1
|
Grzesiak M, Herian M, Kamińska K, Ajersch P. Insight into vitamin D 3 action within the ovary-Basic and clinical aspects. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:99-130. [PMID: 39059995 DOI: 10.1016/bs.apcsb.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Vitamin D3 is a fat-soluble secosteroid predominantly synthesized in the skin or delivered with a diet. Nevertheless, recently it is considered more as a hormone than a vitamin due to its pleiotropic function within the organism ensured by widely distributed vitamin D receptors and metabolic enzymes. Besides the main role in calcium and phosphorus homeostasis, vitamin D3 was shown to regulate many cellular and metabolic processes in normal and cancerous tissues within the immune system, the cardiovascular system, the respiratory system and the endocrine system. The ovary is an important extraskeletal tissue of vitamin D3 action and local metabolism, indicating its role in the regulation of ovarian functions upon physiological and pathological conditions. This chapter reviews firstly the updated information about vitamin D3 metabolism and triggered intracellular pathways. Furthermore, the basic information about ovarian physiology and several aspects of vitamin D3 effects within the ovary are presented. Finally, the special attention is paid into possible mechanism of vitamin D3 action within ovarian pathologies such as premature ovarian failure, polycystic ovary syndrome, and ovarian cancer, considering its clinical application as alternative therapy.
Collapse
Affiliation(s)
- Małgorzata Grzesiak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | | | - Kinga Kamińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Paula Ajersch
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
2
|
Mueller PR, Kershner AJ, Breitrick BI, Keller KN, Radtke RL, Patel RJ, Gierach K, Arvedson J, Moyle-Heyrman GE, Pearson DA. Vitamin D and docosahexaenoic acid inhibit proliferation of the ovarian cancer cell line OVCAR4. Nutr Health 2023:2601060231202565. [PMID: 37728210 DOI: 10.1177/02601060231202565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
BACKGROUND Ovarian cancer is one of the deadliest cancers in women. Improved preventative, diagnostic, and therapeutic strategies are needed. Certain dietary patterns and nutrients such as vitamin D and omega-3 fatty acids are associated with reduced cancer risk, but their effects on ovarian cancer remain to be fully elucidated, and their combined effects have not been explored. AIM To determine the individual and combined effects of the active vitamin D metabolite, calcitriol, and the omega-3 fatty acid, docosahexaenoic acid, on cell growth, and the abundance of the vitamin D receptor (VDR), proteins that modulate cell cycle progression, and apoptotic markers. METHODS OVCAR4 cells, a model of ovarian cancer, were treated with calcitriol, and docosahexaenoic acid, either alone or in combination. Effects on cell growth were determined by the sulforhodamine B assay. Changes in VDR, the cell cycle promotor c-Myc, the cell cycle inhibitor p27 and cleaved PARP, were determined by Western blotting. RESULTS While OVCAR4 cell growth was inhibited by individual treatment with either calcitriol or docosahexaenoic acid, the combined treatment revealed enhanced growth inhibition as compared to either treatment alone. Furthermore, long-term treatment (12 days) yielded stronger growth inhibition at lower concentrations as compared to short-term treatments (3 days). Accompanying this growth inhibition was a decrease in c-Myc, and an increase in p27. CONCLUSIONS The observed reduction in cell growth mediated by calcitriol and docosahexaenoic acid highlights the need for further research utilizing these nutrients, alone and especially in combination, to support ovarian cancer prevention and treatment.
Collapse
Affiliation(s)
- Paul R Mueller
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
- Department of Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Alexandra J Kershner
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Brooke I Breitrick
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Katharina N Keller
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Rebecca L Radtke
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Ruchita J Patel
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Kylie Gierach
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Jon Arvedson
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Georgette E Moyle-Heyrman
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
- Department of Chemistry, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| | - Debra A Pearson
- Department of Human Biology, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
- Department of Chemistry, College of Science Engineering and Technology, University of Wisconsin - Green Bay, Wisconsin, USA
| |
Collapse
|
3
|
Režen T, Rozman D, Kovács T, Kovács P, Sipos A, Bai P, Mikó E. The role of bile acids in carcinogenesis. Cell Mol Life Sci 2022; 79:243. [PMID: 35429253 PMCID: PMC9013344 DOI: 10.1007/s00018-022-04278-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022]
Abstract
AbstractBile acids are soluble derivatives of cholesterol produced in the liver that subsequently undergo bacterial transformation yielding a diverse array of metabolites. The bulk of bile acid synthesis takes place in the liver yielding primary bile acids; however, other tissues have also the capacity to generate bile acids (e.g. ovaries). Hepatic bile acids are then transported to bile and are subsequently released into the intestines. In the large intestine, a fraction of primary bile acids is converted to secondary bile acids by gut bacteria. The majority of the intestinal bile acids undergo reuptake and return to the liver. A small fraction of secondary and primary bile acids remains in the circulation and exert receptor-mediated and pure chemical effects (e.g. acidic bile in oesophageal cancer) on cancer cells. In this review, we assess how changes to bile acid biosynthesis, bile acid flux and local bile acid concentration modulate the behavior of different cancers. Here, we present in-depth the involvement of bile acids in oesophageal, gastric, hepatocellular, pancreatic, colorectal, breast, prostate, ovarian cancer. Previous studies often used bile acids in supraphysiological concentration, sometimes in concentrations 1000 times higher than the highest reported tissue or serum concentrations likely eliciting unspecific effects, a practice that we advocate against in this review. Furthermore, we show that, although bile acids were classically considered as pro-carcinogenic agents (e.g. oesophageal cancer), the dogma that switch, as lower concentrations of bile acids that correspond to their serum or tissue reference concentration possess anticancer activity in a subset of cancers. Differences in the response of cancers to bile acids lie in the differential expression of bile acid receptors between cancers (e.g. FXR vs. TGR5). UDCA, a bile acid that is sold as a generic medication against cholestasis or biliary surge, and its conjugates were identified with almost purely anticancer features suggesting a possibility for drug repurposing. Taken together, bile acids were considered as tumor inducers or tumor promoter molecules; nevertheless, in certain cancers, like breast cancer, bile acids in their reference concentrations may act as tumor suppressors suggesting a Janus-faced nature of bile acids in carcinogenesis.
