51
|
Abstract
Estrogens coordinate and integrate cellular metabolism and mitochondrial activities by direct and indirect mechanisms mediated by differential expression and localization of estrogen receptors (ER) in a cell-specific manner. Estrogens regulate transcription and cell signaling pathways that converge to stimulate mitochondrial function- including mitochondrial bioenergetics, mitochondrial fusion and fission, calcium homeostasis, and antioxidant defense against free radicals. Estrogens regulate nuclear gene transcription by binding and activating the classical genomic estrogen receptors α and β (ERα and ERβ) and by activating plasma membrane-associated mERα, mERβ, and G-protein coupled ER (GPER, GPER1). Localization of ERα and ERβ within mitochondria and in the mitochondrial membrane provides additional mechanisms of regulation. Here we review the mechanisms of rapid and longer-term effects of estrogens and selective ER modulators (SERMs, e.g., tamoxifen (TAM)) on mitochondrial biogenesis, morphology, and function including regulation of Nuclear Respiratory Factor-1 (NRF-1, NRF1) transcription. NRF-1 is a nuclear transcription factor that promotes transcription of mitochondrial transcription factor TFAM (mtDNA maintenance factorFA) which then regulates mtDNA-encoded genes. The nuclear effects of estrogens on gene expression directly controlling mitochondrial biogenesis, oxygen consumption, mtDNA transcription, and apoptosis are reviewed.
Collapse
|
52
|
Rifaï K, Judes G, Idrissou M, Daures M, Bignon YJ, Penault-Llorca F, Bernard-Gallon D. SIRT1-dependent epigenetic regulation of H3 and H4 histone acetylation in human breast cancer. Oncotarget 2018; 9:30661-30678. [PMID: 30093977 PMCID: PMC6078139 DOI: 10.18632/oncotarget.25771] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy in women worldwide. It is well established that the complexity of carcinogenesis involves profound epigenetic deregulations that contribute to the tumorigenesis process. Deregulated H3 and H4 acetylated histone marks are amongst those alterations. Sirtuin-1 (SIRT1) is a class-III histone deacetylase deeply involved in apoptosis, genomic stability, gene expression regulation and breast tumorigenesis. However, the underlying molecular mechanism by which SIRT1 regulates H3 and H4 acetylated marks, and consequently cancer-related gene expression in breast cancer, remains uncharacterized. In this study, we elucidated SIRT1 epigenetic role and analyzed the link between the latter and histones H3 and H4 epigenetic marks in all 5 molecular subtypes of breast cancer. Using a cohort of 135 human breast tumors and their matched normal tissues, as well as 5 human-derived cell lines, we identified H3k4ac as a new prime target of SIRT1 in breast cancer. We also uncovered an inverse correlation between SIRT1 and the 3 epigenetic marks H3k4ac, H3k9ac and H4k16ac expression patterns. We showed that SIRT1 modulates the acetylation patterns of histones H3 and H4 in breast cancer. Moreover, SIRT1 regulates its H3 acetylated targets in a subtype-specific manner. Furthermore, SIRT1 siRNA-mediated knockdown increases histone acetylation levels at 6 breast cancer-related gene promoters: AR, BRCA1, ERS1, ERS2, EZH2 and EP300. In summary, this report characterizes for the first time the epigenetic behavior of SIRT1 in human breast carcinoma. These novel findings point to a potential use of SIRT1 as an epigenetic therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Khaldoun Rifaï
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand 63001, France.,INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France
| | - Gaëlle Judes
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand 63001, France.,INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France
| | - Mouhamed Idrissou
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand 63001, France.,INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France
| | - Marine Daures
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand 63001, France.,INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France
| | - Yves-Jean Bignon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand 63001, France.,INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France
| | - Frédérique Penault-Llorca
- INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France.,Department of Biopathology, Centre Jean Perrin, Clermont-Ferrand 63011, France
| | - Dominique Bernard-Gallon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, Clermont-Ferrand 63001, France.,INSERM, UMR 1240, IMoST Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand 63005, France
| |
Collapse
|
53
|
Tan J, Liu Y, Maimaiti Y, Wang C, Yan Y, Zhou J, Ruan S, Huang T. Combination of SIRT1 and Src overexpression suggests poor prognosis in luminal breast cancer. Onco Targets Ther 2018; 11:2051-2061. [PMID: 29695913 PMCID: PMC5905521 DOI: 10.2147/ott.s162503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objectives 1) Analyze the correlation of SIRT1 and Src with human breast cancer (BC) prognosis; 2) explore the roles of SIRT1 and Src in BC cell proliferation, tumor invasion, and metastasis; and 3) analyze the correlation and interaction between SIRT1 and Src. Materials and methods 1) Tissue microarray was used to analyze the expression of SIRT1 and Src in human BC tissues and the correlation between protein expression and cancer prognosis; 2) CCK8 assay was used to determine the influence of SIRT1 and Src inhibitors on BC cell proliferation; 3) Transwell migration assay and wound healing assay were used to determine the effect of SIRT1 and Src inhibitors on BC cell migration and invasion; and 4) Western blotting was used to analyze the correlation and interaction between SIRT1 and Src. Results 1) Combination of SIRT1 and/or Src positivity is a prognosis factor in BC, especially in luminal type; 2) MCF-7 cell proliferation is suppressed by SIRT1 inhibitor Ex527, and cell migration and invasion were inhibited by Src inhibitor bosutinib; 3) combined with Ex527, bosutinib has a significantly increased effect on MCF-7 cell migration suppression; and 4) there is a positive association between SIRT1 and Src both in BC tissues and in MCF-7 cells. Conclusion 1) SIRT1 and Src overexpression are both correlated with poor prognosis in human BC; 2) SIRT1 + Src (SIRT1 and/or Src positivity) is a fine prognosis model for luminal-type BC; 3) SIRT1 is a copromotor of Src in BC migration and invasion, but not in cell proliferation; and 4) our results suggest a potential interaction or a common regulation pathway between SIRT1 and Src expression and activity.
Collapse
Affiliation(s)
- Jie Tan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuyin Liu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yusufu Maimaiti
- Department of General Surgery, Research Institute of Minimally Invasive, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Changwen Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengnan Ruan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
54
|
Sweeney MF, Sonnenschein C, Soto AM. Characterization of MCF-12A cell phenotype, response to estrogens, and growth in 3D. Cancer Cell Int 2018; 18:43. [PMID: 29559854 PMCID: PMC5859508 DOI: 10.1186/s12935-018-0534-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/05/2018] [Indexed: 01/28/2023] Open
Abstract
Background Three-dimensional cultures of mammary epithelial cells allow for biologically-relevant studies of the development of the mammary gland in rodents and humans under normal and pathological conditions, like carcinogenesis. Under these conditions, mammotropic hormones play significant roles in tissue morphogenesis. Therefore, a system that recreates the normal, hormonally responsive epithelium would be a valuable tool to study the normal state and its transition to carcinogenesis. MCF-12A cells have been claimed to be non-tumorigenic mammary epithelial cells with reported sensitivity to estrogens. In this study, we aimed at characterizing MCF-12A cells for use in a hormone-responsive 3D culture system to determine their usefulness as a tool to identify normal and abnormal microenvironmental cues. Methods MCF-12A cells were single-cell cloned in order to investigate their heterogeneous makeup. The parental cells were then treated with estradiol to investigate proliferative and transcriptional responses through the estrogen receptor alpha. Finally, parental cells and epithelial-like cell-derived clones were seeded in rat-tail collagen I to profile the morphogenesis of multicellular 3D structures. The resultant structures were then analyzed using unsupervised morphometric analysis. Results MCF-12A cells consist of epithelial-like colonies which shed elongated, freely growing cells on the colony's edges. The cells express E-cadherin as well as mesenchymal vimentin but do not express markers associated with myoepithelial cells or fibroblasts. Treatment with estradiol does not affect either the proliferation rate or the induction of gene expression in MCF-12A cells. Parental MCF-12A cells form acini, solid spheres and elongated branching ducts when grown in rat-tail collagen type I matrix, the geometries and distribution of which are altered following the removal of fibroblast-like cells. Conclusions MCF-12A cells are a heterogeneous pseudo-epithelial cell line capable of forming a variety of multicellular structures in 3D culture. We found no indication that the cells display estrogen-responsive characteristics, thus refuting previous studies which reported estrogen responsiveness. We report that MCF-12A cells are not suited for use in studies in which differential behaviors of "normal" and "cancerous" estrogen-responsive cells are to be compared.
Collapse
Affiliation(s)
- Michael F Sweeney
- 1Sackler School of Graduate Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Carlos Sonnenschein
- 2Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Ana M Soto
- 2Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| |
Collapse
|
55
|
Li T, Ma J, Han X, Jia Y, Yuan H, Shui S, Guo D. MicroRNA-320 Enhances Radiosensitivity of Glioma Through Down-Regulation of Sirtuin Type 1 by Directly Targeting Forkhead Box Protein M1. Transl Oncol 2018; 11:205-212. [PMID: 29331678 PMCID: PMC5772006 DOI: 10.1016/j.tranon.2017.12.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/15/2017] [Accepted: 12/15/2017] [Indexed: 12/19/2022] Open
Abstract
Glioma is the most common cancer in human brain system and seriously threatens human health. miRNA-320 has been demonstrated to be closely correlated with the development of glioma. However, its effect and molecular mechanism underlying radioresistance have not been fully elucidated in glioma. Here, RT-qPCR assay was used to assess the expressions of miR-320 and forkhead box protein M1 (FoxM1) mRNA in glioma tumor tissues and cells. The effects of miR-320, FoxM1 and sirtuin type 1 (Sirt1) on radiosensitivity in glioma cells were evaluated by clone formation assay, apoptosis assay, histone H2AX phosphorylation level (γH2AX) detection and caspase 3 activity analysis, respectively. The direct interaction between miR-320 and FoxM1 was detected by luciferase assay. The protein levels of FoxM1, Sirt1 and γH2AX were measured by western blot assay. We found that miR-320 expression was down-regulated and FoxM1 expression was up-regulated in radioresistant glioma tissues and IR-treated glioma cells. miR-320 overexpression dramatically enhanced radiosensitivity, promoted apoptosis, and improved γH2AX expression and caspase 3 activity in glioma cells. Luciferase reporter assay and western blot assay further validated that miR-320 suppressed FoxM1 expression by directly targeting 3' UTR region of FoxM1. Moreover, miR-320 inhibited Sirt1 expression via targeting FoxM1 in glioma cells. Furthermore, overexpression of FoxM1 and Sirt1 strikingly attenuated miR-320-induced increase of radiosensitivity, apoptosis and γH2AX expression in glioma cells. In conclusion, miR-320 enhanced radiosensitivity of glioma cells through down-regulation of Sirt1 by directly targeting FoxM1.
Collapse
Affiliation(s)
- Tengfei Li
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China; Interventional Institute of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Ji Ma
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China; Interventional Institute of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Xinwei Han
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China; Interventional Institute of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yongxu Jia
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Huifeng Yuan
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China; Interventional Institute of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Shaofeng Shui
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China; Interventional Institute of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Dong Guo
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China; Interventional Institute of Zhengzhou University, Zhengzhou, 450052, PR China
| |
Collapse
|
56
|
Liarte S, Alonso-Romero JL, Nicolás FJ. SIRT1 and Estrogen Signaling Cooperation for Breast Cancer Onset and Progression. Front Endocrinol (Lausanne) 2018; 9:552. [PMID: 30319540 PMCID: PMC6170604 DOI: 10.3389/fendo.2018.00552] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/30/2018] [Indexed: 01/10/2023] Open
Abstract
Breast cancer remains a significant female mortality cause. It constitutes a multifactorial disease for which research on environmental factors offers little help in predicting onset or progression. The pursuit for its foundations by analyzing hormonal changes as a motive for disease development, indicates that increased exposure to estrogens associates with increased risk. A prevalent number of breast cancer cases show dependence on the increased activity of the classic nuclear estrogen receptor (ER) for cell proliferation and survival. SIRT1 is a Type III histone deacetylase which is receiving increasing attention due to its ability to perform activities over relevant non-histone proteins and transcription factors. Interestingly, concomitant SIRT1 overexpression is commonly found in ER-positive breast cancer cases. Both proteins had been shown to directly interact, in a process related to altered intracellular signaling and aberrant transcription, then promoting tumor progression. Moreover, SIRT1 activities had been also linked to estrogenic effects through interaction with the G-protein coupled membrane bound estrogen receptor (GPER). This work aims to summarize present knowledge on the interplay between SIRT1 and ER/GPER for breast cancer onset and progression. Lastly, evidences on the ability of SIRT1 to interact with TGFß signaling, a concurrent pathway significantly involved in breast cancer progression, are reported. The potential of this research field for the development of innovative strategies in the assessment of orphan breast cancer subtypes, such as triple negative breast cancer (TNBC), is discussed.
