101
|
Forkhead box transcription factor 1: role in the pathogenesis of diabetic cardiomyopathy. Cardiovasc Diabetol 2016; 15:44. [PMID: 26956801 PMCID: PMC4784400 DOI: 10.1186/s12933-016-0361-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a disorder of the heart muscle in people with diabetes that can occur independent of hypertension or vascular disease. The underlying mechanism of DCM is incompletely understood. Some transcription factors have been suggested to regulate the gene program intricate in the pathogenesis of diabetes prompted cardiac injury. Forkhead box transcription factor 1 is a pleiotropic transcription factor that plays a pivotal role in a variety of physiological processes. Altered FOXO1 expression and function have been associated with cardiovascular diseases, and the important role of FOXO1 in DCM has begun to attract attention. In this review, we focus on the FOXO1 pathway and its role in various processes that have been related to DCM, such as metabolism, oxidative stress, endothelial dysfunction, inflammation and apoptosis.
Collapse
|
102
|
Hendrix MJC, Seftor EA, Seftor REB, Chao JT, Chien DS, Chu YW. Tumor cell vascular mimicry: Novel targeting opportunity in melanoma. Pharmacol Ther 2016; 159:83-92. [PMID: 26808163 PMCID: PMC4779708 DOI: 10.1016/j.pharmthera.2016.01.006] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In 1999, the American Journal of Pathology published an article, entitled "Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry" by Maniotis and colleagues, which ignited a spirited debate for several years and earned the journal's distinction of a "citation classic" (Maniotis et al., 1999). Tumor cell vasculogenic mimicry (VM), also known as vascular mimicry, describes the plasticity of aggressive cancer cells forming de novo vascular networks and is associated with the malignant phenotype and poor clinical outcome. The tumor cells capable of VM share the commonality of a stem cell-like, transendothelial phenotype, which may be induced by hypoxia. Since its introduction as a novel paradigm for melanoma tumor perfusion, many studies have contributed new findings illuminating the underlying molecular pathways supporting VM in a variety of tumors, including carcinomas, sarcomas, glioblastomas, astrocytomas, and melanomas. Of special significance is the lack of effectiveness of angiogenesis inhibitors on tumor cell VM, suggesting a selective resistance by this phenotype to conventional therapy. Facilitating the functional plasticity of tumor cell VM are key proteins associated with vascular, stem cell, extracellular matrix, and hypoxia-related signaling pathways--each deserving serious consideration as potential therapeutic targets and diagnostic indicators of the aggressive, metastatic phenotype. This review highlights seminal findings pertinent to VM, including the effects of a novel, small molecular compound, CVM-1118, currently under clinical development to target VM, and illuminates important molecular pathways involved in the suppression of this plastic, aggressive phenotype, using melanoma as a model.
Collapse
Affiliation(s)
- Mary J C Hendrix
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60614, United States; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States.
| | - Elisabeth A Seftor
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60614, United States
| | - Richard E B Seftor
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60614, United States; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | | | | | | |
Collapse
|
103
|
Hamacher-Brady A, Brady NR. Mitophagy programs: mechanisms and physiological implications of mitochondrial targeting by autophagy. Cell Mol Life Sci 2016; 73:775-95. [PMID: 26611876 PMCID: PMC4735260 DOI: 10.1007/s00018-015-2087-8] [Citation(s) in RCA: 302] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 02/07/2023]
Abstract
Mitochondria are an essential source of ATP for cellular function, but when damaged, mitochondria generate a plethora of stress signals, which lead to cellular dysfunction and eventually programmed cell death. Thus, a major component of maintaining cellular homeostasis is the recognition and removal of dysfunctional mitochondria through autophagy-mediated degradation, i.e., mitophagy. Mitophagy further constitutes a developmental program, and undergoes a high degree of crosstalk with apoptosis. Reduced mitochondrial quality control is linked to disease pathogenesis, suggesting the importance of process elucidation as a clinical target. Recent work has revealed multiple mitophagy programs that operate independently or undergo crosstalk, and require modulated autophagy receptor activities at outer membranes of mitochondria. Here, we review these mitophagy programs, focusing on pathway mechanisms which recognize and target mitochondria for sequestration by autophagosomes, as well as mechanisms controlling pathway activities. Furthermore, we provide an introduction to the currently available methods for detecting mitophagy.
Collapse
Affiliation(s)
- Anne Hamacher-Brady
- Lysosomal Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bioquant, University of Heidelberg, INF 267, BQ0045, 69120, Heidelberg, Germany.
| | - Nathan Ryan Brady
- Systems Biology of Cell Death Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany.
- Bioquant, University of Heidelberg, INF 267, BQ0045, 69120, Heidelberg, Germany.
| |
Collapse
|
104
|
Hesp K, Smant G, Kammenga JE. Caenorhabditis elegans DAF-16/FOXO transcription factor and its mammalian homologs associate with age-related disease. Exp Gerontol 2015; 72:1-7. [DOI: 10.1016/j.exger.2015.09.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 11/29/2022]
|
105
|
Liu YC, Chang PY, Chao CCK. CITED2 silencing sensitizes cancer cells to cisplatin by inhibiting p53 trans-activation and chromatin relaxation on the ERCC1 DNA repair gene. Nucleic Acids Res 2015; 43:10760-81. [PMID: 26384430 PMCID: PMC4678856 DOI: 10.1093/nar/gkv934] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
In this study, we show that silencing of CITED2 using small-hairpin RNA (shCITED2) induced DNA damage and reduction of ERCC1 gene expression in HEK293, HeLa and H1299 cells, even in the absence of cisplatin. In contrast, ectopic expression of ERCC1 significantly reduced intrinsic and induced DNA damage levels, and rescued the effects of CITED2 silencing on cell viability. The effects of CITED2 silencing on DNA repair and cell death were associated with p53 activity. Furthermore, CITED2 silencing caused severe elimination of the p300 protein and markers of relaxed chromatin (acetylated H3 and H4, i.e. H3K9Ac and H3K14Ac) in HEK293 cells. Chromatin immunoprecipitation assays further revealed that DNA damage induced binding of p53 along with H3K9Ac or H3K14Ac at the ERCC1 promoter, an effect which was almost entirely abrogated by silencing of CITED2 or p300. Moreover, lentivirus-based CITED2 silencing sensitized HeLa cell line-derived tumor xenografts to cisplatin in immune-deficient mice. These results demonstrate that CITED2/p300 can be recruited by p53 at the promoter of the repair gene ERCC1 in response to cisplatin-induced DNA damage. The CITED2/p300/p53/ERCC1 pathway is thus involved in the cell response to cisplatin and represents a potential target for cancer therapy.
Collapse
Affiliation(s)
- Yu-Chin Liu
- Tumor Biology Laboratory, Department of Biochemistry and Molecular Biology, Chang Gung University, 259 Wen-Hua first Road, Gueishan, Taoyuan 333, Taiwan, Republic of China Graduate Institute of Biomedical Sciences, Chang Gung University, 259 Wen-Hua first Road, Gueishan,Taoyuan 333, Taiwan, Republic of China
| | - Pu-Yuan Chang
- Tumor Biology Laboratory, Department of Biochemistry and Molecular Biology, Chang Gung University, 259 Wen-Hua first Road, Gueishan, Taoyuan 333, Taiwan, Republic of China
| | - Chuck C-K Chao
- Tumor Biology Laboratory, Department of Biochemistry and Molecular Biology, Chang Gung University, 259 Wen-Hua first Road, Gueishan, Taoyuan 333, Taiwan, Republic of China Graduate Institute of Biomedical Sciences, Chang Gung University, 259 Wen-Hua first Road, Gueishan,Taoyuan 333, Taiwan, Republic of China
| |
Collapse
|
106
|
Cho S, Cho M, Kim J, Kaeberlein M, Lee SJ, Suh Y. Syringaresinol protects against hypoxia/reoxygenation-induced cardiomyocytes injury and death by destabilization of HIF-1α in a FOXO3-dependent mechanism. Oncotarget 2015; 6:43-55. [PMID: 25415049 PMCID: PMC4381577 DOI: 10.18632/oncotarget.2723] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/06/2014] [Indexed: 02/07/2023] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) is a master regulator of hypoxic response and has been a prime therapeutic target for ischemia/reperfusion (I/R)-derived myocardial dysfunction and tissue damage. There is also increasing evidence that HIF-1 plays a central role in regulating aging, both through interactions with key longevity factors including Sirtuins and mTOR, as well as by directly promoting longevity in Caenorhabditis elegans. We investigated a novel function and the underlying mechanism of syringaresinol, a lignan compound, in modulation of HIF-1 and protection against cellular damage and death in a cardiomyocyte model of I/R injury. Syringaresinol caused destabilization of HIF-1α following H/R and then protected against hypoxia/reoxygenation (H/R)-induced cellular damage, apoptosis, and mitochondrial dysfunction in a dose-dependent manner. Knock-down of FOXO3 by specific siRNAs completely abolished the ability of syringaresinol to inhibit HIF-1 stabilization and apoptosis caused by H/R. Syringaresinol stimulated the nuclear localization and activity of FOXO3 leading to increased expression of antioxidant genes and decreased levels of reactive oxygen species (ROS) following H/R. Our results provide a new mechanistic insight into a functional role of syringaresinol against H/R-induced cardiomyocyte injury and death. The degradation of HIF-1α through activation of FOXO3 is a potential therapeutic strategy for ischemia-related diseases.
Collapse
Affiliation(s)
- Siyoung Cho
- R&D Unit, Amorepacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Miook Cho
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Juewon Kim
- R&D Unit, Amorepacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Sang Jun Lee
- R&D Unit, Amorepacific Corporation, Yongin-si, Gyeonggi-do, Korea
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA. Department of Medicine, Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY, USA. Institute for Aging Research, Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
107
|
Klotz LO, Sánchez-Ramos C, Prieto-Arroyo I, Urbánek P, Steinbrenner H, Monsalve M. Redox regulation of FoxO transcription factors. Redox Biol 2015; 6:51-72. [PMID: 26184557 PMCID: PMC4511623 DOI: 10.1016/j.redox.2015.06.019] [Citation(s) in RCA: 550] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/19/2022] Open
Abstract
Transcription factors of the forkhead box, class O (FoxO) family are important regulators of the cellular stress response and promote the cellular antioxidant defense. On one hand, FoxOs stimulate the transcription of genes coding for antioxidant proteins located in different subcellular compartments, such as in mitochondria (i.e. superoxide dismutase-2, peroxiredoxins 3 and 5) and peroxisomes (catalase), as well as for antioxidant proteins found extracellularly in plasma (e.g., selenoprotein P and ceruloplasmin). On the other hand, reactive oxygen species (ROS) as well as other stressful stimuli that elicit the formation of ROS, may modulate FoxO activity at multiple levels, including posttranslational modifications of FoxOs (such as phosphorylation and acetylation), interaction with coregulators, alterations in FoxO subcellular localization, protein synthesis and stability. Moreover, transcriptional and posttranscriptional control of the expression of genes coding for FoxOs is sensitive to ROS. Here, we review these aspects of FoxO biology focusing on redox regulation of FoxO signaling, and with emphasis on the interplay between ROS and FoxOs under various physiological and pathophysiological conditions. Of particular interest are the dual role played by FoxOs in cancer development and their key role in whole body nutrient homeostasis, modulating metabolic adaptations and/or disturbances in response to low vs. high nutrient intake. Examples discussed here include calorie restriction and starvation as well as adipogenesis, obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Lars-Oliver Klotz
- Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-Universität Jena, Dornburger Straße 29, 07743 Jena, Germany.
