1
|
Mauro MS, Martin SL, Dumont J, Shirasu-Hiza M, Canman JC. Patterning, regulation, and role of FoxO/DAF-16 in the early embryo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.594029. [PMID: 38798632 PMCID: PMC11118310 DOI: 10.1101/2024.05.13.594029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Insulin resistance and diabetes are associated with many health issues including higher rates of birth defects and miscarriage during pregnancy. Because insulin resistance and diabetes are both associated with obesity, which also affects fertility, the role of insulin signaling itself in embryo development is not well understood. A key downstream target of the insulin/insulin-like growth factor signaling (IIS) pathway is the forkhead family transcription factor FoxO (DAF-16 in C. elegans ). Here, we used quantitative live imaging to measure the patterning of endogenously tagged FoxO/DAF-16 in the early worm embryo. In 2-4-cell stage embryos, FoxO/DAF-16 initially localized uniformly to all cell nuclei, then became dramatically enriched in germ precursor cell nuclei beginning at the 8-cell stage. This nuclear enrichment in early germ precursor cells required germ fate specification, PI3K (AGE-1)- and PTEN (DAF-18)-mediated phospholipid regulation, and the deubiquitylase USP7 (MATH-33), yet was unexpectedly insulin receptor (DAF-2)- and AKT-independent. Functional analysis revealed that FoxO/DAF-16 acts as a cell cycle pacer for early cleavage divisions-without FoxO/DAF-16 cell cycles were shorter than in controls, especially in germ lineage cells. These results reveal the germ lineage specific patterning, upstream regulation, and cell cycle role for FoxO/DAF-16 during early C. elegans embryogenesis.
Collapse
|
2
|
Chang X, Wang J, Bian J, Liu Z, Guo M, Li Z, Wu Y, Zhai X, Zuo D. 1-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)-3-(2-(dimethylamino)ethyl)imidazolidin-2-one (ZX-42) inhibits cell proliferation and induces apoptosis via inhibiting ALK and its downstream pathways in Karpas299 cells. Toxicol Appl Pharmacol 2022; 450:116156. [PMID: 35803438 DOI: 10.1016/j.taap.2022.116156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 11/19/2022]
Abstract
Anaplastic lymphoma kinase (ALK) belongs to the family of receptor tyrosine kinases. Recently, the incidence of anaplastic large cell lymphoma (ALCL) with ALK rearrangement has raised considerably. The application of ALK-targeted inhibitors such as ceritinib provides an effective therapy for the treatment of ALK-positive cancers. However, with the prolongation of treatment time, the emergence of resistance is inevitable. We found that 1-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-3-methoxyphenyl)-3-(2-(dimethylamino)ethyl)imidazolidin-2-one (ZX-42), a novel ceritinib derivative, could inhibit the proliferation of ALK-positive ALCL cells, induce the apoptosis of Karpas299 cells through the mitochondrial pathway in a caspase-dependent manner. In addition, ZX-42 could suppress ALK and downstream pathways including PI3K/Akt, Erk and JAK3/STAT3 and reduce the nuclear translocation of NFκB by inhibiting TRAF2/IKK/IκB pathway. Taken together, our findings indicate that ZX-42 shows more effective activity than ceritinib against ALK-positive ALCL. We hope this study can provide a direction for the structural modification of ceritinib and lay the foundation for the further development of clinical research in ALK-positive ALCL.
Collapse
Affiliation(s)
- Xing Chang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Junfang Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Jiang Bian
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Zi Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Ming Guo
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| |
Collapse
|
3
|
Abstract
Oxidative stress is caused by homeostasis disrupted by excessively increased reactive oxygen species (ROS) due to intrinsic or extrinsic causes. Among diseases caused by the abnormal induction of ROS, cancer is a representative disease that shows gender specificity in the development and malignancy. Females have the advantage of longer life expectancy than males because of the genetic advantages derived from X chromosomes, the antioxidant protective function by estrogen, and the decrease in exposure to extrinsic risk factors such as alcohol and smoking. This study first examines the ordinary biological responses to oxidative stress and the effects of ROS on the cancer progression and describes the differences in cancer incidence and mortality by gender and the differences in oxidative stress affected by sex hormones. This paper summarized how several important transcription factors regulate ROS-induced stress and in vivo responses, and how their expression is changed by sex hormones. Estrogen is associated with disease resistance and greater mitochondrial function, and reduces mitochondrial damage and ROS production in females than in males. In addition, estrogen affects the activation of nuclear factor-erythroid 2 p45-related factor (NRF) 2 and the regulation of other antioxidant-related transcription factors through NRF2, leading to benefits in females. Because ROS have a variety of molecular targets in cells, the effective cancer treatment requires understanding the potential of ROS and focusing on the characteristics of the research target such as patient's gender. Therefore, this review intends to emphasize the necessity of discussing gender specificity as a new therapeutic approach for efficient regulation of ROS considering individual specificity.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Chemistry, College of Science and Technology, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
4
|
Calissi G, Lam EWF, Link W. Therapeutic strategies targeting FOXO transcription factors. Nat Rev Drug Discov 2021; 20:21-38. [PMID: 33173189 DOI: 10.1038/s41573-020-0088-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
FOXO proteins are transcription factors that are involved in numerous physiological processes and in various pathological conditions, including cardiovascular disease, cancer, diabetes and chronic neurological diseases. For example, FOXO proteins are context-dependent tumour suppressors that are frequently inactivated in human cancers, and FOXO3 is the second most replicated gene associated with extreme human longevity. Therefore, pharmacological manipulation of FOXO proteins is a promising approach to developing therapeutics for cancer and for healthy ageing. In this Review, we overview the role of FOXO proteins in health and disease and discuss the pharmacological approaches to modulate FOXO function.
Collapse
Affiliation(s)
- Giampaolo Calissi
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, UK
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
5
|
Zhou X, Zhang X, Wu Z, Xu X, Guo M, Zhai X, Zuo D, Wu Y. The novel ALK inhibitor ZX-29 induces apoptosis through inhibiting ALK and inducing ROS-mediated endoplasmic reticulum stress in Karpas299 cells. J Biochem Mol Toxicol 2020; 35:e22666. [PMID: 33140567 DOI: 10.1002/jbt.22666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 11/08/2022]
Abstract
It is a well-known fact that 60%-85% of anaplastic large cell lymphoma (ALCL) is mainly driven by the anaplastic lymphoma kinase (ALK) fusion protein. Although ALK-positive ALCL patients respond significantly to ALK inhibitors, the development of resistance is inevitable, which requires the development of new therapeutic strategies for ALK-positive ALCL. Here, we investigated the anticancer activities of N-(2((5-chloro-2-((2-methoxy-6-(4-methylpiperazin-1-yl)pyridin-3yl)amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (ZX-29), a newly synthesized ALK inhibitor, against nucleophosmin-ALK-positive cell line Karpas299. We demonstrated that ZX-29 decreased Karpas299 cells growth and had better cytotoxicity than ceritinib, which was mediated through downregulating the expression of ALK and related proteins, inducing cell cycle arrest, and promoting cell apoptosis. Moreover, ZX-29-induced cell apoptosis by inducing endoplasmic reticulum stress (ERS). In addition, ZX-29 increased the generation of reactive oxygen species (ROS), and cells pretreatment with N-acetyl- l-cysteine could attenuate ZX-29-induced cell apoptosis and ERS. Taken together, ZX-29 inhibited Karpas299 cell proliferation and induced apoptosis through inhibiting ALK and its downstream protein expression and inducing ROS-mediated ERS. Therefore, our results provide evidence for a novel antitumor candidate for the further investigation.
Collapse
Affiliation(s)
- Xuejiao Zhou
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoning Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhuzhu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaobo Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ming Guo
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
6
|
Abstract
Forkhead box O (FOXO) transcription factors regulate diverse biological processes, affecting development, metabolism, stem cell maintenance and longevity. They have also been increasingly recognised as tumour suppressors through their ability to regulate genes essential for cell proliferation, cell death, senescence, angiogenesis, cell migration and metastasis. Mechanistically, FOXO proteins serve as key connection points to allow diverse proliferative, nutrient and stress signals to converge and integrate with distinct gene networks to control cell fate, metabolism and cancer development. In consequence, deregulation of FOXO expression and function can promote genetic disorders, metabolic diseases, deregulated ageing and cancer. Metastasis is the process by which cancer cells spread from the primary tumour often via the bloodstream or the lymphatic system and is the major cause of cancer death. The regulation and deregulation of FOXO transcription factors occur predominantly at the post-transcriptional and post-translational levels mediated by regulatory non-coding RNAs, their interactions with other protein partners and co-factors and a combination of post-translational modifications (PTMs), including phosphorylation, acetylation, methylation and ubiquitination. This review discusses the role and regulation of FOXO proteins in tumour initiation and progression, with a particular emphasis on cancer metastasis. An understanding of how signalling networks integrate with the FOXO transcription factors to modulate their developmental, metabolic and tumour-suppressive functions in normal tissues and in cancer will offer a new perspective on tumorigenesis and metastasis, and open up therapeutic opportunities for malignant diseases.
Collapse
Affiliation(s)
- Yannasittha Jiramongkol
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
| |
Collapse
|
7
|
Wu Z, Zhu Q, Zhang Y, Yin Y, Kang D, Cao R, Tian Q, Lu S, Liu P. EGFR‑associated pathways involved in traditional Chinese medicine (TCM)‑1‑induced cell growth inhibition, autophagy and apoptosis in prostate cancer. Mol Med Rep 2018; 17:7875-7885. [PMID: 29620175 DOI: 10.3892/mmr.2018.8818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 11/16/2017] [Indexed: 11/05/2022] Open
Abstract
Traditional Chinese medicine (TCM) has the synergistic effect of the combination of a single ingredient and a monomer, and systemic and local therapeutic effects in cancer treatment, through which TCM is able to enhance the curative effect and reduce the side effects. The present study analyzed the effect of TCM‑1 (an anti‑cancer TCM) on prostate cancer (PCa) cell lines, and studied in detail the mechanism of cell death induced by TCM‑1 in vitro and in vivo. From the present results, it was identified for the first time, to the best of our knowledge, that TCM‑1 arrested the cell cycle at the G1 phase, decreased cell viability and increased nuclear rupture in a dose‑dependent manner; these effects finally resulted in apoptosis in PCa cells. At the molecular level, the data demonstrated that TCM‑1 competitively acted on epidermal growth factor receptor (EGFR) with EGF, and suppressed the auto‑phosphorylation and activity of EGFR. Inhibition of EGFR further suppressed the downstream phosphatidylinositol 3‑kinase (PI3K)/RAC‑α serine/threonine‑protein kinase (AKT) and RAF proto‑oncogene serine/threonine‑protein kinase/extracellular signal regulated kinase signaling pathways and resulted in a decrease in the phosphorylated‑forkhead box protein O1 (at Ser256, Thr24 and Ser319) expression level, and induced cell growth inhibition and apoptosis by regulating the expression of apoptosis‑and cell cycle‑associated genes. In addition, TCM‑1 markedly inhibited the PI3K/AKT/serine/threonine‑protein kinase mTOR signaling pathway and induced cell autophagy by downregulating the phosphorylation of p70S6K and upregulating the levels of Beclin‑1 and microtubule‑associated protein light chain‑3II. In vivo, the TCM‑1‑treated group exhibited a significant decrease in tumor volume compared with the negative control group in subcutaneous xenograft nude mice by inhibiting EGFR‑associated signaling pathways. Therefore, the bio‑functions of Chinese medicine TCM‑1 in inducing PCa cell growth inhibition, autophagy and apoptosis suggested that TCM‑1 may have clinical potential for the treatment of patients with PCa.
