effect of metabolism enzyme polymorphisms on generation of pro‑carcinogens and breast cancer susceptibility.
Collapse
Affiliation(s)
- Chunjie Wen
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lanxiang Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lijuan Fu
- Institute of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Bing Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Honghao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
36
|
Cavalieri EL, Rogan EG, Zahid M. Critical depurinating DNA adducts: Estrogen adducts in the etiology and prevention of cancer and dopamine adducts in the etiology and prevention of Parkinson's disease. Int J Cancer 2017; 141:1078-1090. [PMID: 28388839 DOI: 10.1002/ijc.30728] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/23/2017] [Accepted: 03/17/2017] [Indexed: 01/19/2023]
Abstract
Endogenous estrogens become carcinogens when dangerous metabolites, the catechol estrogen quinones, are formed. In particular, the catechol estrogen-3,4-quinones can react with DNA to produce an excess of specific depurinating estrogen-DNA adducts. Loss of these adducts leaves apurinic sites in the DNA, generating subsequent cancer-initiating mutations. Unbalanced estrogen metabolism yields excessive catechol estrogen-3,4-quinones, increasing formation of depurinating estrogen-DNA adducts and the risk of initiating cancer. Evidence for this mechanism of cancer initiation comes from various types of studies. High levels of depurinating estrogen-DNA adducts have been observed in women with breast, ovarian or thyroid cancer, as well as in men with prostate cancer or non-Hodgkin lymphoma. Observation of high levels of depurinating estrogen-DNA adducts in high risk women before the presence of breast cancer indicates that adduct formation is a critical factor in breast cancer initiation. Formation of analogous depurinating dopamine-DNA adducts is hypothesized to initiate Parkinson's disease by affecting dopaminergic neurons. Two dietary supplements, N-acetylcysteine and resveratrol complement each other in reducing formation of catechol estrogen-3,4-quinones and inhibiting formation of estrogen-DNA adducts in cultured human and mouse breast epithelial cells. They also inhibit malignant transformation of these cells. In addition, formation of adducts was reduced in women who followed a Healthy Breast Protocol that includes N-acetylcysteine and resveratrol. When initiation of cancer is blocked, promotion, progression and development of the disease cannot occur. These results suggest that reducing formation of depurinating estrogen-DNA adducts can reduce the risk of developing a variety of types of human cancer.
Collapse
Affiliation(s)
- Ercole L Cavalieri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE.,Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE
| | - Eleanor G Rogan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE.,Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE
| | - Muhammad Zahid
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
37
|
Alterations in estrogen signalling pathways upon acquisition of anthracycline resistance in breast tumor cells. PLoS One 2017; 12:e0172244. [PMID: 28196134 PMCID: PMC5308870 DOI: 10.1371/journal.pone.0172244] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/01/2017] [Indexed: 11/19/2022] Open
Abstract
Intrinsic or acquired drug resistance is a major impediment to the successful treatment of women with breast cancer using chemotherapy. We have observed that MCF-7 breast tumor cells selected for resistance to doxorubicin or epirubicin (MCF-7DOX2 and MCF-7EPI cells, respectively) exhibited increased expression of several members of the aldo-keto reductase (AKR) gene family (in particular AKR1C3 and AKR1B10) relative to control MCF-7CC cells selected by propagation in the absence of drug. Normal cellular roles for the AKRs include the promotion of estrogen (E2) synthesis from estrone (E1) and the hydroxylation and detoxification of exogenous xenobiotics such as anthracycline chemotherapy drugs. While hydroxylation of anthracyclines strongly attenuates their cytotoxicity, it is unclear whether the enhanced AKR expression in the above anthracycline-resistant cells promotes E2 synthesis and/or alterations in E2 signalling pathways and whether such changes contribute to enhanced survival and anthracycline resistance. To determine the role of AKRs and E2 pathways in doxorubicin resistance, we examined changes in the expression of E2-related genes and proteins upon acquisition of doxorubicin resistance. We also assessed the effects of AKR overexpression or downregulation or the effects of activators or inhibitors of E2-dependent pathways on previously acquired resistance to doxorubicin. In this study we observed that the enhanced AKR expression upon acquisition of anthracycline resistance was, in fact, associated with enhanced E2 production. However, the expression of estrogen receptor α (ERα) was reduced by 2- to 5-fold at the gene transcript level and 2- to 20-fold at the protein level upon acquisition of anthracycline resistance. This was accompanied by an even stronger reduction in ERα phosphorylation and activity, including highly suppressed expression of two proteins under E2-dependent control (Bcl-2 and cyclin D1). The diminished Bcl-2 and cyclin D1 expression would be expected to reduce the growth rate of the cells, a hypothesis which was confirmed in subsequent cell proliferation experiments. AKR1C3 or AKR1B10 overexpression alone had no effect on doxorubicin sensitivity in MCF-7CC cells, while siRNA-mediated knockdown of AKR1C3 and/or AKR1B10 expression had no significant effect on sensitivity to doxorubicin in MCF-7DOX2 or MCF-7EPI cells. This suggested that enhanced or reduced AKR expression/activity is insufficient to confer anthracycline resistance or sensitivity to breast tumor cells, respectively. Rather, it would appear that AKR overexpression acts in concert with other proteins to confer anthracycline resistance, including reduced E2-dependent expression of both an important apoptosis inhibitor (Bcl-2) and a key protein associated with activation of cell cycle-dependent kinases (cyclin D1).