| | - Cristina Sánchez-Ramos
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier, 4, 28029 Madrid, Spain
| | - Ignacio Prieto-Arroyo
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier, 4, 28029 Madrid, Spain
| | - Pavel Urbánek
- Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-Universität Jena, Dornburger Straße 29, 07743 Jena, Germany
| | - Holger Steinbrenner
- Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-Universität Jena, Dornburger Straße 29, 07743 Jena, Germany
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier, 4, 28029 Madrid, Spain.
| |
Collapse
|
108
|
Poulsen RC, Carr AJ, Hulley PA. Cell proliferation is a key determinant of the outcome of FOXO3a activation. Biochem Biophys Res Commun 2015; 462:78-84. [PMID: 25935481 PMCID: PMC4449364 DOI: 10.1016/j.bbrc.2015.04.112] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/22/2015] [Indexed: 02/02/2023]
Abstract
The FOXO family of forkhead transcription factors have a pivotal role in determining cell fate in response to oxidative stress. FOXO activity can either promote cell survival or induce cell death. Increased FOXO-mediated cell death has been implicated in the pathogenesis of degenerative diseases affecting musculoskeletal tissues. The aim of this study was to determine the conditions under which one member of the FOXO family, FOXO3a, promotes cell survival as opposed to cell death. Treatment of primary human tenocytes with 1 pM hydrogen peroxide for 18 h resulted in increased protein levels of FOXO3a. In peroxide-treated cells cultured in low serum media, FOXO3a inhibited cell proliferation and protected against apoptosis. However in peroxide treated cells cultured in high serum media, cell proliferation was unchanged but level of apoptosis significantly increased. Similarly, in tenocytes transduced to over-express FOXO3a, cell proliferation was inhibited and level of apoptosis unchanged in cells cultured in low serum. However there was a robust increase in cell death in FOXO3a-expressing cells cultured in high serum. Inhibition of cell proliferation in either peroxide-treated or FOXO3a-expressing cells cultured in high serum protected against apoptosis induction. Conversely, addition of a Chk2 inhibitor to peroxide-treated or FOXO3a-expressing cells overrode the inhibitory effect of FOXO3a on cell proliferation and led to increased apoptosis in cells cultured in low serum. This study demonstrates that proliferating cells may be particularly susceptible to the apoptosis-inducing actions of FOXO3a. Inhibition of cell proliferation by FOXO3a may be a critical event in allowing the pro-survival rather than the pro-apoptotic activity of FOXO3a to prevail.
Collapse
Affiliation(s)
- Raewyn C. Poulsen
- Corresponding author. Present address: College of Health, Massey University, Private Bag 11-222, Palmerston North 4410, New Zealand. Fax: +64 6 3505657.
| | | | | |
Collapse
|
109
|
Song J, Yoon D, Christensen RD, Horvathova M, Thiagarajan P, Prchal JT. HIF-mediated increased ROS from reduced mitophagy and decreased catalase causes neocytolysis. J Mol Med (Berl) 2015; 93:857-66. [PMID: 26017143 DOI: 10.1007/s00109-015-1294-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/15/2015] [Accepted: 05/12/2015] [Indexed: 02/02/2023]
Abstract
UNLABELLED During prolonged hypoxia, hypoxia-inducible factors (HIFs) mediate an increase in erythropoiesis, leading to an increased red blood cell (RBC) mass and polycythemia. Upon return to normoxia, the increased RBC mass is abruptly overcorrected by the preferential destruction of hypoxia-formed young RBCs, a phenomenon termed neocytolysis. The molecular and biochemical mechanisms involved in neocytolysis are unknown. We developed a murine model of neocytolysis by exposing mice to 12 % oxygen for 10 days followed by return to normoxia. Upon return to normoxia, there was excessive accumulation of reactive oxygen species (ROS) in RBCs from an increased reticulocyte mitochondrial mass correlating with decreased Bnip3L transcripts (Bnip3L mediates reticulocyte mitophagy) and reduced catalase activity. During hypoxia, upregulated miR-21 resulted in low catalase activity in young RBCs. Furthermore, neocytolysis was attenuated by antioxidants and plasma catalase and blunted in mice that had constitutively high expression of HIFs. Among human neonates studied, we report data supporting the existence of neocytolysis during the first week of life. Together, these experiments indicate that the major mechanisms causing neocytolysis involve (1) production of young RBCs with low catalase during hypoxia and (2) lysis of the young RBCs after return to normoxia, mediated by ROS from an increased mitochondrial mass. KEY MESSAGES We report a mouse model of neocytolysis. Neocytolysis is caused by excessive ROS formation mediated by HIF. ROS is generated from increased mitochondria in reticulocytes. Hypoxia-generated RBCs have low catalase and are preferentially destroyed. Reduced catalase is regulated by increased microRNA-21.
Collapse
Affiliation(s)
- Jihyun Song
- Division of Hematology, School of Medicine, University of Utah, Salt Lake City, UT, 84132, USA
| | | | | | | | | | | |
Collapse
|
110
|
FOXO target gene CTDSP2 regulates cell cycle progression through Ras and p21(Cip1/Waf1). Biochem J 2015; 469:289-98. [PMID: 25990325 PMCID: PMC4613505 DOI: 10.1042/bj20140831] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 05/20/2015] [Indexed: 01/07/2023]
Abstract
Growth factor controlled activity of forkhead box O transcription factors results in altered gene expression, including expression of CTDSP2 (C-terminal domain small phosphatase 2). CTDSP2 can regulate cell cycle progression through Ras and the cyclin-dependent kinase inhibitor p21Cip1/Waf1. Activity of FOXO (forkhead box O) transcription factors is inhibited by growth factor–PI3K (phosphoinositide 3-kinase)–PKB (protein kinase B)/Akt signalling to control a variety of cellular processes including cell cycle progression. Through comparative analysis of a number of microarray datasets we identified a set of genes commonly regulated by FOXO proteins and PI3K–PKB/Akt, which includes CTDSP2 (C-terminal domain small phosphatase 2). We validated CTDSP2 as a genuine FOXO target gene and show that ectopic CTDSP2 can induce cell cycle arrest. We analysed transcriptional regulation after CTDSP2 expression and identified extensive regulation of genes involved in cell cycle progression, which depends on the phosphatase activity of CTDSP2. The most notably regulated gene is the CDK (cyclin-dependent kinase) inhibitor p21Cip1/Waf1 and in the present study we show that p21Cip1/Waf1 is partially responsible for the cell cycle arrest through decreasing cyclin–CDK activity. Our data suggest that CTDSP2 induces p21Cip1/Waf1 through increasing the activity of Ras. As has been described previously, Ras induces p21Cip1/Waf1 through p53-dependent and p53-independent pathways and indeed both p53 and MEK inhibition can mitigate the CTDSP2-induced p21Cip1/Waf1 mRNA up-regulation. In support of Ras activation by CTDSP2, depletion of endogenous CTDSP2 results in reduced Ras activity and thus CTDSP2 seems to be part of a larger set of genes regulated by FOXO proteins, which increase growth factor signalling upon FOXO activation.
Collapse
|
111
|
Wang SH, Cheng CY, Tang PC, Chen CF, Chen HH, Lee YP, Huang SY. Acute heat stress induces differential gene expressions in the testes of a broiler-type strain of Taiwan country chickens. PLoS One 2015; 10:e0125816. [PMID: 25932638 PMCID: PMC4416790 DOI: 10.1371/journal.pone.0125816] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/18/2015] [Indexed: 11/29/2022] Open
Abstract
The expression of testicular genes following acute heat stress has been reported in layer-type roosters, but few similar studies have been conducted on broilers. This study investigated the effect of acute heat stress on the gene expression in the testes of a broiler-type strain of Taiwan country chickens. Roosters were subjected to acute heat stress (38°C) for 4 h, and then exposed to 25°C, with testes collected 0, 2, and 6 h after the cessation of heat stress, using non-heat-stressed roosters as controls (n = 3 roosters per group). The body temperature and respiratory rate increased significantly (p<0.05) during the heat stress. The numbers of apoptotic cells increased 2 h after the acute heat stress (79 ± 7 vs. 322 ± 192, control vs. heat stress; p<0.05), which was earlier than the time of increase in layer-type roosters. Based on a chicken 44 K oligo microarray, 163 genes were found to be expressed significantly different in the testes of the heat-stressed chickens from those of the controls, including genes involved in the response to stimulus, protein metabolism, signal transduction, cell adhesion, transcription, and apoptosis. The mRNA expressions of upregulated genes, including HSP25, HSP90AA1, HSPA2, and LPAR2, and of downregulated genes, including CDH5, CTNNA3, EHF, CIRBP, SLA, and NTF3, were confirmed through quantitative real-time polymerase chain reaction (qRT-PCR). Moreover, numerous transcripts in the testes exhibited distinct expressions between the heat-stressed broiler-type and layer-type chickens. We concluded that the transcriptional responses of testes to acute heat stress may differ between the broiler-type and layer-type roosters. Whether the differential expression patterns associate with the heat-tolerance in the strains require a further exploration.
Collapse
Affiliation(s)
- Shih-Han Wang
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Chuen-Yu Cheng
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Pin-Chi Tang
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan
- Center for the Integrative and Evolutionary Galliformes Genomics, iEGG Center, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Chih-Feng Chen
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan
- Center for the Integrative and Evolutionary Galliformes Genomics, iEGG Center, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Hsin-Hsin Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Yen-Pai Lee
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
| | - San-Yuan Huang
- Department of Animal Science, National Chung Hsing University, Taichung, 40227, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan
- Center for the Integrative and Evolutionary Galliformes Genomics, iEGG Center, National Chung Hsing University, Taichung, 40227, Taiwan
- Center of Nanoscience and Nanotechnology, National Chung Hsing University, Taichung, 40227, Taiwan
- * E-mail:
| |
Collapse
|
112
|
Herrer I, Roselló-Lletí E, Ortega A, Tarazón E, Molina-Navarro MM, Triviño JC, Martínez-Dolz L, Almenar L, Lago F, Sánchez-Lázaro I, González-Juanatey JR, Salvador A, Portolés M, Rivera M. Gene expression network analysis reveals new transcriptional regulators as novel factors in human ischemic cardiomyopathy. BMC Med Genomics 2015; 8:14. [PMID: 25884818 PMCID: PMC4386080 DOI: 10.1186/s12920-015-0088-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 03/10/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Ischemic cardiomyopathy (ICM) is characterized by transcriptomic changes that alter cellular processes leading to decreased cardiac output. Because the molecular network of ICM is largely unknown, the aim of this study was to characterize the role of new transcriptional regulators in the molecular mechanisms underlying the responses to ischemia. METHODS Myocardial tissue explants from ICM patients and control (CNT) subjects were analyzed by RNA-Sequencing (RNA-Seq) and quantitative Real-Time PCR. RESULTS Enrichment analysis of the ICM transcriptomic profile allowed the characterization of novel master regulators. We found that the expression of the transcriptional regulators SP100 (-1.5-fold, p < 0.05), CITED2 (-3.8-fold, p < 0.05), CEBPD (-4.9-fold, p < 0.05) and BCL3 (-3.3-fold, p < 0.05) were lower in ICM than in CNT. To gain insights into the molecular network defined by the transcription factors, we identified CEBPD, BCL3, and HIF1A target genes in the RNA-Seq datasets. We further characterized the biological processes of the target genes by gene ontology annotation. Our results suggest that CEBPD-inducible genes with roles in the inhibition of apoptosis are downregulated and that BCL3-repressible genes are involved in the regulation of cellular metabolism in ICM. Moreover, our results suggest that CITED2 downregulation causes increased expression of HIF1A target genes. Functional analysis of HIF1A target genes revealed that hypoxic and stress response genes are activated in ICM. Finally, we found a significant correlation between the mRNA levels of BCL3 and the mRNA levels of both CEBPD (r = 0.73, p < 0.001) and CITED2 (r = 0.56, p < 0.05). Interestingly, CITED2 mRNA levels are directly related to ejection fraction (EF) (r = 0.54, p < 0.05). CONCLUSIONS Our data indicate that changes in the expression of SP100, CITED2, CEBPD, and BCL3 affect their transcription regulatory networks, which subsequently alter a number of biological processes in ICM patients. The relationship between CITED2 mRNA levels and EF emphasizes the importance of this transcription factor in ICM. Moreover, our findings identify new mechanisms used to interpret gene expression changes in ICM and provide valuable resources for further investigation of the molecular basis of human cardiac ischemic response.