Collapse
Affiliation(s)
- Zhaomeng Wu
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Qingyi Zhu
- Laboratory of Molecular Biology, Jiangsu Province Hospital of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Yu Zhang
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Yingying Yin
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Dan Kang
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Runyi Cao
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Qian Tian
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Shan Lu
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| | - Ping Liu
- Jiangsu Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
8
|
Wu Z, Zhu Q, Yin Y, Kang D, Cao R, Tian Q, Zhang Y, Lu S, Liu P. Traditional Chinese Medicine CFF-1 induced cell growth inhibition, autophagy, and apoptosis via inhibiting EGFR-related pathways in prostate cancer. Cancer Med 2018. [PMID: 29533017 PMCID: PMC5911605 DOI: 10.1002/cam4.1419] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traditional Chinese medicine (TCM) has a combined therapeutic result in cancer treatment by integrating holistic and local therapeutical effects, by which TCM can enhance the curative effect and reduce the side effect. In this study, we analyzed the effect of CFF‐1 (alcohol extract from an anticancer compound Chinese medicine) on prostate cancer (PCa) cell lines and studied in detail the mechanism of cell death induced by CFF‐1 in vitro and in vivo. From our data, we found for the first time that CFF‐1 obviously arrested cell cycle in G1 phase, decreased cell viability and then increased nuclear rupture in a dose‐dependent manner and finally resulted in apoptosis in prostate cancer cells. In molecular level, our data showed that CFF‐1 induced inhibition of EGFR auto‐phosphorylation and inactivation of EGFR. Disruption of EGFR activity in turn suppressed downstream PI3K/AKT and Raf/Erk signal pathways, resulted in the decrease of p‐FOXO1 (Ser256) and regulated the expression of apoptosis‐related and cycle‐related genes. Moreover, CFF‐1 markedly induced cell autophagy through inhibiting PI3K/AKT/mTOR pathway and then up‐regulating Beclin‐1 and LC‐3II and down‐regulating phosphorylation of p70S6K. In vivo, CFF‐1‐treated group exhibited a significant decrease in tumor volume compared with the negative control group in subcutaneous xenograft tumor in nude mice via inhibiting EGFR‐related signal pathways. Thus, bio‐functions of Chinese medicine CFF‐1 in inducing PCa cell growth inhibition, autophagy, and apoptosis suggested that CFF‐1 had the clinical potential to treat patients with prostate cancer.
Collapse
Affiliation(s)
- Zhaomeng Wu
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Qingyi Zhu
- Laboratory of Molecular BiologyJiangsu Province Hospital of TCMNanjingJiangsu210029China
| | - Yingying Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Dan Kang
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Runyi Cao
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Qian Tian
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Yu Zhang
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Shan Lu
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| | - Ping Liu
- Jiangsu Province Key Laboratory for Molecular and Medicine BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingJiangsu210023China
| |
Collapse
|
9
|
Drosophila Kruppel homolog 1 represses lipolysis through interaction with dFOXO. Sci Rep 2017; 7:16369. [PMID: 29180716 PMCID: PMC5703730 DOI: 10.1038/s41598-017-16638-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/15/2017] [Indexed: 12/29/2022] Open
Abstract
Transcriptional coordination is a vital process contributing to metabolic homeostasis. As one of the key nodes in the metabolic network, the forkhead transcription factor FOXO has been shown to interact with diverse transcription co-factors and integrate signals from multiple pathways to control metabolism, oxidative stress response, and cell cycle. Recently, insulin/FOXO signaling has been implicated in the regulation of insect development via the interaction with insect hormones, such as ecdysone and juvenile hormone. In this study, we identified an interaction between Drosophila FOXO (dFOXO) and the zinc finger transcription factor Kruppel homolog 1 (Kr-h1), one of the key players in juvenile hormone signaling. We found that Kr-h1 mutants show delayed larval development and altered lipid metabolism, in particular induced lipolysis upon starvation. Notably, Kr-h1 physically and genetically interacts with dFOXO in vitro and in vivo to regulate the transcriptional activation of insulin receptor (InR) and adipose lipase brummer (bmm). The transcriptional co-regulation by Kr-h1 and dFOXO may represent a broad mechanism by which Kruppel-like factors integrate with insulin signaling to maintain metabolic homeostasis and coordinate organism growth.
Collapse
|
10
|
Sullivan DA, Rocha EM, Aragona P, Clayton JA, Ding J, Golebiowski B, Hampel U, McDermott AM, Schaumberg DA, Srinivasan S, Versura P, Willcox MDP. TFOS DEWS II Sex, Gender, and Hormones Report. Ocul Surf 2017; 15:284-333. [PMID: 28736336 DOI: 10.1016/j.jtos.2017.04.001] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 04/16/2017] [Indexed: 12/21/2022]
Abstract
One of the most compelling features of dry eye disease (DED) is that it occurs more frequently in women than men. In fact, the female sex is a significant risk factor for the development of DED. This sex-related difference in DED prevalence is attributed in large part to the effects of sex steroids (e.g. androgens, estrogens), hypothalamic-pituitary hormones, glucocorticoids, insulin, insulin-like growth factor 1 and thyroid hormones, as well as to the sex chromosome complement, sex-specific autosomal factors and epigenetics (e.g. microRNAs). In addition to sex, gender also appears to be a risk factor for DED. "Gender" and "sex" are words that are often used interchangeably, but they have distinct meanings. "Gender" refers to a person's self-representation as a man or woman, whereas "sex" distinguishes males and females based on their biological characteristics. Both gender and sex affect DED risk, presentation of the disease, immune responses, pain, care-seeking behaviors, service utilization, and myriad other facets of eye health. Overall, sex, gender and hormones play a major role in the regulation of ocular surface and adnexal tissues, and in the difference in DED prevalence between women and men. The purpose of this Subcommittee report is to review and critique the nature of this role, as well as to recommend areas for future research to advance our understanding of the interrelationships between sex, gender, hormones and DED.
Collapse
Affiliation(s)
- David A Sullivan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| | - Eduardo M Rocha
- Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Pasquale Aragona
- Department of Biomedical Sciences, Ocular Surface Diseases Unit, University of Messina, Messina, Sicily, Italy
| | - Janine A Clayton
- National Institutes of Health Office of Research on Women's Health, Bethesda, MD, USA
| | - Juan Ding
- Schepens Eye Research Institute, Massachusetts Eye & Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Blanka Golebiowski
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Ulrike Hampel
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alison M McDermott
- The Ocular Surface Institute, College of Optometry, University of Houston, Houston, TX, USA
| | - Debra A Schaumberg
- Harvard School of Public Health, Boston, MA, USA; University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sruthi Srinivasan
- Centre for Contact Lens Research, School of Optometry, University of Waterloo, Ontario, Canada
| | - Piera Versura
- Department of Specialized, Experimental, and Diagnostic Medicine, University of Bologna, Bologna, Italy
| | - Mark D P Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
11
|
Hammerich KH, Frolov A, Li R, Ittmann M, Ayala GE. Cellular interactions of the phosphorylated form of AKT in prostate cancer. Hum Pathol 2017; 63:98-109. [DOI: 10.1016/j.humpath.2017.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/25/2017] [Accepted: 02/23/2017] [Indexed: 11/15/2022]
|
12
|
Andrisse S, Childress S, Ma Y, Billings K, Chen Y, Xue P, Stewart A, Sonko ML, Wolfe A, Wu S. Low-Dose Dihydrotestosterone Drives Metabolic Dysfunction via Cytosolic and Nuclear Hepatic Androgen Receptor Mechanisms. Endocrinology 2017; 158:531-544. [PMID: 27967242 PMCID: PMC5460775 DOI: 10.1210/en.2016-1553] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022]
Abstract
Androgen excess in women is associated with metabolic dysfunction (e.g., obesity, hyperinsulinemia, insulin resistance, and increased risk of type 2 diabetes) and reproductive dysfunction (e.g., polycystic ovaries, amenorrhea, dysregulated gonadotropin release, and infertility). We sought to identify the effects of androgen excess on glucose metabolic dysfunction and the specific mechanisms of action by which androgens are inducing pathology. We developed a mouse model that displayed pathophysiological serum androgen levels with normal body mass/composition to ensure that the phenotypes were directly from androgens and not an indirect consequence of obesity. We performed reproductive tests, metabolic tests, and hormonal assays. Livers were isolated and examined via molecular, biochemical, and histological analysis. Additionally, a low-dose dihydrotestosterone (DHT) cell model using H2.35 mouse hepatocytes was developed to study androgen effects on hepatic insulin signaling. DHT mice demonstrated impaired estrous cyclicity; few corpora lutea in the ovaries; glucose, insulin, and pyruvate intolerance; and lowered hepatic insulin action. Mechanistically, DHT increased hepatic androgen-receptor binding to phosphoinositide-3-kinase (PI3K)-p85, resulting in dissociation of PI3K-p85 from PI3K-p110, leading to reduced PI3K activity and decreased p-AKT and, thus, lowered insulin action. DHT increased gluconeogenesis via direct transcriptional regulation of gluconeogenic enzymes and coactivators. The hepatocyte model recapitulated the in vivo findings. The DHT-induced hepatocyte insulin resistance was reversed by the androgen-receptor antagonist, flutamide. These findings present a phenotype (i.e., impaired glucose tolerance and disrupted glucose metabolism) in a lean hyperandrogenemia model (low-dose DHT) and data to support 2 molecular mechanisms that help drive androgen-induced impaired glucose metabolism.