Collapse
Affiliation(s)
- Sergio Liarte
- Laboratorio de Oncología Molecular y TGFβ, Instituto Murciano de Investigaciones Biosanitarias Arrixaca, Murcia, Spain
- *Correspondence: Sergio Liarte
| | | | - Francisco José Nicolás
- Laboratorio de Oncología Molecular y TGFβ, Instituto Murciano de Investigaciones Biosanitarias Arrixaca, Murcia, Spain
- Francisco José Nicolás
| |
Collapse
|
57
|
Rifaï K, Judes G, Idrissou M, Daures M, Bignon YJ, Penault-Llorca F, Bernard-Gallon D. Dual SIRT1 expression patterns strongly suggests its bivalent role in human breast cancer. Oncotarget 2017; 8:110922-110930. [PMID: 29340027 PMCID: PMC5762295 DOI: 10.18632/oncotarget.23006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 11/13/2017] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common cancer in women, and the leading cause of cancer death in women worldwide. SIRT1 (silent mating type information regulation 2 homolog) 1 is a class-III histone deacetylase involved in apoptosis regulation, DNA repair and tumorigenesis. However, its role in breast carcinoma remains controversial, as both tumor-suppressive and tumor-promoting functions have been reported. Also, there are very few reports available where expression of SIRT1 is comprehensively analyzed in breast tumors classified by molecular subtype. Here, using a cohort of 50 human breast tumors and their matched normal tissues, we investigated SIRT1 expression levels in the 5 molecular subtypes of breast cancer according to the St Gallen classification (2013). Tumors and their corresponding normal tissue samples were collected from all patients, and SIRT1 mRNA and protein expression levels were then examined by real-time quantitative polymerase chain reaction and immunoblotting, respectively. After statistical analysis, the results showed a dual expression profile of SIRT1 in human breast carcinoma, with significant overexpression in luminal and HER2-enriched subtypes and significantly reduced expression in the triple-negative subtype. We also found an inverse correlation between SIRT1 expression and breast cancer aggressivity. These novel findings suggest that SIRT1 plays a dual role in breast tumors depending on its expression rate and the molecular subtype of the cancer. Our data also point to a potential role for SIRT1 as a prognostic biomarker in breast cancer.
Collapse
Affiliation(s)
- Khaldoun Rifaï
- Centre Jean Perrin, Department of Oncogenetics-CBRV, 63001 Clermont-Ferrand, France.,INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France
| | - Gaëlle Judes
- Centre Jean Perrin, Department of Oncogenetics-CBRV, 63001 Clermont-Ferrand, France.,INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France
| | - Mouhamed Idrissou
- Centre Jean Perrin, Department of Oncogenetics-CBRV, 63001 Clermont-Ferrand, France.,INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France
| | - Marine Daures
- Centre Jean Perrin, Department of Oncogenetics-CBRV, 63001 Clermont-Ferrand, France.,INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France
| | - Yves-Jean Bignon
- Centre Jean Perrin, Department of Oncogenetics-CBRV, 63001 Clermont-Ferrand, France.,INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France.,Centre Jean Perrin, Department of Biopathology, 63011 Clermont-Ferrand, France
| | - Dominique Bernard-Gallon
- Centre Jean Perrin, Department of Oncogenetics-CBRV, 63001 Clermont-Ferrand, France.,INSERM U 1240-IMoST, 63005 Clermont-Ferrand, France
| |
Collapse
|
58
|
Behrouzfar K, Alaee M, Nourbakhsh M, Gholinejad Z, Golestani A. Extracellular NAMPT/visfatin causes p53 deacetylation via NAD production and SIRT1 activation in breast cancer cells. Cell Biochem Funct 2017; 35:327-333. [PMID: 28845527 DOI: 10.1002/cbf.3279] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/09/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023]
Abstract
Visfatin, which is secreted as an adipokine and cytokine, has been implicated in cancer development and progression. In this study, we investigated the NAD-producing ability of visfatin and its relationship with SIRT1 (silent information regulator 2) and p53 to clarify the role of visfatin in breast cancer. MCF-7 breast cancer cells were cultured and treated with visfatin. SIRT1 activity was assessed by measuring fluorescence intensity from fluoro-substrate peptide. To investigate the effect of visfatin on p53 acetylation, SDS-PAGE followed by western blotting was performed using specific antibodies against p53 and its acetylated form. Total NAD was measured both in cell lysate and the extracellular medium by colorimetric method. Visfatin increased both extracellular and intracellular NAD concentrations. It also induced proliferation of breast cancer cells, an effect that was abolished by inhibition of its enzymatic activity. Visfatin significantly increased SIRT1 activity, accompanied by induction of p53 deacetylation. In conclusion, the results show that extracellular visfatin produces NAD that causes upregulation of SIRT1 activity and p53 deacetylation. These findings explain the relationship between visfatin and breast cancer progression.
Collapse
Affiliation(s)
- Kiarash Behrouzfar
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Alaee
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zafar Gholinejad
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
59
|
Jang J, Huh YJ, Cho HJ, Lee B, Park J, Hwang DY, Kim DW. SIRT1 Enhances the Survival of Human Embryonic Stem Cells by Promoting DNA Repair. Stem Cell Reports 2017; 9:629-641. [PMID: 28689995 PMCID: PMC5549766 DOI: 10.1016/j.stemcr.2017.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 12/11/2022] Open
Abstract
Human embryonic stem cells (hESCs) hold great promise for the treatment of many incurable diseases. Sirtuin1 (SIRT1), a class III histone deacetylase, is abundantly expressed in hESCs and is known to regulate early differentiation and telomere elongation. Here, we show that downregulation of SIRT1 promotes cell death in hESCs, but not in differentiated cells, and the SIRT1-inhibition-mediated cell death is preceded by increased DNA damage. This increased DNA damage is at least partially due to decreased levels of DNA repair enzymes such as MSH2, MSH6, and APEX1. Furthermore, SIRT1 inhibition causes p53 activation, which eventually leads to DNA damage-induced apoptosis of hESCs. This study provides valuable insights into the mechanism of SIRT1-mediated hESC survival and should contribute to the development of safe and effective cell therapies. SIRT1 downregulation induces cell death in hESCs but not in differentiated cells SIRT1 inhibition increases DNA damage by reducing the level of DNA repair enzymes SIRT1 inhibition activates p53 and induces two pro-apoptotic genes, PUMA and BAX SIRT1 acts as a guardian of hESCs via genome stability
Collapse
Affiliation(s)
- Jiho Jang
- Department of Physiology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yong Jun Huh
- Department of Physiology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyun-Ju Cho
- Department of Physiology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Boram Lee
- Department of Biomedical Science, CHA University, Seongnam, Kyeonggido 13488, Korea
| | - Jaepil Park
- Department of Physiology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Dong-Youn Hwang
- Department of Biomedical Science, CHA University, Seongnam, Kyeonggido 13488, Korea.
| | - Dong-Wook Kim
- Department of Physiology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea.
| |
Collapse
|
60
|
Mvunta DH, Miyamoto T, Asaka R, Yamada Y, Ando H, Higuchi S, Ida K, Kashima H, Shiozawa T. SIRT1 Regulates the Chemoresistance and Invasiveness of Ovarian Carcinoma Cells. Transl Oncol 2017; 10:621-631. [PMID: 28667895 PMCID: PMC5491457 DOI: 10.1016/j.tranon.2017.05.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND SIRT1 is a longevity gene that forestalls aging and age-related diseases including cancer, and has recently attracted widespread attention due to its overexpression in some cancers. We previously identified the overexpression of SIRT1 in ovarian carcinoma (OvCa) as a poor prognostic factor. However, mechanistic insights into the function of SIRT1 in OvCa have yet to be elucidated. METHODS Quantitative real-time reverse PCR (qRT-PCR) and Western blotting were employed to examine the expression of SIRT1 in a panel of human OvCa cell lines. si-RNA or sh-RNA and cDNA technologies were utilized to knockdown or overexpress SIRT1, respectively. The effects of SIRT1 on proliferation and chemoresistance were examined using a WST-1 assay, and the underlying mechanisms were confirmed using an apoptotic assay, and the quantification of glutathione (GSH), and reactive oxygen species (ROS). The aggressiveness of SIRT1 was analyzed using in vitro invasion and migration assays. RESULTS SIRT1 was more strongly expressed in OvCa cell lines than in the immortalized ovarian epithelium at the gene and protein levels. Stress up-regulated the expression of SIRT1 in dose- and time-dependent manners. SIRT1 significantly enhanced the proliferation (P<.05), chemoresistance (P<.05), and aggressiveness of OvCa cells by up-regulating multiple antioxidant pathways to inhibit oxidative stress. Further study into the overexpression of SIRT1 demonstrated the up-regulation of several stemness-associated genes and enrichment of CD44v9 via an as-yet-unidentified pathway. CONCLUSIONS Our results suggest that SIRT1 plays a role in the acquisition of aggressiveness and chemoresistance by OvCa, and has potential as a therapeutic target for OvCa.
Collapse
Affiliation(s)
- David Hamisi Mvunta
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Tsutomu Miyamoto
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan.
| | - Ryoichi Asaka
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Yasushi Yamada
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Hirofumi Ando
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Shotaro Higuchi
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Koichi Ida
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Hiroyasu Kashima
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Tanri Shiozawa
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| |
Collapse
|
61
|
Zou Q, Tang Q, Pan Y, Wang X, Dong X, Liang Z, Huang D. MicroRNA-22 inhibits cell growth and metastasis in breast cancer via targeting of SIRT1. Exp Ther Med 2017; 14:1009-1016. [PMID: 28781618 PMCID: PMC5526179 DOI: 10.3892/etm.2017.4590] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 04/18/2017] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRs), which are a class of small non-coding RNAs, are key regulators of gene expression via induction of translational repression or mRNA degradation. However, the molecular mechanism of miR-22 underlying the malignant progression of breast cancer, remains to be elucidated. The present study aimed to explore the regulatory mechanism of miR-22 in breast cancer cell growth and metastasis. Reverse transcription-quantitative polymerase chain reaction data revealed that miR-22 was significantly downregulated in breast cancer tissues, compared with adjacent non-tumor tissues. Furthermore, the miR-22 levels were further decreased in stage III–IV, compared with stage I–II breast cancer. In addition, low miR-22 levels were significantly associated with the poor differentiation, metastasis and advanced clinical stages of breast cancer. Sirtuin1 (SIRT1) was demonstrated to act as a direct target gene of miR-22 and its protein expression negatively regulated by miR-22 in the MCF-7 breast cancer cell line. Furthermore, SIRT1 expression levels were significantly upregulated in breast cancer tissues, compared with adjacent non-tumor tissues. SIRT1 levels were observed to be increased in stage III–IV when compared with stage I–II breast cancer. miR-22 overexpression decreased the proliferation, migration and invasion of MCF-7 cells, whereas overexpression of SIRT1 eliminated the suppressive effects of the miR-22 overexpression on the malignant phenotype of MCF-7 cells. The results of the present study therefore suggested that miR-22 demonstrated suppressive effects on breast cancer growth and metastasis via targeting SIRT1, and thus the miR-22/SIRT1 axis may be used as a novel and potential therapeutic target for breast cancer in the future.