Collapse
|
38
|
Kumavath R, Azad M, Devarapalli P, Tiwari S, Kar S, Barh D, Azevedo V, Kumar AP. Novel aromatase inhibitors selection using induced fit docking and extra precision methods: Potential clinical use in ER-alpha-positive breast cancer. Bioinformation 2016; 12:324-331. [PMID: 28293075 PMCID: PMC5320928 DOI: 10.6026/97320630012324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/25/2016] [Accepted: 08/12/2016] [Indexed: 11/29/2022] Open
Abstract
Aromatase (CYP19A1) the key enzyme of estrogen biosynthesis, is often deregulated in breast cancer patients. It catalyzes the conversion of androgen to estrogen, thus responsible for production of estrogen in human body. However, it causes over-production of estrogen which eventually leads to proliferation of breast cancer cells. Identification of new small molecule inhibitors targeted against CYP19A1 therefore, facilitates to increase drug sensitivity of cancer cells. In this scenario, the present study aims to identify new molecules which could block or suppress the activity of aromatase enzyme by molecular docking studies using Schrödinger-Maestro v9.3. In this study we used in silico approach by modeling CYP19A1 protein the strcture was subjected to protein preparation wizard; to add hydrogen and optimize the protonation states of Thr310 and Ser478 and Asp309 residues. Active site of the CYP19A1 protein was identified using SiteMap tool of Scchrodinger package. We further carried out docking studies by means of Glid, with various ligands. Based on glid score, potential ligands were screeened and their interaction with CYP19A1 was identified. The best hits were further screened for Lipinski's rule for drug-likeliness and bioactivity scoring properties. Thus, we report two rubivivaxin and rhodethrin compounds that have successfully satisfied all in silico parameters, necessitating further in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences,Central University of Kerala, Padannakad P.O., Kasaragod-671314, Kerala, India
| | - Manan Azad
- Department of Genomic Science, School of Biological Sciences,Central University of Kerala, Padannakad P.O., Kasaragod-671314, Kerala, India
| | - Pratap Devarapalli
- Department of Genomic Science, School of Biological Sciences,Central University of Kerala, Padannakad P.O., Kasaragod-671314, Kerala, India
| | - Sandeep Tiwari
- Instituto de CiênciasBiológicas, Universidade Federal de Minas Gerais. MG,Brazil
| | - Shreya Kar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore-117599
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri-721172, PurbaMedinipur, West Bengal, India
| | - Vasco Azevedo
- Instituto de CiênciasBiológicas, Universidade Federal de Minas Gerais. MG,Brazil
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore-117599
- National University Cancer Institute, National University Health System, Singapore-119074
- Department of Biological Sciences, University of North Texas, Denton-762035017, Texas, USA
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore-117600
- Faculty of Health Sciences, School of Biomedical Sciences, Curtin University, Bently, Western Australia-6102
| |
Collapse
|
39
|
Minari J, Ogar G, Bello A. Antiproliferative potential of aqueous leaf extract of Mucuna pruriens on DMBA-induced breast cancer in female albino rats. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2016. [DOI: 10.1016/j.ejmhg.2015.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
40
|
Wang S, Dunlap TL, Howell CE, Mbachu OC, Rue EA, Phansalkar R, Chen SN, Pauli GF, Dietz BM, Bolton JL. Hop (Humulus lupulus L.) Extract and 6-Prenylnaringenin Induce P450 1A1 Catalyzed Estrogen 2-Hydroxylation. Chem Res Toxicol 2016; 29:1142-50. [PMID: 27269377 PMCID: PMC4951797 DOI: 10.1021/acs.chemrestox.6b00112] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Humulus lupulus L. (hops) is a popular botanical
dietary supplement used by women as a sleep aid and for postmenopausal
symptom relief. In addition to its efficacy for menopausal symptoms,
hops can also modulate the chemical estrogen carcinogenesis pathway
and potentially protect women from breast cancer. In the present study,
an enriched hop extract and the key bioactive compounds [6-prenylnarigenin
(6-PN), 8-prenylnarigenin (8-PN), isoxanthohumol (IX), and xanthohumol
