51
|
Bahrami E, Witzel M, Racek T, Puchałka J, Hollizeck S, Greif-Kohistani N, Kotlarz D, Horny HP, Feederle R, Schmidt H, Sherkat R, Steinemann D, Göhring G, Schlegelbeger B, Albert MH, Al-Herz W, Klein C. Myb-like, SWIRM, and MPN domains 1 (MYSM1) deficiency: Genotoxic stress-associated bone marrow failure and developmental aberrations. J Allergy Clin Immunol 2017; 140:1112-1119. [PMID: 28115216 DOI: 10.1016/j.jaci.2016.10.053] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/22/2016] [Accepted: 10/17/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Myb-like, SWIRM, and MPN domains 1 (MYSM1) is a transcriptional regulator mediating histone deubiquitination. Its role in human immunity and hematopoiesis is poorly understood. OBJECTIVES We sought to investigate the clinical, cellular, and molecular features in 2 siblings presenting with progressive bone marrow failure (BMF), immunodeficiency, and developmental aberrations. METHODS We performed genome-wide homozygosity mapping, whole-exome and Sanger sequencing, immunophenotyping studies, and analysis of genotoxic stress responses. p38 activation, reactive oxygen species levels, rate of apoptosis and clonogenic survival, and growth in immune and nonimmune cells were assessed. The outcome of allogeneic hematopoietic stem cell transplantation (HSCT) was monitored. RESULTS We report 2 patients with progressive BMF associated with myelodysplastic features, immunodeficiency affecting B cells and neutrophil granulocytes, and complex developmental aberrations, including mild skeletal anomalies, neurocognitive developmental delay, and cataracts. Whole-exome sequencing revealed a homozygous premature stop codon mutation in the gene encoding MYSM1. MYSM1-deficient cells are characterized by increased sensitivity to genotoxic stress associated with sustained induction of phosphorylated p38 protein, increased reactive oxygen species production, and decreased survival following UV light-induced DNA damage. Both patients were successfully treated with allogeneic HSCT with sustained reconstitution of hematopoietic defects. CONCLUSIONS Here we show that MYSM1 deficiency is associated with developmental aberrations, progressive BMF with myelodysplastic features, and increased susceptibility to genotoxic stress. HSCT represents a curative therapy for patients with MYSM1 deficiency.
Collapse
Affiliation(s)
- Ehsan Bahrami
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Maximilian Witzel
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Tomas Racek
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Jacek Puchałka
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Sebastian Hollizeck
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Naschla Greif-Kohistani
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Daniel Kotlarz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Hans-Peter Horny
- Institute for Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Regina Feederle
- Helmholtz Zentrum München, German Research Center for Environmental Health, Core Facility Monoclonal Antibody Development, Munich, Germany
| | - Heinrich Schmidt
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Roya Sherkat
- Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Doris Steinemann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Gudrun Göhring
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | | | - Michael H Albert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, and Department of Pediatrics, Al-Sabah Hospital, Kuwait City, Kuwait
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität, Munich, Germany.