Collapse
Affiliation(s)
- Stanley Andrisse
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shameka Childress
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Yaping Ma
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Katelyn Billings
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Yi Chen
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ping Xue
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ashley Stewart
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Momodou L Sonko
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Chattopadhyay I, Wang J, Qin M, Gao L, Holtz R, Vessella RL, Leach RW, Gelman IH. Src promotes castration-recurrent prostate cancer through androgen receptor-dependent canonical and non-canonical transcriptional signatures. Oncotarget 2017; 8:10324-10347. [PMID: 28055971 PMCID: PMC5354662 DOI: 10.18632/oncotarget.14401] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Progression of prostate cancer (PC) to castration-recurrent growth (CRPC) remains dependent on sustained expression and transcriptional activity of the androgen receptor (AR). A major mechanism contributing to CRPC progression is through the direct phosphorylation and activation of AR by Src-family (SFK) and ACK1 tyrosine kinases. However, the AR-dependent transcriptional networks activated by Src during CRPC progression have not been elucidated. Here, we show that activated Src (Src527F) induces androgen-independent growth in human LNCaP cells, concomitant with its ability to induce proliferation/survival genes normally induced by dihydrotestosterone (DHT) in androgen-dependent LNCaP and VCaP cells. Src induces additional gene signatures unique to CRPC cell lines, LNCaP-C4-2 and CWR22Rv1, and to CRPC LuCaP35.1 xenografts. By comparing the Src-induced AR-cistrome and/or transcriptome in LNCaP to those in CRPC and LuCaP35.1 tumors, we identified an 11-gene Src-regulated CRPC signature consisting of AR-dependent, AR binding site (ARBS)-associated genes whose expression is altered by DHT in LNCaP[Src527F] but not in LNCaP cells. The differential expression of a subset (DPP4, BCAT1, CNTNAP4, CDH3) correlates with earlier PC metastasis onset and poorer survival, with the expression of BCAT1 required for Src-induced androgen-independent proliferation. Lastly, Src enhances AR binding to non-canonical ARBS enriched for FOXO1, TOP2B and ZNF217 binding motifs; cooperative AR/TOP2B binding to a non-canonical ARBS was both Src- and DHT-sensitive and correlated with increased levels of Src-induced phosphotyrosyl-TOP2B. These data suggest that CRPC progression is facilitated via Src-induced sensitization of AR to intracrine androgen levels, resulting in the engagement of canonical and non-canonical ARBS-dependent gene signatures.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Binding Sites
- Cell Line, Tumor
- Cell Proliferation
- Dihydrotestosterone/pharmacology
- Disease Progression
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Phosphorylation
- Promoter Regions, Genetic
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/enzymology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Androgen/drug effects
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Signal Transduction
- Time Factors
- Transcription, Genetic/drug effects
- Transcriptome
- Transfection
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Indranil Chattopadhyay
- Department of Life Sciences, School of Basic and Applied Science, Central University of Tamil Nadu, Thiruvarur, Tamil Nadu, India
| | - Jianmin Wang
- Department of Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Maochun Qin
- Department of Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lingqiu Gao
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Renae Holtz
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Robert W. Leach
- Lewis-Sigler Institute for Integrative Genomics, Princeton, NJ, USA
| | - Irwin H. Gelman
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
14
|
Pal M, Gupta S. Testosterone supplementation improves glucose homeostasis despite increasing hepatic insulin resistance in male mouse model of type 2 diabetes mellitus. Nutr Diabetes 2016; 6:e236. [PMID: 27941939 PMCID: PMC5223134 DOI: 10.1038/nutd.2016.45] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 12/12/2022] Open
Abstract
Clinical studies have revealed that testosterone supplementation had a positive effect on glucose homeostasis in type 2 diabetes mellitus (T2DM), but did not address how testosterone supplementation affected insulin responsiveness in the liver, a key glucose homeostatic organ. In this study, we aimed to study the effect of testosterone supplementation on hepatic insulin responsiveness and glucose homeostasis through liver in male high-fat diet-induced T2DM mice. Testosterone treatment to T2DM animals showed reduced hepatic glucose output. Testosterone inhibited the insulin signaling in liver, thus increased insulin resistance. However, testosterone treatment inactivated GSK3α independent of PI3K/AKT pathway and inhibited FOXO1 By interaction of androgen receptor to FOXO1 and downregulated PEPCK, causing repression of gluconeogenic pathway, which is otherwise upregulated in T2DM, resulted in better glucose homeostasis.
Collapse
Affiliation(s)
- M Pal
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, India
| | - S Gupta
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
15
|
Lee J, Park SH. Tumor-suppressive activity of 1,25-dihydroxyvitamin D3 against kidney cancer cells via up-regulation of FOXO3. Biosci Biotechnol Biochem 2016; 80:1947-53. [PMID: 27181027 DOI: 10.1080/09168451.2016.1184561] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
1,25-Dihydroxyvitamin D3 has been known to have the tumor-suppressive activity in various kinds of tumors. However, the exact effect and working mechanism of 1,25-dihydroxyvitamin D3 on the tumor-suppressive activity in human kidney cancer cells remains poorly understood. 1,25-Dihydroxyvitamin D3 has cytotoxicity to ACHN cells and inhibited ACHN cell proliferation compared to the vehicle control. 1,25-Dihydroxyvitamin D3 increased the expression of the cleaved PARP1, active Caspase3, Bax, and Bim but decreased the expression of Bcl2 in ACHN cells. Moreover, 1,25-dihydroxyvitamin D3 down-regulated the phosphorylated Akt and Erk which might lead to apoptosis through activation of FOXO3 in ACHN cells. Transfection of siRNA against FOXO3 attenuated the pro-apoptotic BimEL expression in ACHN cells treated with 1,25-dihydroxyvitamin D3. These results suggest that FOXO3 is involved in the apoptosis induced by 1,25-dihydroxyvitamin D3.
Collapse
Affiliation(s)
- Jongsung Lee
- a Department of Genetic Engineering , Sungkyunkwan University , Suwon , Korea
| | - See-Hyoung Park
- b Department of Bio and Chemical Engineering , Hongik University , Sejong , Korea
| |
Collapse
|
16
|
Integrated network model provides new insights into castration-resistant prostate cancer. Sci Rep 2015; 5:17280. [PMID: 26603105 PMCID: PMC4658549 DOI: 10.1038/srep17280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 10/28/2015] [Indexed: 12/12/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is the main challenge for prostate cancer treatment. Recent studies have indicated that extending the treatments to simultaneously targeting different pathways could provide better approaches. To better understand the regulatory functions of different pathways, a system-wide study of CRPC regulation is necessary. For this purpose, we constructed a comprehensive CRPC regulatory network by integrating multiple pathways such as the MEK/ERK and the PI3K/AKT pathways. We studied the feedback loops of this network and found that AKT was involved in all detected negative feedback loops. We translated the network into a predictive Boolean model and analyzed the stable states and the control effects of genes using novel methods. We found that the stable states naturally divide into two obvious groups characterizing PC3 and DU145 cells respectively. Stable state analysis further revealed that several critical genes, such as PTEN, AKT, RAF, and CDKN2A, had distinct expression behaviors in different clusters. Our model predicted the control effects of many genes. We used several public datasets as well as FHL2 overexpression to verify our finding. The results of this study can help in identifying potential therapeutic targets, especially simultaneous targets of multiple pathways, for CRPC.
Collapse
|
17
|
Diep CH, Knutson TP, Lange CA. Active FOXO1 Is a Key Determinant of Isoform-Specific Progesterone Receptor Transactivation and Senescence Programming. Mol Cancer Res 2015; 14:141-62. [PMID: 26577046 DOI: 10.1158/1541-7786.mcr-15-0431] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/04/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED Progesterone promotes differentiation coupled to proliferation and prosurvival in the breast, but inhibits estrogen-driven growth in the reproductive tract and ovaries. Herein, it is demonstrated, using progesterone receptor (PR) isoform-specific ovarian cancer model systems, that PR-A and PR-B promote distinct gene expression profiles that differ from PR-driven genes in breast cancer cells. In ovarian cancer models, PR-A primarily regulates genes independently of progestin, while PR-B is the dominant ligand-dependent isoform. Notably, FOXO1 and the PR/FOXO1 target gene p21 (CDKN1A) are repressed by PR-A, but induced by PR-B. In the presence of progestin, PR-B, but not PR-A, robustly induced cellular senescence via FOXO1-dependent induction of p21 and p15 (CDKN2B). Chromatin immunoprecipitation (ChIP) assays performed on PR isoform-specific cells demonstrated that while each isoform is recruited to the same PRE-containing region of the p21 promoter in response to progestin, only PR-B elicits active chromatin marks. Overexpression of constitutively active FOXO1 in PR-A-expressing cells conferred robust ligand-dependent upregulation of the PR-B target genes GZMA, IGFBP1, and p21, and induced cellular senescence. In the presence of endogenous active FOXO1, PR-A was phosphorylated on Ser294 and transactivated PR-B at PR-B target genes; these events were blocked by the FOXO1 inhibitor (AS1842856). PR isoform-specific regulation of the FOXO1/p21 axis recapitulated in human primary ovarian tumor explants treated with progestin; loss of progestin sensitivity correlated with high AKT activity. IMPLICATIONS This study indicates FOXO1 as a critical component for progesterone signaling to promote cellular senescence and reveals a novel mechanism for transcription factor control of hormone sensitivity.
Collapse
Affiliation(s)
- Caroline H Diep
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Todd P Knutson
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Carol A Lange
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota. Department of Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
18
|
Duan X, Kong Z, Liu Y, Zeng Z, Li S, Wu W, Ji W, Yang B, Zhao Z, Zeng G. β-Arrestin2 Contributes to Cell Viability and Proliferation via the Down-Regulation of FOXO1 in Castration-Resistant Prostate Cancer. J Cell Physiol 2015; 230:2371-81. [PMID: 25752515 DOI: 10.1002/jcp.24963] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 02/17/2015] [Indexed: 12/22/2022]
Abstract
β-Arrestin2 has been identified to act as a corepressor of androgen receptor (AR) signaling by binding to AR and serving as a scaffold to affect the activity and expression of AR in androgen-dependent prostate cancer cells; however, little is known regarding its role in castration-resistant prostate cancer (CRPC) progression. Here, our data demonstrated that β-arrestin2 contributes to the cell viability and proliferation of CRPC via the downregulation of FOXO1 activity and expression. Mechanistically, in addition to its requirement for FOXO1 phosphorylation induced by IGF-1, β-arrestin2 could inhibit FOXO1 activity in an Akt-independent manner and delay FOXO1 dephosphorylation through the inhibition of PP2A phosphatase activity and the attenuation of the interaction between FOXO1 and PP2A. Furthermore, β-arrestin2 could downregulate FOXO1 expression via ubiquitylation and proteasomal degradation. Together, our results identified a novel role for β-arrestin2 in the modulation of the CRPC progress through FOXO1. Thus, the characterization of β-arrestin2 may represent an alternative therapeutic target for CRPC treatment.