(XH)] were tested for their effects on estrogen metabolism in breast
cells (MCF-10A and MCF-7). The methoxyestrones (2-/4-MeOE1) were analyzed as biomarkers for the nontoxic P450 1A1 catalyzed
2-hydroxylation and the genotoxic P450 1B1 catalyzed 4-hydroxylation
pathways, respectively. The results indicated that the hop extract
and 6-PN preferentially induced the 2-hydroxylation pathway in both
cell lines. 8-PN only showed slight up-regulation of metabolism in
MCF-7 cells, whereas IX and XH did not have significant effects in
either cell line. To further explore the influence of hops and its
bioactive marker compounds on P450 1A1/1B1, mRNA expression and ethoxyresorufin O-dealkylase (EROD) activity were measured. The results
correlated with the metabolism data and showed that hop extract and
6-PN preferentially enhanced P450 1A1 mRNA expression and increased
P450 1A1/1B1 activity. The aryl hydrocarbon receptor (AhR) activation
by the isolated compounds was tested using xenobiotic response element
(XRE) luciferase construct transfected cells. 6-PN was found to be
an AhR agonist that significantly induced XRE activation and inhibited
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induced
XRE activity. 6-PN mediated induction of EROD activity was also inhibited
by the AhR antagonist CH223191. These data show that the hop extract
and 6-PN preferentially enhance the nontoxic estrogen 2-hydroxylation
pathway through AhR mediated up-regulation of P450 1A1, which further
emphasizes the importance of standardization of botanical extracts
to multiple chemical markers for both safety and desired bioactivity.
Collapse
Affiliation(s)
- Shuai Wang
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Tareisha L Dunlap
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Caitlin E Howell
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Obinna C Mbachu
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Emily A Rue
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Rasika Phansalkar
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Shao-Nong Chen
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Guido F Pauli
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Birgit M Dietz
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| | - Judy L Bolton
- UIC/NIH Center for Botanical Dietary Supplements Research, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago , 833 South Wood Street, Chicago, Illinois 60612-7231, United States
| |
Collapse
|
41
|
Petkov PI, Schultz TW, Donner EM, Honma M, Morita T, Hamada S, Wakata A, Mishima M, Maniwa J, Todorov M, Kaloyanova E, Kotov S, Mekenyan OG. Integrated approach to testing and assessment for predicting rodent genotoxic carcinogenicity. J Appl Toxicol 2016; 36:1536-1550. [DOI: 10.1002/jat.3338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Petko I. Petkov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Terry W. Schultz
- College of Veterinary Medicine; The University of Tennessee; Knoxville TN 37996-4500 USA
| | - E. Maria Donner
- DuPont Haskell Global Centers for Health and Environmental Sciences, Newark; DE USA
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis; National Institute of Health Sciences; Tokyo Japan
| | - Takeshi Morita
- Division of Risk Assessment; National Institute of Health Sciences; Tokyo Japan
| | | | | | - Masayuki Mishima
- Chugai Pharmaceutical Co., Ltd., Fuji Gotemba Research Labs; Shizuoka Japan
| | - Jiro Maniwa
- Clinical Science Division, Research & Development AstraZeneca KK; Osaka Japan
| | - Milen Todorov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Elena Kaloyanova
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Stefan Kotov
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| | - Ovanes G. Mekenyan
- Laboratory of Mathematical Chemistry (LMC); As. Zlatarov University; Bourgas Bulgaria
| |
Collapse
|
42
|
Relation of Serum Estrogen Metabolites with Terminal Duct Lobular Unit Involution Among Women Undergoing Diagnostic Image-Guided Breast Biopsy. Discov Oncol 2016; 7:305-315. [PMID: 27138982 DOI: 10.1007/s12672-016-0265-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/19/2016] [Indexed: 12/11/2022] Open
Abstract