Collapse
Affiliation(s)
- Tadeja Režen
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Damjana Rozman
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tünde Kovács
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
| | - Patrik Kovács
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
| | - Péter Bai
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, University of Debrecen, Egyetem tér 1., Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
| |
Collapse
|
4
|
Dogra S, Neelakantan D, Patel MM, Griesel B, Olson A, Woo S. Adipokine Apelin/APJ Pathway Promotes Peritoneal Dissemination of Ovarian Cancer Cells by Regulating Lipid Metabolism. Mol Cancer Res 2021; 19:1534-1545. [PMID: 34172534 PMCID: PMC11486291 DOI: 10.1158/1541-7786.mcr-20-0991] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/05/2021] [Accepted: 05/11/2021] [Indexed: 11/16/2022]
Abstract
Adipose tissue, which can provide adipokines and nutrients to tumors, plays a key role in promoting ovarian cancer metastatic lesions in peritoneal cavity. The adipokine apelin promotes ovarian cancer metastasis and progression through its receptor APJ, which regulates cell proliferation, energy metabolism, and angiogenesis. The objective of this study was to investigate the functional role and mechanisms of the apelin-APJ pathway in ovarian cancer metastasis, especially in context of tumor cell-adipocyte interactions. When co-cultured in the conditioned media (AdipoCM) derived from 3T3-L1 adipocytes, which express and secrete high apelin, human ovarian cancer cells with high APJ expression showed significant increases in migration and invasion in vitro. We also found that cells expressing high levels of APJ had increased cell adhesion to omentum ex vivo, and preferentially "home-in" on the omentum in vivo. These apelin-induced pro-metastatic effects were reversed by APJ antagonist F13A in a dose-dependent manner. Apelin-APJ activation increased lipid droplet accumulation in ovarian cancer cells, which was further intensified in the presence of AdipoCM and reversed by F13A or APJ knockdown. Mechanistically, this increased lipid uptake was mediated by CD36 upregulation via APJ-STAT3 activation, and the lipids were utilized in promoting fatty acid oxidation via activation of AMPK-CPT1a axis. Together, our studies demonstrate that adipocyte-derived apelin activates APJ-expressing tumor cells in a paracrine manner, promoting lipid uptake and utilization and providing energy for ovarian cancer cell survival at the metastatic sites. Hence, the apelin-APJ pathway presents a novel therapeutic target to curb ovarian cancer metastasis. IMPLICATIONS: Targeting the APJ pathway in high-grade serous ovarian carcinoma is a novel strategy to inhibit peritoneal metastasis.
Collapse
Affiliation(s)
- Samrita Dogra
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Deepika Neelakantan
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Maulin M Patel
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Cardiovascular Biology Department, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Beth Griesel
- Department of Biochemistry and Molecular Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ann Olson
- Department of Biochemistry and Molecular Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
5
|
Is Vitamin D Deficiency Related to Increased Cancer Risk in Patients with Type 2 Diabetes Mellitus? Int J Mol Sci 2021; 22:ijms22126444. [PMID: 34208589 PMCID: PMC8233804 DOI: 10.3390/ijms22126444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/08/2021] [Accepted: 06/12/2021] [Indexed: 12/30/2022] Open
Abstract
There is mounting evidence that type 2 diabetes mellitus (T2DM) is related with increased risk for the development of cancer. Apart from shared common risk factors typical for both diseases, diabetes driven factors including hyperinsulinemia, insulin resistance, hyperglycemia and low grade chronic inflammation are of great importance. Recently, vitamin D deficiency was reported to be associated with the pathogenesis of numerous diseases, including T2DM and cancer. However, little is known whether vitamin D deficiency may be responsible for elevated cancer risk development in T2DM patients. Therefore, the aim of the current review is to identify the molecular mechanisms by which vitamin D deficiency may contribute to cancer development in T2DM patients. Vitamin D via alleviation of insulin resistance, hyperglycemia, oxidative stress and inflammation reduces diabetes driven cancer risk factors. Moreover, vitamin D strengthens the DNA repair process, and regulates apoptosis and autophagy of cancer cells as well as signaling pathways involved in tumorigenesis i.e., tumor growth factor β (TGFβ), insulin-like growth factor (IGF) and Wnt-β-Cathenin. It should also be underlined that many types of cancer cells present alterations in vitamin D metabolism and action as a result of Vitamin D Receptor (VDR) and CYP27B1 expression dysregulation. Although, numerous studies revealed that adequate vitamin D concentration prevents or delays T2DM and cancer development, little is known how the vitamin affects cancer risk among T2DM patients. There is a pressing need for randomized clinical trials to clarify whether vitamin D deficiency may be a factor responsible for increased risk of cancer in T2DM patients, and whether the use of the vitamin by patients with diabetes and cancer may improve cancer prognosis and metabolic control of diabetes.
Collapse
|
6
|
Brożyna AA, Kim TK, Zabłocka M, Jóźwicki W, Yue J, Tuckey RC, Jetten AM, Slominski AT. Association among Vitamin D, Retinoic Acid-Related Orphan Receptors, and Vitamin D Hydroxyderivatives in Ovarian Cancer. Nutrients 2020; 12:E3541. [PMID: 33227893 PMCID: PMC7699234 DOI: 10.3390/nu12113541] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Vitamin D and its derivatives, acting via the vitamin D receptor (VDR) and retinoic acid-related orphan receptors γ and α (RORγ and RORα), show anticancer properties. Since pathological conditions are characterized by disturbances in the expression of these receptors, in this study, we investigated their expression in ovarian cancers (OCs), as well as explored the phenotypic effects of vitamin D hydroxyderivatives and RORγ/α agonists on OC cells. The VDR and RORγ showed both a nuclear and a cytoplasmic location, and their expression levels were found to be reduced in the primary and metastatic OCs in comparison to normal ovarian epithelium, as well as correlated to the tumor grade. This reduction in VDR and RORγ expression correlated with a shorter overall disease-free survival. VDR, RORγ, and RORα were also detected in SKOV-3 and OVCAR-3 cell lines with increased expression in the latter line. 20-Hydroxy-lumisterol3 (20(OH)L3) and synthetic RORα/RORγ agonist SR1078 inhibited proliferation only in the OVCAR-3 line, while 20-hydroxyvitamin-D3 (20(OH)D3) only inhibited SKOV-3 cell proliferation. 1,25(OH)2D3, 20(OH)L3, and SR1078, but not 20(OH)D3, inhibited spheroid formation in SKOV-3 cells. In summary, decreases in VDR, RORγ, and RORα expression correlated with an unfavorable outcome for OC, and compounds targeting these receptors had a context-dependent anti-tumor activity in vitro. We conclude that VDR and RORγ expression can be used in the diagnosis and prognosis of OC and suggest their ligands as potential candidates for OC therapy.