| |
Collapse
|
52
|
Sun YY, Xiao L, Wang D, Ji YC, Yang YP, Ma R, Chen XH. Triptolide inhibits viability and induces apoptosis in liver cancer cells through activation of the tumor suppressor gene p53. Int J Oncol 2017; 50:847-852. [PMID: 28098861 DOI: 10.3892/ijo.2017.3850] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 12/23/2016] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the effect of triptolide on viability and apoptosis along with underlying mechanism in liver cancer cells. CCK-8 assay showed that triptolide treatment for 48 h significantly reduced the viability of HepG2 and QSG7701 cells at 50 µM concentration. Annexin V-FITC and propidium iodide staining showed that triptolide treatment of HepG2 cells at 50 µM concentrations induced apoptosis in 56.45% cells compared to only 2.36% cells in the control cultures. Western blot assay showed that treatment of HepG2 cells with 50 µM concentration of triptolide significantly induced phosphorylation of p53 in a 2 h-treatment. Phosphorylation of histone H2A.X indicator of DNA damage was induced by triptolide treatment after 12 h in HepG2 cells. The level of nuclear p53 in a 6 h-treatment with 0, 10, 20, 30, 40 and 50 µM concentration of triptolide was found to be 15.3, 19.6, 28.5, 43.7, 63.8 and 91.5%, respectively. Treatment of HepG2 cells with triptolide at 50 µM concentration caused a significant increase in the binding potential of p53 to DNA. Triptolide treatment of HepG2 cells caused a significant increase in the expression of p21, Bax and DR5 genes in HepG2 cells. It also increased the expression of miR-34b and miR-34c in HepG2 cells markedly. Treatment of HepG2 cells with p53 inhibitor, pifithrin-α prior to incubation with triptolide significantly prevented induction of cell apoptosis. Suppression of p53 expression by siRNA inhibited the expression of p53 as well as its target genes along with the prevention of apoptosis induction. In conclusion, triptolide inhibits viability and induces apoptosis in liver cancer cells through activation of the tumor suppressor gene p53. Thus, triptolide can be used for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yan-Yan Sun
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Lei Xiao
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yan-Chao Ji
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yu-Peng Yang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Rong Ma
- Department of Gynecological Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150040, P.R. China
| | - Xi-Hai Chen
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
53
|
UBE4B targets phosphorylated p53 at serines 15 and 392 for degradation. Oncotarget 2016; 7:2823-36. [PMID: 26673821 PMCID: PMC4823074 DOI: 10.18632/oncotarget.6555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 11/21/2015] [Indexed: 12/31/2022] Open
Abstract
Phosphorylation of p53 is a key mechanism responsible for the activation of its tumor suppressor functions in response to various stresses. In unstressed cells, p53 is rapidly turned over and is maintained at a low basal level. After DNA damage or other forms of cellular stress, the p53 level increases, and the protein becomes metabolically stable. However, the mechanism of phosphorylated p53 regulation is unclear. In this study, we studied the kinetics of UBE4B, Hdm2, Pirh2, Cop1 and CHIP induction in response to p53 activation. We show that UBE4B coimmunoprecipitates with phosphorylated p53 at serines 15 and 392. Notably, the affinity between UBE4B and Hdm2 is greatly decreased after DNA damage. Furthermore, we observe that UBE4B promotes endogenous phospho-p53(S15) and phospho-p53(S392) degradation in response to IR. We demonstrate that UBE4B and Hdm2 repress p53S15A, p53S392A, and p53-2A(S15A, S392A) functions, including p53-dependent transactivation and growth inhibition. Overall, our results reveal that UBE4B plays an important role in regulating phosphorylated p53 following DNA damage.
Collapse
|
54
|
Song Y, Li N, Gu J, Fu S, Peng Z, Zhao C, Zhang Y, Li X, Wang Z, Li X, Liu G. β-Hydroxybutyrate induces bovine hepatocyte apoptosis via an ROS-p38 signaling pathway. J Dairy Sci 2016; 99:9184-9198. [PMID: 27756472 DOI: 10.3168/jds.2016-11219] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/26/2016] [Indexed: 12/22/2022]
Abstract
β-Hydroxybutyrate (BHB) is an important indicator for metabolic disorders in dairy cows, such as ketosis and fatty liver. Dairy cows with ketosis display oxidative stress that may be associated with high levels of BHB. The purpose of this study was to demonstrate a correlation between the high levels of BHB and oxidative stress in dairy cows with ketosis, and to investigate the molecular mechanisms underlying oxidative damage in bovine hepatocytes. The results showed that dairy cows with ketosis exhibited oxidative stress and liver damage, which was significantly correlated with plasma BHB. Similarly, high concentrations of BHB increased the oxidative stress of cow hepatocytes in vitro, resulting in the phosphorylation and activation of p38 mitogen-activated protein kinase (MAPK), which led to increased expression, nuclear localization, and transcriptional activity of p53 and decreased Nrf2 in bovine hepatocytes. High concentrations of BHB significantly increased the expression of proapoptotic genes and significantly inhibited the expression of antiapoptotic genes. Finally, high concentrations of BHB promoted apoptosis in bovine hepatocytes. N-Acetyl-l-cysteine, glucose, and SB203580 (p38 inhibitor) significantly attenuated BHB-induced apoptotic damage in hepatocytes. These results indicate that BHB induces bovine hepatocyte apoptosis through the ROS-p38-p53/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Na Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Jingmin Gu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Shoupeng Fu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Chenxu Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Yuming Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin, 130062, China.