Higher levels of circulating estrogens and estrogen metabolites (EMs) have been associated with higher breast cancer risk. In breast tissues, reduced levels of terminal duct lobular unit (TDLU) involution, as reflected by higher numbers of TDLUs and acini per TDLU, have also been linked to elevated breast cancer risk. However, it is unknown whether reduced TDLU involution mediates the risk associated with circulating EMs. In a cross-sectional analysis of 94 premenopausal and 92 postmenopausal women referred for clinical breast biopsy at an academic facility in Vermont, we examined the associations of 15 EMs, quantified using liquid chromatography-tandem mass spectrometry, with the number of TDLUs and acini count/TDLU using zero-inflated Poisson regression with a robust variance estimator and ordinal logistic regression models, respectively. All analyses were stratified by menopausal status and adjusted for potential confounders. Among premenopausal women, comparing the highest vs. the lowest tertiles, levels of unconjugated estradiol (risk ratio (RR) = 1.74, 95 % confidence interval (CI) = 1.06-2.87, p trend = 0.03), 2-hydroxyestrone (RR = 1.74, 95 % CI = 1.01-3.01, p trend = 0.04), and 4-hydroxyestrone (RR = 1.74, 95 % CI = 0.99-3.06, p trend = 0.04) were associated with significantly higher TDLU count. Among postmenopausal women, higher levels of estradiol (RR = 2.09, 95 % CI = 1.01-4.30, p trend = 0.04) and 16α-hydroxyestrone (RR = 2.27, 95 % CI = 1.29-3.99, p trend = 0.02) were significantly associated with higher TDLU count. Among postmenopausal women, higher levels of EMs, specifically conjugated estrone and 2- and 4-pathway catechols, were also associated with higher acini count/TDLU. Our data suggest that higher levels of serum EMs are generally associated with lower levels of TDLU involution.
Collapse
|
43
|
Rárová L, Steigerová J, Kvasnica M, Bartůněk P, Křížová K, Chodounská H, Kolář Z, Sedlák D, Oklestkova J, Strnad M. Structure activity relationship studies on cytotoxicity and the effects on steroid receptors of AB-functionalized cholestanes. J Steroid Biochem Mol Biol 2016; 159:154-69. [PMID: 26976651 DOI: 10.1016/j.jsbmb.2016.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/02/2016] [Accepted: 03/10/2016] [Indexed: 01/14/2023]
Abstract
Structure-activity relationship analysis and profiling of a library of AB-functionalized cholestane derivatives closely related to brassinosteroids (BRs) were performed to examine their antiproliferative activities and activities on steroid hormone receptors. Some of the compounds were found to have strong cytotoxic activity in several human normal and cancer cell lines. The presence of a 3-hydroxy or 3-oxo group and 2,3-vicinal diol or 3,4-vicinal diol moiety were found to be necessary for optimum biological activity, as well as a six-membered B ring. According to the profiling of all steroid receptors in both agonist and antagonist mode, the majority of the cholestanes were weakly active or inactive compared to the natural ligands. Estrogenic activity was detected for two compounds, two compounds possessed antagonistic properties on estrogen receptors and seven compounds showed agonistic activity. Two active cholestane derivatives were shown to strongly influence cell viability, proliferation, cell cycle distribution, apoptosis and molecular pathways responsible for these processes in hormone-sensitive/insensitive (MCF7/MDA-MB-468) breast cancer cell lines.
Collapse
Affiliation(s)
- Lucie Rárová
- Department of Chemical Biology and Genetics, Centre of the Region Haná for Biotechnological and Agricultural Research, Palacký University, Šlechtitelů 27, 78371 Olomouc, Czech Republic.
| | - Jana Steigerová
- Laboratory of Molecular Pathology, Institute of Clinical and Molecular Pathology, Faculty of Medicine, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic; Institute of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacký University and Faculty Hospital in Olomouc, Hněvotínská 5, 77900 Olomouc, Czech Republic.
| | - Miroslav Kvasnica
- Laboratory of Growth Regulators, Centre of the Region Haná for Biotechnological and Agricultural Research, Institute of Experimental Botany ASCR & Palacký University, Šlechtitelů 27, 78371 Olomouc, Czech Republic.
| | - Petr Bartůněk
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20 Praha 4, Czech Republic.