Collapse
Affiliation(s)
- Isabel Herrer
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Esther Roselló-Lletí
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Ana Ortega
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Estefanía Tarazón
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - María Micaela Molina-Navarro
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | | | - Luis Martínez-Dolz
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - Luis Almenar
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - Francisca Lago
- Cellular and Molecular Cardiology Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago Compostela, Spain.
| | - Ignacio Sánchez-Lázaro
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago Compostela, Spain.
| | - Antonio Salvador
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - Manuel Portolés
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| |
Collapse
|
113
|
Kim SY, Ko YS, Park J, Choi Y, Park JW, Kim Y, Pyo JS, Yoo YB, Lee JS, Lee BL. Forkhead Transcription Factor FOXO1 Inhibits Angiogenesis in Gastric Cancer in Relation to SIRT1. Cancer Res Treat 2015; 48:345-54. [PMID: 25761483 PMCID: PMC4720104 DOI: 10.4143/crt.2014.247] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 12/05/2014] [Indexed: 01/26/2023] Open
Abstract
Purpose We previously reported that forkhead transcription factors of the O class 1 (FOXO1) expression in gastric cancer (GC) was associated with angiogenesis-related molecules. However, there is little experimental evidence for the direct role of FOXO1 in GC. In the present study, we investigated the effect of FOXO1 on the tumorigenesis and angiogenesis in GC and its relationship with SIRT1. Materials and Methods Stable GC cell lines (SNU-638 and SNU-601) infected with a lentivirus containing FOXO1 shRNA were established for animal studies as well as cell culture experiments. We used xenograft tumors in nude mice to evaluate the effect of FOXO1 silencing on tumor growth and angiogenesis. In addition, we examined the association between FOXO1 and SIRT1 by immunohistochemical tissue array analysis of 471 human GC specimens and Western blot analysis of xenografted tumor tissues. Results In cell culture, FOXO1 silencing enhanced hypoxia inducible factor-1α (HIF-1α) expression and GC cell growth under hypoxic conditions, but not under normoxic conditions. The xenograft study showed that FOXO1 downregulation enhanced tumor growth, microvessel areas, HIF-1α activation and vascular endothelial growth factor (VEGF) expression. In addition, inactivated FOXO1 expression was associated with SIRT1 expression in human GC tissues and xenograft tumor tissues. Conclusion Our results indicate that FOXO1 inhibits GC growth and angiogenesis under hypoxic conditions via inactivation of the HIF-1α–VEGF pathway, possibly in association with SIRT1. Thus, development of treatment modalities aiming at this pathway might be useful for treating GC.
Collapse
Affiliation(s)
- Sue Youn Kim
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Young San Ko
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Jinju Park
- Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yiseul Choi
- Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea ; Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bok Yoo
- Department of Anatomy, Dankook University School of Medicine, Cheonan, Korea
| | - Jae-Seon Lee
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | - Byung Lan Lee
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea ; Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea ; Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
114
|
Korthuis PM, Berger G, Bakker B, Rozenveld-Geugien M, Jaques J, de Haan G, Schuringa JJ, Vellenga E, Schepers H. CITED2-mediated human hematopoietic stem cell maintenance is critical for acute myeloid leukemia. Leukemia 2015; 29:625-35. [PMID: 25184385 DOI: 10.1038/leu.2014.259] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 02/07/2023]
Abstract
As the transcriptional coactivator CITED2 (CBP/p300-interacting-transactivator-with-an ED-rich-tail 2) can be overexpressed in acute myeloid leukemia (AML) cells, we analyzed the consequences of high CITED2 expression in normal and AML cells. CITED2 overexpression in normal CD34(+) cells resulted in enhanced hematopoietic stem and progenitor cell (HSPC) output in vitro, as well as in better hematopoietic stem cell (HSC) engraftability in NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice. This was because of an enhanced quiescence and maintenance of CD34(+)CD38(-) HSCs, due in part to an increased expression of the cyclin-dependent kinase inhibitor CDKN1A. We demonstrated that PU.1 is a critical regulator of CITED2, as PU.1 repressed CITED2 expression in a DNA methyltransferase 3A/B (DNMT3A/B)-dependent manner in normal CD34(+) cells. CD34(+) cells from a subset of AML patients displayed higher expression levels of CITED2 as compared with normal CD34(+) HSPCs, and knockdown of CITED2 in AML CD34(+) cells led to a loss of long-term expansion, both in vitro and in vivo. The higher CITED2 expression resulted from reduced PU.1 activity and/or dysfunction of mutated DNMT3A/B. Collectively, our data demonstrate that increased CITED2 expression results in better HSC maintenance. In concert with low PU.1 levels, this could result in a perturbed myeloid differentiation program that contributes to leukemia maintenance.
Collapse
MESH Headings
- Animals
- Antigens, CD34/genetics
- Antigens, CD34/metabolism
- Cell Proliferation
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methyltransferase 3A
- Female
- Gene Expression Regulation, Leukemic
- Graft Survival
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred NOD
- Mutation
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transplantation, Heterologous
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- P M Korthuis
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - G Berger
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B Bakker
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Rozenveld-Geugien
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J Jaques
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - G de Haan
- Department of Stem Cell Biology, European Research Institute for the Biology of Aging (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J J Schuringa
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E Vellenga
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - H Schepers
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
115
|
Xia W, Xie C, Jiang M, Hou M. Improved survival of mesenchymal stem cells by macrophage migration inhibitory factor. Mol Cell Biochem 2015; 404:11-24. [PMID: 25701358 PMCID: PMC4544672 DOI: 10.1007/s11010-015-2361-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/14/2015] [Indexed: 12/22/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a critical inflammatory cytokine that was recently associated with progenitor cell survival and potently inhibits apoptosis. We examined the protective effect of MIF on hypoxia/serum deprivation (SD)-induced apoptosis of mesenchymal stem cells (MSCs), as well as the possible mechanisms. MSCs were obtained from rat bone marrow and cultured in vitro. Apoptosis was induced by culturing MSCs under hypoxia/SD conditions for up to 24 h and assessed by flow cytometry. Expression levels of c-Met, Akt, and FOXO3a were detected by Western blotting. CD74 expression was detected by qRT-PCR, Western blot, and immunofluorescence. Oxidative stress under hypoxia/SD was examined by detection of reactive oxygen species (ROS) and activity of superoxide dismutase (SOD) and malondialdehyde (MDA). Hypoxia/SD-induced apoptosis was significantly attenuated by recombinant rat MIF in a concentration-dependent manner. MIF induced CD74-asssociated c-Met activation, which was blocked by knocking down CD74 expression using siRNA. MIF also induced Akt and associated FOXO3a phosphorylation, and this effect was abolished by knocking down either CD74 or Akt. In addition, MIF decreased oxidative stress in MSCs, as shown by decreased ROS and MDA, and increased the activity of SOD. Knockdown of CD74, Akt, or FOXO3a largely attenuated the anti-apoptotic effect of MIF and its ability to protect against oxidative stress. MIF protected MSCs from hypoxia/SD-induced apoptosis by interacting with CD74 to stimulate c-Met, leading to downstream PI3K/Akt-FOXO3a signaling and decreased oxidative stress.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | | | | | | |
Collapse
|
116
|
Charitou P, Rodriguez-Colman M, Gerrits J, van Triest M, Groot Koerkamp M, Hornsveld M, Holstege F, Verhoeven-Duif NM, Burgering BMT. FOXOs support the metabolic requirements of normal and tumor cells by promoting IDH1 expression. EMBO Rep 2015; 16:456-66. [PMID: 25648147 DOI: 10.15252/embr.201439096] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 01/08/2015] [Indexed: 01/02/2023] Open
Abstract
FOXO transcription factors are considered bona fide tumor suppressors; however, recent studies showed FOXOs are also required for tumor survival. Here, we identify FOXOs as transcriptional activators of IDH1. FOXOs promote IDH1 expression and thereby maintain the cytosolic levels of α-ketoglutarate and NADPH. In cancer cells carrying mutant IDH1, FOXOs likewise stimulate mutant IDH1 expression and maintain the levels of the oncometabolite 2-hydroxyglutarate, which stimulates cancer cell proliferation and inhibits TET enzymes and histone demethylases. Combined, our data provide a new paradigm for the paradoxical role of FOXOs in both tumor suppression and promotion.
Collapse
Affiliation(s)
- Paraskevi Charitou
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| | - Maria Rodriguez-Colman
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| | - Johan Gerrits
- Department of Medical Genetics, UMC Utrecht, Utrecht, The Netherlands
| | - Miranda van Triest
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| | - Marian Groot Koerkamp
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| | - Marten Hornsveld
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| | - Frank Holstege
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| | | | - Boudewijn M T Burgering
- Department of Molecular Cancer Research, University Medical Center Utrecht, CG Utrecht, The Netherlands
| |
Collapse
|
117
|
SIRT3 interactions with FOXO3 acetylation, phosphorylation and ubiquitinylation mediate endothelial cell responses to hypoxia. Biochem J 2014; 464:157-68. [DOI: 10.1042/bj20140213] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This article reports that hypoxia elicits SIRT3 to deacetylate FOXO3 in endothelial cells. This drives an increase in the expression of mitochondrial antioxidant enzymes, reduces accumulation of reactive oxygen species in mitochondria and thereby confers cellular capacity to adapt to hypoxia.