Collapse
Affiliation(s)
- Anna A. Brożyna
- Department of Human Biology, Institute of Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland
| | - Tae-Kang Kim
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Marzena Zabłocka
- Department of Tumor Pathology and Pathomorphology, Oncology Centre—Prof. Franciszek Łukaszczyk Memorial Hospital in Bydgoszcz, 85-796 Bydgoszcz, Poland;
| | - Wojciech Jóźwicki
- Department of Tumor Pathology and Pathomorphology, Department of Oncology, Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Junming Yue
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Robert C. Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia;
| | - Anton M. Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA;
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Laboratory Service of the VA Medical Center, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Xu J, Chen K, Zhao F, Huang D, Zhang H, Fu Z, Xu J, Wu Y, Lin H, Zhou Y, Lu W, Wu Y, Xia D. Association between vitamin D/calcium intake and 25-hydroxyvitamin D and risk of ovarian cancer: a dose-response relationship meta-analysis. Eur J Clin Nutr 2020; 75:417-429. [PMID: 32814859 DOI: 10.1038/s41430-020-00724-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/20/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The association between vitamin D/calcium and risk of ovarian cancer is still a debatable point. The aim of our study was to systematically investigate the association between vitamin D/calcium, and the risk of ovarian cancer and estimate their dose-response association quantitatively. METHODS PubMed, EMBASE, and Web of Science databases were searched to identify relevant observational studies. Two investigators screened citations and extracted data independently. Data were extracted and the association between vitamin D/calcium and ovarian cancer risk was estimated by calculating pooled relative risks (RRs). Subgroup analyses, publication bias estimation, and dose-response analyses were carried out as well. RESULTS In total, 21 articles involving 980,008 participants were included in our present study. No significant association was observed between total vitamin D intake and ovarian cancer risk (RR: 1.02; 95% CI, 0.89-1.16, p = 0.81). Further subgroup analysis suggested that neither dietary vitamin D intake (RR: 0.80; 95% CI, 0.62-1.03, p = 0.09) nor supplementary vitamin D intake (RR: 0.98; 95% CI, 0.85-1.13, p = 0.80) was associated with the risk of ovarian cancer. As for calcium, total calcium intake was found to be statistically inversely associated with ovarian cancer risk in case-control studies (RR: 0.73; 95% CI, 0.63-0.86, p < 0.001) but not in cohort studies (RR: 1.05; 95% CI, 0.90-1.24, p = 0.52). Besides, supplementation with calcium plus vitamin D was not effective for the prevention of ovarian cancer (p = 0.98). Of note, dose-response analysis based on cohort studies suggested a potential inverse U-shape relationship between calcium intake (including total calcium and dietary calcium) and ovarian cancer risk, which indicated that low dose of calcium intake might reduce ovarian cancer risk while high dose of calcium intake might not. CONCLUSIONS Taken together, vitamin D could not decrease the risk of ovarian cancer. The role of calcium intake was not proven for reducing ovarian cancer risk. Besides, no evidence showed combinative use of calcium and vitamin D have additional benefits for ovarian cancer prevention.
Collapse
Affiliation(s)
- Jiawei Xu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fan Zhao
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, 310058, China
| | - Dongdong Huang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Honghe Zhang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Zhiqin Fu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Jinming Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yongfeng Wu
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, 310058, China.,Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hui Lin
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yexinyi Zhou
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, 310058, China
| | - Weiguo Lu
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Yihua Wu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dajing Xia
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Ji MT, Nie J, Nie XF, Hu WT, Pei HL, Wan JM, Wang AQ, Zhou GM, Zhang ZL, Chang L, Li BY. 1α,25(OH) 2D 3 Radiosensitizes Cancer Cells by Activating the NADPH/ROS Pathway. Front Pharmacol 2020; 11:945. [PMID: 32848720 PMCID: PMC7426479 DOI: 10.3389/fphar.2020.00945] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The radioresistance of tumors affect the outcome of radiotherapy. Accumulating data suggest that 1α,25(OH)2D3 is a potential anti-oncogenic molecule in various cancers. In the present study, we investigated the radiosensitive effects and underlying mechanisms of 1α,25(OH)2D3 in vitro and in vivo. We found that 1α,25(OH)2D3 enhanced the radiosensitivity of lung cancer and ovarian cancer cells by promoting the NADPH oxidase-ROS-apoptosis axis. Compared to the group that only received radiation, the survival fraction and self-renewal capacity of cancer cells treated with a combination of 1α,25(OH)2D3 and radiation were decreased. Both apoptosis and ROS were significantly increased in the combination group compared with the radiation only group. Moreover, N-acetyl-L-cysteine, a scavenger of intracellular ROS, reversed the apoptosis and ROS induced by 1α,25(OH)2D3, indicating that 1α,25(OH)2D3 enhanced the radiosensitivity of cancer cells in vitro by promoting ROS-induced apoptosis. Moreover, our results demonstrated that 1α,25(OH)2D3 promoted the ROS level via activating NADPH oxidase complexes, NOX4, p22phox, and p47phox. In addition, knockdown of the vitamin D receptor (VDR) abolished the radiosensitization of 1α,25(OH)2D3, which confirmed that 1α,25(OH)2D3 radiosensitized tumor cells that depend on VDR. Similarly, our study also evidenced that vitamin D3 enhanced the radiosensitivity of cancer cells in vivo and extended the overall survival of mice with tumors. In summary, these results demonstrate that 1α,25(OH)2D3 enhances the radiosensitivity depending on VDR and activates the NADPH oxidase-ROS-apoptosis axis. Our findings suggest that 1α,25(OH)2D3 in combination with radiation enhances lung and ovarian cell radiosensitivity, potentially providing a novel combination therapeutic strategy.
Collapse
Affiliation(s)
- Min-Tao Ji
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jing Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Xue-Fei Nie
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| | - Wen-Tao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Hai-Long Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jian-Mei Wan
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| | - Ai-Qing Wang
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| | - Guang-Ming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Zeng-Li Zhang
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Centre of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Bing-Yan Li
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| |
Collapse
|
9
|
Role of vitamin D 3 in selected malignant neoplasms. Nutrition 2020; 79-80:110964. [PMID: 32877827 DOI: 10.1016/j.nut.2020.110964] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023]
Abstract
Vitamin D3 is a fat-soluble essential nutrient that affects multiple biologic functions in the organism through calcitriol and the vitamin D3 receptor. This review article focuses on the results of studies on the relationship between the level of vitamin D3 and cancer incidence or mortality, but also on the anticancer properties of vitamin D3 that support its significant role in the prevention, clinical course, and overall survival rates of selected cancers (colorectal, prostate, breast, ovarian, endometrial, bladder, and malignant melanoma). The mechanisms of vitamin D3 action involve, among others, polymorphism of vitamin D3 receptor, cell cycle, caspases, and cancer stem cells. The level of vitamin D3 has been also demonstrated to serve as a biomarker in some cancers, and high levels of vitamin D3 can be conducive to successful cancer therapy.