| | - Kateřina Křížová
- Laboratory of Molecular Pathology, Institute of Clinical and Molecular Pathology, Faculty of Medicine, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic; Institute of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacký University and Faculty Hospital in Olomouc, Hněvotínská 5, 77900 Olomouc, Czech Republic.
| | - Hana Chodounská
- Institute of Organic Chemistry and Biochemistry of the Academy of Sciences of the Czech Republic, v.v.i, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic.
| | - Zdeněk Kolář
- Laboratory of Molecular Pathology, Institute of Clinical and Molecular Pathology, Faculty of Medicine, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic.
| | - David Sedlák
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20 Praha 4, Czech Republic.
| | - Jana Oklestkova
- Laboratory of Growth Regulators, Centre of the Region Haná for Biotechnological and Agricultural Research, Institute of Experimental Botany ASCR & Palacký University, Šlechtitelů 27, 78371 Olomouc, Czech Republic.
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Centre of the Region Haná for Biotechnological and Agricultural Research, Institute of Experimental Botany ASCR & Palacký University, Šlechtitelů 27, 78371 Olomouc, Czech Republic.
| |
Collapse
|
44
|
Phipps AI, Bhatti P, Neuhouser ML, Chen C, Crane TE, Kroenke CH, Ochs-Balcom H, Rissling M, Snively BM, Stefanick ML, Treggiari MM, Watson NF. Pre-diagnostic Sleep Duration and Sleep Quality in Relation to Subsequent Cancer Survival. J Clin Sleep Med 2016; 12:495-503. [PMID: 26612513 PMCID: PMC4795275 DOI: 10.5664/jcsm.5674] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/02/2015] [Indexed: 12/19/2022]
Abstract
STUDY OBJECTIVES Poor sleep quality and short sleep duration have been associated with elevated risk for several cancer types; however, the relationship between sleep and cancer outcomes has not been well characterized. We assessed the association between pre-diagnostic sleep attributes and subsequent cancer survival within the Women's Health Initiative (WHI). METHODS We identified WHI participants in whom a first primary invasive cancer had been diagnosed during follow-up (n = 21,230). Participants provided information on sleep characteristics at enrollment. Cox regression was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for associations between these pre-diagnostic sleep characteristics and cancer-specific survival for all cancers combined and separately for common cancers. Analyses were adjusted for age, study arm, cancer site, marital status, income, smoking, physical activity, and time to diagnosis. RESULTS No individual pre-diagnostic sleep characteristics were found to be significantly associated with cancer survival in analyses of all cancer sites combined; however, women who reported short sleep duration (≤ 6 h sleep/night) combined with frequent snoring (≥ 5 nights/w experienced significantly poorer cancer-specific survival than those who reported 7-8 h of sleep/night and no snoring (HR = 1.32, 95% CI: 1.14-1.54). Short sleep duration (HR = 1.46, 95% CI: 1.07-1.99) and frequent snoring (HR = 1.34, 95% CI: 0.98-1.85) were each associated with poorer breast cancer survival; those reporting short sleep combined with frequent snoring combined had substantially poorer breast cancer survival than those reporting neither (HR = 2.14, 95% CI: 1.47-3.13). CONCLUSIONS Short sleep duration combined with frequent snoring reported prior to cancer diagnosis may influence subsequent cancer survival, particularly breast cancer survival.