Collapse
|
118
|
Feng J, Meng C, Xing D. Aβ induces PUMA activation: a new mechanism for Aβ-mediated neuronal apoptosis. Neurobiol Aging 2014; 36:789-800. [PMID: 25457551 DOI: 10.1016/j.neurobiolaging.2014.10.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 09/02/2014] [Accepted: 10/07/2014] [Indexed: 12/11/2022]
Abstract
p53 upregulated modulator of apoptosis (PUMA) is a promising tumor therapy target because it elicits apoptosis and profound sensitivity to radiation and chemotherapy. However, inhibition of PUMA may be beneficial for curbing excessive apoptosis associated with neurodegenerative disorders. Alzheimer's disease (AD) is a representative neurodegenerative disease in which amyloid-β (Aβ) deposition causes neurotoxicity. The regulation of PUMA during Aβ-induced neuronal apoptosis remains poorly understood. Here, we reported that PUMA expression was significantly increased in the hippocampus of transgenic mice models of AD and hippocampal neurons in response to Aβ. PUMA knockdown protected the neurons against Aβ-induced apoptosis. Furthermore, besides p53, PUMA transactivation was also regulated by forkhead box O3a through p53-independent manner following Aβ treatment. Notably, PUMA contributed to neuronal apoptosis through competitive binding of apoptosis repressor with caspase recruitment domain to activate caspase-8 that cleaved Bid into tBid to accelerate Bax mitochondrial translocation, revealing a novel pathway of Bax activation by PUMA to mediate Aβ-induced neuronal apoptosis. Together, we demonstrated that PUMA activation involved in Aβ-induced apoptosis, representing a drug target to antagonize AD progression.
Collapse
Affiliation(s)
- Jie Feng
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Chengbo Meng
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.
| |
Collapse
|
119
|
Zheng X, Zhai B, Koivunen P, Shin SJ, Lu G, Liu J, Geisen C, Chakraborty AA, Moslehi JJ, Smalley DM, Wei X, Chen X, Chen Z, Beres JM, Zhang J, Tsao JL, Brenner MC, Zhang Y, Fan C, DePinho RA, Paik J, Gygi SP, Kaelin WG, Zhang Q. Prolyl hydroxylation by EglN2 destabilizes FOXO3a by blocking its interaction with the USP9x deubiquitinase. Genes Dev 2014; 28:1429-44. [PMID: 24990963 PMCID: PMC4083087 DOI: 10.1101/gad.242131.114] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The three EglN prolyl hydroxylases (EglN1, EglN2, and EglN3) regulate the stability of the HIF transcription factor. We recently showed that loss of EglN2, however, also leads to down-regulation of Cyclin D1 and decreased cell proliferation in a HIF-independent manner. Here we report that EglN2 can hydroxylate FOXO3a on two specific prolyl residues in vitro and in vivo. Hydroxylation of these sites prevents the binding of USP9x deubiquitinase, thereby promoting the proteasomal degradation of FOXO3a. FOXO transcription factors can repress Cyclin D1 transcription. Failure to hydroxylate FOXO3a promotes its accumulation in cells, which in turn suppresses Cyclin D1 expression. These findings provide new insights into post-transcriptional control of FOXO3a and provide a new avenue for pharmacologically altering Cyclin D1 activity.
Collapse
Affiliation(s)
- Xingnan Zheng
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Bo Zhai
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| | - Sandra J Shin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Gang Lu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Jiayun Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Christoph Geisen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Abhishek A Chakraborty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Javid J Moslehi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - David M Smalley
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Xin Wei
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Zhengming Chen
- Department of Public Health, Division of Biostatistics and Epidemiology, Weill Cornell Medical College, New York, New York 10065, USA
| | - Justine M Beres
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Jing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Jen Lan Tsao
- Fibrogen, Incorporated, San Francisco, California 94158, USA
| | | | - Yuqing Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
120
|
Quercetin attenuates cell apoptosis of oxidant-stressed SK-N-MC cells while suppressing up-regulation of the defensive element, HIF-1α. Neuroscience 2014; 277:780-93. [PMID: 25108166 DOI: 10.1016/j.neuroscience.2014.07.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 07/23/2014] [Accepted: 07/23/2014] [Indexed: 01/25/2023]
Abstract
Evidence is emerging that reactive oxygen species (ROS)-induced oxidative stress has a crucial role in the pathogenesis of neurodegenerative diseases. To find the effective therapies for neurodegenerative diseases, evaluation of the relevant molecular mechanisms is necessary. In the current study, we investigated the effects of hydrogen peroxide (H2O2)-induced oxidative stress on SK-N-MC cell death with focus on HIF-1α, Foxo3a and Notch1 signaling factors. Our results revealed that H2O2 reduced viability of cells through up-regulation of p53 followed by increase in Bax/Bcl2 ratio. In addition, H2O2 increased intracellular levels of HIF-1α, Foxo-3a and Notch intracellular domain (NICD). However, Quercetin decreased cell contents of HIF-1α, Foxo-3a and NICD as well as pro-apoptotic factors including p53 and Bax compared to H2O2-treated cells. Additionally, we found that HIF-1α down-regulation reduced Foxo3a and NICD contents parallel to up-regulation of p53 and Bax and led to further vulnerability to oxidative stress-induced cell death. In contrast, Notch inhibition resulted in HIF-1α/Foxo3a signaling pathway up-regulation, suggesting the bidirectional crosstalk between HIF-1α and Notch1. These results collectively suggest that ROS are involved in activation of both the defensive and pro-apoptotic pathways encompassing HIF-1α and p53, respectively. Regarding the HIF-1α-mediated neuroprotection role, elucidation of the molecular mechanism would certainly be essential for effective drug design against neurodegenerative diseases.
Collapse
|
121
|
Cited2 is required in trophoblasts for correct placental capillary patterning. Dev Biol 2014; 392:62-79. [DOI: 10.1016/j.ydbio.2014.04.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/21/2014] [Accepted: 04/23/2014] [Indexed: 01/14/2023]
|
122
|
Wu WJ, Zhang XK, Zheng XF, Yang YH, Jiang SD, Jiang LS. SHH-dependent knockout of HIF-1 alpha accelerates the degenerative process in mouse intervertebral disc. Int J Immunopathol Pharmacol 2014; 26:601-9. [PMID: 24067457 DOI: 10.1177/039463201302600304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hypoxia-inducible factor-1alpha (HIF-1 alpha) has been reported to have an important role in the metabolism and synthesis of extracellular matrix of the nucleus pulposus cells (NPCs) and was assumed to be involved in the process of intervertebral disc degeneration. The objective of this study was to investigate the role of HIF-1alpha in disc degeneration in vivo using a conditional HIF-1alpha knockout (KO) mouse model. ShhCre transgenic mice were mated with HIF-1 alpha fl/fl mice to generate conditional HIF-1alpha KO mice (HIF-1alpha fl/fl-ShhCre+). Three mice of each genotype (Wide-type and HIF-1alpha KO) at the age of 3 days, 6, and 12 weeks were sacrificed after genotyping. Five lumbar disc samples were harvested from each mouse, with a total of 45 disc samples for each genotype. In situ hybridization and immunohistochemical analysis were used to check the efficacy of HIF-1alpha knockout. Histological grading of the disc degeneration was performed according to the classification system proposed by Boos et al. Picro-sirius red staining, Safranine O/fast green staining and immunohistochemical study were used to evaluate the expression of aggrecan, type-II collagen and vascular endothelial growth factor (VEGF). Histologic analysis revealed more NPC deaths and signs of degeneration in HIF-1alpha KO mice and the degeneration scores of HIF-1alpha KO mice were significantly higher than those of the Wide-type mice at the age of 6 weeks and 12 weeks. There were less expressions of aggrecan, type-II collagen and VEGF in the intervertebral discs of HIF1-alpha KO mice than in those of wild-type mice. Taken together, the results of our study indicated that HIF-1alpha is a pivotal contributor to NPC survival and the homeotasis of extracellular matrix through the HIF-1alpha/VEGF signaling pathway, and plays an important role in the development of disc degeneration.
Collapse
Affiliation(s)
- W J Wu
- Department of Orthopaedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | |
Collapse
|
123
|
Barilovits SJ, Newsom KJ, Bickford JS, Beachy DE, Rhoton-Vlasak A, Nick HS. Characterization of a mechanism to inhibit ovarian follicle activation. Fertil Steril 2014; 101:1450-7. [PMID: 24559722 DOI: 10.1016/j.fertnstert.2014.01.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/14/2014] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To demonstrate that a small molecule can induce the transcription factor Foxo3 in the ovary and lead to inhibition of follicle activation. DESIGN Cell culture, organ culture, and animal studies. SETTING University-based laboratory. ANIMAL(S) 23 female C57BL/6 mice. INTERVENTION(S) Human ovary cells and mouse ovaries in culture treated with 2-deoxyglucose (2-DG) to mimic glucose deprivation, and mice intraperitoneally injected with 100 mg/kg, 300 mg/kg, or 600 mg/kg 2-DG daily for 2 weeks. MAIN OUTCOME MEASURE(S) In cell and organ culture, Foxo3 expression analyzed by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR); in treated animals, expression of genes regulated by nutrient deprivation (Foxo3, ATF4, GRP78, CHOP, ASNS, c-Myc) measured in brain, kidney, and ovary by qRT-PCR; and ovarian follicles histologically classified and counted. RESULT(S) Foxo3 expression is induced by 2-DG at both the mRNA and protein level in human ovarian cell culture, possibly through ATF4-dependent gene regulation. Foxo3 expression is also induced by 2-DG in ovarian organ culture. Treatment of mice with 100 mg/kg 2-DG resulted in a 2.6 fold induction of Foxo3 in the ovary and a 58% decrease in type 3a primary follicles. CONCLUSION(S) Expression of Foxo3 is induced by nutrient deprivation in cell culture, organ culture, and in vivo. In mice, 2-DG treatment results in an inhibition of primordial follicle activation. These data indicate that Foxo3 induction by 2-DG may be useful for fertility preservation.
Collapse
Affiliation(s)
- Sarah J Barilovits
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida; Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida
| | - Kimberly J Newsom
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida
| | - Justin S Bickford
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida
| | - Dawn E Beachy
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida
| | - Alice Rhoton-Vlasak
- Department of Obstetrics and Gynecology, College of Medicine, University of Florida, Gainesville, Florida
| | - Harry S Nick
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida; Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
124
|
Sinha I, Allen JE, Pinto JT, Sinha R. Methylseleninic acid elevates REDD1 and inhibits prostate cancer cell growth despite AKT activation and mTOR dysregulation in hypoxia. Cancer Med 2014; 3:252-64. [PMID: 24515947 PMCID: PMC3987075 DOI: 10.1002/cam4.198] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/22/2013] [Accepted: 12/26/2013] [Indexed: 12/23/2022] Open
Abstract
Methylseleninic acid (MSeA) is a monomethylated selenium metabolite theoretically derived from subsequent β-lyase or transamination reactions of dietary Se-methylselenocysteine that has potent antitumor activity by inhibiting cell proliferation of several cancers. Our previous studies showed that MSeA promotes apoptosis in invasive prostate cancer cells in part by downregulating hypoxia-inducible factor HIF-1α. We have now extended these studies to evaluate the impact of MSeA on REDD1 (an mTOR inhibitor) in inducing cell death of invasive prostate cancer cells in hypoxia. In both PTEN+ and PTEN− prostate cancer cells we show that MSeA elevates REDD1 and phosphorylation of AKT along with p70S6K in hypoxia. Furthermore, REDD1 induction by MSeA is independent of AKT and the mTOR inhibition in prostate cancer cells causes partial resistance to MSeA-induced growth reduction in hypoxia. Our data suggest that MSeA induces REDD1 and inhibits prostate cancer cell growth in hypoxia despite activation of AKT and dysregulation of mTOR. MSeA elevates REDD1 and AKT to promote cell death in invasive prostate cancer cells in hypoxia.