Collapse
Affiliation(s)
- Quanqing Zou
- Department of Breast Surgery, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China.,Department of Hepatobiliary and Endocrine Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Qianli Tang
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yinhua Pan
- Department of Hepatobiliary and Endocrine Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Xuedi Wang
- Department of Hepatobiliary and Endocrine Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Xiaofeng Dong
- Department of Hepatobiliary and Endocrine Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Zhongxiao Liang
- Department of Hepatobiliary and Endocrine Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Dong Huang
- Department of Hepatobiliary and Endocrine Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
62
|
Jin J, Chu Z, Ma P, Meng Y, Yang Y. SIRT1 promotes the proliferation and metastasis of human pancreatic cancer cells. Tumour Biol 2017; 39:1010428317691180. [PMID: 28351313 DOI: 10.1177/1010428317691180] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
SIRT1 plays an important role in human malignant progression, inducing cancer cell proliferation and metastasis by regulating downstream gene expressions. However, little is known about the underlying mechanisms in which SIRT1 promotes pancreatic cancer tumorigenesis. The aim of this study is to investigate the SIRT1 expression levels and biological functions in promoting pancreatic cancer progression. We first investigated the expression of SIRT1 in a series of pancreatic cancer tissues as well as in a panel of pancreatic cancer cell lines. The effect of SIRT1 on cell activity was explored by knockdown experiments. Cell growth was measured using the MTT assay and colony-formation assay. Migration and invasion were tested using transwell assay. Our results showed that the expression of SIRT1 was significantly up-regulated both in pancreatic cancer tissues and cell lines. Knockdown of SIRT1 suppressed cell proliferation and migration of pancreatic cancer cells. This is the first report to disclose the role of SIRT1 in regulation of pancreatic cancer cell proliferation and migration, which may provide a potential therapeutic target for pancreatic cancer patients.
Collapse
Affiliation(s)
- Jianguang Jin
- Department of General Surgery, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Zhijie Chu
- Department of General Surgery, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Pengfei Ma
- Department of General Surgery, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Yuanpu Meng
- Department of General Surgery, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Yanhui Yang
- Department of General Surgery, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
63
|
Wu W, Zhang L, Lin J, Huang H, Shi B, Lin X, Huang Z, Wang C, Qiu J, Wei X. Hypermethylation of the HIC1 promoter and aberrant expression of HIC1/SIRT1 contribute to the development of thyroid papillary carcinoma. Oncotarget 2016; 7:84416-84427. [PMID: 27793057 PMCID: PMC5356670 DOI: 10.18632/oncotarget.12936] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 10/21/2016] [Indexed: 02/07/2023] Open
Abstract
Hypermethylation leading to the loss of hypermethylated in cancer-1 (HIC1) gene expression occurs in many different types of human cancer. HIC1 is a transcriptional repressor that directly binds to the promoter region of NAD-dependent deacetylase sirtuin-1 (SIRT1). SIRT1 functions in cell growth, is anti-apoptotic, protect neurons, functions in senescence, and regulates energy restriction. Epigenetic modification and dysregulation affecting the HIC1/SIRT1 axis is potentially important for the development of malignancies. However, the importance of HIC1 expression in the development of papillary thyroid carcinoma, especially in Chinese patients, is uncertain. Therefore, we assessed the level of methylation in the HIC1 promoter and the mRNA and protein expression levels of HIC1 and SIRT1 in human thyroid papillary carcinoma and tumor adjacent control tissues. The demethylation reagent 5-aza-2'-deoxyctidine (5-aza-dc) and an HIC1 overexpression plasmid were used to manipulate the HIC1/SIRT1 pathway, and the effects on cell senescence, apoptosis, and cell cycle progression were assessed. Compared to normal thyroid tissue, thyroid tumors had lower expression of HIC1 and higher SIRT1 expression. The level of HIC1 methylation was also higher in thyroid carcinoma tissues than adjacent tissues. HIC1 expression was closely correlated with patient age and tumor progression. Restoration of HIC1 expression through an overexpression plasmid or 5-aza-dC treatment reduced SIRT1 expression and cell proliferation, and led to senescence, cell cycle arrest, and apoptosis. Aberrant expression of HIC1/SIRT1 and hypermethylation of the HIC1 promoter may be critical for the development and progression of papillary thyroid cancer.
Collapse
Affiliation(s)
- Wenyi Wu
- Department of general surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Liting Zhang
- Endocrine Department, The 180th Military Hospital of Chinese Peoples, Liberation Army, Quanzhou, Fujian, China
| | - Jianqing Lin
- Department of general surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hanwei Huang
- Endocrine Department, Affiliated Zhongshan Hospital of Guangdong Medical College, Zhongshan, Guangdong, China
| | - Bai Shi
- Department of general surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xingong Lin
- Department of general surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhongxin Huang
- Department of general surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Chaoyang Wang
- Department of general surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jianlong Qiu
- Department of Pathology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiaolong Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
64
|
Gharabaghi MA. Diagnostic investigation of BIRC6 and SIRT1 protein expression level as potential prognostic biomarkers in patients with non-small cell lung cancer. CLINICAL RESPIRATORY JOURNAL 2016; 12:633-638. [PMID: 27768839 DOI: 10.1111/crj.12572] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/08/2016] [Accepted: 09/28/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIM Lung cancer is the major contributor to overall cancer-related mortality. Biomarkers are important in early detection and prognosis, in addition to developing treatment regimes, which may improve the patient survival rates. Therefore, the present study was designed to evaluate the prognostic and diagnostic value of SIRT1/BIRC6 expression in non-small cell lung cancer (NSCLC). METHODS The data on the prognostic impact of SIRT1/BIRC6 in NSCLC were collected from September 11, 2006 to July 10, 2014. Immunoexpressions and real-time quantitative reverse transcription-PCR of SIRT1/BIRC6 were analyzed and the outcomes were correlated with clinicopathological features and patient survivals. RESULTS MRNA level of SIRT1 was upregulated in NSCLC tissues as compared to normal tissues (3.18 ± 0.77 vs. 1.27 ± 0.62; P = .001). BIRC6 mRNA was upregulated in cancer tissues when compared with normal tissues (4.13 ± 0.91 vs. 1.51 ± 0.72; P = .001). SIRT1 protein was overexpressed in 27 patients (67.5%), while normal tissues showed weak or negative staining to SIRT1 (P = .002). Furthermore, these findings suggested that advanced pathological T stage, and poor differentiation were significantly associated with expression of SIRT1 (P < .05). Increased expression of BIRC6 was detected in 75% of patients, while weak or negative expression were detected in normal tissues (P = .001). Furthermore, increased expression of BIRC6 was linked to advanced pathological T stage, poor differentiation, and lymph node metastasis (P < .05). CONCLUSIONS SIRT1 and BIRC6 may be linked to tumor progression and could be useful for the treatment of NSCLC.
Collapse
Affiliation(s)
- Mehrnaz Asadi Gharabaghi
- Advanced Thoracic Research Center, Department of Pulmonary Diseases, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
65
|
Abstract
Protein homeostasis networks are highly regulated systems responsible for maintaining the health and productivity of cells. Whereas therapeutics have been developed to disrupt protein homeostasis, more recently identified techniques have been used to repurpose homeostatic networks to effect degradation of disease-relevant proteins. Here, we review recent advances in the use of small molecules to degrade proteins in a selective manner. First, we highlight all-small-molecule techniques with direct clinical application. Second, we describe techniques that may find broader acceptance in the biomedical research community that require little or no synthetic chemistry. In addition to serving as innovative research tools, these new approaches to control intracellular protein levels offer the potential to develop novel therapeutics targeting proteins that are not currently pharmaceutically vulnerable.
Collapse
Affiliation(s)
- Daniel P Bondeson
- Department of Molecular, Cellular, and Developmental Biology, Department of Chemistry, and Department of Pharmacology, Yale University, New Haven, Connecticut 06511;
| | - Craig M Crews
- Department of Molecular, Cellular, and Developmental Biology, Department of Chemistry, and Department of Pharmacology, Yale University, New Haven, Connecticut 06511;
| |
Collapse
|
66
|
Pruitt K. Molecular and Cellular Changes During Cancer Progression Resulting From Genetic and Epigenetic Alterations. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:3-47. [PMID: 27865461 DOI: 10.1016/bs.pmbts.2016.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumorigenesis is a complex process that involves a persistent dismantling of cellular safeguards and checkpoints. These molecular and cellular changes that accumulate over months or decades lead to a change in the fundamental identity of a cell as it transitions from normal to malignant. In this chapter, we will examine some of the molecular changes in the evolving relationship between the genome and epigenome and highlight some of the key changes that occur as normal cells progress to tumor cells. For many years tumorigenesis was almost exclusively attributed to mutations in protein-coding genes. This notion that mutations in protein-coding genes were a fundamental driver of tumorigenesis enabled the development of several novel therapeutics that targeted the mutant protein or overactive pathway responsible for driving a significant portion of the tumor growth. However, because many therapeutic challenges remained in the face of these advances, it was clear that other pieces to the puzzle had yet to be discovered. Advances in molecular and genomics techniques continued and the study of epigenetics began to expand and helped reshape the view that drivers of tumorigenesis extended beyond mutations in protein-coding genes. Studies in the field of epigenetics began to identify aberrant epigenetic marks which created altered chromatin structures and enabled protein expression in tissues that defied rules governing tissue-specificity. Not only were epigenetic alterations found to enable overexpression of proto-oncogenes, they also led to the silencing of tumor suppressor genes. With these discoveries, it became clear that tumor growth could be stimulated by much more than mutations in protein-coding genes. In fact, it became increasingly clear that much of the human genome, while transcribed, did not lead to proteins. This discovery further led to studies that began to uncover the role of noncoding RNAs in regulating chromatin structure, gene transcription, and tumor biology. In this chapter, some of the key alterations in the genome and epigenome will be explored, and some of the cancer therapies that were developed as a result of these discoveries will be discussed.
Collapse
Affiliation(s)
- K Pruitt
- Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
67
|
Fang M, Guo WR, Park Y, Kang HG, Zarbl H. Enhancement of NAD⁺-dependent SIRT1 deacetylase activity by methylselenocysteine resets the circadian clock in carcinogen-treated mammary epithelial cells. Oncotarget 2016; 6:42879-91. [PMID: 26544624 PMCID: PMC4767478 DOI: 10.18632/oncotarget.6002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/13/2015] [Indexed: 12/29/2022] Open
Abstract
We previously reported that dietary methylselenocysteine (MSC) inhibits N-methyl-N-nitrosourea (NMU)-induced mammary tumorigenesis by resetting circadian gene expression disrupted by the carcinogen at the early stage of tumorigenesis. To investigate the underlying mechanism, we developed a circadian reporter system comprised of human mammary epithelial cells with a luciferase reporter driven by the promoter of human PERIOD 2 (PER2), a core circadian gene. In this in vitro model, NMU disrupted cellular circadian rhythm in a pattern similar to that observed with SIRT1-specific inhibitors; in contrast, MSC restored the circadian rhythms disrupted by NMU and protected against SIRT1 inhibitors. Moreover, NMU inhibited intracellular NAD+/NADH ratio and reduced NAD+-dependent SIRT1 activity in a dose-dependent manner, while MSC restored NAD+/NADH and SIRT1 activity in the NMU-treated cells, indicating that the NAD+-SIRT1 pathway was targeted by NMU and MSC. In rat mammary tissue, a carcinogenic dose of NMU also disrupted NAD+/NADH oscillations and decreased SIRT1 activity; dietary MSC restored NAD+/NADH oscillations and increased SIRT1 activity in the mammary glands of NMU-treated rats. MSC-induced SIRT1 activity was correlated with decreased acetylation of BMAL1 and increased acetylation of histone 3 lysine 9 at the Per2 promoter E-Box in mammary tissue. Changes in SIRT1 activity were temporally correlated with loss or restoration of rhythmic Per2 mRNA expression in NMU-treated or MSC-rescued rat mammary glands, respectively. Together with our previous findings, these results suggest that enhancement of NAD+-dependent SIRT1 activity contributes to the chemopreventive efficacy of MSC by restoring epigenetic regulation of circadian gene expression at early stages of mammary tumorigenesis.