Collapse
|
10
|
Duman İ, Tiftik RN, Ün İ. Effects of Vitamin D Analogs Alfacalcidol and Calcitriol on Cell Proliferation and Migration of HEC1A Endometrial Adenocarcinoma Cells. Nutr Cancer 2020; 73:273-281. [PMID: 32400204 DOI: 10.1080/01635581.2020.1764066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Endometrial carcinoma is the most diagnosed among infiltrating tumor of the female genital tract. Vitamin D has antiproliferative and immunomodulatory properties besides its classical effect on calcium and phosphate. We aimed to demonstrate whether alfacalcidol and calcitriol have any effect on proliferation and migration. Endometrial adenocarcinoma HEC1A was used as a cancer cell line. The effect of alfacalcidol (1α-hydroxyvitamin D3) and calcitriol (1α,25-dihydroxyvitamin D3) on proliferation was demonstrated by real-time cell analysis device and migration was shown by a wound healing model. 10-11-10-9M alfacalcidol and calcitriol reduced both proliferation and migration. In contrast, the high concentration of alfacalcidol and calcitriol (10-8-10-6M) increased proliferation and migration. The proliferative effects of alfacalcidol (0-12 h) immediately started earlier than calcitriol (12-48 h). In contrast, the antiproliferative effects of calcitriol (12-24 h) have begun earlier than alfacalcidol (48-60 h). While the high concentrations of alfacalcidol and calcitriol increased the migration relatively earlier (12-24 h), low concentrations decreased the migration at late times (24-48 h). Lower concentrations of vitamin D prevent proliferation and migration in endometrial adenocarcinoma HEC1A cells. In contrast, high concentrations of vitamin D increase proliferation and migration.
Collapse
Affiliation(s)
- İbrahim Duman
- Department of Medical Pharmacology, Faculty of Medicine, University of Mersin, Mersin, Turkey
| | - R Nalan Tiftik
- Department of Medical Pharmacology, Faculty of Medicine, University of Mersin, Mersin, Turkey
| | - İsmail Ün
- Department of Medical Pharmacology, Faculty of Medicine, University of Mersin, Mersin, Turkey
| |
Collapse
|
11
|
Vitamin D and Ovarian Cancer: Systematic Review of the Literature with a Focus on Molecular Mechanisms. Cells 2020; 9:cells9020335. [PMID: 32024052 PMCID: PMC7072673 DOI: 10.3390/cells9020335] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 12/18/2022] Open
Abstract
Vitamin D is a lipid soluble vitamin involved primarily in calcium metabolism. Epidemiologic evidence indicates that lower circulating vitamin D levels are associated with a higher risk of ovarian cancer and that vitamin D supplementation is associated with decreased cancer mortality. A vast amount of research exists on the possible molecular mechanisms through which vitamin D affects cancer cell proliferation, cancer progression, angiogenesis, and inflammation. We conducted a systematic review of the literature on the effects of vitamin D on ovarian cancer cell.
Collapse
|
12
|
Hudson LG, Gillette JM, Kang H, Rivera MR, Wandinger-Ness A. Ovarian Tumor Microenvironment Signaling: Convergence on the Rac1 GTPase. Cancers (Basel) 2018; 10:cancers10100358. [PMID: 30261690 PMCID: PMC6211091 DOI: 10.3390/cancers10100358] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment for epithelial ovarian cancer is complex and rich in bioactive molecules that modulate cell-cell interactions and stimulate numerous signal transduction cascades. These signals ultimately modulate all aspects of tumor behavior including progression, metastasis and therapeutic response. Many of the signaling pathways converge on the small GTPase Ras-related C3 botulinum toxin substrate (Rac)1. In addition to regulating actin cytoskeleton remodeling necessary for tumor cell adhesion, migration and invasion, Rac1 through its downstream effectors, regulates cancer cell survival, tumor angiogenesis, phenotypic plasticity, quiescence, and resistance to therapeutics. In this review we discuss evidence for Rac1 activation within the ovarian tumor microenvironment, mechanisms of Rac1 dysregulation as they apply to ovarian cancer, and the potential benefits of targeting aberrant Rac1 activity in this disease. The potential for Rac1 contribution to extraperitoneal dissemination of ovarian cancer is addressed.
Collapse
Affiliation(s)
- Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Jennifer M Gillette
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Huining Kang
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Melanie R Rivera
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Angela Wandinger-Ness
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
13
|
Guo H, Guo J, Xie W, Yuan L, Sheng X. The role of vitamin D in ovarian cancer: epidemiology, molecular mechanism and prevention. J Ovarian Res 2018; 11:71. [PMID: 30157901 PMCID: PMC6114234 DOI: 10.1186/s13048-018-0443-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Vitamin D is a fat-soluble prohormone best known for its role in maintaining calcium homeostasis. Large numbers of epidemiological studies have shown that vitamin D plays an important role in cancer prevention by regulating cellular proliferation and metabolism. Studies of the cellular mechanism of vitamin D in ovarian cancer strongly suggest that it exhibits protective and antitumorigenic activities through genomic and nongenomic signal transduction pathways. These results indicate that vitamin D deficiency results in an increase in the risk of developing ovarian cancer and that vitamin supplements may potentially be an efficient way of preventing cancer. Consequently, this review describes the epidemiology, molecular mechanism and evidence linking vitamin D deficiency to ovarian cancer.
Collapse
Affiliation(s)
- Hui Guo
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jing Guo
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenli Xie
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.,Shandong University, Jinan, Shandong, China
| | - Lingqin Yuan
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Xiugui Sheng
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China. .,Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Guangdong, China.
| |
Collapse
|
14
|
Yuan Q, Song J, Yang W, Wang H, Huo Q, Yang J, Yu X, Liu Y, Xu C, Bao H. The effect of CA125 on metastasis of ovarian cancer: old marker new function. Oncotarget 2018. [PMID: 28637006 PMCID: PMC5564824 DOI: 10.18632/oncotarget.18388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
CA125 has been used extensively to screen for neoplasms, especially in ovarian cancer. The serum CA125 level can be used as a better prognosis evaluation and it may dynamic monitoring the disease progression. We explored the effect of CA125 on ovarian cancer cell migration and its underlying mechanism. Transwell assays showed that exposure to 0.2 μg/ml or 0.4 μg/ml CA125 for 48 h increased migration of A2780 and OVCAR-3 ovarian cancer cells. This effect of CA125 was blocked addition of 200 ng/ml DKK-1, a Wnt pathway inhibitor. Conversely, addition of CA125 reversed the inhibitory effect of Wnt inhibition in A2780 cells pretreated with DKK-1. Examination of CA125 levels in serum from 97 ovarian cancer patients revealed no relationship between a patient's age or CA125 level currently used clinically for ovarian cancer diagnosis and metastasis. However, using receiver operating characteristic (ROC) curves, we identified a new cut-off value for the serum CA125 concentration (82.9 U/ml) that is predictive of metastasis. The area under the curve is 0.632. This new cut-off value has the potential to serve as a clinically useful indicator of metastasis in ovarian cancer patients.