| |
Collapse
|
55
|
Russo A, Pagliara V, Albano F, Esposito D, Sagar V, Loreni F, Irace C, Santamaria R, Russo G. Regulatory role of rpL3 in cell response to nucleolar stress induced by Act D in tumor cells lacking functional p53. Cell Cycle 2016; 15:41-51. [PMID: 26636733 DOI: 10.1080/15384101.2015.1120926] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many chemotherapeutic drugs cause nucleolar stress and p53-independent pathways mediating the nucleolar stress response are emerging. Here, we demonstrate that ribosomal stress induced by Actinomycin D (Act D) is associated to the up-regulation of ribosomal protein L3 (rpL3) and its accumulation as ribosome-free form in lung and colon cancer cell lines devoid of p53. Free rpL3 regulates p21 expression at transcriptional and post-translational levels through a molecular mechanism involving extracellular-signal-regulated kinases1/2 (ERK1/2) and mouse double minute-2 homolog (MDM2). Our data reveal that rpL3 participates to cell response acting as a critical regulator of apoptosis and cell migration. It is noteworthy that silencing of rpL3 abolishes the cytotoxic effects of Act D suggesting that the loss of rpL3 makes chemotherapy drugs ineffective while rpL3 overexpression associates to a strong increase of Act D-mediated inhibition of cell migration. Taking together our results show that the efficacy of Act D chemotherapy depends on rpL3 status revealing new specific targets involved in the molecular pathways activated by Act D in cancers lacking of p53. Hence, the development of treatments aimed at upregulating rpL3 may be beneficial for the treatment of these cancers.
Collapse
Affiliation(s)
- Annapina Russo
- a Department of Pharmacia , University of Naples "Federico II," ; Naples , Italy
| | - Valentina Pagliara
- a Department of Pharmacia , University of Naples "Federico II," ; Naples , Italy
| | - Francesco Albano
- a Department of Pharmacia , University of Naples "Federico II," ; Naples , Italy
| | - Davide Esposito
- b Department of Molecular Medicine and Medical Biotechnology , University of Naples "Federico II," Naples , Italy.,c Department of Oncological Sciences , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Vinay Sagar
- d Department of Biology , University of Rome "Tor Vergata," Rome , Italy
| | - Fabrizio Loreni
- d Department of Biology , University of Rome "Tor Vergata," Rome , Italy
| | - Carlo Irace
- a Department of Pharmacia , University of Naples "Federico II," ; Naples , Italy
| | - Rita Santamaria
- a Department of Pharmacia , University of Naples "Federico II," ; Naples , Italy
| | - Giulia Russo
- a Department of Pharmacia , University of Naples "Federico II," ; Naples , Italy
| |
Collapse
|
56
|
Pan JH, Lee KY, Kim JH, Shin H, Lee JH, Kim YJ. Prunus mume Sieb. et Zucc. fruit ameliorates alcoholic liver injury in mice by inhibiting apoptosis and inflammation through oxidative stress. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
57
|
Yuan F, Chen X, Liu J, Feng W, Wu X, Chen SY. Up-regulation of Siah1 by ethanol triggers apoptosis in neural crest cells through p38 MAPK-mediated activation of p53 signaling pathway. Arch Toxicol 2016; 91:775-784. [PMID: 27270636 DOI: 10.1007/s00204-016-1746-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/01/2016] [Indexed: 01/22/2023]
Abstract