Collapse
Affiliation(s)
- Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Zhenzhen Kong
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Yang Liu
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Zhiwen Zeng
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shujue Li
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Wenqi Wu
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Weidong Ji
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Bicheng Yang
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Zhijian Zhao
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Wang F, Marshall CB, Ikura M. Forkhead followed by disordered tail: The intrinsically disordered regions of FOXO3a. INTRINSICALLY DISORDERED PROTEINS 2015; 3:e1056906. [PMID: 28232890 DOI: 10.1080/21690707.2015.1056906] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 12/22/2022]
Abstract
Forkhead box Class O is one of 19 subfamilies of the Forkhead box family, comprising 4 human transcription factors: FOXO1, FOXO3a, FOXO4, and FOXO6, which are involved in many crucial cellular processes. FOXO3a is a tumor suppressor involved in multiple physiological and pathological processes, and plays essential roles in metabolism, cell cycle arrest, DNA repair, and apoptosis. In its role as a transcription factor, the FOXO3a binds a consensus Forkhead response element DNA sequence, and recruits transcriptional coactivators to activate gene transcription. FOXO3a has additional functions, such as regulating p53-mediated apoptosis and activating kinase ATM. With the exception of the structured DNA-binding forkhead domain, most of the FOXO3a sequence comprises intrinsically disordered regions (IDRs), including 3 regions (CR1-3) that are conserved within the FOXO subfamily. Numerous studies have demonstrated that these IDRs directly mediate many of the diverse functions of FOXO3a. These regions contain post-translational modification and protein-protein interaction sites that integrate upstream signals to maintain homeostasis. Thus, the FOXO3a IDRs are emerging as key mediators of diverse regulatory processes, and represent an important target for the future development of therapeutics for FOXO3a-related diseases.
Collapse
Affiliation(s)
- Feng Wang
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Center, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Present affiliation: Department of Biochemistry; Vanderbilt University School of Medicine; Nashville, TN USA
| | - Christopher B Marshall
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Center, Department of Medical Biophysics, University of Toronto , Toronto, Ontario, Canada
| | - Mitsuhiko Ikura
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Center, Department of Medical Biophysics, University of Toronto , Toronto, Ontario, Canada
| |
Collapse
|
20
|
Katsogiannou M, Ziouziou H, Karaki S, Andrieu C, Henry de Villeneuve M, Rocchi P. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev 2015; 41:588-97. [PMID: 25981454 DOI: 10.1016/j.ctrv.2015.05.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 12/17/2022]
Abstract
Prostate cancer has become a real public health issue in industrialized countries, mainly due to patients' relapse by castration-refractory disease after androgen ablation. Castration-resistant prostate cancer is an incurable and highly aggressive terminal stage of prostate cancer, seriously jeopardizing the patient's quality of life and lifespan. The management of castration-resistant prostate cancer is complex and has opened new fields of research during the last decade leading to an improved understanding of the biology of the disease and the development of new therapies. Most advanced tumors resistant to therapy still maintain the androgen receptor-pathway, which plays a central role for survival and growth of most castration-resistant prostate cancers. Many mechanisms induce the emergence of the castration resistant phenotype through this pathway. However some non-related AR pathways like neuroendocrine cells or overexpression of anti-apoptotic proteins like Hsp27 are described to be involved in CRPC progression. More recently, loss of expression of tumor suppressor gene, post-transcriptional modification using miRNA, epigenetic alterations, alternatif splicing and gene fusion became also hallmarks of castration-resistant prostate cancer. This review presents an up-to-date overview of the androgen receptor-related mechanisms as well as the latest evidence of the non-AR-related mechanisms underlying castration-resistant prostate cancer progression.
Collapse
Affiliation(s)
- Maria Katsogiannou
- Inserm, UMR1068, CRCM, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; Aix-Marseille Université, F-13284 Marseille, France; CNRS, UMR7258, CRCM, Marseille F-13009, France.
| | - Hajer Ziouziou
- Inserm, UMR1068, CRCM, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; Aix-Marseille Université, F-13284 Marseille, France; CNRS, UMR7258, CRCM, Marseille F-13009, France
| | - Sara Karaki
- Inserm, UMR1068, CRCM, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; Aix-Marseille Université, F-13284 Marseille, France; CNRS, UMR7258, CRCM, Marseille F-13009, France
| | - Claudia Andrieu
- Inserm, UMR1068, CRCM, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; Aix-Marseille Université, F-13284 Marseille, France; CNRS, UMR7258, CRCM, Marseille F-13009, France
| | - Marie Henry de Villeneuve
- Inserm, UMR1068, CRCM, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; Aix-Marseille Université, F-13284 Marseille, France; CNRS, UMR7258, CRCM, Marseille F-13009, France
| | - Palma Rocchi
- Inserm, UMR1068, CRCM, Marseille F-13009, France; Institut Paoli-Calmettes, Marseille F-13009, France; Aix-Marseille Université, F-13284 Marseille, France; CNRS, UMR7258, CRCM, Marseille F-13009, France.
| |
Collapse
|
21
|
Abstract
Progesterone and progesterone receptors (PRs) are essential for the development and cyclical regulation of hormone-responsive tissues including the breast and reproductive tract. Altered functions of PR isoforms contribute to the pathogenesis of tumors that arise in these tissues. In the breast, progesterone acts in concert with estrogen to promote proliferative and pro-survival gene programs. In sharp contrast, progesterone inhibits estrogen-driven growth in the uterus and protects the ovary from neoplastic transformation. Progesterone-dependent actions and associated biology in diverse tissues and tumors are mediated by two PR isoforms, PR-A and PR-B. These isoforms are subject to altered transcriptional activity or expression levels, differential crosstalk with growth factor signaling pathways, and distinct post-translational modifications and cofactor-binding partners. Herein, we summarize and discuss the recent literature focused on progesterone and PR isoform-specific actions in breast, uterine, and ovarian cancers. Understanding the complexity of context-dependent PR actions in these tissues is critical to developing new models that will allow us to advance our knowledge base with the goal of revealing novel and efficacious therapeutic regimens for these hormone-responsive diseases.
Collapse
Affiliation(s)
- Caroline H Diep
- HematologyOncology, and Transplantation DivisionDepartments of MedicinePharmacologyMasonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer Cardiology Research Building, 2231 6th Street SE, Minneapolis, Minnesota 55455, USADivision of Physiology and GrowthDepartment of Animal Science, University of Minnesota, Minneapolis, Minnesota 55108, USA
| | - Andrea R Daniel
- HematologyOncology, and Transplantation DivisionDepartments of MedicinePharmacologyMasonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer Cardiology Research Building, 2231 6th Street SE, Minneapolis, Minnesota 55455, USADivision of Physiology and GrowthDepartment of Animal Science, University of Minnesota, Minneapolis, Minnesota 55108, USA
| | - Laura J Mauro
- HematologyOncology, and Transplantation DivisionDepartments of MedicinePharmacologyMasonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer Cardiology Research Building, 2231 6th Street SE, Minneapolis, Minnesota 55455, USADivision of Physiology and GrowthDepartment of Animal Science, University of Minnesota, Minneapolis, Minnesota 55108, USA
| | - Todd P Knutson
- HematologyOncology, and Transplantation DivisionDepartments of MedicinePharmacologyMasonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer Cardiology Research Building, 2231 6th Street SE, Minneapolis, Minnesota 55455, USADivision of Physiology and GrowthDepartment of Animal Science, University of Minnesota, Minneapolis, Minnesota 55108, USA
| | - Carol A Lange
- HematologyOncology, and Transplantation DivisionDepartments of MedicinePharmacologyMasonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer Cardiology Research Building, 2231 6th Street SE, Minneapolis, Minnesota 55455, USADivision of Physiology and GrowthDepartment of Animal Science, University of Minnesota, Minneapolis, Minnesota 55108, USA HematologyOncology, and Transplantation DivisionDepartments of MedicinePharmacologyMasonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer Cardiology Research Building, 2231 6th Street SE, Minneapolis, Minnesota 55455, USADivision of Physiology and GrowthDepartment of Animal Science, University of Minnesota, Minneapolis, Minnesota 55108, USA
| |
Collapse
|
22
|
Koyama T, Rodrigues MA, Athanasiadis A, Shingleton AW, Mirth CK. Nutritional control of body size through FoxO-Ultraspiracle mediated ecdysone biosynthesis. eLife 2014; 3:e03091. [PMID: 25421296 PMCID: PMC4337420 DOI: 10.7554/elife.03091] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 10/18/2014] [Indexed: 01/21/2023] Open
Abstract
Despite their fundamental importance for body size regulation, the mechanisms that stop growth are poorly understood. In Drosophila melanogaster, growth ceases in response to a peak of the molting hormone ecdysone that coincides with a nutrition-dependent checkpoint, critical weight. Previous studies indicate that insulin/insulin-like growth factor signaling (IIS)/Target of Rapamycin (TOR) signaling in the prothoracic glands (PGs) regulates ecdysone biosynthesis and critical weight. Here we elucidate a mechanism through which this occurs. We show that Forkhead Box class O (FoxO), a negative regulator of IIS/TOR, directly interacts with Ultraspiracle (Usp), part of the ecdysone receptor. While overexpressing FoxO in the PGs delays ecdysone biosynthesis and critical weight, disrupting FoxO-Usp binding reduces these delays. Further, feeding ecdysone to larvae eliminates the effects of critical weight. Thus, nutrition controls ecdysone biosynthesis partially via FoxO-Usp prior to critical weight, ensuring that growth only stops once larvae have achieved a target nutritional status.
Collapse
Affiliation(s)
- Takashi Koyama
- Development, Evolution and the Environment
Laboratory, Instituto Gulbenkian de
Ciência, Oeiras, Portugal
| | - Marisa A Rodrigues
- Development, Evolution and the Environment
Laboratory, Instituto Gulbenkian de
Ciência, Oeiras, Portugal
| | - Alekos Athanasiadis
- Protein-Nucleic Acids Interactions Laboratory,
Instituto Gulbenkian de Ciência,
Oeiras, Portugal
| | - Alexander W Shingleton
- Department of Biology, Lake Forest
College, Lake
Forest, United States
- Department of Zoology, Michigan State
University, East
Lansing, United States
| | - Christen K Mirth
- Development, Evolution and the Environment
Laboratory, Instituto Gulbenkian de
Ciência, Oeiras, Portugal
| |
Collapse
|
23
|
Yang J, Li T, Gao C, Lv X, Liu K, Song H, Xing Y, Xi T. FOXO1 3′UTR functions as a ceRNA in repressing the metastases of breast cancer cells via regulating miRNA activity. FEBS Lett 2014; 588:3218-24. [DOI: 10.1016/j.febslet.2014.07.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/03/2014] [Accepted: 07/03/2014] [Indexed: 01/17/2023]
|
24
|
Semaphorin 3A upregulates FOXO 3a-dependent MelCAM expression leading to attenuation of breast tumor growth and angiogenesis. Oncogene 2014; 34:1584-95. [PMID: 24727891 DOI: 10.1038/onc.2014.79] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 01/19/2014] [Accepted: 02/18/2014] [Indexed: 12/16/2022]
Abstract
Semaphorin 3A (Sema 3A), a member of semaphorin family, serves as a guidance clue during embryonic development and is known as a candidate tumor suppressor that attenuates breast tumor progression by binding with its co-receptor, neuropilin-1 (NRP-1). However, the underlying mechanism by which Sema 3A suppresses breast tumor growth is still unexplored. In this study, we report that Sema 3A regulates phosphorylation and nuclear translocation of phosphatase and tensin homolog (PTEN) and FOXO 3a. Moreover, Sema 3A controls NRP-1-mediated PTEN-dependent FOXO 3a activation. Overexpression of PTEN and FOXO 3a enhances Sema 3A-induced attenuation of breast cancer cell migration. Chromatin immunoprecipitation and electrophoretic mobility shift assay data revealed that FOXO 3a regulates MelCAM at the transcriptional level. Furthermore, Sema 3A induces NRP-1-mediated MelCAM expression through PTEN and FOXO 3a. The data also showed that vascular endothelial growth factor-induced angiogenesis is inhibited by Sema 3A. Loss of or gain in function study revealed that Sema 3A modulates phosphorylation of PTEN and FOXO 3a and expression of MelCAM, leading to suppression of tumor growth and angiogenesis using in vivo mice model. Clinical specimen analysis revealed that reduced expression of Sema 3A and p-PTEN are correlated with enhanced breast cancer progression, further strengthening our in vitro and in vivo findings. Correlation of relapse-free survival of breast cancer patients (n=2878) with expression levels of Sema 3A, NRP-1, FOXO 3a and MelCAM were studied by Kaplan-Meier analysis. Statistical analysis revealed a close association between reduced expression of Sema 3A and MelCAM with that of poor patient's survival. Our study demonstrated a novel mechanism of regulation of tumor suppression by Sema 3A in coordination with a chain of tumor-suppressor genes, which in turn inhibits breast cancer cell migration, tumor growth and angiogenesis.