Collapse
Affiliation(s)
- Mingzhu Fang
- Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,NIEHS Center for Environmental Exposures and Disease, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Wei-Ren Guo
- Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Youngil Park
- Veterinary Drugs & Biologics Division, Animal and Plant Quarantine Agency, Anyang 430-757, Republic of Korea
| | - Hwan-Goo Kang
- Veterinary Drugs & Biologics Division, Animal and Plant Quarantine Agency, Anyang 430-757, Republic of Korea
| | - Helmut Zarbl
- Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,NIEHS Center for Environmental Exposures and Disease, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
68
|
Li K, Ying M, Feng D, Du J, Chen S, Dan B, Wang C, Wang Y. Brachyury promotes tamoxifen resistance in breast cancer by targeting SIRT1. Biomed Pharmacother 2016; 84:28-33. [PMID: 27621036 DOI: 10.1016/j.biopha.2016.09.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/21/2016] [Accepted: 09/06/2016] [Indexed: 12/01/2022] Open
Abstract
Tamoxifen is effective for treating estrogen receptor-alpha (ERα)-positive breast cancers. However, few molecular mediators of tamoxifen resistance have been elucidated. In the present study, we determine the underlying roles of Brachyury in tamoxifen resistance. Loss- and gain-of-function assay are utilized to confirm the oncogenic roles of Brachyury in breast cancer. Compared with the normal MCF10A cells, Brachyury is commonly overexpressed in breast cancer cell lines. Knockdown of Brachyury inhibits tamoxifen resistance, whereas overexpression of Brachyury enhances tamoxifen resistance as demonstrated increased cell viability and reduced cell apoptosis. Mechanistically, we demonstrate for the first time that Brachyury mediates tamoxifen resistance by regulating Sirtuin-1 (SIRT1). Collectively, our data, as a proof of principle, indicate that Brachyury is a candidate marker for predicting the clinical efficacy of tamoxifen and targeting SIRT1 could overcome resistance to tamoxifen in breast cancer cells.
Collapse
Affiliation(s)
- Kaichun Li
- Department of Oncology, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, PR China
| | - Mingzhen Ying
- Department of Oncology, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, PR China
| | - Dan Feng
- Department of Oncology, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, PR China
| | - Jie Du
- Department of Oncology, Tianyou Hospital Affiliated to Tongji University, Shanghai 200331, PR China
| | - Shiyu Chen
- Department of Oncology, Tianyou Hospital Affiliated to Tongji University, Shanghai 200331, PR China
| | - Bing Dan
- Department of Oncology, Tianyou Hospital Affiliated to Tongji University, Shanghai 200331, PR China
| | - Cuihua Wang
- Department of Oncology, Tianyou Hospital Affiliated to Tongji University, Shanghai 200331, PR China
| | - Yajie Wang
- Department of Oncology, Changhai Hospital Affiliated to The Second Military Medical University, Shanghai 200433, PR China.
| |
Collapse
|
69
|
Lee WY, Lee WT, Cheng CH, Chen KC, Chou CM, Chung CH, Sun MS, Cheng HW, Ho MN, Lin CW. Repositioning antipsychotic chlorpromazine for treating colorectal cancer by inhibiting sirtuin 1. Oncotarget 2016; 6:27580-95. [PMID: 26363315 PMCID: PMC4695010 DOI: 10.18632/oncotarget.4768] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/24/2015] [Indexed: 02/06/2023] Open
Abstract
Investigating existing drugs for repositioning can enable overcoming bottlenecks in the drug development process. Here, we investigated the effect and molecular mechanism of the antipsychotic drug chlorpromazine (CPZ) and identified its potential for treating colorectal cancer (CRC). Human CRC cell lines harboring different p53 statuses were used to investigate the inhibitory mechanism of CPZ. CPZ effectively inhibited tumor growth and induced apoptosis in CRC cells in a p53-dependent manner. Activation of c-jun N-terminal kinase (JNK) was crucial for CPZ-induced p53 expression and the subsequent induction of tumor apoptosis. Induction of p53 acetylation at lysine382 was involved in CPZ-mediated tumor apoptosis, and this induction was attenuated by sirtuin 1 (SIRT1), a class III histone deacetylase. By contrast, knocking down SIRT1 sensitized tumor cells to CPZ treatment. Moreover, CPZ induced the degradation of SIRT1 protein participating downstream of JNK, and JNK suppression abrogated CPZ-mediated SIRT1 downregulation. Clinical analysis revealed a significant association between high SIRT1 expression and poor outcome in CRC patients. These data suggest that SIRT1 is an attractive therapeutic target for CRC and that CPZ is a potential repositioned drug for treating CRC.
Collapse
Affiliation(s)
- Wen-Ying Lee
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wai-Theng Lee
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ku-Chung Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ming Chou
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chu-Hung Chung
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Min-Siou Sun
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Wei Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Ni Ho
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
70
|
Zhang L, Yuan Y, Lu KH, Zhang L. Identification of recurrent focal copy number variations and their putative targeted driver genes in ovarian cancer. BMC Bioinformatics 2016; 17:222. [PMID: 27230211 PMCID: PMC4881176 DOI: 10.1186/s12859-016-1085-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/14/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Genomic regions with recurrent DNA copy number variations (CNVs) are generally believed to encode oncogenes and tumor suppressor genes (TSGs) that drive cancer growth. However, it remains a challenge to delineate the key cancer driver genes from the regions encoding a large number of genes. RESULTS In this study, we developed a new approach to CNV analysis based on spectral decomposition of CNV profiles into focal CNVs and broad CNVs. We performed an analysis of CNV data of 587 serous ovarian cancer samples on multiple platforms. We identified a number of novel focal regions, such as focal gain of ESR1, focal loss of LSAMP, prognostic site at 3q26.2 and losses of sub-telomere regions in multiple chromosomes. Furthermore, we performed network modularity analysis to examine the relationships among genes encoded in the focal CNV regions. Our results also showed that the recurrent focal gains were significantly associated with the known oncogenes and recurrent losses associated with TSGs and the CNVs had a greater effect on the mRNA expression of the driver genes than that of the non-driver genes. CONCLUSIONS Our results demonstrate that spectral decomposition of CNV profiles offers a new way of understanding the role of CNVs in cancer.
Collapse
Affiliation(s)
- Liangcai Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1410, Houston, TX, 77401, USA
- Department of Statistics, Rice University, Houston, TX, USA
- Department of Biophysics, College of Bioinformatics Sciences and Technology, Harbin Medical University, Harbin, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1410, Houston, TX, 77401, USA
- Department of Statistics, Rice University, Houston, TX, USA
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1410, Houston, TX, 77401, USA.
| |
Collapse
|
71
|
Wątroba M, Szukiewicz D. The role of sirtuins in aging and age-related diseases. Adv Med Sci 2016; 61:52-62. [PMID: 26521204 DOI: 10.1016/j.advms.2015.09.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 09/02/2015] [Accepted: 09/11/2015] [Indexed: 02/09/2023]
Abstract
Sirtuins, initially described as histone deacetylases and gene silencers in yeast, are now known to have much more functions and to be much more abundant in living organisms. Sirtuins gained much attention when they were first acknowledged to be responsible for some beneficial and longevity-promoting effects of calorie restriction in many species of animals - from fruit flies to mammals. In this paper, we discuss some detailed molecular mechanisms of inducing these effects, and wonder if they could be possibly mimicked without actually applying calorie restriction, through induction of sirtuin activity. It is known now that sirtuins, when adjusting the pattern of cellular metabolism to nutrient availability, can regulate many metabolic functions significant from the standpoint of aging research - including DNA repair, genome stability, inflammatory response, apoptosis, cell cycle, and mitochondrial functions. While carrying out these regulations, sirtuins cooperate with many transcription factors, including PGC-1a, NFKB, p53 and FoxO. This paper contains some considerations about possible use of facilitating activity of the sirtuins in prevention of aging, metabolic syndrome, chronic inflammation, and other diseases.
Collapse
|
72
|
Clinicopathological significance of SIRT1 expression in colorectal cancer: A systematic review and meta analysis. Int J Surg 2016; 26:32-7. [DOI: 10.1016/j.ijsu.2016.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/13/2015] [Accepted: 01/01/2016] [Indexed: 12/16/2022]
|
73
|
Giguère SSB, Guise AJ, Jean Beltran PM, Joshi PM, Greco TM, Quach OL, Kong J, Cristea IM. The Proteomic Profile of Deleted in Breast Cancer 1 (DBC1) Interactions Points to a Multifaceted Regulation of Gene Expression. Mol Cell Proteomics 2015; 15:791-809. [PMID: 26657080 DOI: 10.1074/mcp.m115.054619] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 01/01/2023] Open
Abstract
Deleted in breast cancer 1 (DBC1) has emerged as an important regulator of multiple cellular processes, ranging from gene expression to cell cycle progression. DBC1 has been linked to tumorigenesis both as an inhibitor of histone deacetylases, HDAC3 and sirtuin 1, and as a transcriptional cofactor for nuclear hormone receptors. However, despite mounting interest in DBC1, relatively little is known about the range of its interacting partners and the scope of its functions. Here, we carried out a functional proteomics-based investigation of DBC1 interactions in two relevant cell types, T cells and kidney cells. Microscopy, molecular biology, biochemistry, and mass spectrometry studies allowed us to assess DBC1 mRNA and protein levels, localization, phosphorylation status, and protein interaction networks. The comparison of DBC1 interactions in these cell types revealed conserved regulatory roles for DBC1 in gene expression, chromatin organization and modification, and cell cycle progression. Interestingly, we observe previously unrecognized DBC1 interactions with proteins encoded by cancer-associated genes. Among these interactions are five components of the SWI/SNF complex, the most frequently mutated chromatin remodeling complex in human cancers. Additionally, we identified a DBC1 interaction with TBL1XR1, a component of the NCoR complex, which we validated by reciprocal isolation. Strikingly, we discovered that DBC1 associates with proteins that regulate the circadian cycle, including DDX5, DHX9, and SFPQ. We validated this interaction by colocalization and reciprocal isolation. Functional assessment of this association demonstrated that DBC1 protein levels are important for regulating CLOCK and BMAL1 protein oscillations in synchronized T cells. Our results suggest that DBC1 is integral to the maintenance of the circadian molecular clock. Furthermore, the identified interactions provide a valuable resource for the exploration of pathways involved in DBC1-associated tumorigenesis.