Seven in absentia homolog 1 (Siah1) is one of the E3 ubiquitin ligases and plays a key role in regulating target protein degradation. This study was designed to test the hypothesis that Siah1 mediates ethanol-induced apoptosis in NCCs through p38 MAPK-mediated activation of the p53 signaling pathway. We found that exposure of NCCs to ethanol resulted in the increases in the total protein levels of p53 and the phosphorylation of p53 at serine 15. Ethanol exposure also resulted in a significant increase in the phosphorylation of p38 MAPK. Knock-down of Siah1 dramatically reduced the ethanol-induced increase in the phosphorylation of p38 MAPK. Knock-down of Siah1 by siRNA or down-regulation of p38 MAPK by either siRNA or inhibitor significantly diminished ethanol-induced accumulations of p53 and the phosphorylation of p53. In addition, ethanol exposure resulted in a significant increase in the expression of p53 downstream targets and apoptosis in NCCs, which can be significantly diminished by down-regulation of Siah1 with siRNA. Knock-down of p38 MAPK by siRNA also dramatically reduced the ethanol-induced apoptosis. These results demonstrate that Siah1 plays a crucial role in ethanol-induced apoptosis in NCCs, and that the up-regulation of Siah1 by ethanol can trigger apoptosis through p38 MAPK-mediated activation of the p53 signaling pathway.
Collapse
Affiliation(s)
- Fuqiang Yuan
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40292, USA.,University of Louisville Alcohol Research Center, Louisville, KY, 40292, USA
| | - Xiaopan Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40292, USA.,University of Louisville Alcohol Research Center, Louisville, KY, 40292, USA
| | - Jie Liu
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40292, USA.,University of Louisville Alcohol Research Center, Louisville, KY, 40292, USA
| | - Wenke Feng
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40292, USA.,University of Louisville Alcohol Research Center, Louisville, KY, 40292, USA.,Department of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, 40292, USA. .,University of Louisville Alcohol Research Center, Louisville, KY, 40292, USA.
| |
Collapse
|
58
|
Li N, Lorenzi F, Kalakouti E, Normatova M, Babaei-Jadidi R, Tomlinson I, Nateri AS. FBXW7-mutated colorectal cancer cells exhibit aberrant expression of phosphorylated-p53 at Serine-15. Oncotarget 2016; 6:9240-56. [PMID: 25860929 PMCID: PMC4496214 DOI: 10.18632/oncotarget.3284] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 02/05/2015] [Indexed: 12/22/2022] Open
Abstract
FBXW7 mutations occur in a variety of human cancers including colorectal cancer (CRC). Elucidating its mechanism of action has become crucial for cancer therapy; however, it is also complicated by the fact that FBXW7 can influence many pathways due to its role as an E3-ubiquitin ligase in proteasome degradation. FBXW7 and TP53 are tumour suppressors intensively implicated in colorectal carcinogenesis. Deletion mutations in these two genes in animal models mark the progression from adenoma to carcinoma. Although still largely unknown, the last defense mechanism against CRC at the molecular level could be through a synergistic effect of the two genes. The underlying mechanism requires further investigation. In our laboratory, we have used a phospho-kinase profiler array to illustrate a potential molecular link between FBXW7 and p53 in CRC cells. In vitro and in vivo assessments demonstrated aberrant induction of phosphorylated p53 at Serine 15 [phospho-p53(Ser15)] in human FBXW7-deficient CRC cells as compared to their FBXW7-wild-type counterparts. FBXW7 loss in HCT116 cells promoted resistance to oxaliplatin. Immunoblotting data further confirmed that reduction of phospho-p53(Ser15) may contribute to the decreased efficacy of therapy in FBXW7-mutated CRC cells. The findings may suggest the applicability of phospho-p53(Ser15) as an indicative marker of FBXW7-mutations. Phospho-p53(Ser15) regulation by FBXW7 E3-ligase activity could provide important clues for understanding FBXW7 behavior in tumour progression and grounds for its clinical applicability thereafter.