Collapse
Affiliation(s)
- Amanda I. Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Epidemiology Department, School of Public Health, University of Washington, Seattle, WA
| | - Parveen Bhatti
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Epidemiology Department, School of Public Health, University of Washington, Seattle, WA
| | - Marian L. Neuhouser
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Tracy E. Crane
- Health Promotion Sciences Division, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | - Candyce H. Kroenke
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Heather Ochs-Balcom
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY
| | | | - Beverly M. Snively
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Marcia L. Stefanick
- Stanford Prevention Research Center, Stanford University, School of Medicine, Palo Alto, CA
| | - Miriam M. Treggiari
- Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR
| | | |
Collapse
|
45
|
Hultman MT, Song Y, Tollefsen KE. 17α-Ethinylestradiol (EE2) effect on global gene expression in primary rainbow trout (Oncorhynchus mykiss) hepatocytes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 169:90-104. [PMID: 26519835 DOI: 10.1016/j.aquatox.2015.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/06/2015] [Accepted: 10/06/2015] [Indexed: 06/05/2023]
Abstract
The potential impact of endocrine disrupting chemicals (EDCs) in the aquatic environment has driven the development of screening assays to evaluate the estrogenic properties of chemicals and their effects on aquatic organisms such as fish. However, obtaining full concentration-response relationships in animal (in vivo) exposure studies are laborious, costly and unethical, hence a need for developing feasible alternative (non-animal) methods. Use of in vitro bioassays such as primary fish hepatocytes, which retain many of the native properties of the liver, has been proposed for in vitro screening of estrogen receptor (ER) agonists and antagonists. The aim of present study was to characterize the molecular mode of action (MoA) of the ER agonist 17α-ethinylestradiol (EE2) in primary rainbow trout (Oncorhynchus mykiss) hepatocytes. A custom designed salmonid 60,000-feature (60k) oligonucleotide microarray was used to characterize the potential MoAs after 48h exposure to EE2. The microarray analysis revealed several concentration-dependent gene expression alterations including classical estrogen sensitive biomarker gene expression (e.g. estrogen receptor α, vitellogenin, zona radiata). Gene Ontology (GO) analysis displayed transcriptional changes suggesting interference of cellular growth, fatty acid and lipid metabolism potentially mediated through the estrogen receptor (ER), which were proposed to be associated with modulation of genes involved in endocrine function and reproduction. Pathway analysis supported the identified GOs and revealed modulation of additional genes associated with apoptosis and cholesterol biosynthesis. Differentially expressed genes (DEGs) related to impaired lipid metabolism (e.g. peroxisome proliferator-activated receptor α and γ), growth (e.g. insulin growth factor protein 1), phase I and II biotransformation (e.g. cytochrome P450 1A, sulfotransferase, UDP-glucuronosyltransferase and glutathione S-transferase) provided additional insight into the MoA of EE2 in primary fish hepatocytes. Results from the present study suggest that biotransformation, estrogen receptor-mediated responses, lipid homeostasis, growth and cancer/apoptosis in primary fish hepatocytes may be altered after short-term exposure to ER-agonists such as EE2. In many cases the observed changes were similar to those reported for estrogen-exposed fish in vivo. In conclusion, global transcriptional analysis demonstrated that EE2 affected a number of toxicologically relevant pathways associated with an estrogenic MoA in the rainbow trout hepatocytes.
Collapse
Affiliation(s)
- Maria T Hultman
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway; Faculty of Environmental Science & Technology, Department for Environmental Sciences, Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway.
| | - You Song
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway
| | - Knut Erik Tollefsen
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway; Faculty of Environmental Science & Technology, Department for Environmental Sciences, Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway
| |
Collapse
|
46
|
Santos MA, Florencio-Silva R, Teixeira CP, Sasso GRDS, Marinho DS, Simões RS, Simões MJ, Carbonel AF. Effects of early and late treatment with soy isoflavones in the mammary gland of ovariectomized rats. Climacteric 2015; 19:77-84. [PMID: 26606166 DOI: 10.3109/13697137.2015.1094783] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
INTRODUCTION Soy isoflavones have been shown to be an alternative to hormone therapy at menopause, without causing side-effects such as breast cancer. However, the effects of early and late treatment with isoflavones on the mammary gland remain controversial. OBJECTIVE To investigate the effects of early and late treatment with soy isoflavones on the mammary gland of ovariectomized rats. METHODS Thirty 3-month-old rats were ovariectomized and divided equally into groups: Control, treated with vehicle solution; or with 150 mg/kg/body weight of isoflavones by gavage; or subcutaneously treated with 10 μg/kg/body weight with 17β-estradiol. Treatments started 3 days (early treatment) or 30 days (late treatment) after ovariectomy and lasted for 30 consecutive days. Thereafter, the animals were euthanized and the mammary glands were removed and processed for paraffin embedding. Sections were stained with hematoxylin and eosin for histomorphometry or subjected to immunohistochemical detection of Ki-67 and VEGF-A. RESULTS The ductal, lobular and total epithelial fractions were similar between controls and the early/late isoflavone groups, but they were significantly higher in the groups treated with estradiol. In both epithelial and stromal regions, the immunoreactivity of VEGF-A and the percentage of Ki-67-positive cells were significantly higher in the groups treated with estradiol, while they were similar in the early/late isoflavone groups and control groups. CONCLUSION Our results indicate that early and late treatment with soy isoflavones at the dose of 150 mg/kg/body weight does not show proliferative and angiogenic effects on the mammary gland of ovariectomized rats.