Collapse
Affiliation(s)
- Indu Sinha
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Penn State Hershey Cancer Institute, Hershey, Pennsylvania
| | | | | | | |
Collapse
|
125
|
Miller BA, Hoffman NE, Merali S, Zhang XQ, Wang J, Rajan S, Shanmughapriya S, Gao E, Barrero CA, Mallilankaraman K, Song J, Gu T, Hirschler-Laszkiewicz I, Koch WJ, Feldman AM, Madesh M, Cheung JY. TRPM2 channels protect against cardiac ischemia-reperfusion injury: role of mitochondria. J Biol Chem 2014; 289:7615-29. [PMID: 24492610 DOI: 10.1074/jbc.m113.533851] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cardiac TRPM2 channels were activated by intracellular adenosine diphosphate-ribose and blocked by flufenamic acid. In adult cardiac myocytes the ratio of GCa to GNa of TRPM2 channels was 0.56 ± 0.02. To explore the cellular mechanisms by which TRPM2 channels protect against cardiac ischemia/reperfusion (I/R) injury, we analyzed proteomes from WT and TRPM2 KO hearts subjected to I/R. The canonical pathways that exhibited the largest difference between WT-I/R and KO-I/R hearts were mitochondrial dysfunction and the tricarboxylic acid cycle. Complexes I, III, and IV were down-regulated, whereas complexes II and V were up-regulated in KO-I/R compared with WT-I/R hearts. Western blots confirmed reduced expression of the Complex I subunit and other mitochondria-associated proteins in KO-I/R hearts. Bioenergetic analyses revealed that KO myocytes had a lower mitochondrial membrane potential, mitochondrial Ca(2+) uptake, ATP levels, and O2 consumption but higher mitochondrial superoxide levels. Additionally, mitochondrial Ca(2+) uniporter (MCU) currents were lower in KO myocytes, indicating reduced mitochondrial Ca(2+) uptake was likely due to both lower ψm and MCU activity. Similar to isolated myocytes, O2 consumption and ATP levels were also reduced in KO hearts. Under a simulated I/R model, aberrant mitochondrial bioenergetics was exacerbated in KO myocytes. Reactive oxygen species levels were also significantly higher in KO-I/R compared with WT-I/R heart slices, consistent with mitochondrial dysfunction in KO-I/R hearts. We conclude that TRPM2 channels protect the heart from I/R injury by ameliorating mitochondrial dysfunction and reducing reactive oxygen species levels.
Collapse
|
126
|
Dwyer DS, Weeks K, Aamodt EJ. Drug discovery based on genetic and metabolic findings in schizophrenia. Expert Rev Clin Pharmacol 2014; 1:773-89. [PMID: 24410607 DOI: 10.1586/17512433.1.6.773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent progress in the genetics of schizophrenia provides the rationale for re-evaluating causative factors and therapeutic strategies for this disease. Here, we review the major candidate susceptibility genes and relate the aberrant function of these genes to defective regulation of energy metabolism in the schizophrenic brain. Disturbances in energy metabolism potentially lead to neurodevelopmental deficits, impaired function of the mature nervous system and failure to maintain neurites/dendrites and synaptic connections. Current antipsychotic drugs do not specifically address these underlying deficits; therefore, a new generation of more effective medications is urgently needed. Novel targets for future drug discovery are identified in this review. The coordinated application of structure-based drug design, systems biology and research on model organisms may greatly facilitate the search for next-generation antipsychotic drugs.
Collapse
Affiliation(s)
- Donard S Dwyer
- Professor and Director of Basic Research, Departments of Psychiatry and Pharmacology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.
| | | | | |
Collapse
|
127
|
Tooze RM. A replicative self-renewal model for long-lived plasma cells: questioning irreversible cell cycle exit. Front Immunol 2013; 4:460. [PMID: 24385976 PMCID: PMC3866514 DOI: 10.3389/fimmu.2013.00460] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/02/2013] [Indexed: 12/23/2022] Open
Abstract
Plasma cells are heterogenous in terms of their origins, secretory products, and lifespan. A current paradigm is that cell cycle exit in plasma cell differentiation is irreversible, following a pattern familiar in short-lived effector populations in other hemopoietic lineages. This paradigm no doubt holds true for many plasma cells whose lifespan can be measured in days following the completion of differentiation. Whether this holds true for long-lived bone marrow plasma cells that are potentially maintained for the lifespan of the organism is less apparent. Added to this the mechanisms that establish and maintain cell cycle quiescence in plasma cells are incompletely defined. Gene expression profiling indicates that in the transition of human plasmablasts to long-lived plasma cells a range of cell cycle regulators are induced in a pattern that suggests a quiescence program with potential for cell cycle re-entry. Here a model of relative quiescence with the potential for replicative self-renewal amongst long-lived plasma cells is explored. The implications of such a mechanism would be diverse, and the argument is made here that current evidence is not sufficiently strong that the possibility should be disregarded.
Collapse
Affiliation(s)
- Reuben M Tooze
- Section of Experimental Haematology, Leeds Institute of Cancer and Pathology, University of Leeds , Leeds , UK ; Haematological Malignancy Diagnostic Service, Leeds Teaching Hospitals NHS Trust , Leeds , UK
| |
Collapse
|
128
|
Yang J, Carra S, Zhu WG, Kampinga HH. The regulation of the autophagic network and its implications for human disease. Int J Biol Sci 2013; 9:1121-33. [PMID: 24339733 PMCID: PMC3858585 DOI: 10.7150/ijbs.6666] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 06/28/2013] [Indexed: 12/19/2022] Open
Abstract
Autophagy has attracted a lot of attention in recent years. More and more proteins and signaling pathways have been discovered that somehow feed into the autophagy regulatory pathways. Regulation of autophagy is complex and condition-specific, and in several diseases, autophagic fluxes are changed. Here, we review the most well-established concepts in this field as well as the reported signaling pathways or components which steer the autophagy machinery. Furthermore, we will highlight how autophagic fluxes are changed in various diseases either as cause for or as response to deal with an altered cellular homeostasis and how modulation of autophagy might be used as potential therapy for such diseases.
Collapse
Affiliation(s)
- Jing Yang
- 1. Department of Cell Biology; University Medical Center Groningen, University of Groningen; Groningen, The Netherlands. ; 2. Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing 100191, China
| | | | | | | |
Collapse
|
129
|
The interaction between Toll-like receptor 4 signaling pathway and hypoxia-inducible factor 1α in lung ischemia-reperfusion injury. J Surg Res 2013; 188:290-7. [PMID: 24472280 DOI: 10.1016/j.jss.2013.11.1086] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 11/03/2013] [Accepted: 11/15/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND Lung ischemia-reperfusion injury (LIRI) is the life-threatening complication occurring after lung transplantation. Toll-like receptor 4 (TLR4) signaling pathway and hypoxia-inducible factor-1α (HIF-1α) are intimately involved in the development and progression of various inflammatory and hypoxia diseases; however, the relationship of them in LIRI in vivo is still far from clear. MATERIALS AND METHODS Forty-five Sprague-Dawley rats were randomly distributed in nine groups: (1) Sham group, (2) LIRI group, (3) LIRI + saline control group, (4) LIRI + dimethyl Sulfoxide control group, (5) LIRI + lipopolysaccharide group, (6) LIRI + TAK-242 group (TAK-242 is a TLR4 inhibitor, ethyl (6R)-6- [N-(2-chloro-4-fluorophenyl)sulfamoyl]cyclohex-1-ene-1-carboxylate), (7) LIRI + thioredoxin group (thioredoxin is an apoptosis signal-regulating kinase 1 (ASK1) inhibitor), (8) LIRI + SB203580 group (SB203580 is a p38 inhibitor), and (9) LIRI + chetomin group (chetomin is a HIF-1α inhibitor). The interaction between TLR4 signaling pathway (including TLR4, myeloid differentiation primary response gene 88 (MyD88), TIR-domain-containing adapter-inducing interferon-β (TRIF), ASK1, and p38) and HIF-1α and the role of TLR4-dependent HIF-1α were analyzed. RESULTS In LIRI, HIF-1α accumulation was induced in a TLR4-dependent fashion, and MyD88, but not TRIF, and activation of ASK1 and p38 were found to be critical for TLR4-mediated HIF-1α accumulation. HIF-1α protein played a critical role in TLR4-mediated lung injury of LIRI (including inflammation, cell apoptosis, and lung damage). HIF-1α protein upregulated TLR4 expression of LIRI in a positive feedback manner. CONCLUSIONS We identify that the TLR4-HIF-1 loop may be existed in LIRI. Therefore, we suggest that the interaction between them may represent a novel therapeutic target for the development of novel target-based therapies of LIRI.
Collapse
|
130
|
Li Q, Hakimi P, Liu X, Yu WM, Ye F, Fujioka H, Raza S, Shankar E, Tang F, Dunwoodie SL, Danielpour D, Hoppel CL, Ramírez-Bergeron DL, Qu CK, Hanson RW, Yang YC. Cited2, a transcriptional modulator protein, regulates metabolism in murine embryonic stem cells. J Biol Chem 2013; 289:251-63. [PMID: 24265312 DOI: 10.1074/jbc.m113.497594] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CREB-binding protein (CBP)/p300 interacting transactivator with glutamic acid (Glu) and aspartic acid (Asp)-tail 2 (Cited2) was recently shown to be essential for gluconeogenesis in the adult mouse. The metabolic function of Cited2 in mouse embryonic stem cells (mESCs) remains elusive. In the current study, the metabolism of glucose was investigated in mESCs, which contained a deletion in the gene for Cited2 (Cited2(Δ/-)). Compared with its parental wild type counterpart, Cited2(Δ/-) ESCs have enhanced glycolysis, alternations in mitochondria morphology, reduced glucose oxidation, and decreased ATP content. Cited2 is recruited to the hexokinase 1 (HK1) gene promoter to regulate transcription of HK1, which coordinates glucose metabolism in wild type ESCs. Reduced glucose oxidation and enhanced glycolytic activity in Cited2(Δ/-) ESCs correlates with defective differentiation during hypoxia, which is reflected in an increased expression of pluripotency marker (Oct4) and epiblast marker (Fgf5) and decreased expression of lineage specification markers (T, Gata-6, and Cdx2). Knockdown of hypoxia inducible factor-1α in Cited2(Δ/-) ESCs re-initiates the expression of differentiation markers T and Gata-6. Taken together, a deletion of Cited2 in mESCs results in abnormal mitochondrial morphology and impaired glucose metabolism, which correlates with a defective cell fate decision.