Collapse
Affiliation(s)
- Ningning Li
- Cancer Genetics & Stem Cell Group, Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.,Department of Neurodegenerative Disease, Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Federica Lorenzi
- Cancer Genetics & Stem Cell Group, Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Eliana Kalakouti
- Cancer Genetics & Stem Cell Group, Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.,Hillingdon Hospital, Uxbridge UB8 3NN, UK
| | - Makhliyo Normatova
- Cancer Genetics & Stem Cell Group, Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Roya Babaei-Jadidi
- Cancer Genetics & Stem Cell Group, Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Ian Tomlinson
- Molecular and Population Genetics Laboratory, the Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Abdolrahman S Nateri
- Cancer Genetics & Stem Cell Group, Cancer Biology Unit, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
59
|
Stulpinas A, Imbrasaitė A, Krestnikova N, Šarlauskas J, Čėnas N, Kalvelytė AV. Study of Bioreductive Anticancer Agent RH-1-Induced Signals Leading the Wild-Type p53-Bearing Lung Cancer A549 Cells to Apoptosis. Chem Res Toxicol 2015; 29:26-39. [DOI: 10.1021/acs.chemrestox.5b00336] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Aurimas Stulpinas
- Vilnius University Institute of Biochemistry, Mokslininku
st. 12, LT-08662 Vilnius, Lithuania
| | - Aušra Imbrasaitė
- Vilnius University Institute of Biochemistry, Mokslininku
st. 12, LT-08662 Vilnius, Lithuania
| | - Natalija Krestnikova
- Vilnius University Institute of Biochemistry, Mokslininku
st. 12, LT-08662 Vilnius, Lithuania
| | - Jonas Šarlauskas
- Vilnius University Institute of Biochemistry, Mokslininku
st. 12, LT-08662 Vilnius, Lithuania
| | - Narimantas Čėnas
- Vilnius University Institute of Biochemistry, Mokslininku
st. 12, LT-08662 Vilnius, Lithuania
| | | |
Collapse
|
60
|
Lenox AR, Bhootada Y, Gorbatyuk O, Fullard R, Gorbatyuk M. Unfolded protein response is activated in aged retinas. Neurosci Lett 2015; 609:30-5. [PMID: 26467812 DOI: 10.1016/j.neulet.2015.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/04/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022]
Abstract
An unfolded protein response (UPR) in addition to oxidative stress and the inflammatory response is known to be activated in age-related ocular disorders, such as macular degeneration, diabetic retinopathy, glaucoma, and cataracts. Therefore, we aimed to investigate whether healthy aged retinas display UPR hallmarks, in order to establish a baseline for the activated UPR markers for age-related ocular diseases. Using western blotting, we determined that the hallmarks of the UPR PERK arm, phosphorylated (p) eIF2a, ATF4, and GADD34, were significantly altered in aged vs. young rat retinas. The cleaved pATF6 (50) and CHOP proteins were dramatically upregulated in the aged rodent retinas, indicating the activation of the ATF6 UPR arm. The UPR activation was associated with a drop in rhodopsin expression and in the NRF2 and HO1 levels, suggesting a decline in the anti-oxidant defense in aged retinas. Moreover, we observed down-regulation of anti-inflammatory IL-10 and IL-13 and upregulation of pro-inflammatory RANTES in the healthy aged retinas, as measured using the Bio-plex assay. Our results suggest that cellular homeostasis in normal aged retinas is compromised, resulting in the concomitant activation of the UPR, oxidative stress, and inflammatory signaling. This knowledge brings us closer to understanding the cellular mechanisms of the age-related retinopathies and ocular disorders characterized by an ongoing UPR, and highlight the UPR signaling molecules that should be validated as potential therapeutic targets.
