1
|
Hu C, Zhang B, Zhao S. METTL3-mediated N6-methyladenosine modification stimulates mitochondrial damage and ferroptosis of kidney tubular epithelial cells following acute kidney injury by modulating the stabilization of MDM2-p53-LMNB1 axis. Eur J Med Chem 2023; 259:115677. [PMID: 37542992 DOI: 10.1016/j.ejmech.2023.115677] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 08/07/2023]
Abstract
N6-methyladenosine (m6A) and MELLT3 assume a role in the development of acute kidney injury (AKI). However, their mechanism in AKI remains under-explored. On this basis, this study explored the mechanism of MELLT3 in mitochondrial damage and ferroptosis of kidney tubular epithelial cells after AKI. HK-2 cells were induced by lipopolysaccharide (LPS) to simulate AKI, followed by gain and loss of function of genes, detection of mitochondrial damage and ferroptosis indicators, and analysis of gene interactions. An AKI mouse model was developed using the cecal ligation and puncture (CLP) method to investigate the effect of METTL3 knockdown on kidney injury. MDM2 and LMNB1 were upregulated and p53 was downregulated in LPS-treated HK-2 cells. Mechanistically, the E3 ubiquitin ligase MDM2 increased p53 ubiquitination to activate LMNB1. METTL3 knockdown decreased m6A methylation of MDM2, thus diminishing YTHDF1-mediated MDM2 mRNA stability and translation in LPS-treated HK-2 cells. Knockdown of LMNB1, MDM2, or METTL3 reduced NO, MDA, iron ion, and ROS levels as well as mitochondrial damage and raised SOD, GSH, XCT, GPX4, FPN1, and TFR1 levels in LPS-treated HK-2 cells. The in vivo results showed that METTL3 knockdown reduced renal injury and ferroptosis in CLP mice. METTL3 knockdown prevents mitochondrial damage and ferroptosis of kidney tubular epithelial cells after AKI via the MDM2-p53-LMNB1 axis.
Collapse
Affiliation(s)
- Chenghuan Hu
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China; Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
| | - Buyao Zhang
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Shuangping Zhao
- Department of Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China; Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Li X, Sun T, Liu J, Wei S, Yang Y, Liu J, Zhang B, Li W. Phloretin alleviates doxorubicin-induced cardiotoxicity through regulating Hif3a transcription via targeting transcription factor Fos. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155046. [PMID: 37659297 DOI: 10.1016/j.phymed.2023.155046] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Doxorubicin (Dox), a chemotherapeutic agent known for its efficacy, has been associated with the development of severe cardiotoxicity, commonly referred to as doxorubicin-induced cardiotoxicity (DIC). The role and mechanism of action of phloretin (Phl) in cardiovascular diseases are well-established; however, its specific function and underlying mechanism in the context of DIC have yet to be fully elucidated. OBJECTIVE This research aimed to uncover the protective effect of Phl against DIC in vivo and in vitro, while also providing a comprehensive understanding of the underlying mechanisms involved. METHODS DIC cell and murine models were established. The action targets and mechanism of Phl against DIC were comprehensively examined by systematic network pharmacology, molecular docking, transcriptomics technologies, transcription factor (TF) prediction, and experimental validation. RESULTS Phl relieved Dox-induced cell apoptosis in vitro and in vivo. Through network pharmacology analysis, a total of 554 co-targeted genes of Phl and Dox were identified. Enrichment analysis revealed several key pathways including the PI3K-Akt signaling pathway, Apoptosis, and the IL-17 signaling pathway. Protein-protein interaction (PPI) analysis identified 24 core co-targeted genes, such as Fos, Jun, Hif1a, which were predicted to bind well to Phl based on molecular docking. Transcriptomics analysis was performed to identify the top 20 differentially expressed genes (DEGs), and 202 transcription factors (TFs) were predicted for these DEGs. Among these TFs, 10 TFs (Fos, Jun, Hif1a, etc.) are also the co-targeted genes, and 3 TFs (Fos, Jun, Hif1a) are also the core co-targeted genes. Further experiments validated the finding that Phl reduced the elevated levels of Hif3a (one of the top 20 DEGs) and Fos (one of Hif3a's predicted TFs) induced by Dox. Moreover, the interaction between Fos protein and the Hif3a promoter was confirmed through luciferase reporter assays. CONCLUSION Phl actively targeted and down-regulated the Fos protein to inhibit its binding to the promoter region of Hif3a, thereby providing protection against DIC.
Collapse
Affiliation(s)
- Xiangyun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China; School of Pharmacy, Central South University, Changsha, Hunan 410078, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiaqin Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Yuanying Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China; School of Pharmacy, Central South University, Changsha, Hunan 410078, China.