Collapse
Affiliation(s)
- Qin Yuan
- School of Laboratory Science, Tianjin Medical University, Tianjin, China
| | - Jiayin Song
- The Department of Clinical Laboratory, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Weiwei Yang
- The Department of Laboratory Science, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Hongyan Wang
- School of Laboratory Science, Tianjin Medical University, Tianjin, China
| | - Qianyu Huo
- School of Laboratory Science, Tianjin Medical University, Tianjin, China
| | - Jie Yang
- School of Laboratory Science, Tianjin Medical University, Tianjin, China
| | - Xiaoxu Yu
- The Department of Laboratory Science, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yunde Liu
- School of Laboratory Science, Tianjin Medical University, Tianjin, China
| | - Chen Xu
- The Department of Laboratory Science, Tianjin Fourth Central Hospital, Tianjin, China
| | - Huijing Bao
- School of Laboratory Science, Tianjin Medical University, Tianjin, China
| |
Collapse
|
15
|
Deuster E, Jeschke U, Ye Y, Mahner S, Czogalla B. Vitamin D and VDR in Gynecological Cancers-A Systematic Review. Int J Mol Sci 2017; 18:ijms18112328. [PMID: 29113037 PMCID: PMC5713297 DOI: 10.3390/ijms18112328] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/26/2022] Open
Abstract
In recent years, a vast amount of studies have centered on the role of vitamin D in the pathogenesis of certain types of cancers such as breast, colorectal and lung cancer. Increasing evidence suggests that vitamin D and its receptor play a crucial role in the development of gynecological cancers. In this review, we systematically analyzed the effect of vitamin D and the vitamin D receptor on endometrial, ovarian, cervical, vulvar and vaginal cancer. Our literature research shows that vitamin D levels and vitamin-D-related pathways affect the risk of gynecological cancers. Numerous ecological studies give evidence on the inverse relationship between UVB exposure and gynecological cancer risk. However, epidemiologic research is still inconclusive for endometrial and ovarian cancer and insufficient for rarer types of gynecological cancers. The vitamin D receptor (VDR) is upregulated in all gynecological cancers, indicating its influence on cancer etiology. The VDR polymorphism FokI (rs2228570) seems to increase the risk of ovarian cancer. Other nuclear receptors, such as the RXR, also influence gynecological cancers. Although there is limited knowledge on the role of the VDR/RXR on the survival of endometrial, cervical, vulvar or vaginal cancer patients, some studies showed that both receptors influence survival. Therefore, we suggest that further studies should focus on the vitamin D- and its hetero dimer receptor RXR in gynecological cancers.
Collapse
Affiliation(s)
- Eileen Deuster
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr 15, Munich 81377, Germany.
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr 15, Munich 81377, Germany.
| | - Yao Ye
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr 15, Munich 81377, Germany.
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr 15, Munich 81377, Germany.
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr 15, Munich 81377, Germany.
| |
Collapse
|
16
|
Quarni W, Lungchukiet P, Tse A, Wang P, Sun Y, Kasiappan R, Wu JY, Zhang X, Bai W. RIPK1 binds to vitamin D receptor and decreases vitamin D-induced growth suppression. J Steroid Biochem Mol Biol 2017; 173:157-167. [PMID: 28159673 PMCID: PMC5538941 DOI: 10.1016/j.jsbmb.2017.01.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 01/07/2017] [Accepted: 01/30/2017] [Indexed: 01/26/2023]
Abstract
Receptor interacting protein kinase 1 (RIPK1) is an enzyme acting downstream of tumor necrosis factor alpha to control cell survival and death. RIPK1 expression has been reported to cause drug resistance in cancer cells, but so far, no published studies have investigated the role of RIPK1 in vitamin D signaling. In the present study, we investigated whether RIPK1 plays any roles in 1,25-dihydroxyvitamin D3 (1,25D3)-induced growth suppression. In our studies, RIPK1 decreased the transcriptional activity of vitamin D receptor (VDR) in luciferase reporter assays independent of its kinase activity, suggesting a negative role of RIPK1 in 1,25D3 action. RIPK1 also formed a complex with VDR, and deletion analyses mapped the RIPK1 binding region to the C-terminal ligand-binding domain of the VDR. Subcellular fractionation analyses indicated that RIPK1 increased VDR retention in the cytoplasm, which may account for its inhibition of VDR transcriptional activity. Consistent with the reporter analyses, 1,25D3-induced growth suppression was more pronounced in RIPK1-null MEFs and RIPK1-knockdown ovarian cancer cells than in control cells. Our studies have defined RIPK1 as a VDR repressor, projecting RIPK1 depletion as a potential strategy to increase the potency of 1,25D3 and its analogs for cancer intervention.
Collapse
Affiliation(s)
- Waise Quarni
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States
| | - Panida Lungchukiet
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States
| | - Anfernee Tse
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States
| | - Pei Wang
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States
| | - Yuefeng Sun
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States
| | - Ravi Kasiappan
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States
| | - Jheng-Yu Wu
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, United States
| | - Xiaohong Zhang
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, United States
| | - Wenlong Bai
- The Departments of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612, United States; Oncological Sciences, University of South Florida College of Medicine, Tampa, FL 33612, United States; Programs of Cancer Biology & Evolution, H. Lee Moffitt Cancer Center, Tampa, FL 33612, United States.
| |
Collapse
|
17
|
Guo J, Cai J, Zhang Y, Zhu Y, Yang P, Wang Z. Establishment of two ovarian cancer orthotopic xenograft mouse models for in vivo imaging: A comparative study. Int J Oncol 2017; 51:1199-1208. [PMID: 28902355 DOI: 10.3892/ijo.2017.4115] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 07/12/2017] [Indexed: 11/06/2022] Open
Abstract
Orthotopic tumor animal models are optimal for preclinical research of novel therapeutic interventions. The aim of the present study was to compare two types of ovarian cancer orthotopic xenograft (OCOX) mouse models, i.e. cellular orthotopic injection (COI) and surgical orthotopic implantation (SOI), regarding xenograft formation rate, in vivo imaging, tumor growth and metastasis, and tumor microenvironment. The tumor formation and progression were monitored by bioluminescent in vivo imaging. Cell proliferation and migration abilities were detected by EdU and scratch assays, respectively. Expression of α-SMA, CD34, MMP2, MMP9, vimentin, E-cadherin and Ki67 in tumor samples were detected by immunohistochemistry. As a result, we successfully established COI- and SOI-OCOX mouse models using ovarian cancer cell lines ES2 and SKOV3. The tumor formation rate in the COI and SOI models were 87.5 and 100%, respectively. Suspected tumor cell leakage occurred in 37.5% of the COI models. The SOI xenografts grew faster, held larger primary tumors, and were more metastatic than the COI xenografts. The migration and proliferation properties of the cells that generated SOI xenografts were significantly starker than those deriving COI xenografts in vitro. The tumor cells in SOI xenografts exhibited a mesenchymal phenotype and proliferated more actively than those in the COI xenografts. Additionally, compared with the COI tumors, the SOI tumors contained more cancer associated fibroblasts, matrix metallopeptidase 2 and 9. In conclusion, SOI is a feasible and reliable technique to establish OCOX mouse models mimicking the clinical process of ovarian cancer growth and metastasis, although SOI is more technically difficult and time-consuming than COI.