Collapse
|
25
|
LEE SUNHWA, KANG YONGJUNG, SUNG BOKYUNG, KIM DONGHWAN, LIM HYUNSOOK, KIM HYERIM, KIM SEONGJIN, YOON JEONGHYUN, MOON HYUNGRYONG, CHUNG HAEYOUNG, KIM NAMDEUK. MHY-449, a novel dihydrobenzofuro[4,5-b][1,8] naphthyridin-6-one derivative, induces apoptotic cell death through modulation of Akt/FoxO1 and ERK signaling in PC3 human prostate cancer cells. Int J Oncol 2014; 44:905-11. [DOI: 10.3892/ijo.2014.2257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/23/2013] [Indexed: 11/05/2022] Open
|
26
|
Zhu J, Jiang X, Chehab FF. FoxO4 interacts with the sterol regulatory factor SREBP2 and the hypoxia inducible factor HIF2α at the CYP51 promoter. J Lipid Res 2013; 55:431-42. [PMID: 24353279 DOI: 10.1194/jlr.m043521] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The late steps of cholesterol biosynthesis are oxygen demanding, requiring eleven oxygen molecules per synthesized cholesterol molecule. A key enzymatic reaction, which occurs at the top of the Bloch and Kandutsch-Russell pathways, is the demethylation of lanosterol and dihydrolanosterol (DHL). This reaction is catalyzed by lanosterol 14α demethylase (CYP51) and requires three oxygen molecules. Thus, it is the first step in the distal pathway to be susceptible to oxygen deprivation. Having previously identified that the forkhead transcription factor 4 (FoxO4) represses CYP51 expression, we aimed to characterize its role at the CYP51 promoter. Hypoxia-treated 3T3L1 cells showed decreased cholesterol biosynthesis, accumulation of lanosterol/DHL, and stimulation of FoxO4 expression and its cytoplasmic translocation to the nucleus. Transfection assays with a CYP51 promoter reporter gene revealed that FoxO4 and sterol regulatory element binding protein (SREBP)2 exert a stimulatory effect, whereas FoxO4 and the hypoxia inducible factor (HIF)2α repress CYP51 promoter activity. Electromobility shift, chromatin immunoprecipitation, pull-down, and coimmunoprecipitation assays show that FoxO4 interacts with SREBP2 and HIF2α to modulate CYP51 promoter activity. We also show an inverse correlation between FoxO4 and CYP51 in adipose tissue of ob/ob mice and mouse fetal cortical neurons exposed to hypoxia. Overall, these studies demonstrate a role for FoxO4 in the regulation of CYP51 expression.
Collapse
Affiliation(s)
- Jun Zhu
- Departments of Laboratory Medicine University of California, San Francisco, CA 94143
| | | | | |
Collapse
|
27
|
Haflidadóttir BS, Larne O, Martin M, Persson M, Edsjö A, Bjartell A, Ceder Y. Upregulation of miR-96 enhances cellular proliferation of prostate cancer cells through FOXO1. PLoS One 2013; 8:e72400. [PMID: 23951320 PMCID: PMC3741168 DOI: 10.1371/journal.pone.0072400] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/10/2013] [Indexed: 12/21/2022] Open
Abstract
Aberrant expression of miR-96 in prostate cancer has previously been reported. However, the role and mechanism of action of miR-96 in prostate cancer has not been determined. In this study, the diagnostic and prognostic properties of miR-96 expression levels were investigated by qRT-PCR in two well documented prostate cancer cohorts. The miR-96 expression was found to be significantly higher in prostate cancer patients and correlate with WHO grade, and decreased overall survival time; patients with low levels of miR-96 lived 1.5 years longer than patients with high miR-96 levels. The therapeutic potential was further investigated in vitro, showing that ectopic levels of miR-96 enhances growth and cellular proliferation in prostate cancer cells, implying that miR-96 has oncogenic properties in this setting. We demonstrate that miR-96 expression decreases the transcript and protein levels of FOXO1 by binding to one of two predicted binding sites in the FOXO1 3'UTR sequence. Blocking this binding site completely inhibited the growth enhancement conveyed by miR-96. This finding was corroborated in a large external prostate cancer patient cohort where miR-96 expression inversely correlated to FOXO1 expression. Taken together these findings indicate that miR-96 plays a key role in prostate cancer cellular proliferation and can enhance prostate cancer progression. This knowledge might be utilized for the development of novel therapeutic tools for prostate cancer.
Collapse
Affiliation(s)
| | - Olivia Larne
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Malmö, Sweden
| | - Myriam Martin
- Department of Laboratory Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Margareta Persson
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Malmö, Sweden
| | - Anders Edsjö
- Department of Laboratory Medicine, Center for Molecular Pathology, Lund University, Malmö, Sweden
| | - Anders Bjartell
- Department of Clinical Sciences, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Yvonne Ceder
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Malmö, Sweden
- * E-mail:
| |
Collapse
|
28
|
Bohrer LR, Liu P, Zhong J, Pan Y, Angstman J, Brand LJ, Dehm SM, Huang H. FOXO1 binds to the TAU5 motif and inhibits constitutively active androgen receptor splice variants. Prostate 2013; 73:1017-27. [PMID: 23389878 PMCID: PMC3915545 DOI: 10.1002/pros.22649] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 01/08/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND Aberrant activation of the androgen receptor (AR) is a major factor highly relevant to castration-resistant progression of prostate cancer (PCa). FOXO1, a key downstream effector of PTEN, inhibits androgen-independent activation of the AR. However, the underlying mechanism remains elusive. METHODS The inhibitory effect of FOXO1 on full-length and constitutively active splice variants of the AR was examined by luciferase reporter assays and real-time reverse transcription polymerase chain reaction (RT-qPCR). In vitro protein binding assays and western blot analyses were used to determine the regions in FOXO1 and AR responsible for their interaction. RESULTS We found that a putative transcription repression domain in the NH2-terminus of FOXO1 is dispensable for FOXO1 inhibition of the AR. In vitro protein binding assays showed that FOXO1 binds to the transcription activation unit 5 (TAU5) motif in the AR NH2-terminal domain (NTD), a region required for recruitment of p160 activators including SRC-1. Ectopic expression of SRC-1 augmented transcriptional activity of some, but not all AR splice variants examined. Forced expression of FOXO1 blocked the effect of SRC-1 on AR variants' transcriptional activity by decreasing the binding of SRC-1 to the AR NTD. Ectopic expression of FOXO1 inhibited expression of endogenous genes activated primarily by alternatively spliced AR variants in human castration-resistant PCa 22Rv1 cells. CONCLUSIONS FOXO1 binds to the TAU5 motif in the AR NTD and inhibits ligand-independent activation of AR splice variants, suggesting the PTEN/FOXO1 pathway as a potential therapeutic target for inhibition of aberrant AR activation and castration-resistant PCa growth.
Collapse
Affiliation(s)
- Laura R. Bohrer
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Ping Liu
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jian Zhong
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yunqian Pan
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - James Angstman
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Lucas J. Brand
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Graduate Programin Microbiology, Immunology, and Cancer Biology, Universityof Minnesota, Minneapolis, Minnesota
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
- Correspondence to: Scott M. Dehm and Haojie Huang, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455. ;
| | - Haojie Huang
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
- Correspondence to: Scott M. Dehm and Haojie Huang, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455. ;
| |
Collapse
|
29
|
Mediwala SN, Sun H, Szafran AT, Hartig SM, Sonpavde G, Hayes TG, Thiagarajan P, Mancini MA, Marcelli M. The activity of the androgen receptor variant AR-V7 is regulated by FOXO1 in a PTEN-PI3K-AKT-dependent way. Prostate 2013; 73:267-77. [PMID: 22821817 PMCID: PMC3961010 DOI: 10.1002/pros.22566] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 06/27/2012] [Indexed: 11/07/2022]
Abstract
BACKGROUND The androgen receptor (AR) AR-V7 splice isoform is a constitutively active outlaw transcription factor. Transition of prostate cancer (PC) to the castration-resistant phenotype correlates with AR-V7 accumulation, suggesting that PC progression in patients refractory to conventional therapy is due to the activity of this AR isoform. The mechanism of AR-V7 constitutive activation is not known. METHODS We analyzed potential signaling pathways associated with AR-V7 constitutive activation in PTEN (-) PC-3 and LNCaP cells. We used transient and stable transfection, reporter gene assay, RNAi technology together with a number of kinase inhibitors to determine if AR-V7 activation is linked to a kinase-dependent signaling pathway. RESULTS In these cell lines, AR-V7 transcriptional activity was inhibited by LY294002, Wortmanin, and AKT inhibitor II. Analysis of the contributing mechanisms demonstrated the involvement of the Phosphatidylinositol 3-kinase (PI3K)-AKT-FOXO1 signaling pathway, and a significant reduction of AR-V7 constitutive activity under conditions of PTEN reactivation. CONCLUSIONS Our study identifies a pathway regulating AR-V7 constitutive activity and potential therapeutic targets for the treatment of castration-resistant PC.