Collapse
Affiliation(s)
- Qiang Li
- From the Departments of Biochemistry
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Du J, Li Q, Tang F, Puchowitz MA, Fujioka H, Dunwoodie SL, Danielpour D, Yang YC. Cited2 is required for the maintenance of glycolytic metabolism in adult hematopoietic stem cells. Stem Cells Dev 2013; 23:83-94. [PMID: 24083546 DOI: 10.1089/scd.2013.0370] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mammalian adult hematopoietic stem cells (HSCs) reside in the hypoxic bone marrow microenvironment and display a distinct metabolic phenotype compared with their progenitors. It has been proposed that HSCs generate energy mainly through anaerobic glycolysis in a pyruvate dehydrogenase kinase (Pdk)-dependent manner. Cited2 is an essential regulator for HSC quiescence, apoptosis, and function. Herein, we show that conditional deletion of Cited2 in murine HSCs results in elevated levels of reactive oxygen species, decreased cellular glutathione content, increased mitochondrial activity, and decreased glycolysis. At the molecular level, Cited2 deficiency significantly reduced the expression of genes involved in metabolism, such as Pdk2, Pdk4, and lactate dehydrogenases B and D (LDHB and LDHD). Cited2-deficient HSCs also exhibited increased Akt signaling, concomitant with elevated mTORC1 activity and phosphorylation of FoxOs. Further, inhibition of PI3/Akt, but not mTORC1, partially rescued the repression of Pdk4 caused by deletion of Cited2. Altogether, our results suggest that Cited2 is required for the maintenance of adult HSC glycolytic metabolism likely through regulating Pdk2, Pdk4, LDHB, LDHD, and Akt activity.
Collapse
Affiliation(s)
- Jinwei Du
- 1 Department of Biochemistry and Comprehensive Cancer Center, Case Western Reserve University , Cleveland, Ohio
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Wang F, Marshall CB, Ikura M. Transcriptional/epigenetic regulator CBP/p300 in tumorigenesis: structural and functional versatility in target recognition. Cell Mol Life Sci 2013; 70:3989-4008. [PMID: 23307074 PMCID: PMC11113169 DOI: 10.1007/s00018-012-1254-4] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 11/08/2012] [Accepted: 12/20/2012] [Indexed: 01/19/2023]
Abstract
In eukaryotic cells, gene transcription is regulated by sequence-specific DNA-binding transcription factors that recognize promoter and enhancer elements near the transcriptional start site. Some coactivators promote transcription by connecting transcription factors to the basal transcriptional machinery. The highly conserved coactivators CREB-binding protein (CBP) and its paralog, E1A-binding protein (p300), each have four separate transactivation domains (TADs) that interact with the TADs of a number of DNA-binding transcription activators as well as general transcription factors (GTFs), thus mediating recruitment of basal transcription machinery to the promoter. Most promoters comprise multiple activator-binding sites, and many activators contain tandem TADs, thus multivalent interactions may stabilize CBP/p300 at the promoter, and intrinsically disordered regions in CBP/p300 and many activators may confer adaptability to these multivalent complexes. CBP/p300 contains a catalytic histone acetyltransferase (HAT) domain, which remodels chromatin to 'relax' its superstructure and enables transcription of proximal genes. The HAT activity of CBP/p300 also acetylates some transcription factors (e.g., p53), hence modulating the function of key transcriptional regulators. Through these numerous interactions, CBP/p300 has been implicated in complex physiological and pathological processes, and, in response to different signals, can drive cells towards proliferation or apoptosis. Dysregulation of the transcriptional and epigenetic functions of CBP/p300 is associated with leukemia and other types of cancer, thus it has been recognized as a potential anti-cancer drug target. In this review, we focus on recent exciting findings in the structural mechanisms of CBP/p300 involving multivalent and dynamic interactions with binding partners, which may pave new avenues for anti-cancer drug development.
Collapse
Affiliation(s)
- Feng Wang
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9 Canada
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7 Canada
- Present Address: Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Christopher B. Marshall
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9 Canada
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7 Canada
| | - Mitsuhiko Ikura
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9 Canada
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7 Canada
| |
Collapse
|
133
|
Abstract
PURPOSE OF REVIEW Transcription co-regulator Cited2 is essential for mouse development. Recent work has shown that Cited2 plays important roles in normal hematopoiesis in fetal liver and adult bone marrow. This review focuses on the function of Cited2 in the maintenance of hematopoietic stem cells (HSCs) and its potential role in the metabolic regulation of HSCs. RECENT FINDINGS Fetal liver cells from Cited2 null embryos give rise to reduced numbers of hematopoietic colonies and display significantly impaired hematopoietic reconstitution capacity. In adult mice, conditional deletion of Cited2 markedly reduces the number of HSCs and compromises hematopoietic reconstitution in mice receiving a transplant of Cited2 deficient bone marrow cells. Additional deletion of Ink4a/Arf or p53 in a Cited2-deficient background restores HSC functionality. Meanwhile, Cited2 deficient HSCs display loss of quiescence, which can be partially rescued by additional deletion of hypoxia inducible factor-1α. SUMMARY Cited2 is an essential regulator in fetal liver and adult hematopoiesis. Further studies into the function of Cited2 and the underlying mechanism in the metabolic regulation of HSCs will provide a better understanding of the connection between energy metabolism and HSC quiescence and self-renewal. Investigations of the pathologic role of Cited2 in leukemogenesis may yield useful information in developing effective therapeutic strategies.
Collapse
|
134
|
Charitou P, Burgering BMT. Forkhead box(O) in control of reactive oxygen species and genomic stability to ensure healthy lifespan. Antioxid Redox Signal 2013; 19:1400-19. [PMID: 22978495 DOI: 10.1089/ars.2012.4921] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Transcription factors of the Forkhead box O class (FOXOs) are associated with lifespan and play a role in age-related diseases. FOXOs, therefore, serve as a paradigm for developing an understanding as to how age-related diseases, such as cancer and diabetes interconnect with lifespan. Understanding the regulatory inputs on FOXO may reveal how changes in these regulatory signaling pathways affect disease and lifespan. RECENT ADVANCES Numerous regulators of FOXO have now been described and a clear and evolutionary conserved role has emerged for phosphoinositide-3 kinase/protein kinase B (also known as c-Akt or AKT) signaling and c-jun N-terminal kinase signaling. Analysis of FOXO function in the context of these signaling pathways has shown the importance of FOXO-mediated transcriptional regulation on cell cycle progression and other cell fates, such as cell metabolism, stress resistance, and apoptosis in mediating disease and lifespan. CRITICAL ISSUES Persistent DNA damage is also tightly linked to disease and aging; yet, data on a possible link between DNA damage and FOXO have been limited. Here, we discuss possible connections between FOXO and the DNA damage response in the context of the broader role of connecting lifespan and disease. FUTURE DIRECTIONS Understanding the role of lifespan in diseases onset may provide unique and generic possibilities to intervene in disease processes to ensure a healthy lifespan.
Collapse
Affiliation(s)
- Paraskevi Charitou
- Molecular Cancer Research, University Medical Center Utrecht , Utrecht, The Netherlands
| | | |
Collapse
|
135
|
Yeh YH, Yang YC, Hsieh MY, Yeh YC, Li TK. A negative feedback of the HIF-1α pathway via interferon-stimulated gene 15 and ISGylation. Clin Cancer Res 2013; 19:5927-39. [PMID: 24056783 DOI: 10.1158/1078-0432.ccr-13-0018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The IFN-stimulated gene 15 (ISG15)- and ubiquitin-conjugation pathways play roles in mediating hypoxic and inflammatory responses. To identify interaction(s) between these two tumor microenvironments, we investigated the effect of ISG15 on the activity of the master hypoxic transcription factor HIF-1α. EXPERIMENTAL DESIGN IFN and desferoxamine treatments were used to induce the expression of ISGs and HIF-1α, respectively. Interactions between HIF-1α and the ISG15 and ISGylation system were studied using knockdown of mRNA expression, immunoblotting, coimmunoprecipitation, and pull-down analyses. Effects of the ISG15 and ISGylation system on the HIF-1α-directed processes were examined using reporter, reverse transcription polymerase chain reaction (RT-PCR), and tumorigenic growth assays. RESULTS We found that the level of the free form of HIF-1α is differentially regulated by IFN treatment, and that the free ISG15 level is lower under hypoxia. Mechanism-directed studies have shown that HIF-1α not only interacts physically with ISG15, but is also ISGylated in multiple domains. ISG15 expression disrupts the functional dimerization of HIF-1α and -1β. Subsequently, expression of the ISG15 and/or ISGylation system attenuates HIF-1α-mediated gene expression and tumorigenic growth. CONCLUSION In summary, our results revealed cross-talk between inflammatory and hypoxic pathways through the ISGylation of HIF-1α. On the basis of these results, we propose a novel negative feedback loop for the HIF-1α-mediated pathway involving the regulation of HIF-1α via IFN-induced ISGylation.
Collapse
Affiliation(s)
- Yen-Hsiu Yeh
- Authors' Affiliations: Department and Graduate Institute of Microbiology, College of Medicine; Center for Biotechnology, National Taiwan University, Taipei; and Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
136
|
Wang Y, Tian C, Zheng JC. FoxO3a contributes to the reprogramming process and the differentiation of induced pluripotent stem cells. Stem Cells Dev 2013; 22:2954-63. [PMID: 23815557 DOI: 10.1089/scd.2013.0044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Induced pluripotent stem (iPS) cells, which are morphologically and functionally similar with embryonic stem (ES) cells, have been successfully generated from somatic cells through defined reprogramming transcription factors. Forkhead class O3a (FoxO3a) has been recently reported to play an important role in the homeostasis and maintenance of certain types of stem cells; however, the role of FoxO3a in the reprogramming process and differentiation of iPS cells remains unclear. In this study, we investigate the function of FoxO3a during the reprogramming process and characterize the properties of iPS cells from FoxO3a-wild type and -null mouse embryonic fibroblasts (MEFs). Our results show that the FoxO3a-null iPS cells are similar to the wild-type iPS cells in the levels of ES cell markers, alkaline phosphatase activity, and formation of teratoma in vivo. The reprogramming process is delayed in the FoxO3a-null MEFs compared to the wild-type MEFs; whereas the overexpression of FoxO3a partially recovers the impaired reprogramming efficiency in the null group. More importantly, FoxO3a deficiency impairs the neuronal lineage differentiation potential of iPS cells in vitro. These results suggest that FoxO3a affects the reprogramming kinetics and the neuronal lineage differentiation potential of the resulting iPS cells. Therefore, this study demonstrates a novel function of FoxO3a in cell reprogramming, which will help the development of alternative strategies for generating iPS cells.