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Institute of Clinical Pharmacy, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
3
|
Li C, Lei C, Wang K, Tan M, Xu F, Wang J, Zheng Y. MADS2 regulates priming defence in postharvest peach through combined salicylic acid and abscisic acid signaling. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:3787-3806. [PMID: 35266534 DOI: 10.1093/jxb/erac099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
MADS-box genes play well-documented roles in plant development, but relatively little is known regarding their involvement in defence responses. In this study, pre-treatment of peach (Prunus persica) fruit with β-aminobutyric acid (BABA) activated resistance against Rhizopus stolonifer, leading to a significant delay in the symptomatic appearance of disease. This was associated with an integrated defence response that included a H2O2 burst, ABA accumulation, and callose deposition. cDNA library screening identified nucleus-localized MADS2 as an interacting partner with NPR1, and this was further confirmed by yeast two-hybrid, luciferase complementation imaging, and co-immunoprecipitation assays. The DNA-binding activity of NPR1 conferred by the NPR1-MADS2 complex was required for the transcription of SA-dependent pathogenesis-related (PR) and ABA-inducible CalS genes in order to gain the BABA-induced resistance, in which MAPK1-induced post-translational modification of MADS2 was also involved. In accordance with this, overexpression of PpMADS2 in Arabidopsis potentiated the transcription of a group of PR genes and conferred fungal resistance in the transgenic plants. Conversely, Arabidopsis mads2-knockout lines showed high sensitivity to the fungal pathogen. Our results indicate that MADS2 positively participates in BABA-elicited defence in peach through a combination of SA-dependent NPR1 activation and ABA signaling-induced callose accumulation, and that this defence is also related to the post-translational modification of MADS2 by MAPK1 for signal amplification.
Collapse
Affiliation(s)
- Chunhong Li
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095 Jiangsu, P.R. China
- College of Biology and Food Science, Chongqing Three Gorges University, Chongqing 404000, P.R. China
| | - Changyi Lei
- College of Biology and Food Science, Chongqing Three Gorges University, Chongqing 404000, P.R. China
| | - Kaituo Wang
- College of Biology and Food Science, Chongqing Three Gorges University, Chongqing 404000, P.R. China
| | - Meilin Tan
- College of Biology and Food Science, Chongqing Three Gorges University, Chongqing 404000, P.R. China
| | - Feng Xu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, P.R. China
| | - Jinsong Wang
- College of Biology and Food Science, Chongqing Three Gorges University, Chongqing 404000, P.R. China
| | - Yonghua Zheng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095 Jiangsu, P.R. China
| |
Collapse
|
4
|
Gartz M, Beatka M, Prom MJ, Strande JL, Lawlor MW. Cardiomyocyte-produced miR-339-5p mediates pathology in Duchenne muscular dystrophy cardiomyopathy. Hum Mol Genet 2021; 30:2347-2361. [PMID: 34270708 PMCID: PMC8600005 DOI: 10.1093/hmg/ddab199] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/19/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disease characterized by severe, progressive muscle wasting. Cardiomyopathy has emerged as a leading cause of death in patients with DMD. The mechanisms contributing to DMD cardiac disease remain under investigation and specific therapies available are lacking. Our prior work has shown that DMD-iPSC-derived cardiomyocytes (DMD-iCMs) are vulnerable to oxidative stress injury and chronic exposure to DMD-secreted exosomes impaired the cell's ability to protect against stress. In this study, we sought to examine a mechanism by which DMD cardiac exosomes impair cellular response through altering important stress-responsive genes in the recipient cells. Here, we report that DMD-iCMs secrete exosomes containing altered microRNA (miR) profiles in comparison to healthy controls. In particular, miR-339-5p was upregulated in DMD-iCMs, DMD exosomes and mdx mouse cardiac tissue. Restoring dystrophin in DMD-iCMs improved the cellular response to stress and was associated with downregulation of miR-339-5p, suggesting that it is disease-specific. Knockdown of miR-339-5p was associated with increased expression of MDM2, GSK3A and MAP2K3, which are genes involved in important stress-responsive signaling pathways. Finally, knockdown of miR-339-5p led to mitochondrial protection and a reduction in cell death in DMD-iCMs, indicating miR-339-5p is involved in direct modulation of stress-responsiveness. Together, these findings identify a potential mechanism by which exosomal miR-339-5p may be modulating cell signaling pathways that are important for robust stress responses. Additionally, these exosomal miRs may provide important disease-specific targets for future therapeutic advancements for the management and diagnosis of DMD cardiomyopathy.
Collapse
Affiliation(s)
- Melanie Gartz
- Department of Cell Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Margaret Beatka
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mariah J Prom
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer L Strande
- Department of Cell Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael W Lawlor
- Department of Cell Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
5
|
Klein AM, de Queiroz RM, Venkatesh D, Prives C. The roles and regulation of MDM2 and MDMX: it is not just about p53. Genes Dev 2021; 35:575-601. [PMID: 33888565 PMCID: PMC8091979 DOI: 10.1101/gad.347872.120] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, Klein et al. discuss the p53-independent roles of MDM2 and MDMX. First, they review the structural and functional features of MDM2 and MDMX proteins separately and together that could be relevant to their p53-independent activities. Following this, they summarize how these two proteins are regulated and how they can function in cells that lack p53. Most well studied as proteins that restrain the p53 tumor suppressor protein, MDM2 and MDMX have rich lives outside of their relationship to p53. There is much to learn about how these two proteins are regulated and how they can function in cells that lack p53. Regulation of MDM2 and MDMX, which takes place at the level of transcription, post-transcription, and protein modification, can be very intricate and is context-dependent. Equally complex are the myriad roles that these two proteins play in cells that lack wild-type p53; while many of these independent outcomes are consistent with oncogenic transformation, in some settings their functions could also be tumor suppressive. Since numerous small molecules that affect MDM2 and MDMX have been developed for therapeutic outcomes, most if not all designed to prevent their restraint of p53, it will be essential to understand how these diverse molecules might affect the p53-independent activities of MDM2 and MDMX.