Collapse
Affiliation(s)
- Sanjay N. Mediwala
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
- Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Huiying Sun
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
- Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Adam T. Szafran
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Sean M. Hartig
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Guru Sonpavde
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
- Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Teresa G. Hayes
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
- Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Perumal Thiagarajan
- Departments of Medicine, Pathology, Baylor College of Medicine, Houston TX 77030 (USA)
- Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Michael A. Mancini
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
| | - Marco Marcelli
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
- Departments of Medicine, Molecular and Cellular Biology, Baylor College of Medicine, Houston TX 77030 (USA)
- Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston TX 77030 (USA)
| |
Collapse
|
30
|
Kumar N, Chornokur G. Molecular Targeted Therapies Using Botanicals for Prostate Cancer Chemoprevention. TRANSLATIONAL MEDICINE (SUNNYVALE, CALIF.) 2012; Suppl 2:005. [PMID: 24527269 PMCID: PMC3920581 DOI: 10.4172/2161-1025.s2-005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In spite of the large number of botanicals demonstrating promise as potential cancer chemopreventive agents, most have failed to prove effectiveness in clinical trials. Critical requirements for moving botanical agents to recommendation for clinical use include adopting a systematic, molecular-target based approach and utilizing the same ethical and rigorous methods that are used to evaluate other pharmacological agents. Preliminary data on a mechanistic rationale for chemoprevention activity as observed from epidemiological, in vitro and preclinical studies, phase I data of safety in suitable cohorts, duration of intervention based on time to progression of pre-neoplastic disease to cancer and using a valid panel of biomarkers representing the hypothesized carcinogenesis pathway for measuring efficacy must inform the design of clinical trials. Botanicals have been shown to influence multiple biochemical and molecular cascades that inhibit mutagenesis, proliferation, induce apoptosis, suppress the formation and growth of human cancers, thus modulating several hallmarks of carcinogenesis. These agents appear promising in their potential to make a dramatic impact in cancer prevention and treatment, with a significantly superior safety profile than most agents evaluated to date. The goal of this paper is to provide models of translational research based on the current evidence of promising botanicals with a specific focus on targeted therapies for PCa chemoprevention.
Collapse
Affiliation(s)
- Nagi Kumar
- Department of Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Florida, USA ; University of South Florida College of Medicine, Florida, USA
| | - Ganna Chornokur
- Department of Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Florida, USA
| |
Collapse
|
31
|
Rajabi H, Ahmad R, Jin C, Joshi MD, Guha M, Alam M, Kharbanda S, Kufe D. MUC1-C oncoprotein confers androgen-independent growth of human prostate cancer cells. Prostate 2012; 72:1659-68. [PMID: 22473899 PMCID: PMC3413781 DOI: 10.1002/pros.22519] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 02/29/2012] [Indexed: 01/27/2023]
Abstract
BACKGROUND The mucin 1 (MUC1) heterodimeric oncoprotein is overexpressed in human prostate cancers with aggressive pathologic and clinical features. However, few insights are available regarding the functional role of MUC1 in prostate cancer. METHODS Effects of MUC1-C on androgen receptor (AR) expression were determined by RT-PCR, immunoblotting and AR promoter activation. Coimmunoprecipitations, direct binding assays, and chromatin immunoprecipitation (ChIP) studies were performed to assess the interaction between MUC1-C and AR. Cells were analyzed for invasion, growth in androgen-depleted medium, and sensitivity to MUC1-C inhibitors. RESULTS The present studies in androgen-dependent LNCaP and LAPC4 prostate cancer cells demonstrate that the oncogenic MUC1-C subunit suppresses AR expression. The results show that MUC1-C activates a posttranscriptional mechanism involving miR-135b-mediated downregulation of AR mRNA levels. The results further demonstrate that MUC1-C forms a complex with AR through a direct interaction between the MUC1-C cytoplasmic domain and the AR DNA-binding domain (DBD). In addition, MUC1-C associates with AR in a complex that occupies the PSA promoter. The interaction between MUC1-C and AR is associated with induction of the epithelial-mesenchymal transition (EMT) and increased invasion. MUC1-C also conferred growth in androgen-depleted medium and resistance to bicalutamide treatment. Moreover, expression of MUC1-C resulted in sensitivity to the MUC1-C inhibitor GO-203 with inhibition of growth in vitro. GO-203 treatment also inhibited growth of established tumor xenografts in nude mice. CONCLUSIONS These findings indicate that MUC1-C suppresses AR expression in prostate cancer cells and confers a more aggressive androgen-independent phenotype that is sensitive to MUC1-C inhibition.
Collapse
Affiliation(s)
- Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kasiappan R, Shen Z, Tse AKW, Jinwal U, Tang J, Lungchukiet P, Sun Y, Kruk P, Nicosia SV, Zhang X, Bai W. 1,25-Dihydroxyvitamin D3 suppresses telomerase expression and human cancer growth through microRNA-498. J Biol Chem 2012; 287:41297-309. [PMID: 23055531 DOI: 10.1074/jbc.m112.407189] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Telomerase is an essential enzyme that counteracts the telomere attrition accompanying DNA replication during cell division. Regulation of the promoter activity of the gene encoding its catalytic subunit, the telomerase reverse transcriptase, is established as the dominant mechanism conferring the high telomerase activity in proliferating cells, such as embryonic stem and cancer cells. This study reveals a new mechanism of telomerase regulation through non-coding small RNA by showing that microRNA-498 (miR-498) induced by 1,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) decreases the mRNA expression of the human telomerase reverse transcriptase. MiR-498 was first identified in a microarray analysis as the most induced microRNA by 1,25(OH)(2)D(3) in ovarian cancer cells and subsequently validated by quantitative polymerase chain reaction assays in multiple human cancer types. A functional vitamin D response element was defined in the 5-prime regulatory region of the miR-498 genome, which is occupied by the vitamin D receptor and its coactivators. Further studies showed that miR-498 targeted the 3-prime untranslated region of human telomerase reverse transcriptase mRNA and decreased its expression. The levels of miR-498 expression were decreased in malignant human ovarian tumors as well as human ovarian cancer cell lines. The ability of 1,25(OH)(2)D(3) to decrease human telomerase reverse transcriptase mRNA and to suppress ovarian cancer growth was compromised when miR-498 was depleted using the sponges in cell lines and mouse tumor models. Taken together, our studies define a novel mechanism of telomerase regulation by small non-coding RNAs and identify miR-498 as an important mediator for the anti-tumor activity of 1,25(OH)(2)D(3).
Collapse
Affiliation(s)
- Ravi Kasiappan
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida 33612-4799, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wu Z, Sun H, Zeng W, He J, Mao X. Upregulation of MircoRNA-370 induces proliferation in human prostate cancer cells by downregulating the transcription factor FOXO1. PLoS One 2012; 7:e45825. [PMID: 23029264 PMCID: PMC3445500 DOI: 10.1371/journal.pone.0045825] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/24/2012] [Indexed: 01/17/2023] Open
Abstract
Forkhead box protein O1 (FOXO1), a key member of the FOXO family of transcription factors, acts as a tumor suppressor and has been associated with various key cellular functions, including cell growth, differentiation, apoptosis and angiogenesis. Therefore, it is puzzling why FOXO protein expression is downregulated in cancer cells. MicroRNAs, non-coding 20∼22 nucleotide single-stranded RNAs, result in translational repression or degradation and gene silencing of their target genes, and significantly contribute to the regulation of gene expression. In the current study, we report that miR-370 expression was significantly upregulated in five prostate cancer cell lines, compared to normal prostatic epithelial (PrEC) cells. Ectopic expression of miR-370 induced proliferation and increased the anchorage-independent growth and colony formation ability of DU145 and LNCaP prostate cancer cells, while inhibition of miR-370 reduced proliferation, anchorage-independent growth and colony formation ability. Furthermore, upregulation of miR-370 promoted the entry of DU145 and LNCaP prostate cancer cells into the G1/S cell cycle transition, which was associated with downregulation of the cyclin-dependent kinase (CDK) inhibitors, p27Kip1 and p21Cip1, and upregulation of the cell-cycle regulator cyclin D1 mRNA. Additionally, we demonstrated that miR-370 can downregulate expression of FOXO1 by directly targeting the FOXO1 3′-untranslated region. Taken together, our results suggest that miR-370 plays an important role in the proliferation of human prostate cancer cells, by directly suppressing the tumor suppressor FOXO1.
Collapse
Affiliation(s)
- Ziqing Wu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Huabin Sun
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, P. R. China
| | - Weixia Zeng
- Laura Biotech Co, Ltd., Guangzhou, Guangdong Province, P. R. China
| | - Jincan He
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xiangming Mao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
- * E-mail:
| |
Collapse
|
34
|
Khandelwal P, Liu S, Sullivan DA. Androgen regulation of gene expression in human meibomian gland and conjunctival epithelial cells. Mol Vis 2012; 18:1055-67. [PMID: 22605918 PMCID: PMC3351406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 04/24/2012] [Indexed: 10/26/2022] Open
Abstract
PURPOSE Androgens exert a significant influence on the structure, function and/or pathophysiology of the meibomian gland and conjunctiva. We sought to determine whether this hormone action involves the regulation of epithelial cell gene expression in these tissues. METHODS Immortalized human meibomian gland and conjunctival epithelial cells were treated with placebo or dihydrotestosterone (DHT) and processed for molecular biologic procedures. Gene expression was evaluated with BeadChips and data were analyzed with bioinformatic and statistical software. RESULTS Androgen treatment significantly influenced the expression of approximately 3,000 genes in immortalized human meibomian gland and conjunctival epithelial cells. The nature of DHT action on gene activity was predominantly cell-specific. Similarly, DHT exerted a significant, but primarily cell-specific, influence on many gene ontologies and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. These included groups of genes related, for example, to lipid dynamics, innate immunity, cell cycle, Janus kinase (JAK)-signal transducer and activator of transcription (stat) cascades, oxidative phosphorylation, the proteasome, and mammalian target of rapamycin (mTOR), Wnt, and peroxisome proliferator-activated receptor (PPAR) signaling. CONCLUSIONS Our findings support our hypothesis that androgens regulate gene expression in human meibomian gland and conjunctival epithelial cells. Our ongoing studies are designed to determine whether many of these genes are translated and play a role in the health and well being of the eye.
Collapse
|
35
|
Inhibition of cyclin-dependent kinase phosphorylation of FOXO1 and prostate cancer cell growth by a peptide derived from FOXO1. Neoplasia 2012; 13:854-63. [PMID: 21969818 DOI: 10.1593/neo.11594] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/26/2011] [Accepted: 06/27/2011] [Indexed: 11/18/2022] Open
Abstract
Increasing evidence suggests that FOXO1 possesses a tumor suppressor function. Inactivation of FOXO1 has been documented in many types of human cancer, and restoring the activity of FOXO1 holds promise for cancer treatment. In this study, we identified a FOXO1-derived peptide termed FO1-6nls that inhibits cyclin-dependent kinases 1 and 2 (CDK1/2)-mediated phosphorylation of FOXO1 at the serine 249 residue in vitro and in vivo. Overexpression of FO1-6nls in prostate cancer (PCa) cells not only blocked CDK1-induced cytoplasmic localization of FOXO1 but also augmented FOXO1's transcriptional activity. This effect of FO1-6nls requires its binding to CDK1 and CDK2. Moreover, the ectopic expression of FO1-6nls inhibited the growth of PTEN-positive DU145 PCa cells. Importantly, the growth-inhibitory function of FO1-6nls is dependent on FOXO1. Finally, the ectopic expression of FO1-6nls overcame CDK1-mediated inhibition of FOXO1-induced apoptosis of PCa cells. These results indicate that the FOXO1-derived peptide FO1-6nls can restore FOXO1's tumor suppressor function by specifically opposing CDK1/2-mediated phosphorylation and inhibition of FOXO1 and hence may have a therapeutic potential for the treatment of PCa.