Collapse
Affiliation(s)
- Jing Guo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Yunxia Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Yapei Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Ping Yang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| |
Collapse
|
18
|
Hseu YC, Tsai TJ, Korivi M, Liu JY, Chen HJ, Lin CM, Shen YC, Yang HL. Antitumor properties of Coenzyme Q 0 against human ovarian carcinoma cells via induction of ROS-mediated apoptosis and cytoprotective autophagy. Sci Rep 2017; 7:8062. [PMID: 28808311 PMCID: PMC5556069 DOI: 10.1038/s41598-017-08659-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022] Open
Abstract
Coenzyme Q0 (CoQ0, 2,3-dimethoxy-5-methyl-1,4-benzoquinone) has been reported to exert anticancer properties against human breast/lung cancer cells. This study investigated the in vitro and in vivo anticancer properties of CoQ0 on human ovarian carcinoma (SKOV-3) cells and xenografted nude mice, and revealed the underlying molecular mechanism. CoQ0 induced G2/M arrest through downregulation of cyclin B1/A and CDK1/K2 expressions. CoQ0-induced autophagy as a survival mechanism was evidenced by increased accumulation of LC3-II, GFP-LC3 puncta, AVOs formation and Beclin-1/Bcl-2 dysregulation. Increased TUNEL-positive cells and Annexin-V/PI stained cells indicated CoQ0-induced late apoptosis. Both mitochondrial (caspase-3, PARP and Bax/Bcl-2 dysregulation) and ER stress (caspase-12 and Hsp70) signals are involved in execution of apoptosis. Interestingly, CoQ0-induced apoptosis/autophagy is associated with suppression of HER-2/neu and PI3K/AKT signalling cascades. CoQ0 triggered intracellular ROS production, whereas antioxidant N-acetylcysteine prevented CoQ0-induced apoptosis, but not autophagy. Inhibition of apoptosis by Z-VAD-FMK suppressed CoQ0-induced autophagy (diminished LC3-II/AVOs), indicates CoQ0-induced apoptosis led to evoke autophagy. Contrary, inhibition of autophagy by 3-MA/CQ potentiated CoQ0-induced apoptosis (increased DNA fragmentation/PARP cleavage). Furthermore, CoQ0 treatment to SKOV-3 xenografted nude mice reduced tumor incidence and burden. Histopathological analyses confirmed that CoQ0 modulated xenografted tumor progression by apoptosis induction. Our findings emphasize that CoQ0 triggered ROS-mediated apoptosis and cytoprotective autophagy.
Collapse
Affiliation(s)
- You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan
| | - Tai-Jung Tsai
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Mallikarjuna Korivi
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Jer-Yuh Liu
- Graduate Institute of Cancer Biology, China Medical University, Taichung, 40402, Taiwan
| | - Hui-Jye Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, 402, Taiwan
| | - Chung-Ming Lin
- Department of Biotechnology, Ming Chuan University, Taoyuan, 333, Taiwan
| | - Yi-Chun Shen
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
19
|
Louka ML, Fawzy AM, Naiem AM, Elseknedy MF, Abdelhalim AE, Abdelghany MA. Vitamin D and K signaling pathways in hepatocellular carcinoma. Gene 2017; 629:108-116. [PMID: 28764978 DOI: 10.1016/j.gene.2017.07.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is a primary liver malignancy, and is now the six most common in between malignancies. Early diagnosis of HCC with prompt treatment increases the opportunity of patients to survive. With the advances in understanding the molecular biology of HCC, new therapeutic strategies to treat HCC have emerged. There is a growing consensus that vitamins are important for the control of various cancers. Biochemical evidence clearly indicates that HCC cells are responsive to the inhibitory effect of vitamin D, vitamin D analogues and vitamin K. In this review, we summarize the mechanisms used by vitamin D and K to influence the development of HCC and the latest development of vitamin analogues for potential HCC therapy.
Collapse
Affiliation(s)
- Manal L Louka
- Medical Biochemistry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Ahmed M Fawzy
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Abdelrahman M Naiem
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mustafa F Elseknedy
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Ahmed E Abdelhalim
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mohamed A Abdelghany
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| |
Collapse
|
20
|
Murray A, Madden SF, Synnott NC, Klinger R, O'Connor D, O'Donovan N, Gallagher W, Crown J, Duffy MJ. Vitamin D receptor as a target for breast cancer therapy. Endocr Relat Cancer 2017; 24:181-195. [PMID: 28213567 DOI: 10.1530/erc-16-0463] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 01/24/2023]
Abstract
Considerable epidemiological evidence suggests that high levels of circulating vitamin D (VD) are associated with a decreased incidence and increased survival from cancer, i.e., VD may possess anti-cancer properties. The aim of this investigation was therefore to investigate the anti-cancer potential of a low calcaemic vitamin D analogue, i.e., inecalcitol and compare it with the active form of vitamin D, i.e., calcitriol, in a panel of breast cancer cell lines (n = 15). Using the MTT assay, IC50 concentrations for response to calcitriol varied from 0.12 µM to >20 µM, whereas those for inecalcitol were significantly lower, ranging from 2.5 nM to 63 nM (P = 0.001). Sensitivity to calcitriol and inecalcitol was higher in VD receptor (VDR)-positive compared to VDR-negative cell lines (P = 0.0007 and 0.0080, respectively) and in ER-positive compared to ER-negative cell lines (P = 0.043 and 0.005, respectively). Using RNA-seq analysis, substantial but not complete overlap was found between genes differentially regulated by calcitriol and inecalcitol. In particular, significantly enriched gene ontology terms such as cell surface signalling and cell communication were found after treatment with inecalcitol but not with calcitriol. In contrast, ossification and bone morphogenesis were found significantly enriched after treatment with calcitriol but not with inecalcitol. Our preclinical results suggest that calcitriol and inecalcitol can inhibit breast cancer cell line growth, especially in cells expressing ER and VDR. As inecalcitol is significantly more potent than calcitriol and has low calcaemic potential, it should be further investigated for the treatment of breast cancer.