Collapse
Affiliation(s)
- Alyssa M Klein
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, New York 10032, USA
| | | | - Divya Venkatesh
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
6
|
Lam B, Roudier E. Considering the Role of Murine Double Minute 2 in the Cardiovascular System? Front Cell Dev Biol 2020; 7:320. [PMID: 31921839 PMCID: PMC6916148 DOI: 10.3389/fcell.2019.00320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/21/2019] [Indexed: 01/26/2023] Open
Abstract
The E3 ubiquitin ligase Murine double minute 2 (MDM2) is the main negative regulator of the tumor protein p53 (TP53). Extensive studies over more than two decades have confirmed MDM2 oncogenic role through mechanisms both TP53-dependent and TP53-independent oncogenic function. These studies have contributed to designate MDM2 as a therapeutic target of choice for cancer treatment and the number of patents for MDM2 antagonists has increased immensely over the last years. However, the question of the physiological functions of MDM2 has not been fully resolved yet, particularly when expressed and regulated physiologically in healthy tissue. Cardiovascular complications are almost an inescapable side-effect of anti-cancer therapies. While several MDM2 antagonists are entering phase I, II and even III of clinical trials, this review proposes to bring awareness on the physiological role of MDM2 in the cardiovascular system.
Collapse
Affiliation(s)
- Brian Lam
- Angiogenesis Research Group, School of Kinesiology and Health Sciences, Muscle Health Research Center, Faculty of Health, York University, Toronto, ON, Canada
| | - Emilie Roudier
- Angiogenesis Research Group, School of Kinesiology and Health Sciences, Muscle Health Research Center, Faculty of Health, York University, Toronto, ON, Canada
| |
Collapse
|
7
|
Meijles DN, Zoumpoulidou G, Markou T, Rostron KA, Patel R, Lay K, Handa BS, Wong B, Sugden PH, Clerk A. The cardiomyocyte "redox rheostat": Redox signalling via the AMPK-mTOR axis and regulation of gene and protein expression balancing survival and death. J Mol Cell Cardiol 2019; 129:118-129. [PMID: 30771309 PMCID: PMC6497135 DOI: 10.1016/j.yjmcc.2019.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/16/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) play a key role in development of heart failure but, at a cellular level, their effects range from cytoprotection to induction of cell death. Understanding how this is regulated is crucial to develop novel strategies to ameliorate only the detrimental effects. Here, we revisited the fundamental hypothesis that the level of ROS per se is a key factor in the cellular response by applying different concentrations of H2O2 to cardiomyocytes. High concentrations rapidly reduced intracellular ATP and inhibited protein synthesis. This was associated with activation of AMPK which phosphorylated and inhibited Raptor, a crucial component of mTOR complex-1 that regulates protein synthesis. Inhibition of protein synthesis by high concentrations of H2O2 prevents synthesis of immediate early gene products required for downstream gene expression, and such mRNAs (many encoding proteins required to deal with oxidant stress) were only induced by lower concentrations. Lower concentrations of H2O2 promoted mTOR phosphorylation, associated with differential recruitment of some mRNAs to the polysomes for translation. Some of the upregulated genes induced by low H2O2 levels are cytoprotective. We identified p21Cip1/WAF1 as one such protein, and preventing its upregulation enhanced the rate of cardiomyocyte apoptosis. The data support the concept of a "redox rheostat" in which different degrees of ROS influence cell energetics and intracellular signalling pathways to regulate mRNA and protein expression. This sliding scale determines cell fate, modulating survival vs death.
Collapse
Affiliation(s)
- Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London SW17 0RE, UK; School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK.
| | - Georgia Zoumpoulidou
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Thomais Markou
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Kerry A Rostron
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK
| | - Rishi Patel
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Kenneth Lay
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Balvinder S Handa
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Bethany Wong
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Peter H Sugden
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Angela Clerk
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
8
|
Strategies of biochemical adaptation for hibernation in a South American marsupial, Dromiciops gliroides: 3. Activation of pro-survival response pathways. Comp Biochem Physiol B Biochem Mol Biol 2017; 224:26-31. [PMID: 29248591 DOI: 10.1016/j.cbpb.2017.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022]
Abstract
The South American marsupial, monito del monte (Dromiciops gliroides) uses both daily torpor and multi-day hibernation to survive in its southern Chile native environment. The present study leverages multiplex technology to assess the contributions of key stress-inducible cell cycle regulators and heat shock proteins to hibernation in liver, heart, and brain of monito del monte in a comparison of control versus 4day hibernating conditions. The data indicate that MDM2, a stress-responsive ubiquitin ligase, plays a crucial role in marsupial hibernation since all three tissues showed statistically significant increases in MDM2 levels during torpor (1.6-1.8 fold). MDM2 may have a cytoprotective action to deal with ischemia/reperfusion stress and is also involved in a nutrient sensing pathway where it could help regulate the metabolic switch to fatty acid oxidation during torpor. Elevated levels of stress-sensitive cell cycle regulators including ATR (2.32-3.91 fold), and the phosphorylated forms of p-Chk1 (Ser345) (1.92 fold), p-Chk2 (Thr68) (2.20 fold) and p21 (1.64 fold) were observed in heart and liver during hibernation suggesting that the cell cycle is likely suppressed to conserve energy while animals are in torpor. Upregulation of heat shock proteins also occurred as a cytoprotective strategy with increased levels of hsp27 (2.00 fold) and hsp60 (1.72-2.76 fold) during hibernation. The results suggest that cell cycle control and selective chaperone action are significant components of hibernation in D. gliroides and reveal common molecular responses to those seen in eutherian hibernators.