Collapse
|
36
|
Kumar N, Crocker T, Smith T, Pow-Sang J, Spiess PE, Connors S, Chornukur G, Dickinson SI, Bai W, Williams CR, Salup R, Fu W. Prostate Cancer Chemoprevention Targeting High Risk Populations: Model for Trial Design and Outcome Measures. ACTA ACUST UNITED AC 2012; 2011. [PMID: 22422102 DOI: 10.4172/1948-5956.s3-007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inspite of the large number of promising nutrient-derived agents demonstrating promise as potential chemopreventive agents, most have failed to prove effectiveness in clinical trials. Critical requirements for moving nutrient-derived agents to recommendation for clinical use include adopting a systematic, molecular-mechanism based approach and utilizing the same ethical and rigorous methods such as are used to evaluate other pharmacological agents. Preliminary data on a mechanistic rationale for chemoprevention activity as observed from epidemiological, in vitro and preclinical studies, phase I data of safety in high-risk cohorts are required to inform design of phase II clinical trials. Additionally, a valid panel of biomarkers representing the hypothesized carcinogenesis pathway for measuring efficacy must be utilized to evaluate effectiveness in these trials. The goal of this paper is to provide a model, using a systematic approach for evaluating the safety, effectiveness and mechanism of action of a well characterized nutrient-derived agent-isoflavones - in a phase II clinical trial for prostate cancer (CaP) chemoprevention, targeting a population of African American (AA) and Caucasian men. Based on our previous observations, we hypothesize that the effects of isoflavones on prostate carcinogenesis are mainly mediated through the down regulation of androgen receptor (AR) and AR activity in AA men is higher due to its shorter length of Glutamine repeats in its N-terminus. We thus believe that isoflavones will exert a stronger protective effect for CaP in AA men and cause a higher activation of FOXO factors and their target genes. The aim of the study is to evaluate the comparative effectiveness of the study agent and placebo, in addition to a comparison of the effectiveness and safety in African American men compared to Caucasian men treated with this agent.
Collapse
Affiliation(s)
- Nagi Kumar
- Department of Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, College of Medicine, Tampa, Florida
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lam EWF, Shah K, Brosens JJ. The diversity of sex steroid action: the role of micro-RNAs and FOXO transcription factors in cycling endometrium and cancer. J Endocrinol 2012; 212:13-25. [PMID: 21382987 DOI: 10.1530/joe-10-0480] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The rise and fall in ovarian oestrogen and progesterone production orchestrates a series of events that are indispensable for reproduction, including ovulation, implantation, decidualisation and menstruation. In the uterus, these events involve extensive tissue remodelling, characterised by waves of endometrial cell proliferation, differentiation, recruitment of inflammatory cells, apoptosis, tissue breakdown, menstruation and regeneration. The ability of ovarian hormones to trigger such diverse physiological responses is foremost dependent upon interaction of activated steroid receptors with specific transcription factors, such as Forkhead box class O (FOXO) proteins, involved in cell fate decisions. Furthermore, micro-RNAs (miRNAs), small non-coding RNAs that function as posttranscriptional regulators of gene expression, have emerged as a major regulator system of steroid hormone responses in the female reproductive tract. Consequently, increasing evidence shows that deregulated uterine miRNA expression underpins a spectrum of common reproductive disorders, ranging from implantation failure to endometriosis. Furthermore, by targeting FOXO transcription factors and other key regulators of tissue homeostasis, oncogenic endometrial miRNAs promote tumourigenesis and cancer progression.
Collapse
Affiliation(s)
- Eric W-F Lam
- Cancer Research-UK Laboratories, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.
| | | | | |
Collapse
|
38
|
Saraon P, Jarvi K, Diamandis EP. Molecular alterations during progression of prostate cancer to androgen independence. Clin Chem 2011; 57:1366-75. [PMID: 21956922 DOI: 10.1373/clinchem.2011.165977] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Prostate cancer is the most commonly diagnosed cancer among men in North America and is a leading cause of death. Standard treatments include androgen deprivation therapy, which leads to improved clinical outcomes. However, over time, most tumors become androgen independent and no longer respond to hormonal therapies. Several mechanisms have been implicated in the progression of prostate cancer to androgen independence. CONTENT Most tumors that have become androgen independent still rely on androgen receptor (AR) signaling. Mechanisms that enhance AR signaling in androgen-depleted conditions include: AR gene amplification, AR mutations, changes in the balance of AR cofactors, increases in steroidogenic precursors, and activation via "outlaw" pathways. Along with AR signaling, various other AR-independent "bypass" pathways have been shown to operate aberrantly during androgen independence. Changes in the epigenetic signatures and microRNA concentrations have also been implicated in the development of androgen-independent prostate cancer. SUMMARY Understanding of the molecular mechanisms that lead to the development of androgen-independent prostate cancer will allow for improved therapeutic strategies that target key pathways and molecules that are essential for these cells to survive.
Collapse
Affiliation(s)
- Punit Saraon
- Samuel Lunenfeld Research Institute and Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | | | | |
Collapse
|
39
|
Kajihara T, Tochigi H, Prechapanich J, Uchino S, Itakura A, Brosens JJ, Ishihara O. Androgen signaling in decidualizing human endometrial stromal cells enhances resistance to oxidative stress. Fertil Steril 2011; 97:185-91. [PMID: 22078782 DOI: 10.1016/j.fertnstert.2011.10.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 10/13/2011] [Accepted: 10/15/2011] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the effect of androgens on the expression of genes involved in oxidative stress resistance in decidualized human endometrial stromal cells (HESCs). DESIGN In vitro experiment. SETTING University hospital. PATIENT(S) Premenopausal women undergoing hysterectomy for uterine fibroids. INTERVENTION(S) Human endometrial stromal cells isolated from hysterectomy specimens were decidualized with 8-bromo-cyclic adenosine monophosphate (8-br-cAMP) and P in the presence or absence of dihydrotestosterone (DHT) at various concentrations. Hydrogen peroxide was used as a source of reactive oxygen species. MAIN OUTCOME MEASURE(S) Prolactin secretion, apoptosis, FOXO1, and the free radical scavengers superoxide dismutase 2 (SOD2) and SOD1 protein expression. RESULT(S) Prolactin production was induced in HESCs in response to 8-br-cAMP and P. Dihydrotestosterone further enhanced the secretion of PRL in cells treated with 8-br-cAMP plus P. The effect of DHT was blocked by the antiandrogen flutamide. Dihydrotestosterone enhanced resistance to oxidative stress-induced apoptosis on decidualized HESCs. Moreover, DHT enhanced FOXO1 expression in parallel with increased SOD2 protein but not with SOD1. CONCLUSION(S) Androgens might play a critical role in the decidualization process at the time of embryo implantation and trophoblast invasion by promoting resistance to oxidative stress.
Collapse
Affiliation(s)
- Takeshi Kajihara
- Department of Obstetrics and Gynecology, Saitama Medical University, Moroyama, Iruma-gun, Saitama, Japan.
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Dysregulation of phosphatidyl inositol signaling occurs in many cancers and other disorders. The lipid and protein phosphatase, PTEN (Phosphatase and Tensin homology protein on chromosome 10), is a known tumor suppressor whose function is frequently lost in various malignancies due to mutations in the coding region or genomic deletions. Recently, another lipid phosphatase, Inositol Polyphosphate 4-phosphatase type II (INPP4B), has emerged as a potential tumor suppressor in prostate, breast, and ovarian cancers and possibly in leukemia. We will review its structure and function, crosstalk with androgen receptor signaling, and regulation of INPP4B expression, as well as existing data about its role in cancer.
Collapse
|
41
|
Almeida M. Unraveling the role of FoxOs in bone--insights from mouse models. Bone 2011; 49:319-27. [PMID: 21664311 PMCID: PMC3143252 DOI: 10.1016/j.bone.2011.05.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 05/17/2011] [Accepted: 05/24/2011] [Indexed: 12/11/2022]
Abstract
The FoxO subfamily of forkhead transcription factors plays a critical role in a variety of physiological processes including metabolism, differentiation, proliferation, apoptosis and protection from stress. FoxO activity is inhibited by growth factors and the insulin signaling pathways and stimulated by nutrient depletion and a plethora of reactive oxygen species (ROS)-induced post-translational modifications. Recent studies have uncovered a fundamental role for FoxOs in skeletal homeostasis. In cells of the osteoblast lineage, FoxOs modulate redox balance, protein synthesis, and differentiation through the activation of specific gene programs and interaction with other transcription factors and co-factors such as β-catenin, ATF-4, and Runx2. FoxO activation also attenuates osteoclastogenesis through both cell autonomous and indirect mechanisms. In this review I discuss recent advances in the understanding of FoxO specific actions in osteoblast progenitors, osteoblasts, and osteoclast, as well as the implications of FoxO activation for age-related skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA.
| |
Collapse
|
42
|
Munekata K, Sakamoto K. Forkhead transcription factor Foxo1 is essential for adipocyte differentiation. In Vitro Cell Dev Biol Anim 2011; 45:642-51. [PMID: 19585174 DOI: 10.1007/s11626-009-9230-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 06/26/2009] [Indexed: 01/04/2023]
Abstract
We analyzed the physiological role of forkhead box class O 1 (Foxo1) in adipocyte differentiation by suppressing Foxo1 mRNAwith siRNA specific for Foxo1.Mouse 3T3-L1 preadipocytes infected with an adenovirus expressing Foxo1-siRNA showed a marked decrease in lipid droplet formation when induced to differentiate into adipocytes. Adipocyte differentiation was most severely inhibited by exposing cells to Foxo1-siRNA before induction of differentiation. The incorporation of fluorescent-labeled glucose and fatty acid was significantly inhibited in cells deficient in Foxo1. RTPCR revealed that downregulation of Foxo1 decreased the expression of the transcription factors, PPAR-γ and C/EBP-α. By comparison, Foxo1-siRNA did not affect the expression of C/EBP-β or C/EBP-δ during the early period of adipocyte differentiation. These results indicate that Foxo1 plays an essential role in adipocyte differentiation, especially at the very early stage of terminal adipocyte differentiation.