Collapse
Affiliation(s)
- Alyson Murray
- UCD School of MedicineConway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Stephen F Madden
- Division of Population Health SciencesRoyal College of Surgeons in Ireland, Dublin, Ireland
| | - Naoise C Synnott
- UCD School of MedicineConway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Rut Klinger
- UCD School of Biomolecular and Biomedical ScienceUCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Darran O'Connor
- Department of Molecular & Cellular TherapeuticsRoyal College of Surgeons Ireland, Dublin, Ireland
| | - Norma O'Donovan
- National Institute for Cellular Biotechnology (NICB)Dublin City University, Dublin, Ireland
| | - William Gallagher
- UCD School of Biomolecular and Biomedical ScienceUCD Conway Institute, University College Dublin, Dublin, Ireland
| | - John Crown
- Department of Medical OncologySt. Vincent's University Hospital, Dublin, Ireland
| | - Michael J Duffy
- UCD School of MedicineConway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- UCD Clinical Research CentreSt. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
21
|
Duffy MJ, Murray A, Synnott NC, O'Donovan N, Crown J. Vitamin D analogues: Potential use in cancer treatment. Crit Rev Oncol Hematol 2017; 112:190-197. [PMID: 28325259 DOI: 10.1016/j.critrevonc.2017.02.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/15/2016] [Accepted: 02/14/2017] [Indexed: 01/06/2023] Open
Abstract
The vitamin D receptor (VDR) is a member of the thyroid-steroid family of nuclear transcription factors. Following binding of the active form of vitamin D, i.e., 1,25(OH)2D3 (also known as calcitriol) and interaction with co-activators and co-repressors, VDR regulates the expression of several different genes. Although relatively little work has been carried out on VDR in human cancers, several epidemiological studies suggest that low circulating levels of vitamin D are associated with both an increased risk of developing specific cancer types and poor outcome in patients with specific diagnosed cancers. These associations apply especially in colorectal and breast cancer. Consistent with these findings, calcitriol as well as several of its synthetic analogues have been shown to inhibit tumor cell growth in vitro and in diverse animal model systems. Indeed, some of these vitamin D analogues with low calcemic inducing activity (e.g., EB1089, inecalcitol, paricalcitol) have progressed to clinical trials in patients with cancer. Preliminary results from these trials suggest that these vitamin D analogues have minimal toxicity, but clear evidence of efficacy remains to be shown. Although evidence of efficacy for mono-treatment with vitamin D analogues is currently lacking, several studies have reported that supplementation with calcitriol or the presence of high endogenous circulating levels of vitamin D enhances response to standard therapies.
Collapse
Affiliation(s)
- Michael J Duffy
- UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin 4, Ireland; UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| | - Alyson Murray
- UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin 4, Ireland; UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Naoise C Synnott
- UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin 4, Ireland; UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Norma O'Donovan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - John Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
22
|
Uberti F, Morsanuto V, Lattuada D, Colciaghi B, Cochis A, Bulfoni A, Colombo P, Bolis G, Molinari C. Protective effects of vitamin D3 on fimbrial cells exposed to catalytic iron damage. J Ovarian Res 2016; 9:34. [PMID: 27317433 PMCID: PMC4912710 DOI: 10.1186/s13048-016-0243-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/31/2016] [Indexed: 12/13/2022] Open
Abstract
Background Recently, vitamin D3 (1alpha, 25-dihydroxyvitamin D) has shown its capability to take part in many extraskeletal functions and its serum levels have been related to patient survival rate and malignancy of many types of neoplasms, including ovarian cancers. Catalytic iron is a free circulating form of iron that is able to generate reactive oxygen species and consequently to promote a number of cellular and tissutal dysfunctions including tumorigenesis. In fertile women an important source of catalytic iron is derived from retrograde menstruation. Epithelial secretory cells from fimbriae of fallopian tubes are greatly exposed to catalytic iron derived from menstrual reflux and so represent the site of origin for most serous ovarian cancers. The aim of this study was to assess whether vitamin D3 can play a role in counteracting catalytic iron-induced oxidative stress in cells from fimbriae of fallopian tubes. Methods The cells, isolated from women undergoing isteroannessiectomy, were treated with catalytic iron 50-75-100 mM and vitamin D3 at a concentration ranging from 0.01 to 10 nM to study cell viability, radical oxygen species production, p53, pan-Ras, Ki67 and c-Myc protein expressions through Western Blot, and immunocytochemistry or immunofluorescence analysis. Results The pre-treatment with vitamin D3 1 nM showed its beneficial effects that consists in a significant decrease in ROS production. In addition a novel finding is represented by the demonstration that pre-treatment with vitamin D3 is also able to significantly counteract tumoral biomarkers activation, such as p53, pan-Ras, Ki67 and c-Myc, and consequently the catalytic iron-induced cellular injury. Conclusions This study demonstrates for the first time that vitamin D3 plays an important role in preventing catalytic iron-dependent oxidative stress in cultured fimbrial cells. These results support the hypothesis that vitamin D3 could counteract carcinogenic changes induced by catalytic iron.
Collapse
Affiliation(s)
- Francesca Uberti
- Physiology Laboratory, Department of Translational Medicine, UPO - University of Eastern Piedmont, Via Solaroli 17, Novara, 28100, Italy.
| | - Vera Morsanuto
- Physiology Laboratory, Department of Translational Medicine, UPO - University of Eastern Piedmont, Via Solaroli 17, Novara, 28100, Italy
| | - Debora Lattuada
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Barbara Colciaghi
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Andrea Cochis
- Department of Biomedical, Surgical and Dental Sciences, Milan State University, via Beldiletto 1, Milan, 20142, Italy
| | - Alessandro Bulfoni
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Paola Colombo
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Giorgio Bolis
- Department of Obstetrics and Gynecology, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Claudio Molinari
- Physiology Laboratory, Department of Translational Medicine, UPO - University of Eastern Piedmont, Via Solaroli 17, Novara, 28100, Italy
| |
Collapse
|
23
|
Liu L, Hu Z, Zhang H, Hou Y, Zhang Z, Zhou G, Li B. Vitamin D postpones the progression of epithelial ovarian cancer induced by 7, 12-dimethylbenz [a] anthracene both in vitro and in vivo. Onco Targets Ther 2016; 9:2365-75. [PMID: 27143932 PMCID: PMC4846061 DOI: 10.2147/ott.s100581] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose Ovarian cancer is the most lethal malignancy of the female reproductive system, and the prevention and treatment of ovarian carcinoma are still far from optimal. Epidemiological studies reported that ovarian cancer risk was inversely associated with low level of 25-hydroxy vitamin D [25(OH)]. Therefore, this study focuses on exploring the chemoprevention of vitamin D on epithelial ovarian cancer induced by 7, 12-dimethylbenz [a] anthracene (DMBA). Methods The mouse ovarian surface epithelial cells were isolated from estrus mice by mild trypsinization and maintained in completed culture medium by repeated passaging. The malignant transformation of mouse ovarian surface epithelial cells was induced by DMBA in vitro. DMBA was directly injected into the bursa of mouse ovary to produce optimized in vivo ovarian cancer model. Results The results indicate that 1α,25 dihydroxyvitamin D3 may delay malignant transformation of mouse ovarian surface epithelial cells induced by DMBA and significantly decreased the colony formation rate from 18.4% to 3.2% (P<0.05). There was a negative correlation between incidence of DMBA-induced tumor and 25-hydroxy vitamin D level (R2=0.978, P<0.05). Vitamin D3 can delay the progression of ovarian cancer induced by DMBA, and the administration of vitamin D3 during the whole process worked more effectively than the administration only during tumor initiation or promotion. Moreover, we found the vitamin D3 increased the expression of E-cadherin and vitamin D receptor while it decreased the expression of β-catenin. Conclusion We succeeded in establishment of epithelial ovarian cancer models both in vitro and in vivo. The DMBA-implanted model in mice yields high incidence and specificity of epithelial derived tumors. We also found that vitamin D delays the progression of ovarian cancer. However, spontaneous epithelial ovarian carcinoma models are still to be explored for testing the preventive effects of vitamin D on epithelial ovarian cancer.