Collapse
|
9
|
Zhang J, Cui L, Han X, Zhang Y, Zhang X, Chu X, Zhang F, Zhang Y, Chu L. Protective effects of tannic acid on acute doxorubicin-induced cardiotoxicity: Involvement of suppression in oxidative stress, inflammation, and apoptosis. Biomed Pharmacother 2017; 93:1253-1260. [DOI: 10.1016/j.biopha.2017.07.051] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 07/01/2017] [Accepted: 07/11/2017] [Indexed: 01/10/2023] Open
|
10
|
Parameswaran S, Sharma RK. Expression of calcineurin, calpastatin and heat shock proteins during ischemia and reperfusion. Biochem Biophys Rep 2015; 4:207-214. [PMID: 30338302 PMCID: PMC6189699 DOI: 10.1016/j.bbrep.2015.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/24/2022] Open
Abstract
Objective Calcineurin (CaN) interacts with calpains (Calpn) and causes cellular damage eventually leading to cell death. Calpastatin (Calp) is a specific Calpn inhibitor, along with CaN stimulation has been implicated in reduced cell death and self-repair. Molecular chaperones, heat shock proteins (Hsp70 and Hsp90) acts as regulators in Calpn signaling. This study aims to elucidate the role of CaN, Calp and Hsps during induced ischemia and reperfusion in primary cardiomyocyte cultures (murine). Methods and results Protein expression was analyzed concurrently with viability using flow cytometry (FACS) in ischemia- and reperfusion-induced murine cardiomyocyte cultures. The expression of Hsp70 and Hsp90, both being molecular chaperones, increased during ischemia with a concurrent increase in death of cells expressing these proteins. The relative expression of Hsp70 and Hsp90 during ischemia with respect to CaN was enhanced in comparison to Calp. Reperfusion slightly decreased the number of cells expressing these chaperones. There was no increase in death of cells co-expressing Hsp70 and Hsp90 along with CaN and Calp. CaN expression peaked during ischemia and subsequent reperfusion reduced its expression and cell death. Calp expression increased both during ischemia and subsequent reperfusion but cell death decreased during reperfusion. Conclusion The present study adds to the existing knowledge that Hsp70, Hsp90, CaN and Calp interact with each other and play significant role in cardio protection. Differential expression of calcineurin and calpastatin during ischemia and reperfusion. Enhanced ischemia induced cell death in cells expressing Hsp70 and Hsp90. Cardio protective role of calcineurin, calpastatin, Hsp70 and Hsp90.
Collapse
Key Words
- CaN, calcineurin
- Calcineurin
- Calp, Calpastatin
- Calpastatin
- Calpn, calpain
- FACS, flow cytometry
- FITC, fluorescein isothiocyanate
- HMWCaMBP, high molecular weight calmodulin-binding protein
- Heat shock proteins
- I/R, Ischemia and Reperfusion
- Ischemia
- NDB, nutrient deficient buffer
- NMCC, primary neonatal mouse cardiomyocyte culture
- PE, R-phycoerythrin
- Primary cardiomyocyte culture
- Reperfusion
Collapse
|
11
|
Parameswaran S, Sharma RK. Ischemia and reperfusion induce differential expression of calpastatin and its homologue high molecular weight calmodulin-binding protein in murine cardiomyocytes. PLoS One 2014; 9:e114653. [PMID: 25486053 PMCID: PMC4259361 DOI: 10.1371/journal.pone.0114653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/12/2014] [Indexed: 12/26/2022] Open
Abstract
In the heart, calpastatin (Calp) and its homologue high molecular weight calmodulin-binding protein (HMWCaMBP) regulate calpains (Calpn) by inhibition. A rise in intracellular myocardial Ca2+ during cardiac ischemia activates Calpn thereby causing damage to myocardial proteins, which leads to myocyte death and consequently to loss of myocardial structure and function. The present study aims to elucidate expression of Calp and HMWCaMBP with respect to Calpn during induced ischemia and reperfusion in primary murine cardiomyocyte cultures. Ischemia and subsequently reperfusion was induced in ∼80% confluent cultures of neonatal murine cardiomyocytes (NMCC). Flow cytometric analysis (FACS) has been used for analyzing protein expression concurrently with viability. Confocal fluorescent microscopy was used to observe protein localization. We observed that ischemia induces increased expression of Calp, HMWCaMBP and Calpn. Calpn expressing NMCC on co-expressing Calp survived ischemic induction compared to NMCC co-expressing HMWCaMBP. Similarly, living cells expressed Calp in contrast to dead cells which expressed HMWCaMBP following reperfusion. A significant difference in the expression of Calp and its homologue HMWCaMBP was observed in localization studies during ischemia. The current study adds to the existing knowledge that HMWCaMBP could be a putative isoform of Calp. NMCC on co-expressing Calp and Calpn-1 survived ischemic and reperfusion inductions compared to NMCC co-expressing HMWCaMBP and Calpn-1. A significant difference in expression of Calp and HMWCaMBP was observed in localization studies during ischemia.