Collapse
Affiliation(s)
| | - R Florencio-Silva
- b Morphology and Genetics , Universidade Federal De São Paulo , São Paulo
| | - C P Teixeira
- b Morphology and Genetics , Universidade Federal De São Paulo , São Paulo
| | | | - D Souza Marinho
- b Morphology and Genetics , Universidade Federal De São Paulo , São Paulo
| | - R S Simões
- c Gynecology, Universidade De São Paulo , São Paulo , Brazil
| | - M J Simões
- b Morphology and Genetics , Universidade Federal De São Paulo , São Paulo
| | - A Ferraz Carbonel
- b Morphology and Genetics , Universidade Federal De São Paulo , São Paulo
| |
Collapse
|
47
|
Go RE, Hwang KA, Choi KC. Cytochrome P450 1 family and cancers. J Steroid Biochem Mol Biol 2015; 147:24-30. [PMID: 25448748 DOI: 10.1016/j.jsbmb.2014.11.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/28/2014] [Accepted: 11/03/2014] [Indexed: 02/07/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcriptional factor that dimerizes with aryl hydrocarbon receptor nuclear translocator (ARNT). This complex binds to xenobiotics response element (XREs), and then starts the expressions of downstream genes including cytochrome P450 (CYP) 1 family members: CYP1A1, CYP1A2 and CYP1B1. Role of CYP1 family is involved in the metabolism of endogenous hormones, xenobiotics and drug. The expression of CYP1 family is regulated by estradiol (E2) or xenobiotics in diverse cancers. In breast cancers expressing estrogen receptors (ERs), level of CYP1B1 is increased by E2 and reversed by an estrogen receptor antagonist, ICI 182,780 or 4-hydrotamoxifen, which indicates that the expression of CYP1 family in downstream region of AhR is regulated by an activation of ERα. In metabolic pathways, E2 is converted into 4-hydroxyestradiol by CYP1B1, which can be converted into mainly estradiol-3,4-quinone, a potential carcinogen, by peroxidase. Increased expression of CYP1 family indicates the possibility of carcinogenesis by exposure of xenobiotics in endometrial and ovarian cancers. Apart from roles of CYP1 family in relation with ER pathway, CYP1 family is over-expressed in ER independent cancers. CYP1A1 exhibits hydroxylase activity in oxidation of arachidonic acid, which has been transformed to 12(R)-hydrxyeicosatetraenoic (HETEs), a potent activator of AhR activity. On the basis of results, phytoestrogens and dexamethasone are provided as cancer therapy regulating the expression of CYP1 family. Thus, this review focuses on the role(s) of CYP1 family in ER-dependent or ER-independent cancers and the potential for cancer therapy to target CYP1 family in these cancers.
Collapse
Affiliation(s)
- Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763,Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763,Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763,Republic of Korea.
| |
Collapse
|
48
|
Niwa T, Murayama N, Imagawa Y, Yamazaki H. Regioselective hydroxylation of steroid hormones by human cytochromes P450. Drug Metab Rev 2015; 47:89-110. [PMID: 25678418 DOI: 10.3109/03602532.2015.1011658] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This article reviews in vitro metabolic activities [including Michaelis constants (Km), maximal velocities (Vmax) and Vmax/Km] and drug-steroid interactions [such as induction and cooperativity (activation)] of cytochromes P450 (P450 or CYP) in human tissues, including liver and adrenal gland, for 14 kinds of endogenous steroid compounds, including allopregnanolone, cholesterol, cortisol, cortisone, dehydroepiandrosterone, estradiol, estrone, pregnenolone, progesterone, testosterone and bile acids (cholic acid). First, we considered the drug-metabolizing P450s. 6β-Hydroxylation of many steroids, including cortisol, cortisone, progesterone and testosterone, was catalyzed primarily by CYP3A4. CYP1A2 and CYP3A4, respectively, are likely the major hepatic enzymes responsible for 2-/4-hydroxylation and 16α-hydroxylation of estradiol and estrone, steroids that can contribute to breast cancer risk. In contrast, CYP1A1 and CYP1B1 predominantly metabolized estrone and estradiol to 2- and 4-catechol estrogens, which are endogenous ultimate carcinogens if formed in the breast. Some metabolic activities of CYP3A4, including dehydroepiandrosterone 7β-/16α-hydroxylation, estrone 2-hydroxylation and testosterone 6β-hydroxylation, were higher than those for polymorphically expressed CYP3A5. Next, we considered typical steroidogenic P450s. CYP17A1, CYP19A1 and CYP27A1 catalyzed steroid synthesis, including hydroxylation at 17α, 19 and 27 positions, respectively. However, it was difficult to predict which hepatic drug-metabolizing P450 or steroidogenic P450 will be mainly responsible for metabolizing each steroid hormone in vivo based on these results. Further research is required on the metabolism of steroid hormones by various P450s and on prediction of their relative contributions to in vivo metabolism. The findings collected here provide fundamental and useful information on the metabolism of steroid compounds.