Collapse
Affiliation(s)
- Lizhi Liu
- School of Public Health, Medical College of Soochow University, Suzhou, People's Republic of China
| | - Zhiyong Hu
- Department of Chronic Disease Management, Lishui Center for Disease Control and Prevention, Lishui, People's Republic of China
| | - Hemei Zhang
- Department of Chronic Disease Management, Wenzhou Center for Disease Control and Prevention, Wenzhou, People's Republic of China
| | - Yongfeng Hou
- School of Public Health, Medical College of Soochow University, Suzhou, People's Republic of China
| | - Zengli Zhang
- Department of Labor Hygiene and Environmental Health, School of Public Health, Soochow University, Suzhou, People's Republic of China
| | - Guangming Zhou
- School of Radiation Medicine and Protection, Soochow University, Suzhou, People's Republic of China
| | - Bingyan Li
- School of Public Health, Medical College of Soochow University, Suzhou, People's Republic of China; School of Radiation Medicine and Protection, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
24
|
Abdelbaset-Ismail A, Pedziwiatr D, Suszyńska E, Sluczanowska-Glabowska S, Schneider G, Kakar SS, Ratajczak MZ. Vitamin D3 stimulates embryonic stem cells but inhibits migration and growth of ovarian cancer and teratocarcinoma cell lines. J Ovarian Res 2016; 9:26. [PMID: 27091127 PMCID: PMC4835879 DOI: 10.1186/s13048-016-0235-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022] Open
Abstract
Background Deficiency in Vitamin D3 (cholecalciferol) may predispose to some malignancies, including gonadal tumors and in experimental models vitamin D3 has been proven to inhibit the growth of cancer cells. To learn more about the potential role of vitamin D3 in cancerogenesis, we evaluated the expression and functionality of the vitamin D receptor (VDR) and its role in metastasis of ovarian cancer cells and of murine and human teratocarcinoma cell lines. Methods In our studies we employed murine embrynic stem cells (ESD3), murine (P19) and human (NTERA-2) teratocarcimona cells lines, human ovarian cancer cells (A2780) as well as purified murine and human purified very small embryonic like stem cells (VSELs). We evaluated expression of Vitamin D3 receptor (VDR) in these cells as well as effect of vitamin D3 exposure on cell proliferation and migration. Results We here provide also more evidence for the role of vitamin D3 in germline-derived malignancies, and this evidence supports the proposal that vitamin D3 treatment inhibits growth and metastatic potential of several germline-derived malignancies. We also found that the ESD3 murine immortalized embryonic stem cell line and normal, pluripotent, germline-marker-positive very small embryonic-like stem cells (VSELs) isolated from adult tissues are stimulated by vitamin D3, which suggests that vitamin D3 affects the earliest stages of embryogenesis. Conclusions We found that however all normal and malignant germ-line derived cells express functional VDR, Vitamin D3 differently affects their proliferation and migration. We postulate that while Vitamin D3 as anticancer drug inhibits proliferation of malignant cells, it may protect normal stem cells that play an important role in development and tissue/organ regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13048-016-0235-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ahmed Abdelbaset-Ismail
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Daniel Pedziwiatr
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Ewa Suszyńska
- Department of Physiology Pomeranian Medical University, Szczecin, Poland
| | | | - Gabriela Schneider
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Sham S Kakar
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA. .,Department of Regenerative Medicine Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
25
|
Targeting of free fatty acid receptor 1 in EOC: A novel strategy to restrict the adipocyte-EOC dependence. Gynecol Oncol 2016; 141:72-9. [DOI: 10.1016/j.ygyno.2016.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 02/15/2016] [Accepted: 02/21/2016] [Indexed: 12/14/2022]
|
26
|
Williams JD, Aggarwal A, Swami S, Krishnan AV, Ji L, Albertelli MA, Feldman BJ. Tumor Autonomous Effects of Vitamin D Deficiency Promote Breast Cancer Metastasis. Endocrinology 2016; 157:1341-7. [PMID: 26934299 PMCID: PMC4816742 DOI: 10.1210/en.2015-2036] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Patients with breast cancer (BCa) frequently have preexisting vitamin D deficiency (low serum 25-hydroxyvitamin D) when their cancer develops. A number of epidemiological studies show an inverse association between BCa risk and vitamin D status in humans, although some studies have failed to find an association. In addition, several studies have reported that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic indicators. However, it is unknown whether this association is mechanistically causative and, if so, whether it results from systemic or tumor autonomous effects of vitamin D signaling. We found that ablation of vitamin D receptor expression within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We show that vitamin D signaling inhibits the expression of the tumor progression gene Id1, and this pathway is abrogated in vitamin D deficiency in vivo in 2 murine models of BCa. These findings are relevant to humans, because we discovered that the mechanism of VDR regulation of Inhibitor of differentiation 1 (ID1) is conserved in human BCa cells, and there is a negative correlation between serum 25-hydroxyvitamin D levels and the level of ID1 in primary tumors from patients with BCa.
Collapse
Affiliation(s)
- Jasmaine D Williams
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| | - Abhishek Aggarwal
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| | - Srilatha Swami
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| | - Aruna V Krishnan
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| | - Lijuan Ji
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| | - Megan A Albertelli
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| | - Brian J Feldman
- Department of Pediatrics (J.D.W., A.A., A.V.K., L.J., B.J.F.), Stanford School of Medicine, Stanford, California 94305; Department of Medicine (S.S.), Stanford School of Medicine, Stanford, California 94305; Department of Comparative Medicine (M.A.A.), Stanford School of Medicine, Stanford, California 94305; and Stanford Cancer Institute (B.J.F.), Stanford School of Medicine, Stanford, California 94305
| |
Collapse
|