Collapse
Affiliation(s)
- Sreejit Parameswaran
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Rajendra K. Sharma
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- * E-mail:
| |
Collapse
|
12
|
Senescence induction in renal carcinoma cells by Nutlin-3: a potential therapeutic strategy based on MDM2 antagonism. Cancer Lett 2014; 353:211-9. [DOI: 10.1016/j.canlet.2014.07.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 12/16/2022]
|
13
|
Tong L, Wu S. ROS and p53 in regulation of UVB-induced HDM2 alternative splicing. Photochem Photobiol 2014; 91:221-4. [PMID: 24986024 DOI: 10.1111/php.12306] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/29/2014] [Indexed: 11/30/2022]
Abstract
Alternative splicing plays an important role in proteasome diversity and gene expression regulation in eukaryotic cells. Hdm2, the human homolog of mdm2 (murine double minute oncogene 2), is known to be an oncogene as its role in suppression of p53. Hdm2 alternative splicing, occurs in both tumor and normal tissues, is believed to be a response of cells for cellular stress, and thus modulate p53 activity. Therefore, understanding the regulation of hdm2 splicing is critical in elucidating the mechanisms of tumor development and progression. In this study, we determined the effect of ultraviolet B light (UVB) on alternative splicing of hdm2. Our data indicated that UVB (50 mJ cm(-2)) alone is not a good inducer of alternative splicing of hdm2. The less effectiveness could be due to the induction of ROS and p53 by UVB because removing ROS by L-NAC (10 mm) in p53 null cells could lead to alternative splicing of hdm2 upon UVB irradiation.
Collapse
Affiliation(s)
- Lingying Tong
- Department of Chemistry and Biochemistry, and Molecular and Cellular Biology Program, Edison Biotechnology Institute, Ohio University, Athens, OH
| | | |
Collapse
|
14
|
Abstract
Proper protein turnover is required for cardiac homeostasis and, accordingly, impaired proteasomal function appears to contribute to heart disease. Specific proteasomal degradation mechanisms underlying cardiovascular biology and disease have been identified, and such cellular pathways have been proposed to be targets of clinical relevance. This review summarizes the latest literature regarding the specific E3 ligases involved in heart biology, and the general ways that the proteasome regulates protein quality control in heart disease. The potential for therapeutic intervention in Ubiquitin Proteasome System function in heart disease is discussed.
Collapse
Affiliation(s)
- Julia Pagan
- Department of Translational Medical Sciences, Via Sergio Pansini, 5, 80131 Naples, Italy
| | | | | | | |
Collapse
|
15
|
Caputo F, Vegliante R, Ghibelli L. Redox modulation of the DNA damage response. Biochem Pharmacol 2012; 84:1292-306. [PMID: 22846600 DOI: 10.1016/j.bcp.2012.07.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 01/09/2023]
Abstract
Lesions to DNA trigger the DNA-damage response (DDR), a complex, multi-branched cell-intrinsic process targeted to DNA repair, or elimination of the damaged cells by apoptosis. DDR aims at reducing permanence of mutated cells, decreasing the risk of tumor development: the more stringent the response, the lower the likelihood that sub-lethally damaged, unrepaired cells survive and proliferate. Accordingly, leakage often occurs in tumor cells with compromised DDR, accumulating mutations and accelerating tumor progression. Oxidations mediate DNA damage upon different insults such as UV, X and γ radiation, pollutants, poisons, or endogenous disequilibria, producing different types of lesions that trigger DDR, which can be alleviated by antioxidants. But reactive oxygen species (ROS), and the enzymes involved in their production or scavenging, also participate in DDR signaling, modulating the activity of key enzymes, and regulating the stringency of DDR. Accordingly, antioxidant enzymes such as superoxide dismutase play intimate and complex roles in tumor development, exceeding the basal roles of preventing the initial DNA damage. Likewise, it is emerging that dietary antioxidants help controlling tumor onset and progression by preventing DNA damage and by acting on cell cycle checkpoints, opening a novel and promising frontier to anticancer therapy.