Collapse
Affiliation(s)
- Sophie S B Giguère
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Amanda J Guise
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Pierre M Jean Beltran
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Preeti M Joshi
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Todd M Greco
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Olivia L Quach
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Jeffery Kong
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Ileana M Cristea
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| |
Collapse
|
74
|
Sirtuin 1 promotes the growth and cisplatin resistance of endometrial carcinoma cells: a novel therapeutic target. J Transl Med 2015; 95:1363-73. [PMID: 26367491 DOI: 10.1038/labinvest.2015.119] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 07/12/2015] [Accepted: 07/13/2015] [Indexed: 11/08/2022] Open
Abstract
Sirtuin 1 (SIRT1), originally identified as a longevity gene, is induced by caloric restriction, and regulates various cellular functions including DNA repair, cell survival and metabolism via the deacetylation of target proteins such as histone and p53. These functions are considered to act dualistically as preventing or facilitating cancer. This study aimed to clarify the expression and role of SIRT1 in endometrial carcinoma. Because a high-calorie diet was a well-known risk factor for endometrial carcinoma, we first hypothesized that SIRT1 might be downregulated in normal endometrial glandular cells of obese women. However, no correlation was observed between the expression of SIRT1 and body mass index (BMI). In contrast, regardless of BMI, the immunohistochemical expression of SIRT1 was significantly higher in endometrial carcinoma (108 cases) than in normal endometria (60 cases) (P<0.05), and its overexpression was associated with a shorter survival (P<0.05). Our experiments in vivo revealed that SIRT1 accelerated the proliferation of endometrial carcinoma cell lines (HHUA, HEC151, and HEC1B). SIRT1 overexpression significantly enhanced the resistance for cisplatin and paclitaxel in HHUA cells. Although p53 is an important target protein for SIRT1, the selective SIRT1 inhibitor (EX527) significantly suppressed the proliferation and cisplatin resistance of three endometrial carcinoma cell lines regardless of the p53 mutation status. In addition, SIRT1 overexpression in HHUA cells accelerated tumor growth and cisplatin resistance in nude mice, and EX527 significantly suppressed the growth of tumors of HHUA and HEC1B cells. No adverse effect of EX527 was observed in these mice. In conclusion, SIRT1 is involved in the acquisition of the aggressive behavior associated with endometrial carcinoma, and the SIRT1 inhibitor, EX527, may be a useful agent for the treatment of this malignancy.
Collapse
|
75
|
Abstract
Sirtinol, a Schiff base derived from 2-hydroxy-1-naphthaldehyde, is an inhibitor of sirtuin proteins, a family of deacetylases active in gene regulation and relevant to the study of cancer growth. The formation of copper(II) and zinc(II) complexes of sirtinol is investigated by spectroscopic and structural methods. The molecular structure of this protein inhibitor allows for coordination of first-row transition metals in both tridentate and bidentate fashion. In addition, assays in cultured breast cancer cells reveal that CuII(sirtinol-H)2 and previously reported FeIII(sirtinol-H)(NO3)2 present enhanced cytotoxicity when compared to the free ligand, and that the ferric complex causes an increase in intracellular oxidative stress. Transition metal coordination in the biological milieu could therefore contribute additional effects to the biological profile of sirtinol.
Collapse
Affiliation(s)
- Eman A Akam
- Department of Chemistry and Biochemistry, University of Arizona, Tucson AZ, USA
| | - Ritika Gautam
- Department of Chemistry and Biochemistry, University of Arizona, Tucson AZ, USA
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, University of Arizona, Tucson AZ, USA
| |
Collapse
|
76
|
Kim H, Lee KH, Park IA, Chung YR, Im SA, Noh DY, Han W, Moon HG, Jung YY, Ryu HS. Expression of SIRT1 and apoptosis-related proteins is predictive for lymph node metastasis and disease-free survival in luminal A breast cancer. Virchows Arch 2015; 467:563-70. [PMID: 26280894 DOI: 10.1007/s00428-015-1815-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 06/29/2015] [Accepted: 07/17/2015] [Indexed: 12/18/2022]
Abstract
Luminal A breast cancer can present with early, unexpected lymph node metastasis, and sentinel lymph node biopsy has been reported false negative in some cases. We aimed to construct a biomarker-based model that predicts lymph node metastasis in luminal A breast cancer, using expression of silent mating type information regulation 2 homolog 1 (SIRT1) and apoptosis-related factors, which are known to be closely related. We selected tissue samples of 278 cases of luminal A invasive ductal carcinoma, constructed tissue microarrays, and performed immunohistochemical staining for SIRT1 and four apoptosis-related proteins. In constructing the best predictive model for lymph node metastasis, six clinicopathological parameters and five molecular markers were considered. Independent factors predictive of lymph node metastasis were pT stage (OR 1.829, p = 0.027), lymphovascular invasion (OR 4.128, p < 0.001), and decreased expression of caspase-3 (OR 0.535, p = 0.034) and of SIRT1 (OR 0.526, p = 0.053). A combination nuclear grade, lymphovascular invasion, increased B-cell lymphoma 2 (Bcl-2) expression, and reduced expression of caspase-3 and of SIRT1 yielded the strongest predictive performance for lymph node metastasis with an area under the curve (AUC) of 0.696. This combination was also predictive of shortened disease-free survival (73.1 vs. 67.7 months, p = 0.003). Our data support a role of SIRT1 protein as tumor suppressor in luminal A breast cancer, in association with apoptosis-related proteins. Our model based upon a combination of these biomarkers is expected to increase accuracy of prediction of lymph node metastasis in luminal A breast cancer. This might serve as a valuable tool in determining the optimal surgical strategy in breast cancer patients.
Collapse
Affiliation(s)
- Hyojin Kim
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Yul Ri Chung
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
| | - Dong-Young Noh
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Yoon Yang Jung
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
77
|
Santolla MF, Avino S, Pellegrino M, De Francesco EM, De Marco P, Lappano R, Vivacqua A, Cirillo F, Rigiracciolo DC, Scarpelli A, Abonante S, Maggiolini M. SIRT1 is involved in oncogenic signaling mediated by GPER in breast cancer. Cell Death Dis 2015. [PMID: 26225773 PMCID: PMC4650744 DOI: 10.1038/cddis.2015.201] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A number of tumors exhibit an altered expression of sirtuins, including NAD+-dependent histone deacetylase silent information regulator 1 (SIRT1) that may act as a tumor suppressor or tumor promoter mainly depending on the tumor types. For instance, in breast cancer cells SIRT1 was shown to exert an essential role toward the oncogenic signaling mediated by the estrogen receptor-α (ERα). In accordance with these findings, the suppression of SIRT1 led to the inhibition of the transduction pathway triggered by ERα. As the regulation of SIRT1 has not been investigated in cancer cells lacking ER, in the present study we ascertained the expression and function of SIRT1 by estrogens in ER-negative breast cancer cells and cancer-associated fibroblasts obtained from breast cancer patients. Our results show that 17β-estradiol (E2) and the selective ligand of GPER, namely G-1, induce the expression of SIRT1 through GPER and the subsequent activation of the EGFR/ERK/c-fos/AP-1 transduction pathway. Moreover, we demonstrate that SIRT1 is involved in the pro-survival effects elicited by E2 through GPER, like the prevention of cell cycle arrest and cell death induced by the DNA damaging agent etoposide. Interestingly, the aforementioned actions of estrogens were abolished silencing GPER or SIRT1, as well as using the SIRT1 inhibitor Sirtinol. In addition, we provide evidence regarding the involvement of SIRT1 in tumor growth stimulated by GPER ligands in breast cancer cells and xenograft models. Altogether, our data suggest that SIRT1 may be included in the transduction network activated by estrogens through GPER toward the breast cancer progression.
Collapse
Affiliation(s)
- M F Santolla
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - S Avino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - M Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - E M De Francesco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - P De Marco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - R Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - A Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - F Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - D C Rigiracciolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - A Scarpelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - S Abonante
- Breast Cancer Unit, Regional Hospital, Cosenza, Italy
| | - M Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
78
|
Song CL, Tang H, Ran LK, Ko BCB, Zhang ZZ, Chen X, Ren JH, Tao NN, Li WY, Huang AL, Chen J. Sirtuin 3 inhibits hepatocellular carcinoma growth through the glycogen synthase kinase-3β/BCL2-associated X protein-dependent apoptotic pathway. Oncogene 2015; 35:631-41. [PMID: 25915842 DOI: 10.1038/onc.2015.121] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/10/2015] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
SIRT3 is a class III histone deacetylase that has been implicated in a variety of cancers. The role of SIRT3 in hepatocellular carcinoma (HCC) remains elusive. In this study, we found that SIRT3 expression was frequently repressed in HCC and its downregulation was closely associated with tumor grade and size. Ectopic expression of SIRT3 inhibited cell growth and induced apoptosis in HCC cells, whereas depletion of SIRT3 in immortalized hepatocyte promoted cell growth and decreased epirubicin-induced apoptosis. Mechanistic studies revealed that SIRT3 deacetylated and activated glycogen synthase kinase-3β (GSK-3β), which subsequently induced expression and mitochondrial translocation of the pro-apoptotic protein BCL2-associated X protein (Bax) to promote apoptosis. GSK-3β inhibitor or gene silencing of BAX reversed SIRT3-induced growth inhibition and apoptosis. Furthermore, SIRT3 overexpression also suppressed tumor growth in vivo. Together, this study reveals a role of SIRT3/GSK-3β/Bax signaling pathway in the suppression of HCC growth, and also suggests that targeting this pathway may represent a potential therapeutic approach for HCC treatment.
Collapse
Affiliation(s)
- C-L Song
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - H Tang
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - L-K Ran
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - B C B Ko
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chirosciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Z-Z Zhang
- Department of Infectious Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - X Chen
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - J-H Ren
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - N-N Tao
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - W-Y Li
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - A-L Huang
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Zhejiang, China
| | - J Chen
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
79
|
Chung YR, Kim H, Park SY, Park IA, Jang JJ, Choe JY, Jung YY, Im SA, Moon HG, Lee KH, Suh KJ, Kim TY, Noh DY, Han W, Ryu HS. Distinctive role of SIRT1 expression on tumor invasion and metastasis in breast cancer by molecular subtype. Hum Pathol 2015; 46:1027-35. [PMID: 26004371 DOI: 10.1016/j.humpath.2015.03.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/23/2015] [Accepted: 03/30/2015] [Indexed: 01/25/2023]
Abstract
The aim of this study was to evaluate silent mating type information regulation 2 homolog 1 (SIRT1) expression levels by subtype and evaluate its predictive power of axillary lymph node metastasis (LNM) and its association with clinical outcome. A total of 427 patients diagnosed with invasive ductal carcinoma were chosen, immunohistochemical staining for SIRT1 expression was performed on tissue microarrays, and in vitro experiments with each intrinsic subtype of human breast cancer cell line were carried out. Increased expression of SIRT1 in hormone receptor-positive breast cancer and HER2 breast cancer subtype significantly correlated with lower risks of LNM. On the contrary, in triple-negative breast cancer, increased SIRT1 expression was more frequently observed in LNM-positive subgroup than LNM-negative subgroup. Combination of statistically significant, independent parameters including SIRT1 revealed predictive performance for LNM with area under the curve of 0.602, 0.587, and 0.726 for hormone receptor-positive breast cancer, HER2 breast cancer, and triple-negative breast cancer subtype, respectively. Inhibition of SIRT1 expression with small interfering RNA suppressed tumor invasion in MDA-MB-231, specifically. This is the first study to examine SIRT1 expression in breast cancer by subtype, and we have observed the potentially different role of SIRT1 gene having tumor-suppressive or tumor-promoting influence depending on the subtype; thus, different associations between SIRT1 expression and prognosis by subtype should be considered in its target therapy.