Collapse
Affiliation(s)
- Keisuke Munekata
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
43
|
El-Haibi CP, Singh R, Sharma PK, Singh S, Lillard JW. CXCL13 mediates prostate cancer cell proliferation through JNK signalling and invasion through ERK activation. Cell Prolif 2011; 44:311-9. [PMID: 21645150 DOI: 10.1111/j.1365-2184.2011.00757.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES The focus of this study was to determine the dedicator of cytokinesis 2 (DOCK2), extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase-1 (JNK) and Akt signals involved in CXCL13-mediated prostate cancer (PCa) cell invasion and proliferation. MATERIALS AND METHODS Androgen-sensitive (LNCaP), hormone-refractory (PC3) cells and normal cells (RWPE-1) were used to determine CXCL13-mediated PCa cell invasion and proliferation. Immuno-blotting, fast activated cell-based (FACE) ELISA, caspase activity, cell invasion and proliferation assays were performed to ascertain some of the signalling events involved in PCa cell proliferation and invasion. RESULTS Unlike androgen-sensitive LNCaP cells, we report for the first time that the hormone-refractory cell line, PC3, expresses DOCK2. CXCL13-mediated LNCaP and PC3 cell invasion was regulated by Akt and ERK1/2 activation in a DOCK2-independent fashion. CXCL13 also promoted LNCaP cell proliferation in a JNK-dependent fashion even in the absence of DOCK2. In contrast, CXCL13 induced PC3 cell proliferation through JNK activation, which required DOCK2. CONCLUSIONS Our results show CXCL13-mediated PCa cell invasion requires Akt and ERK1/2 activation and suggests a new role for DOCK2 in proliferation of hormone-refractory CXCR5-positive PCa cells.
Collapse
Affiliation(s)
- C P El-Haibi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
44
|
Yang Y, Tse AKW, Li P, Ma Q, Xiang S, Nicosia SV, Seto E, Zhang X, Bai W. Inhibition of androgen receptor activity by histone deacetylase 4 through receptor SUMOylation. Oncogene 2011; 30:2207-18. [PMID: 21242980 PMCID: PMC3093431 DOI: 10.1038/onc.2010.600] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 10/20/2010] [Accepted: 11/23/2010] [Indexed: 01/15/2023]
Abstract
The transcriptional activity of the androgen receptor (AR) is regulated by both ligand binding and post-translational modifications, including acetylation and small ubiquitin-like modifier (SUMO)ylation. Histone deacetylases (HDACs) are known to catalyze the removal of acetyl groups from both histones and non-histone proteins. In this study, we report that HDAC4 binds to and inhibits the activity of the AR. This inhibition was found to depend on the SUMOylation, instead of deacetylation, of the AR. Consistently, HDAC4 increases the level of AR SUMOylation in both whole-cell and cell-free assay systems, raising the possibility that the deacetylase may act as an E3 ligase for AR SUMOylation. Knock down of HDAC4 increases the activity of endogenous AR and androgen induction of prostate-specific antigen expression and prostate cancer cell growth, which is associated with decreased SUMOylation of the receptor. Overall, the studies identify HDAC4 as a positive regulator for AR SUMOylation, revealing a deacetylase-independent mechanism of HDAC action in prostate cancer cells.
Collapse
Affiliation(s)
- Y Yang
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, FL 33612-4799, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhang H, Pan Y, Zheng L, Choe C, Lindgren B, Jensen ED, Westendorf JJ, Cheng L, Huang H. FOXO1 inhibits Runx2 transcriptional activity and prostate cancer cell migration and invasion. Cancer Res 2011; 71:3257-67. [PMID: 21505104 PMCID: PMC3108023 DOI: 10.1158/0008-5472.can-10-2603] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Prostate cancer patients with regional lymph node involvement at radical prostatectomy often experience disease progression to other organs, with the bone as the predominant site. The transcription factor Runx2 plays an important role in bone formation and prostate cancer cell migration, invasion, and metastasis. Here we showed that the forkhead box O (FOXO1) protein, a key downstream effector of the tumor suppressor PTEN, inhibits the transcriptional activity of Runx2 in prostate cancer cells. This inhibition was enhanced by PTEN but diminished by active Akt. FOXO1 bound to Runx2 in vitro and in vivo and suppressed Runx2's activity independent of its transcriptional function. FOXO1 inhibited Runx2-promoted migration of prostate cancer cells, whereas silencing of endogenous FOXO1 enhanced prostate cancer cell migration in a Runx2-dependent manner. Forced expression of FOXO1 also inhibited Runx2-promoted prostate cancer cell invasion. Finally, we found that expression of PTEN and the level of FOXO1 in the nucleus is inversely correlated with expression of Runx2 in a cohort of prostate cancer specimens from patients with lymph node and bone metastasis. These data reveal FOXO1 as a critical negative regulator of Runx2 in prostate cancer cells. Inactivation of FOXO1 due to frequent loss of PTEN in prostate cancer cells may leave the oncogenic activities of Runx2 unchecked, thereby driving promiscuous expression of Runx2 target genes involved in cell migration and invasion and favoring prostate cancer progression.
Collapse
Affiliation(s)
- Haijun Zhang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Yunqian Pan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
- Department of Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794
| | - Li Zheng
- Department of Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794
| | - Chungyoul Choe
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Bruce Lindgren
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - Eric D. Jensen
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN 55455
| | - Jennifer J. Westendorf
- Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Haojie Huang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
- Department of Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794
| |
Collapse
|
46
|
Akt, FoxO and regulation of apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:1978-86. [PMID: 21440011 DOI: 10.1016/j.bbamcr.2011.03.010] [Citation(s) in RCA: 789] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 03/09/2011] [Accepted: 03/11/2011] [Indexed: 12/14/2022]
Abstract
Forkhead box O (FoxO) transcription factors are downstream targets of the serine/threonine protein kinase B (PKB)/Akt. The Akt kinase regulates processes of cellular proliferation and survival. Phosphorylation of FoxOs by Akt inhibits transcriptional functions of FoxOs and contributes to cell survival, growth and proliferation. Emerging evidence suggests involvement of FoxOs in diverse intracellular signaling pathways with critical roles in a number of physiological as well as pathological conditions including cancer. The FoxO signaling is regulated by their interactions with other intracellular proteins as well as their post-translational modifications such as phosphorylation. FoxOs promote cell growth inhibitory and/or apoptosis signaling by either inducing expression of multiple pro-apoptotic members of the Bcl2-family of mitochondria-targeting proteins, stimulating expression of death receptor ligands such as Fas ligand and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), or enhancing levels of various cyclin-dependent kinase inhibitors (CDKIs). Coupled with their ability to cross-talk with p53, FoxOs represent an important class of tumor suppressors in a variety of cancers. This review summarizes our current understanding of mechanisms by which Akt and FoxOs regulate cell growth and survival that in turn offers opportunities for development of novel strategies to combat cancer. This article is part of a Special Issue entitled: P13K-AKT-FOxO axis in cancer and aging.
Collapse
|
47
|
Daitoku H, Sakamaki JI, Fukamizu A. Regulation of FoxO transcription factors by acetylation and protein-protein interactions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:1954-60. [PMID: 21396404 DOI: 10.1016/j.bbamcr.2011.03.001] [Citation(s) in RCA: 223] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/06/2010] [Revised: 02/09/2011] [Accepted: 03/02/2011] [Indexed: 12/20/2022]
Abstract
The forkhead box O transcription factors convert a variety of external stimuli, including growth factors, nutrients, and oxidative stress, into diverse biological responses through modulation of specific gene expression. Forkhead box O regulation is principally achieved by two distinct mechanisms: post-translational modifications and protein-protein interactions. Among several modifications of forkhead box O factors, we focus on reversible acetylation, describing past research and current advances. In the latter part of this review, we also provide an overview of forkhead box O-binding partners that control the transcriptional activity of forkhead box O factors. These two layers of regulation mostly overlap and thereby enable a more precise fine-tuning of forkhead box O functions involved in metabolism, longevity, and tumor suppression. This article is part of a Special Issue entitled: PI3K-AKT-FoxO axis in cancer and aging.
Collapse
Affiliation(s)
- Hiroaki Daitoku
- Life Science Center, Tsukuba Advanced Research Aliance, University of Tsukuba, Lbaraki, Japan
| | | | | |
Collapse
|
48
|
Abstract
Androgens are critical for normal prostate development and function, as well as prostate cancer initiation and progression. Androgens function mainly by regulating target gene expression through the androgen receptor (AR). Many studies have shown that androgen-AR signaling exerts actions on key events during prostate carcinogenesis. In this review, androgen action in distinct aspects of prostate carcinogenesis, including (i) cell proliferation, (ii) cell apoptosis, and (iii) prostate cancer metastasis will be discussed.
Collapse
Affiliation(s)
- Diping Wang
- Department of Urology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | |
Collapse
|
49
|
Carvalho M, Schwudke D, Sampaio JL, Palm W, Riezman I, Dey G, Gupta GD, Mayor S, Riezman H, Shevchenko A, Kurzchalia TV, Eaton S. Survival strategies of a sterol auxotroph. Development 2010; 137:3675-85. [PMID: 20940226 DOI: 10.1242/dev.044560] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The high sterol concentration in eukaryotic cell membranes is thought to influence membrane properties such as permeability, fluidity and microdomain formation. Drosophila cannot synthesize sterols, but do require them for development. Does this simply reflect a requirement for sterols in steroid hormone biosynthesis, or is bulk membrane sterol also essential in Drosophila? If the latter is true, how do they survive fluctuations in sterol availability and maintain membrane homeostasis? Here, we show that Drosophila require both bulk membrane sterol and steroid hormones in order to complete adult development. When sterol availability is restricted, Drosophila larvae modulate their growth to maintain membrane sterol levels within tight limits. When dietary sterol drops below a minimal threshold, larvae arrest growth and development in a reversible manner. Strikingly, membrane sterol levels in arrested larvae are dramatically reduced (dropping sixfold on average) in most tissues except the nervous system. Thus, sterols are dispensable for maintaining the basic membrane biophysical properties required for cell viability; these functions can be performed by non-sterol lipids when sterols are unavailable. However, bulk membrane sterol is likely to have essential functions in specific tissues during development. In tissues in which sterol levels drop, the overall level of sphingolipids increases and the proportion of different sphingolipid variants is altered. These changes allow survival, but not growth, when membrane sterol levels are low. This relationship between sterols and sphingolipids could be an ancient and conserved principle of membrane homeostasis.
Collapse
Affiliation(s)
- Maria Carvalho
- Max-Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse-108, 01307 Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chung WH, Dao RL, Chen LK, Hung SI. The role of genetic variants in human longevity. Ageing Res Rev 2010; 9 Suppl 1:S67-78. [PMID: 20708717 PMCID: PMC7105197 DOI: 10.1016/j.arr.2010.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 08/03/2010] [Indexed: 01/04/2023]
Abstract
Human longevity is a complex phenotype with a strong genetic predisposition. Increasing evidence has revealed the genetic antecedents of human longevity. This article aims to review the data of various case/control association studies that examine the difference in genetic polymorphisms between long-lived people and younger subjects across different human populations. There are more than 100 candidate genes potentially involved in human longevity; this article particularly focuses on genes of the insulin/IGF-1 pathway, FOXO3A, FOXO1A, lipoprotein metabolism (e.g., APOE and PON1), and cell-cycle regulators (e.g., TP53 and P21). Since the confirmed genetic components for human longevity are few to date, further precise assessment of the genetic contributions is required. Gaining a better understanding of the contribution of genetics to human longevity may assist in the design of improved treatment methods for age-related diseases, delay the aging process, and, ultimately, prolong the human lifespan.
Collapse
|