Collapse
Affiliation(s)
- Fanny Caputo
- Dipartimento di Scienze e Tecnologie Chimiche, Universita' di Roma Tor Vergata, Roma, Italy
| | | | | |
Collapse
|
16
|
Shearn CT, Reigan P, Petersen DR. Inhibition of hydrogen peroxide signaling by 4-hydroxynonenal due to differential regulation of Akt1 and Akt2 contributes to decreases in cell survival and proliferation in hepatocellular carcinoma cells. Free Radic Biol Med 2012; 53:1-11. [PMID: 22580126 PMCID: PMC3377776 DOI: 10.1016/j.freeradbiomed.2012.04.021] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 04/18/2012] [Accepted: 04/22/2012] [Indexed: 11/16/2022]
Abstract
Dysregulation of cell signaling by electrophiles such as 4-hydroxynonenal (4-HNE) is a key component in the pathogenesis of chronic inflammatory liver disease. Another consequence of inflammation is the perpetuation of oxidative damage by the production of reactive oxidative species such as hydrogen peroxide. Previously, we have demonstrated Akt2 as a direct target of 4-HNE in hepatocellular carcinoma cells. In the present study, we used the hepatocellular carcinoma cell line HepG2 as model to understand the combinatorial effects of 4-HNE and hydrogen peroxide. We demonstrate that 4-HNE inhibits hydrogen peroxide-mediated phosphorylation of Akt1 but not Akt2. Pretreatment of HepG2 cells with 4-HNE prevented hydrogen peroxide stimulation of Akt-dependent phosphorylation of downstream targets and intracellular Akt activity compared with untreated control cells. Using biotin hydrazide capture, it was confirmed that 4-HNE treatment resulted in carbonylation of Akt1, which was not observed in untreated control cells. Using a synthetic GSK3α/β peptide as a substrate, treatment of recombinant human myristoylated Akt1 (rAkt1) with 20 or 40 μΜ 4-HNE inhibited rAkt1 activity by 29 and 60%, respectively. We further demonstrate that 4-HNE activates Erk via a PI3 kinase and PP2A-dependent mechanism leading to increased Jnk phosphorylation. At higher concentrations, 4-HNE decreased both cell survival and proliferation as evidenced by MTT assays and EdU incorporation as well as decreased expression of cyclin D1 and β-catenin, an effect only moderately increased by the addition of hydrogen peroxide. The ability of 4-HNE to exert combinatorial effects on Erk, Jnk, and Akt-dependent cell survival pathways provides additional insight into the mechanisms of cellular damage associated with chronic inflammation.
Collapse
Affiliation(s)
| | | | - Dennis R. Petersen
- To whom correspondence should be addressed: Dennis Petersen, University of Colorado Denver, School of Pharmacy, Department of Pharmaceutical Sciences, 12850 East Montview Blvd Box C238, Building V20 Room 2131, Ph. 303-724-3397, Fax 303-724-7266,
| |
Collapse
|
17
|
Abstract
The canonical pathway of regulation of the GCK (germinal centre kinase) III subgroup member, MST3 (mammalian Sterile20-related kinase 3), involves a caspase-mediated cleavage between N-terminal catalytic and C-terminal regulatory domains with possible concurrent autophosphorylation of the activation loop MST3(Thr178), induction of serine/threonine protein kinase activity and nuclear localization. We identified an alternative ‘non-canonical’ pathway of MST3 activation (regulated primarily through dephosphorylation) which may also be applicable to other GCKIII (and GCKVI) subgroup members. In the basal state, inactive MST3 co-immunoprecipitated with the Golgi protein GOLGA2/gm130 (golgin A2/Golgi matrix protein 130). Activation of MST3 by calyculin A (a protein serine/threonine phosphatase 1/2A inhibitor) stimulated (auto)phosphorylation of MST3(Thr178) in the catalytic domain with essentially simultaneous cis-autophosphorylation of MST3(Thr328) in the regulatory domain, an event also requiring the MST3(341–376) sequence which acts as a putative docking domain. MST3(Thr178) phosphorylation increased MST3 kinase activity, but this activity was independent of MST3(Thr328) phosphorylation. Interestingly, MST3(Thr328) lies immediately C-terminal to a STRAD (Sterile20-related adaptor) pseudokinase-like site identified recently as being involved in binding of GCKIII/GCKVI members to MO25 scaffolding proteins. MST3(Thr178/Thr328) phosphorylation was concurrent with dissociation of MST3 from GOLGA2/gm130 and association of MST3 with MO25, and MST3(Thr328) phosphorylation was necessary for formation of the activated MST3–MO25 holocomplex.
Collapse
|
18
|
Jia Y, Zhou F, Deng P, Fan Q, Li C, Liu Y, Fu X, Zhou Y, Xu X, Sun X. Interleukin 6 protects H(2)O(2)-induced cardiomyocytes injury through upregulation of prohibitin via STAT3 phosphorylation. Cell Biochem Funct 2012; 30:426-31. [PMID: 22431190 DOI: 10.1002/cbf.2820] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 02/15/2012] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Hydrogen peroxide (H(2)O(2)) is a potent reactive oxygen species that causes cardiomyocytes injury. As an important cytokine, interleukin 6 (IL-6) has cardioprotective effects as it plays an essential role in the late phase of preconditioning. Our work is to investigate if IL-6 preconditioning has protective effects on neonatal rat ventricular cardiomyocytes in response to H(2)O(2) and its underlying mechanism. METHODS Gel-based comparative proteomic approach along with small interfering RNA (siRNA) and Western blot analysis was used to analyse mechanisms of IL-6 preconditioning on H(2)O(2)-induced neonatal rat ventricular cardiomyocytes injury. RESULTS IL-6 preconditioning protected cardiomyocytes against H(2)O(2)-induced cell death. Proteomic analysis showed that IL-6 pretreatment further increased the expression of prohibitin and improved the viability of cardiomyocytes exposed to H(2)O(2). Knocking down of prohibitin with siRNA abrogated this protection by increasing apoptosis rate. Tyrosine kinase inhibitor AG490 decreased signal transducers and activators of transcription 3 (STAT3) phosphorylation and down-regulated prohibitin expression in cardiomyocytes pretreated with IL-6 and exposed to H(2)O(2), which further dampened the protective effects of IL-6 preconditioning. CONCLUSION Our results provide direct evidence that prohibitin is a protective factor of IL-6 preconditioning in H(2)O(2)-induced neonatal rat ventricular cardiomyocytes death. The upregulation of prohibitin by IL-6 is, at least, partially regulated through STAT3 phosphorylation.