Collapse
Affiliation(s)
- Yongxiang Wang
- 1 Laboratory of Neuroimmunology and Regenerative Therapy, University of Nebraska Medical Center , Omaha, Nebraska
| | | | | |
Collapse
|
137
|
Krivoruchko A, Storey KB. Anoxia-responsive regulation of the FoxO transcription factors in freshwater turtles, Trachemys scripta elegans. Biochim Biophys Acta Gen Subj 2013; 1830:4990-8. [PMID: 23850471 DOI: 10.1016/j.bbagen.2013.06.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 06/10/2013] [Accepted: 06/29/2013] [Indexed: 11/25/2022]
Abstract
BACKGROUND The forkhead class O (FoxO) transcription factors are important regulators of multiple aspects of cellular metabolism. We hypothesized that activation of these transcription factors could play crucial roles in low oxygen survival in the anoxia-tolerant turtle, Trachemys scripta elegans. METHODS Two FoxOs, FoxO1 and FoxO3, were examined in turtle tissues in response to 5 and 20h of anoxic submergence using techniques of RT-PCR, western immunoblotting and DNA-binding assays to assess activation. Transcript levels of FoxO-responsive genes were also quantified using RT-PCR. RESULTS FoxO1 was anoxia-responsive in the liver, with increases in transcript levels, protein levels, nuclear levels and DNA-binding of 1.7-4.8fold in response to anoxia. Levels of phosphorylated FoxO1 also decreased to 57% of control values in response to 5h of anoxia, indicating activation. FoxO3 was activated in the heart, kidney and liver in response to anoxia, with nuclear levels increasing by 1.5-3.7fold and DNA-binding activity increasing by 1.3-2.9fold. Transcript levels of two FoxO-target genes, p27kip1 and catalase, also rose by 2.4-2.5fold in the turtle liver under anoxia. CONCLUSIONS The results suggest that the FoxO transcription factors are activated in response to anoxia in T. scripta elegans, potentially contributing to the regulation of stress resistance and metabolic depression. GENERAL SIGNIFICANCE This study provides the first demonstration of activation of FoxOs in a natural model for vertebrate anoxia tolerance, further improving understanding of how tissues can survive without oxygen.
Collapse
Affiliation(s)
- Anastasia Krivoruchko
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | | |
Collapse
|
138
|
van Boxtel R, Gomez-Puerto C, Mokry M, Eijkelenboom A, van der Vos KE, Nieuwenhuis EES, Burgering BMT, Lam EWF, Coffer PJ. FOXP1 acts through a negative feedback loop to suppress FOXO-induced apoptosis. Cell Death Differ 2013; 20:1219-29. [PMID: 23832113 DOI: 10.1038/cdd.2013.81] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 12/27/2022] Open
Abstract
Transcriptional activity of Forkhead box transcription factor class O (FOXO) proteins can result in a variety of cellular outcomes depending on cell type and activating stimulus. These transcription factors are negatively regulated by the phosphoinositol 3-kinase (PI3K)-protein kinase B (PKB) signaling pathway, which is thought to have a pivotal role in regulating survival of tumor cells in a variety of cancers. Recently, it has become clear that FOXO proteins can promote resistance to anti-cancer therapeutics, designed to inhibit PI3K-PKB activity, by inducing the expression of proteins that provide feedback at different levels of this pathway. We questioned whether such a feedback mechanism may also exist directly at the level of FOXO-induced transcription. To identify critical modulators of FOXO transcriptional output, we performed gene expression analyses after conditional activation of key components of the PI3K-PKB-FOXO signaling pathway and identified FOXP1 as a direct FOXO transcriptional target. Using chromatin immunoprecipitation followed by next-generation sequencing, we show that FOXP1 binds enhancers that are pre-occupied by FOXO3. By sequencing the transcriptomes of cells in which FOXO is specifically activated in the absence of FOXP1, we demonstrate that FOXP1 can modulate the expression of a specific subset of FOXO target genes, including inhibiting expression of the pro-apoptotic gene BIK. FOXO activation in FOXP1-knockdown cells resulted in increased cell death, demonstrating that FOXP1 prevents FOXO-induced apoptosis. We therefore propose that FOXP1 represents an important modulator of FOXO-induced transcription, promoting cellular survival.
Collapse
Affiliation(s)
- R van Boxtel
- Department of Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that are responsible for fine-tuning the spatial and temporal expression of a broad range of genes both during development and in adult tissues. This function is engrained in their ability to integrate a multitude of cellular and environmental signals and to act with remarkable fidelity. Several key members of the FOXA, FOXC, FOXM, FOXO and FOXP subfamilies are strongly implicated in cancer, driving initiation, maintenance, progression and drug resistance. The functional complexities of FOX proteins are coming to light and have established these transcription factors as possible therapeutic targets and putative biomarkers for specific cancers.
Collapse
Affiliation(s)
- Eric W-F Lam
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | | | | | | |
Collapse
|
140
|
Hagenbuchner J, Ausserlechner MJ. Mitochondria and FOXO3: breath or die. Front Physiol 2013; 4:147. [PMID: 23801966 PMCID: PMC3687139 DOI: 10.3389/fphys.2013.00147] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/31/2013] [Indexed: 01/02/2023] Open
Abstract
Forkhead box O (FOXO) transcription factors are regulators of cell-type specific apoptosis and cell cycle arrest but also control longevity and reactive oxygen species (ROS). ROS-control by FOXO is mediated by transcriptional activation of detoxifying enzymes such as Superoxide dismutase 2 (SOD2), Catalase or Sestrins or by the repression of mitochondrial respiratory chain proteins resulting in reduced mitochondrial activity. FOXO3 also regulates the adaptation to hypoxia by reducing mitochondrial mass and oxygen consumption during HIF-1α activation. In neuronal tumor cells, FOXO3 triggers ROS-accumulation as a consequence of transient mitochondrial outer membrane permeabilization, which is essential for FOXO3-induced apoptosis in these cells. Cellular ROS levels are affected by the FOXO-targets Bim, BclxL, and Survivin. All three proteins localize to mitochondria and affect mitochondrial membrane potential, respiration and cellular ROS levels. Bim-activation by FOXO3 causes mitochondrial depolarization resulting in a transitory decrease of respiration and ROS production. Survivin, on the other hand, actively changes mitochondrial architecture, respiration-efficacy and energy metabolism. This ability distinguishes Survivin from other anti-apoptotic proteins such as BclxL, which inhibits ROS by inactivating Bim but does not alter mitochondrial function. Importantly, FOXO3 simultaneously also activates ROS-detoxification via induction of SESN3. In this paper we discuss the hypothesis that the delicate balance between ROS-accumulation by Bim-triggered mitochondrial damage, mitochondrial architecture and ROS-detoxifying proteins determines cell fate. We provide evidence for a FOXO self-reactivating loop and for novel functions of FOXO3 in controlling mitochondrial respiration of neuronal cells, which further supports the current view that FOXO transcription factors are information-integrating sentinels of cellular stress and critical modulators of cell homeostasis.
Collapse
Affiliation(s)
- Judith Hagenbuchner
- Department of Pediatrics II, Medical University Innsbruck Innsbruck, Austria ; Tyrolean Cancer Research Institute Innsbruck, Austria
| | | |
Collapse
|
141
|
Bol GM, Raman V, van der Groep P, Vermeulen JF, Patel AH, van der Wall E, van Diest PJ. Expression of the RNA helicase DDX3 and the hypoxia response in breast cancer. PLoS One 2013; 8:e63548. [PMID: 23696831 PMCID: PMC3656050 DOI: 10.1371/journal.pone.0063548] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 04/03/2013] [Indexed: 12/11/2022] Open
Abstract
Aims DDX3 is an RNA helicase that has antiapoptotic properties, and promotes proliferation and transformation. In addition, DDX3 was shown to be a direct downstream target of HIF-1α (the master regulatory of the hypoxia response) in breast cancer cell lines. However, the relation between DDX3 and hypoxia has not been addressed in human tumors. In this paper, we studied the relation between DDX3 and the hypoxic responsive proteins in human breast cancer. Methods and Results DDX3 expression was investigated by immunohistochemistry in breast cancer in comparison with hypoxia related proteins HIF-1α, GLUT1, CAIX, EGFR, HER2, Akt1, FOXO4, p53, ERα, COMMD1, FER kinase, PIN1, E-cadherin, p21, p27, Transferrin receptor, FOXO3A, c-Met and Notch1. DDX3 was overexpressed in 127 of 366 breast cancer patients, and was correlated with overexpression of HIF-1α and its downstream genes CAIX and GLUT1. Moreover, DDX3 expression correlated with hypoxia-related proteins EGFR, HER2, FOXO4, ERα and c-Met in a HIF-1α dependent fashion, and with COMMD1, FER kinase, Akt1, E-cadherin, TfR and FOXO3A independent of HIF-1α. Conclusions In invasive breast cancer, expression of DDX3 was correlated with overexpression of HIF-1α and many other hypoxia related proteins, pointing to a distinct role for DDX3 under hypoxic conditions and supporting the oncogenic role of DDX3 which could have clinical implication for current development of DDX3 inhibitors.
Collapse
Affiliation(s)
- Guus M. Bol
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Venu Raman
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Petra van der Groep
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Division of Internal Medicine and Dermatology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Jeroen F. Vermeulen
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Arvind H. Patel
- MRC, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Elsken van der Wall
- Division of Internal Medicine and Dermatology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
| | - Paul J. van Diest
- Departments of Pathology, University Medical Center Utrecht Cancer Center, Utrecht, The Netherlands
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
142
|
Peck B, Ferber EC, Schulze A. Antagonism between FOXO and MYC Regulates Cellular Powerhouse. Front Oncol 2013; 3:96. [PMID: 23630664 PMCID: PMC3635031 DOI: 10.3389/fonc.2013.00096] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 04/08/2013] [Indexed: 11/13/2022] Open
Abstract
Alterations in cellular metabolism are a key feature of the transformed phenotype. Enhanced macromolecule synthesis is a prerequisite for rapid proliferation but may also contribute to induction of angiogenesis, metastasis formation, and tumor progression, thereby leading to a poorer clinical outcome. Metabolic adaptations enable cancer cells to survive in suboptimal growth conditions, such as the limited supply of nutrient and oxygen often found in the tumor microenvironment. Metabolic changes, including activation of glycolysis and inhibition of mitochondrial ATP production, are induced under hypoxia to promote survival in low oxygen. FOXO3a, a transcription factor that is inhibited by the phosphatidylinositol 3-kinase/Akt pathway and is upregulated in hypoxia, has emerged as an important negative regulator of MYC function. Recent studies have revealed that FOXO3a acts as a negative regulator of mitochondrial function through inhibition of MYC. Ablation of FOXO3a prevents the inhibition of mitochondrial function induced by hypoxia and results in enhanced oxidative stress. This review will focus on the antagonism between FOXO3a and MYC and discuss their role in cellular bioenergetics, reactive oxygen metabolism, and adaptation to hypoxia, raising questions about the role of FOXO proteins in cancer.
Collapse
Affiliation(s)
- Barrie Peck
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute London, UK
| | | | | |
Collapse
|
143
|
Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell 2013; 48:158-67. [PMID: 23102266 DOI: 10.1016/j.molcel.2012.09.025] [Citation(s) in RCA: 1928] [Impact Index Per Article: 160.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/28/2012] [Accepted: 09/21/2012] [Indexed: 12/11/2022]
Abstract
Historically, mitochondrial reactive oxygen species (mROS) were thought to exclusively cause cellular damage and lack a physiological function. Accumulation of ROS and oxidative damage have been linked to multiple pathologies, including neurodegenerative diseases, diabetes, cancer, and premature aging. Thus, mROS were originally envisioned as a necessary evil of oxidative metabolism, a product of an imperfect system. Yet few biological systems possess such flagrant imperfections, thanks to the persistent optimization of evolution, and it appears that oxidative metabolism is no different. More and more evidence suggests that mROS are critical for healthy cell function. In this Review, we discuss this evidence following some background on the generation and regulation of mROS.