Collapse
Affiliation(s)
- Yul Ri Chung
- Department of Pathology, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Hyojin Kim
- Department of Pathology, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Soo Young Park
- Department of Pathology, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Ja June Jang
- Department of Pathology, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Ji-Young Choe
- Department of Pathology, Seoul National University Bundang Hospital, 463-707 Seongnam-si, Gyeonggido, Republic of Korea
| | - Yoon Yang Jung
- Department of Pathology, Samsung Medical Center, Sungkyungkwan University School of Medicine, 135-710 Seoul, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Koung Jin Suh
- Department of Internal Medicine, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University Hospital, 110-744 Seoul, Republic of Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, 110-744 Seoul, Republic of Korea.
| |
Collapse
|
80
|
Maiese K. SIRT1 and stem cells: In the forefront with cardiovascular disease, neurodegeneration and cancer. World J Stem Cells 2015; 7:235-242. [PMID: 25815111 PMCID: PMC4369483 DOI: 10.4252/wjsc.v7.i2.235] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 12/10/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, nervous system disorders, and cancer in association with other diseases such as diabetes mellitus result in greater than sixty percent of the global annual deaths. These noncommunicable diseases also affect at least one-third of the population in low and middle-income countries and lead to hypertension, elevated cholesterol, malignancy, and neurodegenerative disorders such as Alzheimer’s disease and stroke. With the climbing lifespan of the world’s population, increased prevalence of these disorders is expected requiring the development of new therapeutic strategies against these disabling disease entities. Targeting stem cell proliferation for cardiac disease, vascular disorders, cancer, and neurodegenerative disorders is receiving great enthusiasm, especially those that focus upon SIRT1, a mammalian homologue of the yeast silent information regulator-2. Modulation of the cellular activity of SIRT1 can involve oversight by nicotinamide/nicotinic acid mononucleotide adenylyltransferase, mammalian forkhead transcription factors, mechanistic of rapamycin pathways, and cysteine-rich protein 61, connective tissue growth factor, and nephroblastoma over-expressed gene family members that can impact cytoprotective outcomes. Ultimately, the ability of SIRT1 to control the programmed cell death pathways of apoptosis and autophagy can determine not only cardiac, vascular, and neuronal stem cell development and longevity, but also the onset of tumorigenesis and the resistance against chemotherapy. SIRT1 therefore has a critical role and holds exciting prospects for new therapeutic strategies that can offer reparative processes for cardiac, vascular, and nervous system degenerative disorders as well as targeted control of aberrant cell growth during cancer.
Collapse
|
81
|
Burton K, Shaw L, Morey LM. Differential effect of estradiol and bisphenol A on Set8 and Sirt1 expression in prostate cancer. Toxicol Rep 2015; 2:817-823. [PMID: 28962417 PMCID: PMC5598099 DOI: 10.1016/j.toxrep.2015.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/19/2014] [Accepted: 01/28/2015] [Indexed: 11/28/2022] Open
Abstract
Exposure to estrogenic compounds has been shown to epigenetically reprogram the prostate and may contribute to prostate cancer. The goal of this study was to determine the effect of physiological doses of estradiol and bisphenol A (BPA) on the expression of histone modifying enzymes (HMEs) in prostate cancer. Using two human prostate cancer cell lines we examined the expression of Set8, a histone methyltransferase, and Sirt1, a histone deacetylase, after exposure to estrogen or BPA. These experiments were carried out in the presence of natural hormones to understand the impact of additional exposure to estrogen or BPA on HME expression. We found differential expression of the HMEs in the different models and between the different compounds. Further, we determined that the changes in gene expression occurred via estrogen receptor signaling using the ER antagonist, ICI 182,780 (fulvestrant). Interestingly we found that the combination of ICI with estrogen or BPA greatly affected the expression of Set8, even when the hormone alone had no effect. This study demonstrates that the effects of estrogen and BPA on HME expression vary and that the presence of both the estrogen receptor and androgen receptor may be important for therapeutic intervention.
Collapse
Affiliation(s)
- Kevin Burton
- Department of Biology, Canisius College, Buffalo, NY 14208, United States
| | - Lisa Shaw
- Department of Biology, Canisius College, Buffalo, NY 14208, United States
| | - Lisa M Morey
- Department of Biology, Canisius College, Buffalo, NY 14208, United States
| |
Collapse
|
82
|
Histone deacetylase III as a potential therapeutic target for the treatment of lethal sepsis. J Trauma Acute Care Surg 2015; 77:913-9; discussion 919. [PMID: 25051385 DOI: 10.1097/ta.0000000000000347] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND We have recently demonstrated that inhibition of histone deacetylase (HDAC) Class I, II, and IV with nonspecific HDAC inhibitors improves survival in a mouse model of lethal cecal ligation and puncture (CLP). However, the consequence of HDAC Class III inhibition is unknown in this model. The aims of the present study were to explore the effect of EX-527, a selective Sirtuin 1 (SIRT1) inhibitor, on survival in the lethal model of CLP-sepsis and to assess the impact of the treatment on inflammatory cytokine production, coagulopathy, and bone marrow atrophy during severe sepsis. METHODS For Experiment I, C57BL/6J mice were subjected to CLP and, 1 hour later, intraperitoneally injected with either EX-527 dissolved in dimethyl sulfoxide (DMSO) or DMSO only. Survival was monitored for 10 days. For Experiment II, 1 hour after CLP animals were randomly treated with (1) DMSO vehicle and (2) EX-527. Peritoneal fluid and blood samples were collected for measurement of cytokines, and blood was also used to evaluate coagulation status using thrombelastography. In addition, long bones (femurs and tibias) were examined to determine morphologic changes in the marrow by hematoxylin and eosin staining. For Experiment III, normal primary splenocytes were cultured and treated with lipopolysaccharide in the presence or absence of EX-527 to assess cytokine production. RESULTS EX-527 significantly improved survival (50% vs. 0% survival as compared to vehicle, p = 0.0007) and attenuated levels of cytokines tumor necrosis factor α and interleukin 6 in the blood and the peritoneal fluid compared with the vehicle control. It also decreased tumor necrosis factor α and interleukin 6 production by splenocytes in vitro. Selective inhibition of SIRT1 was associated with significant improvements in fibrin cross-linkage, platelet function, and clot rigidity but had no significant impact on the clot initiation parameters. Moreover, inhibition of SIRT1 was associated with a significant decrease in bone marrow atrophy. CONCLUSION Selective inhibition of Class III HDAC SIRT1 significantly improves survival, attenuates cytokine levels and sepsis-associated coagulopathy, and decreases bone marrow atrophy in a lethal mouse septic model.
Collapse
|
83
|
Kim TH, Kim HS, Kang YJ, Yoon S, Lee J, Choi WS, Jung JH, Kim HS. Psammaplin A induces Sirtuin 1-dependent autophagic cell death in doxorubicin-resistant MCF-7/adr human breast cancer cells and xenografts. Biochim Biophys Acta Gen Subj 2015; 1850:401-10. [PMID: 25445714 DOI: 10.1016/j.bbagen.2014.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/16/2014] [Accepted: 11/06/2014] [Indexed: 11/26/2022]
|
84
|
Chen X, Sun K, Jiao S, Cai N, Zhao X, Zou H, Xie Y, Wang Z, Zhong M, Wei L. High levels of SIRT1 expression enhance tumorigenesis and associate with a poor prognosis of colorectal carcinoma patients. Sci Rep 2014; 4:7481. [PMID: 25500546 PMCID: PMC4265776 DOI: 10.1038/srep07481] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/26/2014] [Indexed: 02/08/2023] Open
Abstract
SIRT1, a NAD+ dependent class III deacetylase, takes part in many important biological processes. Previous studies show that SIRT1 is overexpressed in some cancers and plays an essential role in tumorigenesis. However, the association between SIRT1 and colorectal cancer (CRC) is still unclear. We found that many CRC specimens had strong SIRT1 expression, which had an obvious correlation with poor prognosis of CRC patients. Meanwhile, SIRT1 expression had a co-localization with CD133, a current universal marker to characterize colorectal cancer stem cells (CSCs). In vitro studies also revealed that SIRT1 was overexpressed in colorectal CSC-like cells. Moreover, SIRT1 deficiency decreased percentage of CD133+ cells, attenuated the abilities of colony and sphere formation, and inhibited tumorigenicity in vivo in CRC cells. Further study demonstrated that the expressions of several stemness-associated genes, including Oct4, Nanog, Cripto, Tert and Lin28, were reduced by SIRT1 knockdown in CRC cells. Taken together, our findings suggest that SIRT1 plays a crucial role in keeping the characteristics of CSCs cells. SIRT1 is a potential independent prognostic factor of CRC patients after tumor resection with curative intent, and will contribute to providing a promising new approach to target at CSCs in CRC treatment.
Collapse
Affiliation(s)
- Xiaojing Chen
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Sun
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shufan Jiao
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Cai
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Zhao
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hanbing Zou
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuexia Xie
- Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengshi Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lixin Wei
- 1] Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China [2] Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
85
|
Clark-Knowles KV, Dewar-Darch D, Jardine KE, McBurney MW. Modulation of tumorigenesis by dietary intervention is not mediated by SIRT1 catalytic activity. PLoS One 2014; 9:e112406. [PMID: 25380034 PMCID: PMC4224430 DOI: 10.1371/journal.pone.0112406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 10/14/2014] [Indexed: 12/31/2022] Open
Abstract
The protein deacetylase SIRT1 is involved in the regulation of a large number of cellular processes that are thought to be required for cancer initiation and progression. Both SIRT1 activity and tumorigenesis can be influenced by dietary fat and polyphenolics. We set out to determine whether dietary modulations of tumorigenesis are mediated by SIRT1 catalytic functions. We introduced a mammary gland tumor-inducing transgene, MMTV-PyMT, into stocks of mice bearing a H355Y point mutation in the Sirt1 gene that abolishes SIRT1 catalytic activity. Tumor latency was reduced in animals fed a high fat diet but this effect was not dependent on SIRT1 activity. Resveratrol had little effect on tumor formation except in animals heterozygous for the mutant Sirt1 gene. We conclude that the effects of these dietary interventions on tumorigenesis are not mediated by modulation of SIRT1 catalytic activity.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Antineoplastic Agents, Phytogenic/pharmacology
- Biocatalysis
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Diet, High-Fat
- Heterozygote
- Male
- Mammary Neoplasms, Experimental/diet therapy
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Tumor Virus, Mouse/genetics
- Mice, Transgenic
- Point Mutation
- Resveratrol
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Stilbenes/pharmacology
- Tumor Burden/drug effects
- Tumor Burden/genetics
Collapse
Affiliation(s)
| | - Danielle Dewar-Darch
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Karen E. Jardine
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael W. McBurney
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
86
|
Vrtačnik P, Ostanek B, Mencej-Bedrač S, Marc J. The many faces of estrogen signaling. Biochem Med (Zagreb) 2014; 24:329-42. [PMID: 25351351 PMCID: PMC4210253 DOI: 10.11613/bm.2014.035] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 08/14/2014] [Indexed: 12/21/2022] Open
Abstract
Estrogens have long been known as important regulators of the female reproductive functions; however, our understanding of the role estrogens play in the human body has changed significantly over the past years. It is now commonly accepted that estrogens and androgens have important functions in both female and male physiology and pathology. This is in part due to the local synthesis and action of estrogens that broadens the role of estrogen signaling beyond that of the endocrine system. Furthermore, there are several different mechanisms through which the three estrogen receptors (ERs), ERα, ERβ and G protein-coupled estrogen receptor 1 (GPER1) are able to regulate target gene transcription. ERα and ERβ are mostly associated with the direct and indirect genomic signaling pathways that result in target gene expression. Membrane-bound GPER1 is on the other hand responsible for the rapid non-genomic actions of estrogens that activate various protein-kinase cascades. Estrogen signaling is also tightly connected with another important regulatory entity, i.e. epigenetic mechanisms. Posttranslational histone modifications, microRNAs (miRNAs) and DNA methylation have been shown to influence gene expression of ERs as well as being regulated by estrogen signaling. Moreover, several coregulators of estrogen signaling also exhibit chromatin-modifying activities further underlining the importance of epigenetic mechanisms in estrogen signaling. This review wishes to highlight the newer aspects of estrogen signaling that exceed its classical endocrine regulatory role, especially emphasizing its tight intertwinement with epigenetic mechanisms.