Collapse
Affiliation(s)
- Austin R Lenox
- University of Alabama at Birmingham, Department of Vision Sciences, United States
| | - Yogesh Bhootada
- University of Alabama at Birmingham, Department of Vision Sciences, United States
| | - Oleg Gorbatyuk
- University of Alabama at Birmingham, Department of Vision Sciences, United States
| | - Roderick Fullard
- University of Alabama at Birmingham, Department of Vision Sciences, United States
| | - Marina Gorbatyuk
- University of Alabama at Birmingham, Department of Vision Sciences, United States.
| |
Collapse
|
61
|
O6-Methylguanine-DNA methyltransferase (MGMT) gene expression is associated with ultraviolet B (UVB)-induced cell growth inhibition and recovery. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0308-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
62
|
Lambert IH, Enghoff MS, Brandi ML, Hoffmann EK. Regulation of p53 in NIH3T3 mouse fibroblasts following hyperosmotic stress. Physiol Rep 2015; 3:3/6/e12412. [PMID: 26056062 PMCID: PMC4510620 DOI: 10.14814/phy2.12412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The aim of this project was to analyze the regulation of p53 expression in NIH3T3 fibroblasts under the influence of increasing hyperosmotic stress. Expression of p53 showed a biphasic response pattern in NIH3T3 cells under increasing osmotic stress (337 mOsm to 737 mOsm) with a maximum at 587 mOsm. Under isotonic conditions p53 expression increased after addition of the proteasome inhibitor MG132 indicating that cellular p53 levels in unperturbed cells is kept low by proteasomal degradation. However, under hypertonic conditions p53 synthesis as well as p53 degradation were significantly reduced and it is demonstrated that the increase in p53 expression observed when tonicity is increased from 337 to 587 mOsm reflects that degradation is more inhibited than synthesis, whereas the decrease in p53 expression at higher tonicities reflects that synthesis is more inhibited than degradation. The activity of the p53 regulating proteins p38 MAP kinase and the ubiquitin ligase MDM2 were studied as a function of increasing osmolarity. MDM2 protein expression was unchanged at all osmolarities, whereas MDM2 phosphorylation (Ser166) increased at osmolarities up to 537 mOsm and remained constant at higher osmolarities. Phosphorylation of p38 increased at osmolarities up to 687 mOsm which correlated with an increased phosphorylation of p53 (Ser15) and the decreased p53 degradation. Caspase-3 activity increased gradually with hypertonicity and at 737 mOsm both Caspase-3 activity and annexin V binding are high even though p53 expression and activity are low, indicating that initiation of apoptosis under severe hypertonic conditions is not strictly controlled by p53.
Collapse
Affiliation(s)
- Ian Henry Lambert
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Stine Enghoff
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Luise Brandi
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Else Kay Hoffmann
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
63
|
Chen CC, Kim KH, Lau LF. The matricellular protein CCN1 suppresses hepatocarcinogenesis by inhibiting compensatory proliferation. Oncogene 2015; 35:1314-23. [PMID: 26028023 PMCID: PMC4666840 DOI: 10.1038/onc.2015.190] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 04/23/2015] [Accepted: 05/01/2015] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide, and is on the rise in the United States. Previous studies showed that the matricellular protein CCN1 (CYR61) is induced during hepatic injuries and functions to restrict and resolve liver fibrosis. Here we show that CCN1 suppresses hepatocarcinogenesis by inhibiting carcinogen-induced compensatory hepatocyte proliferation, thus limiting the expansion of damaged and potentially oncogenic hepatocytes. Consistent with tumor suppression, CCN1 expression is down-regulated in human HCC. Ccn1ΔHep mice with hepatocyte-specific deletion of Ccn1 suffer increased HCC tumor multiplicity induced by the hepatocarcinogen diethylnitrosoamine (DEN). Knockin mice (Ccn1dm/dm) that express an integrin α6β1-binding defective CCN1 phenocopied Ccn1ΔHep mice, indicating that CCN1 acts through its α6β1 binding sites in this context. CCN1 effectively inhibits EGFR-dependent hepatocyte proliferation through integrin α6-mediated accumulation of reaction oxygen species (ROS), thereby triggering p53 activation and cell cycle block. Consequently, Ccn1dm/dm mice exhibit diminished p53 activation and elevated compensatory hepatocyte proliferation, resulting in increased HCC. Furthermore, we show that a single dose of the EGFR inhibitor erlotinib delivered prior to DEN-induced injury was sufficient to block compensatory proliferation and annihilate development of HCC nodules observed 8 months later, suggesting potential chemoprevention by targeting CCN1-inhibitable EGFR-dependent hepatocyte proliferation. Together, these results show that CCN1 is an injury response protein that functions not only to restrict fibrosis in the liver, but also to suppress hepatocarcinogenesis by inhibiting EGFR-dependent hepatocyte compensatory proliferation.