Collapse
Affiliation(s)
- Laura A Sena
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
144
|
Morris BJ. Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med 2013; 56:133-71. [PMID: 23104101 DOI: 10.1016/j.freeradbiomed.2012.10.525] [Citation(s) in RCA: 290] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/31/2012] [Accepted: 10/05/2012] [Indexed: 12/14/2022]
Abstract
Sirtuins are a class of NAD(+)-dependent deacetylases having beneficial health effects. This extensive review describes the numerous intracellular actions of the seven mammalian sirtuins, their protein targets, intracellular localization, the pathways they modulate, and their role in common diseases of aging. Selective pharmacological targeting of sirtuins is of current interest in helping to alleviate global disease burden. Since all sirtuins are activated by NAD(+), strategies that boost NAD(+) in cells are of interest. While most is known about SIRT1, the functions of the six other sirtuins are now emerging. Best known is the involvement of sirtuins in helping cells adapt energy output to match energy requirements. SIRT1 and some of the other sirtuins enhance fat metabolism and modulate mitochondrial respiration to optimize energy harvesting. The AMP kinase/SIRT1-PGC-1α-PPAR axis and mitochondrial sirtuins appear pivotal to maintaining mitochondrial function. Downregulation with aging explains much of the pathophysiology that accumulates with aging. Posttranslational modifications of sirtuins and their substrates affect specificity. Although SIRT1 activation seems not to affect life span, activation of some of the other sirtuins might. Since sirtuins are crucial to pathways that counter the decline in health that accompanies aging, pharmacological agents that boost sirtuin activity have clinical potential in treatment of diabetes, cardiovascular disease, dementia, osteoporosis, arthritis, and other conditions. In cancer, however, SIRT1 inhibitors could have therapeutic value. Nutraceuticals such as resveratrol have a multiplicity of actions besides sirtuin activation. Their net health benefit and relative safety may have originated from the ability of animals to survive environmental changes by utilizing these stress resistance chemicals in the diet during evolution. Each sirtuin forms a key hub to the intracellular pathways affected.
Collapse
Affiliation(s)
- Brian J Morris
- Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, Building F13, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
145
|
Keränen MAI, Tuuminen R, Syrjälä S, Krebs R, Walkinshaw G, Flippin LA, Arend M, Koskinen PK, Nykänen AI, Lemström KB. Differential effects of pharmacological HIF preconditioning of donors versus recipients in rat cardiac allografts. Am J Transplant 2013; 13:600-10. [PMID: 23331861 DOI: 10.1111/ajt.12064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 11/08/2012] [Accepted: 11/08/2012] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion injury (IRI) induces hypoxia-inducible factor-1 (HIF-1) in the myocardium, but the consequences remain elusive. We investigated HIF-1 activation during cold and warm ischemia and IRI in rat hearts and cardiac syngrafts. We also tested the effect of HIF-α stabilizing prolyl hydroxylase inhibitor (FG-4497) on IRI or allograft survival. Ex vivo ischemia of the heart increased HIF-1α expression in a time- and temperature-dependent fashion. Immunohistochemistry localized HIF-1α to all cardiac cell types. After reperfusion, HIF-1α immunoreactivity persisted in smooth muscle cells and cardiomyocytes in the areas with IRI. This was accompanied with a transient induction of protective HIF-1 downstream genes. Donor FG-4497 pretreatment for 4 h enhanced IRI in cardiac allografts as evidenced by an increase in cardiac troponin T release, cardiomyocyte apoptosis, and activation of innate immunity. Recipient FG-4497 pretreatment for 4 h decreased infiltration of ED1(+) macrophages, and mildly improved the long-term allograft survival. In syngrafts donor FG-4497 pretreatment increased activation of innate immunity, but did not induce myocardial damage. We conclude that the HIF-1 pathway is activated in heart transplants. We suggest that pharmacological HIF-α preconditioning of cardiac allografts donors would not lead to clinical benefit, while in recipients it may result in antiinflammatory effects and prolonged allograft survival.
Collapse
Affiliation(s)
- M A I Keränen
- Transplantation Laboratory, Haartman Institute, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Harris IS, Blaser H, Moreno J, Treloar AE, Gorrini C, Sasaki M, Mason JM, Knobbe CB, Rufini A, Hallé M, Elia AJ, Wakeham A, Tremblay ML, Melino G, Done S, Mak TW. PTPN12 promotes resistance to oxidative stress and supports tumorigenesis by regulating FOXO signaling. Oncogene 2013; 33:1047-54. [DOI: 10.1038/onc.2013.24] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/21/2012] [Accepted: 12/23/2012] [Indexed: 02/01/2023]
|
147
|
Liang C, Chen W, Zhi X, Ma T, Xia X, Liu H, Zhang Q, Hu Q, Zhang Y, Bai X, Liang T. Serotonin promotes the proliferation of serum-deprived hepatocellular carcinoma cells via upregulation of FOXO3a. Mol Cancer 2013; 12:14. [PMID: 23418729 PMCID: PMC3601970 DOI: 10.1186/1476-4598-12-14] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 02/14/2013] [Indexed: 12/13/2022] Open
Abstract
Background Peripheral serotonin is involved in tumorigenesis and induces a pro-proliferative effect in hepatocellular carcinoma (HCC) cells; however, the intracellular mechanisms by which serotonin exerts a mitogenic effect remain unclear. In this research, we examined whether FOXO3a, a transcription factor at the interface of crucial cellular processes, plays a role downstream of serotonin in HCC cells. Results The cell viability and expression of FOXO3a was assessed in three HCC cell lines (Huh7, HepG2 and Hep3B) during serum deprivation in the presence or absence of serotonin. Serum free media significantly inhibited HCC proliferation and led to reduced expression and nuclear accumulation of FOXO3a. Knockdown of FOXO3a enhanced the ability of serum deprivation to inhibit HCC cells proliferation. And overexpression of non-phosphorylated FOXO3a in HCC cells reversed serum-deprivation-induced growth inhibition. Serotonin reversed the serum-deprivation-induced inhibition of cell proliferation and upregulated FOXO3a in Huh7 cells; however, serotonin had no effect on the proliferation of serum-deprived HepG2 or Hep3B cells. In addition to proliferation, serotonin also induced phosphorylation of AKT and FOXO3a in serum-deprived Huh7 cells but not in HepG2 and Hep3B cells. However, the phosphorylation of FOXO3a induced by serotonin did not export FOXO3a from nucleus to cytoplasm in serum-deprived Huh7 cells. Consequently, we demonstrated that serotonin promoted the proliferation of Huh7 cells by increasing the expression of FOXO3a. We also provide preliminary evidence that different expression levels of the 5-HT2B receptor (5-HT2BR) may contribute to the distinct effects of serotonin in different serum-deprived HCC cells. Conclusions This study demonstrates that FOXO3a functions as a growth factor in serum-deprived HCC cells and serotonin promotes the proliferation of serum-deprived HCC cells via upregulation of FOXO3a, in the presence of sufficient levels of the serotonin receptor 5-HT2BR. Drugs targeting the serotonin-5-HT2BR-FOXO3a pathway may provide a novel target for anticancer therapy.
Collapse
Affiliation(s)
- Chao Liang
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Leiser SF, Fletcher M, Begun A, Kaeberlein M. Life-span extension from hypoxia in Caenorhabditis elegans requires both HIF-1 and DAF-16 and is antagonized by SKN-1. J Gerontol A Biol Sci Med Sci 2013; 68:1135-44. [PMID: 23419779 DOI: 10.1093/gerona/glt016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stabilization of the hypoxia-inducible factor (HIF-1) protein extends longevity in Caenorhabditis elegans. However, stabilization of mammalian HIF-1α has been implicated in tumor growth and cancer development. Consequently, for the hypoxic response to benefit mammalian health, we must determine the components of the response that contribute to longevity, and separate them from those that cause harm in mammals. Here, we subject adult worms to low oxygen environments. We find that growth in hypoxia increases longevity in wild-type worms but not in animals lacking HIF-1 or DAF-16. Conversely, hypoxia shortens life span in combination with overexpression of the antioxidant stress response protein SKN-1. When combined with mutations in other longevity pathways or dietary restriction, hypoxia extends life span but to varying extents. Collectively, our results show that hypoxia modulates longevity in a complex manner, likely involving components in addition to HIF-1.
Collapse
Affiliation(s)
- Scott F Leiser
- Department of Pathology, University of Washington, Seattle, WA 98195.
| | | | | | | |
Collapse
|
149
|
Calnan DR, Webb AE, White JL, Stowe TR, Goswami T, Shi X, Espejo A, Bedford MT, Gozani O, Gygi SP, Brunet A. Methylation by Set9 modulates FoxO3 stability and transcriptional activity. Aging (Albany NY) 2012; 4:462-79. [PMID: 22820736 PMCID: PMC3433933 DOI: 10.18632/aging.100471] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The FoxO family of transcription factors plays an important role in longevity and tumor suppression by regulating the expression of a wide range of target genes. FoxO3 has recently been found to be associated with extreme longevity in humans and to regulate the homeostasis of adult stem cell pools in mammals, which may contribute to longevity. The activity of FoxO3 is controlled by a variety of post-translational modifications that have been proposed to form a ‘code’ affecting FoxO3 subcellular localization, DNA binding ability, protein-protein interactions and protein stability. Lysine methylation is a crucial post-translational modification on histones that regulates chromatin accessibility and is a key part of the ‘histone code’. However, whether lysine methylation plays a role in modulating FoxO3 activity has never been examined. Here we show that the methyltransferase Set9 directly methylates FoxO3 in vitro and in cells. Using a combination of tandem mass spectrometry and methyl-specific antibodies, we find that Set9 methylates FoxO3 at a single residue, lysine 271, a site previously known to be deacetylated by Sirt1. Methylation of FoxO3 by Set9 decreases FoxO3 protein stability, while moderately increasing FoxO3 transcriptional activity. The modulation of FoxO3 stability and activity by methylation may be critical for fine-tuning cellular responses to stress stimuli, which may in turn affect FoxO3's ability to promote tumor suppression and longevity.
Collapse
|
150
|
Abstract
Erythroid ontogeny is characterized by overlapping waves of primitive and definitive erythroid lineages that share many morphologic features during terminal maturation but have marked differences in cell size and globin expression. In the present study, we compared global gene expression in primitive, fetal definitive, and adult definitive erythroid cells at morphologically equivalent stages of maturation purified from embryonic, fetal, and adult mice. Surprisingly, most transcriptional complexity in erythroid precursors is already present by the proerythroblast stage. Transcript levels are markedly modulated during terminal erythroid maturation, but housekeeping genes are not preferentially lost. Although primitive and definitive erythroid lineages share a large set of nonhousekeeping genes, annotation of lineage-restricted genes shows that alternate gene usage occurs within shared functional categories, as exemplified by the selective expression of aquaporins 3 and 8 in primitive erythroblasts and aquaporins 1 and 9 in adult definitive erythroblasts. Consistent with the known functions of Aqp3 and Aqp8 as H2O2 transporters, primitive, but not definitive, erythroblasts preferentially accumulate reactive oxygen species after exogenous H2O2 exposure. We have created a user-friendly Web site (http://www.cbil.upenn.edu/ErythronDB) to make these global expression data readily accessible and amenable to complex search strategies by the scientific community.
Collapse
|