Collapse
Affiliation(s)
- Yuhua Jia
- Nanfang hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Portbury AL, Ronnebaum SM, Zungu M, Patterson C, Willis MS. Back to your heart: ubiquitin proteasome system-regulated signal transduction. J Mol Cell Cardiol 2012; 52:526-37. [PMID: 22085703 PMCID: PMC3294005 DOI: 10.1016/j.yjmcc.2011.10.023] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 10/28/2011] [Accepted: 10/30/2011] [Indexed: 12/19/2022]
Abstract
Awareness of the regulation of cell signaling by post-translational ubiquitination has emerged over the past 2 decades. Like phosphorylation, post-translational modification of proteins with ubiquitin can result in the regulation of numerous cellular functions, for example, the DNA damage response, apoptosis, cell growth, and the innate immune response. In this review, we discuss recently published mechanisms by which the ubiquitin proteasome system regulates key signal transduction pathways in the heart, including MAPK JNK, calcineurin, FOXO, p53, and estrogen receptors α and β. We then explore how ubiquitin proteasome system-specific regulation of these signal transduction pathways plays a role in the pathophysiology of common cardiac diseases, such as cardiac hypertrophy, heart failure, ischemia reperfusion injury, and diabetes. This article is part of a Special Section entitled "Post-translational Modification."
Collapse
Affiliation(s)
- Andrea L. Portbury
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
| | - Sarah M. Ronnebaum
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
| | - Makhosazane Zungu
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Cam Patterson
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Departments of Cell and Developmental Biology, Medicine, and Pharmacology, University of North Carolina, Chapel Hill, NC
| | - Monte S. Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| |
Collapse
|
20
|
Preventive effects of fasudil on adriamycin-induced cardiomyopathy: Possible involvement of inhibition of RhoA/ROCK pathway. Food Chem Toxicol 2011; 49:2975-82. [DOI: 10.1016/j.fct.2011.06.080] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 06/03/2011] [Accepted: 06/27/2011] [Indexed: 11/17/2022]
|
21
|
Knöll R, Linke WA, Zou P, Miocic S, Kostin S, Buyandelger B, Ku CH, Neef S, Bug M, Schäfer K, Knöll G, Felkin LE, Wessels J, Toischer K, Hagn F, Kessler H, Didié M, Quentin T, Maier LS, Teucher N, Unsöld B, Schmidt A, Birks EJ, Gunkel S, Lang P, Granzier H, Zimmermann WH, Field LJ, Faulkner G, Dobbelstein M, Barton PJR, Sattler M, Wilmanns M, Chien KR. Telethonin deficiency is associated with maladaptation to biomechanical stress in the mammalian heart. Circ Res 2011; 109:758-69. [PMID: 21799151 DOI: 10.1161/circresaha.111.245787] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Telethonin (also known as titin-cap or t-cap) is a 19-kDa Z-disk protein with a unique β-sheet structure, hypothesized to assemble in a palindromic way with the N-terminal portion of titin and to constitute a signalosome participating in the process of cardiomechanosensing. In addition, a variety of telethonin mutations are associated with the development of several different diseases; however, little is known about the underlying molecular mechanisms and telethonin's in vivo function. OBJECTIVE Here we aim to investigate the role of telethonin in vivo and to identify molecular mechanisms underlying disease as a result of its mutation. METHODS AND RESULTS By using a variety of different genetically altered animal models and biophysical experiments we show that contrary to previous views, telethonin is not an indispensable component of the titin-anchoring system, nor is deletion of the gene or cardiac specific overexpression associated with a spontaneous cardiac phenotype. Rather, additional titin-anchorage sites, such as actin-titin cross-links via α-actinin, are sufficient to maintain Z-disk stability despite the loss of telethonin. We demonstrate that a main novel function of telethonin is to modulate the turnover of the proapoptotic tumor suppressor p53 after biomechanical stress in the nuclear compartment, thus linking telethonin, a protein well known to be present at the Z-disk, directly to apoptosis ("mechanoptosis"). In addition, loss of telethonin mRNA and nuclear accumulation of this protein is associated with human heart failure, an effect that may contribute to enhanced rates of apoptosis found in these hearts. CONCLUSIONS Telethonin knockout mice do not reveal defective heart development or heart function under basal conditions, but develop heart failure following biomechanical stress, owing at least in part to apoptosis of cardiomyocytes, an effect that may also play a role in human heart failure.