Collapse
Affiliation(s)
- Toshiro Niwa
- School of Pharmacy, Shujitsu University , Okayama , Japan and
| | | | | | | |
Collapse
|
49
|
Pogash TJ, El-Bayoumy K, Amin S, Gowda K, de Cicco RL, Barton M, Su Y, Russo IH, Himmelberger JA, Slifker M, Manni A, Russo J. Oxidized derivative of docosahexaenoic acid preferentially inhibit cell proliferation in triple negative over luminal breast cancer cells. In Vitro Cell Dev Biol Anim 2014; 51:121-7. [PMID: 25413005 DOI: 10.1007/s11626-014-9822-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/15/2014] [Indexed: 02/02/2023]
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) exert an anticancer effect by affecting multiple cellular mechanisms leading to inhibition of proliferation and induction of apoptosis. It is well known that breast cancer comprises distinct molecular subtypes which differ in their responsiveness to therapeutic and preventive agents. We tested the hypothesis that n-3FA may preferentially affect triple-negative breast cancer cells for which no targeted intervention is presently available. The in vitro antiproliferative effects of n-3 PUFA docosahexaenoic acid (DHA) and its metabolite, 4-OH-DHA as well as its putative metabolite 4-OXO-DHA, were tested in five triple-negative human basal breast cell lines at different stages of transformation (MCF-10F, trMCF, bsMCF, MDA-MB-231, and BT-549) and three luminal breast cancer cell lines (MCF-7, T-47D, and SK-BR-3). Cell proliferation was measured with the tetrazolium MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) assay. DHA and its oxidized derivatives significantly inhibited cell proliferation (20-90% reduction) of both basal and luminal breast cancer cell lines. The inhibitory effect was more pronounced on triple-negative basal breast cancer cell lines as compared to luminal breast cancer cell lines after 4-OXO-DHA treatment. Our data provide novel information regarding the preferential antitumor effect of oxidized derivatives of DHA on basal type breast cancer.
Collapse
Affiliation(s)
- Thomas J Pogash
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tan X, Chen M. Association between Catechol-O-methyltransferase rs4680 (G>A) polymorphism and lung cancer risk. Diagn Pathol 2014; 9:192. [PMID: 25280560 PMCID: PMC4196007 DOI: 10.1186/s13000-014-0192-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/23/2014] [Indexed: 02/07/2023] Open
Abstract
Background The association between the Val158Met polymorphism in the catechol-O-methyltransferase (COMT) gene and lung cancer risk remains controversial and inconclusive. Therefore, the meta-analysis was performed to provide a quality reevaluation of the association between the COMT Val158Met polymorphism and the risk of lung cancer. Methods Two major public databases (Pubmed and Embase) and several Chinese databases were searched for eligible studies. Pooled odds ratios (OR) and 95% confidence intervals (CI) were calculated to estimate the strength of the association. Results Five publications, including six individual studies with a total of 4,043 subjects (1,796 cases and 2,247 controls) regarding the association of COMT Val158Met polymorphism with lung cancer susceptibility were included in this meta-analysis. Overall, pooled analysis indicated that there was no significant association between COMT Val158Met polymorphism and lung cancer susceptibility under all genetic models. Likewise, no association was observed in the stratified analysis by ethnicity and control source, either. However, Val158Met polymorphism was shown to increase lung cancer risk among women (AG vs. GG, OR = 1.190, 95% CI = 1.001–1.422, p = 0.049). Conclusion These findings suggested that the COMT l58Val/Met polymorphism confer genetic susceptibility to lung cancer among women. However, no evidence was found for the association with lung cancer risk in ethnicity and smoking status. Virtual slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/13000_2014_192
Collapse
Affiliation(s)
| | - Mingwu Chen
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|