Collapse
Affiliation(s)
- Peter Vrtačnik
- University of Ljubljana, Faculty of Pharmacy, Department of Clinical Biochemistry, Ljubljana, Slovenia
| | - Barbara Ostanek
- University of Ljubljana, Faculty of Pharmacy, Department of Clinical Biochemistry, Ljubljana, Slovenia
| | - Simona Mencej-Bedrač
- University of Ljubljana, Faculty of Pharmacy, Department of Clinical Biochemistry, Ljubljana, Slovenia
| | - Janja Marc
- University of Ljubljana, Faculty of Pharmacy, Department of Clinical Biochemistry, Ljubljana, Slovenia
| |
Collapse
|
87
|
Artsi H, Cohen-Kfir E, Gurt I, Shahar R, Bajayo A, Kalish N, Bellido TM, Gabet Y, Dresner-Pollak R. The Sirtuin1 activator SRT3025 down-regulates sclerostin and rescues ovariectomy-induced bone loss and biomechanical deterioration in female mice. Endocrinology 2014; 155:3508-15. [PMID: 24949665 PMCID: PMC11651357 DOI: 10.1210/en.2014-1334] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/13/2014] [Indexed: 01/28/2023]
Abstract
Estrogen deficiency leads to rapid bone loss and skeletal fragility. Sclerostin, encoded by the sost gene, and a product of the osteocyte, is a negative regulator of bone formation. Blocking sclerostin increases bone mass and strength in animals and humans. Sirtuin1 (Sirt1), a player in aging and metabolism, regulates bone mass and inhibits sost expression by deacetylating histone 3 at its promoter. We asked whether a Sirt1-activating compound could rescue ovariectomy (OVX)-induced bone loss and biomechanical deterioration in 9-week-old C57BL/6 mice. OVX resulted in a substantial decrease in skeletal Sirt1 expression accompanied by an increase in sclerostin. Oral administration of SRT3025, a Sirt1 activator, at 50 and 100 mg/kg·d for 6 weeks starting 6 weeks after OVX fully reversed the deleterious effects of OVX on vertebral bone mass, microarchitecture, and femoral biomechanical properties. Treatment with SRT3025 decreased bone sclerostin expression and increased cortical periosteal mineralizing surface and serum propeptide of type I procollagen, a bone formation marker. In vitro, in the murine long bone osteocyte-Y4 osteocyte-like cell line SRT3025 down-regulated sclerostin and inactive β-catenin, whereas a reciprocal effect was observed with EX-527, a Sirt1 inhibitor. Sirt1 activation by Sirt1-activating compounds is a potential novel pathway to down-regulate sclerostin and design anabolic therapies for osteoporosis concurrently ameliorating other metabolic and age-associated conditions.
Collapse
Affiliation(s)
- Hanna Artsi
- Endocrinology and Metabolism Service (H.A., E.C.-K., I.G., R.D.-P.), Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; Laboratory of Bone Biomechanics (R.S., N.K.), Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, 76100 Israel; Bone Laboratory (A.B.), The Hebrew University of Jerusalem, Jerusalem, 91120 Israel; Department of Anatomy and Cell Biology (T.M.B.), Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana 46202; and Department of Anatomy and Anthropology (Y.G.), Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978 Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Solak KA, Wijnolts FMJ, Nijmeijer SM, Blaauboer BJ, van den Berg M, van Duursen MBM. Excessive levels of diverse phytoestrogens can modulate steroidogenesis and cell migration of KGN human granulosa-derived tumor cells. Toxicol Rep 2014; 1:360-372. [PMID: 28962252 PMCID: PMC5598505 DOI: 10.1016/j.toxrep.2014.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 06/10/2014] [Accepted: 06/10/2014] [Indexed: 12/17/2022] Open
Abstract
Phytoestrogens are plant-derived estrogen-like compounds that are increasingly used for their suggested health promoting properties, even by healthy, young women. However, scientific concerns exist regarding potential adverse effects on female reproduction. In this study, naringenin (NAR), 8-prenylnaringenin (8-PN), genistein (GEN), coumestrol (COU), quercetin (QUE) and resveratrol (RSV) up-regulated steroidogenic acute regulatory protein (StaR) mRNA levels in KGN human granulosa-like tumor cells. Most of the phytoestrogens tested also increased CYP19A1 (aromatase) mRNA levels via activation of ovary-specific I.3 and II promoters. Yet, only NAR (3 and 10 μM), COU (10 and 30 μM) and QUE (10 μM) also statistically significantly induced aromatase activity in KGN cells after 24 h. 8-PN, aromatase inhibitor letrozole and estrogen receptor antagonist ICI 182,780 concentration-dependently inhibited aromatase activity with IC50 values of 8 nM, 10 nM and 72 nM, respectively. Co-exposure with ICI 182,780 (0.1 μM) statistically significantly attenuated the induction of aromatase activity by QUE and COU, but not NAR. Cell cycle status and proliferation of KGN cells were not affected by any of the phytoestrogens tested. Nonetheless, the migration of KGN cells was significantly reduced with approximately 30% by COU, RSV and QUE and 46% by GEN at 10 μM, but not NAR and 8-PN. Our results indicate that phytoestrogens can affect various pathways in granulosa-like cells in vitro at concentrations that can be found in plasma upon supplement intake. This implies that phytoestrogens may interfere with ovarian function and caution is in place regarding the use of supplements with high contents of phytoestrogens.
Collapse
Affiliation(s)
- Kamila A Solak
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Fiona M J Wijnolts
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Sandra M Nijmeijer
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Bas J Blaauboer
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Martin van den Berg
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Majorie B M van Duursen
- Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
89
|
Resveratrol promotes degradation of the human bile acid transporter ASBT (SLC10A2). Biochem J 2014; 459:301-12. [PMID: 24498857 DOI: 10.1042/bj20131428] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The sodium/bile acid co-transporter ASBT [apical sodium-dependent bile acid transporter; SLC10A2 (solute carrier family 10 member 2)] plays a key role in the enterohepatic recycling of the bile acids and indirectly contributes to cholesterol homoeostasis. ASBT inhibitors reportedly lower plasma triglyceride levels and increase HDL (high-density lipoprotein) cholesterol levels. RSV (resveratrol), a major constituent of red wine, is known to lower LDL (low-density lipoprotein) cholesterol levels, but its mechanism of action is still unclear. In the present study, we investigated the possible involvement of ASBT in RSV-mediated cholesterol-lowering effects. We demonstrate that RSV inhibits ASBT protein expression and function via a SIRT1 (sirtuin 1)-independent mechanism. The effect was specific to ASBT since other transporters involved in cholesterol homoeostasis, NTCP (SLC10A1), OSTα (SLC51A) and ABCG1 (ATP-binding cassette G1), remained unaffected. ASBT inhibition by RSV was reversed by proteasome inhibitors (MG-132 and lactacystin) and the ubiquitin inhibitor LDN57444, suggesting involvement of the ubiquitin-proteasome pathway. Immunoprecipitation revealed high levels of ubiquitinated ASBT after RSV treatment. Phosphorylation at Ser335 and Thr339 was shown previously to play a role in proteosomal degradation of rat ASBT. However, mutation at corresponding residues in rat ASBT revealed that phosphorylation does not contribute to RSV-mediated degradation of ASBT. Combined, our data indicate that RSV promotes ASBT degradation via the ubiquitin-proteasome pathway without requiring phosphorylation. We conclude that regulation of ASBT expression by RSV may have clinical relevance with regard to the observed cholesterol-lowering effects of RSV.
Collapse
|
90
|
Hwang BJ, Madabushi A, Jin J, Lin SYS, Lu AL. Histone/protein deacetylase SIRT1 is an anticancer therapeutic target. Am J Cancer Res 2014; 4:211-221. [PMID: 24959376 PMCID: PMC4065402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 04/23/2014] [Indexed: 06/03/2023] Open
Abstract
SIRT1, a member of the NAD(+)-dependent histone/protein deacetylase family, is involved in chromatin remodeling, DNA repair, and stress response and is a potential drug target. 5-fluorouracil (FU) and the SN1-type DNA methylating agent temozolomide (TMZ) are anticancer agents. In this study, we demonstrate that sirt1 knockout mouse embryonic fibroblast cells are more sensitive to FU and DNA methylating agents than normal cells. Based on these findings, the chemotherapy efficacy of SIRT1 inhibitors in combination with FU or TMZ were tested with human breast cancer cells. We found that treatments combining SIRT1 inhibitors with FU or TMZ show synergistic reduction of cell viability and colony formation of breast cancer cells. Thus, inhibition of SIRT1 activity provides a novel anticancer strategy.
Collapse
Affiliation(s)
- Bor-Jang Hwang
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimore, MD 21201, USA
| | - Amrita Madabushi
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimore, MD 21201, USA
- Current address: Department of Natural & Physical Sciences, Life Sciences Institute; Baltimore City Community CollegeBaltimore, MD 21201
| | - Jin Jin
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimore, MD 21201, USA
| | - Shiou-Yuh S Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimore, MD 21201, USA
| | - A-Lien Lu
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimore, MD 21201, USA
- University of Maryland Greenebaum Cancer CenterBaltimore, MD 21201, USA
| |
Collapse
|
91
|
Nwachukwu JC, Srinivasan S, Bruno NE, Parent AA, Hughes TS, Pollock JA, Gjyshi O, Cavett V, Nowak J, Garcia-Ordonez RD, Houtman R, Griffin PR, Kojetin DJ, Katzenellenbogen JA, Conkright MD, Nettles KW. Resveratrol modulates the inflammatory response via an estrogen receptor-signal integration network. eLife 2014; 3:e02057. [PMID: 24771768 PMCID: PMC4017646 DOI: 10.7554/elife.02057] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/05/2014] [Indexed: 12/21/2022] Open
Abstract
Resveratrol has beneficial effects on aging, inflammation and metabolism, which are thought to result from activation of the lysine deacetylase, sirtuin 1 (SIRT1), the cAMP pathway, or AMP-activated protein kinase. In this study, we report that resveratrol acts as a pathway-selective estrogen receptor-α (ERα) ligand to modulate the inflammatory response but not cell proliferation. A crystal structure of the ERα ligand-binding domain (LBD) as a complex with resveratrol revealed a unique perturbation of the coactivator-binding surface, consistent with an altered coregulator recruitment profile. Gene expression analyses revealed significant overlap of TNFα genes modulated by resveratrol and estradiol. Furthermore, the ability of resveratrol to suppress interleukin-6 transcription was shown to require ERα and several ERα coregulators, suggesting that ERα functions as a primary conduit for resveratrol activity.DOI: http://dx.doi.org/10.7554/eLife.02057.001.
Collapse
Affiliation(s)
- Jerome C Nwachukwu
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | - Sathish Srinivasan
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | - Nelson E Bruno
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | | | - Travis S Hughes
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, United States
| | - Julie A Pollock
- Department of Chemistry, University of Illinois, Urbana, United States
| | - Olsi Gjyshi
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | - Valerie Cavett
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | - Jason Nowak
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | - Ruben D Garcia-Ordonez
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, United States
| | - René Houtman
- Nuclear Receptor Group, PamGene International, Den Bosch, Netherlands
| | - Patrick R Griffin
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, United States
| | - Douglas J Kojetin
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, United States
| | | | - Michael D Conkright
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| | - Kendall W Nettles
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, United States
| |
Collapse
|
92
|
Elangovan S, Pathania R, Ramachandran S, Ananth S, Padia RN, Lan L, Singh N, Martin PM, Hawthorn L, Prasad PD, Ganapathy V, Thangaraju M. The niacin/butyrate receptor GPR109A suppresses mammary tumorigenesis by inhibiting cell survival. Cancer Res 2013; 74:1166-78. [PMID: 24371223 DOI: 10.1158/0008-5472.can-13-1451] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
GPR109A, a G-protein-coupled receptor, is activated by niacin and butyrate. Upon activation in colonocytes, GPR109A potentiates anti-inflammatory pathways, induces apoptosis, and protects against inflammation-induced colon cancer. In contrast, GPR109A activation in keratinocytes induces flushing by activation of Cox-2-dependent inflammatory signaling, and the receptor expression is upregulated in human epidermoid carcinoma. Thus, depending on the cellular context and tissue, GPR109A functions either as a tumor suppressor or a tumor promoter. However, the expression status and the functional implications of this receptor in the mammary epithelium are not known. Here, we show that GPR109A is expressed in normal mammary tissue and, irrespective of the hormone receptor status, its expression is silenced in human primary breast tumor tissues, breast cancer cell lines, and in tumor tissues of three different murine mammary tumor models. Functional expression of this receptor in human breast cancer cell lines decreases cyclic AMP production, induces apoptosis, and blocks colony formation and mammary tumor growth. Transcriptome analysis revealed that GPR109A activation inhibits genes, which are involved in cell survival and antiapoptotic signaling, in human breast cancer cells. In addition, deletion of Gpr109a in mice increased tumor incidence and triggered early onset of mammary tumorigenesis with increased lung metastasis in MMTV-Neu mouse model of spontaneous breast cancer. These findings suggest that GPR109A is a tumor suppressor in mammary gland and that pharmacologic induction of this gene in tumor tissues followed by its activation with agonists could be an effective therapeutic strategy to treat breast cancer.