Collapse
Affiliation(s)
- Ralph Knöll
- Imperial College, National Heart & Lung Institute, British Heart Foundation, Centre for Research Excellence, Myocardial Genetics, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang Z, Li B. Mdm2 links genotoxic stress and metabolism to p53. Protein Cell 2011; 1:1063-72. [PMID: 21213101 DOI: 10.1007/s13238-010-0140-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 11/30/2010] [Indexed: 01/06/2023] Open
Abstract
Mouse double minute 2 (Mdm2) gene was isolated from a cDNA library derived from transformed mouse 3T3 cells, and was classified as an oncogene as it confers 3T3 and Rat2 cells tumorigenicity when overexpressed. It encodes a nucleocytoplasmic shuttling ubiquitin E3 ligase, with its main target being tumor suppressor p53, which is mutated in more than 50% of human primary tumors. Mdm2's oncogenic activity is mainly mediated by p53, which is activated by various stresses, especially genotoxic stress, via Atm (ataxia telangiectasia mutated) and Atr (Atm and Rad3-related). Activated p53 inhibits cell proliferation, induces apoptosis or senescence, and maintains genome integrity. Mdm2 is also a target gene of p53 transcription factor. Thus, Mdm2 and p53 form a feedback regulatory loop. External and internal cues, through multiple signaling pathways, can act on Mdm2 to regulate p53 levels and cell proliferation, death, and senescence. This review will focus on how Mdm2 is regulated under genotoxic stress, and by the Akt1-mTOR-S6K1 pathway that is activated by insulin, growth factors, amino acids, or energy status.
Collapse
Affiliation(s)
- Zhongfeng Wang
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | | |
Collapse
|
23
|
Morimoto Y, Bando YK, Shigeta T, Monji A, Murohara T. Atorvastatin prevents ischemic limb loss in type 2 diabetes: role of p53. J Atheroscler Thromb 2010; 18:200-8. [PMID: 21123956 DOI: 10.5551/jat.6437] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Diabetic peripheral artery disease (PAD) is prone to be aggressive and recent reports have demonstrated that p53 accumulation may be responsible for impaired wound healing in diabetes. Statins has been demonstrated to facilitate p53 degradation by activating its specific ubiquitin ligase, MDM2. The aim of this study was to determine whether atorvastatin (ATR) improves the outcome of diabetic PAD through MDM2-mediated reduction of p53. METHODS Male KK/Ay mice (9 weeks old) were treated with ATR (2 mg/kg/day p.o.) or vehicle for 2 weeks and subjected to ischemic hindlimb operation to generate a diabetic PAD model. Incidences of amputation and changes of p53/MDM2 signaling in each ischemic limb were assessed 2 weeks after the operation (at 13 weeks of age). Effects of ATR on the insulin resistance of age-matched (13-week-old) and unoperated KK/Ay mice were assessed by the glucose tolerance test, circulating adiponectin concentration, and changes in insulin signaling (IRS-1/Akt phosphorylation). RESULTS In intact KK/Ay, ATR treatment mitigated insulin resistance without affecting cholesterol levels. All diabetic PAD models exhibited autoamputation (100%); however, ATR treatment partially restored the limb loss (41.7%). The p53 expression level in the ischemic limb of ATR-treated KK/Ay was significantly decreased and MDM2 phosphorylation level was markedly increased in tandem with the activation of Akt. Hypoxia mimetic iron chelator deferroxamine promoted p53 accumulation in H9c2 myoblast cells by suppressing the Akt/MDM2 pathway, which was restored by ATR. CONCLUSIONS ATR was found to restore ischemic limb loss in diabetes by augmenting p53 degradation through direct activation of the Akt/MDM2 pathway in skeletal muscle.
Collapse
Affiliation(s)
- Yasutsugu Morimoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | | | | | | | | |
Collapse
|
24
|
Giglio S, Mancini F, Pellegrino M, Di Conza G, Puxeddu E, Sacchi A, Pontecorvi A, Moretti F. Regulation of MDM4 (MDMX) function by p76(MDM2): a new facet in the control of p53 activity. Oncogene 2010; 29:5935-45. [PMID: 20697359 DOI: 10.1038/onc.2010.324] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Under basal growth conditions, p53 function is tightly controlled by the members of MDM family, MDM2 and MDM4. The Mdm2 gene codes, in addition to the full-length p90(MDM2), for a short protein, p76(MDM2) that lacks the p53-binding domain. Despite this property and at variance with p90(MDM2), this protein acts positively toward p53, although the molecular mechanism remains elusive. Here, we report that p76(MDM2) antagonizes MDM4 inhibitory function. We show that p76(MDM2) possesses intrinsic ubiquitinating and degrading activity, and through these activities controls MDM4 levels. Furthermore, the presence of p76(MDM2) decreases the association of MDM4 with p53 and p90(MDM2), and antagonizes p53 degradation by the heterodimer MDM4/p90(MDM2). The p76(MDM2)-mediated regulation of MDM4 occurs in the cytoplasm, under basal growth conditions. Conversely, upon DNA damage, phosphorylation of MDM4Ser403 dissociates p76(MDM2) and prevents MDM4 degradation. The overall negative control of MDM4 by p76(MDM2) reflects on p53 function as p76(MDM2) impairs MDM4-mediated inhibition of p53 activity. In agreement with the positive role of p76(MDM2) toward p53, the p76(MDM2)/p90(MDM2) ratio significantly decreases in a group of thyroid tumor samples compared with normal counterparts. Overall, these findings reveal a new mechanism in the control of p53 basal activity that may account for the distinct sensitivity of tissues to stress signals depending on the balance among MDM proteins. Moreover, these data suggest an oncosuppressive function for a product of the Mdm2 gene.
Collapse
Affiliation(s)
- S Giglio
- Institute of Neurobiology and Molecular Medicine, CNR/Fondazione Santa Lucia, Roma, Italy
| | | | | | | | | | | | | | | |
Collapse
|