Collapse
Affiliation(s)
- C-C Chen
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - K-H Kim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - L F Lau
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
64
|
Takaguri A, Kamato M, Satoh Y, Ohtsuki K, Satoh K. Effect of alteration of caveolin-1 expression on doxorubicin-induced apoptosis in H9c2 cardiac cells. Cell Biol Int 2015; 39:1053-60. [DOI: 10.1002/cbin.10478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 03/31/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Akira Takaguri
- Division of Pharmacology; Hokkaido Pharmaceutical University School of Pharmacy; 7-1 Katsuraoka Otaru 047-0264 Japan
| | - Maiko Kamato
- Division of Pharmacology; Hokkaido Pharmaceutical University School of Pharmacy; 7-1 Katsuraoka Otaru 047-0264 Japan
| | - Yoshiaki Satoh
- Division of Pharmacology; Hokkaido Pharmaceutical University School of Pharmacy; 7-1 Katsuraoka Otaru 047-0264 Japan
| | - Kazuaki Ohtsuki
- Division of Pharmacology; Hokkaido Pharmaceutical University School of Pharmacy; 7-1 Katsuraoka Otaru 047-0264 Japan
| | - Kumi Satoh
- Division of Pharmacology; Hokkaido Pharmaceutical University School of Pharmacy; 7-1 Katsuraoka Otaru 047-0264 Japan
| |
Collapse
|
65
|
Clark R, Lee J, Lee SH. Synergistic anticancer activity of capsaicin and 3,3'-diindolylmethane in human colorectal cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:4297-4304. [PMID: 25876645 DOI: 10.1021/jf506098s] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. A promising area of cancer research is focused on chemoprevention by nutritional compounds. Epidemiological studies have shown a strong negative correlation between fruit, vegetable, and spice intake and rates of cancer. Although individual active compounds have demonstrated significant anticancer activity, an emerging area of research is focusing on the combination of multiple dietary compounds that act synergistically on cancer to exert greater effects. The current study evaluated the potential synergistic effects of capsaicin, an active compound from red chili peppers, in combination with 3,3'-diindolylmethane (DIM), from cruciferous vegetables. A synergistic induction of apoptosis and inhibition of cell proliferation was observed in human colorectal cancer cells treated with the combination of capsaicin and DIM. It was also observed that these two compounds activated transcriptional activity of NF-κB and p53 synergistically. Combination treatment stabilized nuclear p53 and up- or down-regulated expression of several target genes that are downstream of NF-κB and p53. The present study suggests capsaicin and DIM work synergistically to inhibit cell proliferation and induce apoptosis in colorectal cancer through modulating transcriptional activity of NF-κB, p53, and target genes associated with apoptosis.