Collapse
Affiliation(s)
- Selvakumar Elangovan
- Authors' Affiliations: Departments of Biochemistry and Molecular Biology, Biostatistics and Epidemiology, and Pathology; Cancer Center; Vision Science Discovery Institute, Georgia Regents University; Augusta, Georgia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Clark-Knowles KV, Dewar-Darch D, Jardine KE, McBurney MW. SIRT1 catalytic activity has little effect on tumor formation and metastases in a mouse model of breast cancer. PLoS One 2013; 8:e82106. [PMID: 24278473 PMCID: PMC3836945 DOI: 10.1371/journal.pone.0082106] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/27/2013] [Indexed: 12/23/2022] Open
Abstract
The protein deacetylase SIRT1 has been implicated in the regulation of a large number of cellular processes that are thought to be required for cancer initiation and progression. There are conflicting data that make it unclear whether Sirt1 functions as an oncogene or tumor suppressor. To assess the effect of SIRT1 on the emergence and progression of mammary tumors, we crossed mice that harbor a point mutation that abolishes SIRT1 catalytic activity with mice carrying the polyoma middle T transgene driven by the murine mammary tumor virus promoter (MMTV-PyMT). The absence of SIRT1 catalytic activity neither accelerated nor blocked the formation of tumors and metastases in this model. There was a lag in tumor latency that modestly extended survival in Sirt1 mutant mice that we attribute to a delay in mammary gland development and not to a direct effect of SIRT1 on carcinogenesis. These results are consistent with previous evidence suggesting that Sirt1 is not a tumor promoter or a tumor suppressor.
Collapse
Affiliation(s)
| | - Danielle Dewar-Darch
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Karen E. Jardine
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael W. McBurney
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
94
|
Knight JRP, Allison SJ, Milner J. Active regulator of SIRT1 is required for cancer cell survival but not for SIRT1 activity. Open Biol 2013; 3:130130. [PMID: 24258275 PMCID: PMC3843821 DOI: 10.1098/rsob.130130] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/25/2013] [Indexed: 01/30/2023] Open
Abstract
The NAD(+)-dependent deacetylase SIRT1 is involved in diverse cellular processes, and has also been linked with multiple disease states. Among these, SIRT1 expression negatively correlates with cancer survival in both laboratory and clinical studies. Active regulator of SIRT1 (AROS) was the first reported post-transcriptional regulator of SIRT1 activity, enhancing SIRT1-mediated deacetylation and downregulation of the SIRT1 target p53. However, little is known regarding the role of AROS in regulation of SIRT1 during disease. Here, we report the cellular and molecular effects of RNAi-mediated AROS suppression, comparing this with the role of SIRT1 in a panel of human cell lines of both cancerous and non-cancerous origins. Unexpectedly, AROS is found to vary in its modulation of p53 acetylation according to cell context. AROS suppresses p53 acetylation only following the application of cell damaging stress, whereas SIRT1 suppresses p53 under all conditions analysed. This supplements the original characterization of AROS but indicates that SIRT1 activity can persist following suppression of AROS. We also demonstrate that knockdown of AROS induces apoptosis in three cancer cell lines, independent of p53 activation. Importantly, AROS is not required for the viability of three non-cancer cell lines indicating a putative role for AROS in specifically promoting cancer cell survival.
Collapse
|
95
|
Shimada K, Anai S, Fujii T, Tanaka N, Fujimoto K, Konishi N. Syndecan-1 (CD138) contributes to prostate cancer progression by stabilizing tumour-initiating cells. J Pathol 2013; 231:495-504. [DOI: 10.1002/path.4271] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Keiji Shimada
- Department of Pathology; Nara Medical University School of Medicine; Japan
| | - Satoshi Anai
- Department of Urology; Nara Medical University School of Medicine; Japan
| | - Tomomi Fujii
- Department of Pathology; Nara Medical University School of Medicine; Japan
| | - Nobumichi Tanaka
- Department of Urology; Nara Medical University School of Medicine; Japan
| | - Kiyohide Fujimoto
- Department of Urology; Nara Medical University School of Medicine; Japan
| | - Noboru Konishi
- Department of Pathology; Nara Medical University School of Medicine; Japan
| |
Collapse
|
96
|
Abstract
Innate resistance to various therapeutic interventions is a hallmark of cancer. In recent years, acquired resistance has emerged as a daunting challenge to targeted cancer therapy, which abolishes the efficacy of otherwise successful targeting drugs. Cancer cells gain the resistance property through a variety of mechanisms in primary and metastatic cancers, involving cellular intrinsic and extrinsic factors. Increasing evidence suggests that the mammalian stress response gene sirtuin 1 (SIRT1) plays a critical role in multiple aspects of cancer drug resistance. SIRT1 decreases drug penetration, confers proliferation and antiapoptotic survival advantages to cancer cells, facilitates acquired resistance through genetic mutations, promotes the survival of cancer stem cells, and changes the tumor microenvironment for resistance in cell-autonomous and -nonautonomous manners. This article provides an overview of research advances in the roles of SIRT1 in cancer drug resistance and highlights the prospect of targeting SIRT1 as a new strategy to overcome cancer drug resistance and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | | |
Collapse
|
97
|
Davison CA, Durbin SM, Thau MR, Zellmer VR, Chapman SE, Diener J, Wathen C, Leevy WM, Schafer ZT. Antioxidant enzymes mediate survival of breast cancer cells deprived of extracellular matrix. Cancer Res 2013; 73:3704-15. [PMID: 23771908 DOI: 10.1158/0008-5472.can-12-2482] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metastasis by cancer cells relies upon the acquisition of the ability to evade anoikis, a cell death process elicited by detachment from extracellular matrix (ECM). The molecular mechanisms that ECM-detached cancer cells use to survive are not understood. Striking increases in reactive oxygen species (ROS) occur in ECM-detached mammary epithelial cells, threatening cell viability by inhibiting ATP production, suggesting that ROS must be neutralized if cells are to survive ECM-detachment. Here, we report the discovery of a prominent role for antioxidant enzymes, including catalase and superoxide dismutase, in facilitating the survival of breast cancer cells after ECM-detachment. Enhanced expression of antioxidant enzymes in nonmalignant mammary epithelial cells detached from ECM resulted in ATP elevation and survival in the luminal space of mammary acini. Conversely, silencing antioxidant enzyme expression in multiple breast cancer cell lines caused ATP reduction and compromised anchorage-independent growth. Notably, antioxidant enzyme-deficient cancer cells were compromised in their ability to form tumors in mice. In aggregate, our results reveal a vital role for antioxidant enzyme activity in maintaining metabolic activity and anchorage-independent growth in breast cancer cells. Furthermore, these findings imply that eliminating antioxidant enzyme activity may be an effective strategy to enhance susceptibility to cell death in cancer cells that may otherwise survive ECM-detachment.
Collapse
Affiliation(s)
- Calli A Davison
- Department of Biological Sciences; Notre Dame Integrated Imaging Facility, and Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Mellini P, Kokkola T, Suuronen T, Salo HS, Tolvanen L, Mai A, Lahtela-Kakkonen M, Jarho EM. Screen of pseudopeptidic inhibitors of human sirtuins 1-3: two lead compounds with antiproliferative effects in cancer cells. J Med Chem 2013; 56:6681-95. [PMID: 23927550 DOI: 10.1021/jm400438k] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the past few years sirtuins have gained growing attention for their involvement in many biological processes such as cellular metabolism, apoptosis, aging and inflammation. In this contribution, we report the synthesis of a library of thioacetylated pseudopeptides that were screened against human sirtuins 1-3 to reveal their in vitro inhibition activities. Molecular modeling studies were performed to acquire data about the binding modes of the inhibitors. Three sirtuin inhibitors were subjected to cellular studies, and all of them showed an increase in acetylation of Lys382 of p53 after DNA damage. Furthermore, two of the compounds were able to inhibit both A549 lung carcinoma and MCF-7 breast carcinoma cell growth in micromolar concentration with the ability to arrest cancer cell cycle in the G1 phase.
Collapse
Affiliation(s)
- Paolo Mellini
- School of Pharmacy and ∥Department of Neurology, Institute of Clinical Medicine , University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Dalamaga M. Obesity, insulin resistance, adipocytokines and breast cancer: New biomarkers and attractive therapeutic targets. World J Exp Med 2013; 3:34-42. [PMID: 24520544 PMCID: PMC3905595 DOI: 10.5493/wjem.v3.i3.34] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/08/2013] [Accepted: 08/17/2013] [Indexed: 02/06/2023] Open
Abstract
Worldwide, breast cancer (BC) represents the most common type of non-skin human malignancy and the second leading cause of cancer-related deaths amid women in Western countries. Obesity and its metabolic complications have rapidly become major global health issues and are associated with increased risk for cancer, especially BC in postmenopausal women. Adipose tissue is considered as a genuine endocrine organ secreting a variety of bioactive adipokines, such as leptin, adiponectin, resistin and nicotinamide phosphoribosyl-transferase/visfatin. Recent evidence has indicated that the constellation of obesity, insulin resistance and adipokines is associated with the risk and prognosis of postmenopausal BC. Direct evidence is growing rapidly supporting the stimulating and/or inhibiting role of adipokines in the process of development and progression of BC. Adipokines could exert their effects on the normal and neoplastic mammary tissue by endocrine, paracrine and autocrine mechanisms. Recent studies support a role of adipokines as novel risk factors and potential diagnostic and prognostic biomarkers in BC. This editorial aims at providing important insights into the potential pathophysiological mechanisms linking adipokines to the etiopathogenesis of BC in the context of a dysfunctional adipose tissue and insulin resistance in obesity. A better understanding of these mechanisms may be important for the development of attractive preventive and therapeutic strategies against obesity-related breast malignancy.
Collapse
|
100
|
MicroRNA-126 inhibits osteosarcoma cells proliferation by targeting Sirt1. Tumour Biol 2013; 34:3871-7. [PMID: 23877372 DOI: 10.1007/s13277-013-0974-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 06/24/2013] [Indexed: 12/31/2022] Open
Abstract
Numerous studies have recently suggested that miRNAs contribute to the development of various types of human cancer as well as to their proliferation and metastasis. The aim of this study was to investigate the functional significance of miR-126 and to identify its possible target genes in osteosarcoma (OS) cells. Here, we found that expression level of miR-126 was reduced in osteosarcoma cells in comparison with the adjacent normal tissues. The enforced expression of miR-126 was able to inhibit cell proliferation in U2OS and MG63 cells, while miR-126 antisense oligonucleotides (antisense miR-126) promoted cell proliferation. At the molecular level, our results further revealed that expression of Sirt1, a member of histone deacetylase, was negatively regulated by miR-126. Therefore, the data reported here demonstrate that miR-126 is an important regulator in osteosarcoma, which will contribute to better understanding of the important misregulated miRNAs in osteosarcoma cells.
Collapse
|