Collapse
Affiliation(s)
- Ruth Clark
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| | - Jihye Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
66
|
Kurotani R, Shima R, Miyano Y, Sakahara S, Matsumoto Y, Shibata Y, Abe H, Kimura S. SCGB3A2 Inhibits Acrolein-Induced Apoptosis through Decreased p53 Phosphorylation. Acta Histochem Cytochem 2015; 48:61-8. [PMID: 26019375 PMCID: PMC4427566 DOI: 10.1267/ahc.14065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/21/2015] [Indexed: 12/02/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a major global health problem with increasing morbidity and mortality rates, is anticipated to become the third leading cause of death worldwide by 2020. COPD arises from exposure to cigarette smoke. Acrolein, which is contained in cigarette smoke, is the most important risk factor for COPD. It causes lung injury through altering apoptosis and causes inflammation by augmenting p53 phosphorylation and producing reactive oxygen species (ROS). Secretoglobin (SCGB) 3A2, a secretory protein predominantly present in the epithelial cells of the lungs and trachea, is a cytokine-like small molecule having anti-inflammatory, antifibrotic, and growth factor activities. In this study, the effect of SCGB3A2 on acrolein-related apoptosis was investigated using the mouse fibroblast cell line MLg as the first step in determining the possible therapeutic value of SCGB3A2 in COPD. Acrolein increased the production of ROS and phosphorylation of p53 and induced apoptosis in MLg cells. While the extent of ROS production induced by acrolein was not affected by SCGB3A2, p53 phosphorylation was significantly decreased by SCGB3A2. These results demonstrate that SCGB3A2 inhibited acrolein-induced apoptosis through decreased p53 phosphorylation, not altered ROS levels.
Collapse
Affiliation(s)
- Reiko Kurotani
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University
| | - Reika Shima
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University
| | - Yuki Miyano
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University
| | - Satoshi Sakahara
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University
| | - Yoshie Matsumoto
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University
| | - Yoko Shibata
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine
| | - Hiroyuki Abe
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University
| | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health
| |
Collapse
|
67
|
Chen D, Stueckle TA, Luanpitpong S, Rojanasakul Y, Lu Y, Wang L. Gene expression profile of human lung epithelial cells chronically exposed to single-walled carbon nanotubes. NANOSCALE RESEARCH LETTERS 2015; 10:12. [PMID: 25852310 PMCID: PMC4314466 DOI: 10.1186/s11671-014-0707-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/23/2014] [Indexed: 05/07/2023]
Abstract
A rapid increase in utility of engineered nanomaterials, including carbon nanotubes (CNTs), has raised a concern over their safety. Based on recent evidence from animal studies, pulmonary exposure of CNTs may lead to nanoparticle accumulation in the deep lung without effective clearance which could interact with local lung cells for a long period of time. Physicochemical similarities of CNTs to asbestos fibers may contribute to their asbestos-like carcinogenic potential after long-term exposure, which has not been well addressed. More studies are needed to identify and predict the carcinogenic potential and mechanisms for promoting their safe use. Our previous study reported a long-term in vitro exposure model for CNT carcinogenicity and showed that 6-month sub-chronic exposure of single-walled carbon nanotubes (SWCNT) causes malignant transformation of human lung epithelial cells. In addition, the transformed cells induced tumor formation in mice and exhibited an apoptosis resistant phenotype, a key characteristic of cancer cells. Although the potential role of p53 in the transformation process was identified, the underlying mechanisms of oncogenesis remain largely undefined. Here, we further examined the gene expression profile by using genome microarrays to profile molecular mechanisms of SWCNT oncogenesis. Based on differentially expressed genes, possible mechanisms of SWCNT-associated apoptosis resistance and oncogenesis were identified, which included activation of pAkt/p53/Bcl-2 signaling axis, increased gene expression of Ras family for cell cycle control, Dsh-mediated Notch 1, and downregulation of apoptotic genes BAX and Noxa. Activated immune responses were among the major changes of biological function. Our findings shed light on potential molecular mechanisms and signaling pathways involved in SWCNT oncogenic potential.
Collapse
Affiliation(s)
- Dongquan Chen
- />Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Todd A Stueckle
- />Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505 USA
| | - Sudjit Luanpitpong
- />Department of Pharmaceutical Sciences and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506 USA
| | - Yon Rojanasakul
- />Department of Pharmaceutical Sciences and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506 USA
| | - Yongju Lu
- />Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201 USA
| | - Liying Wang
- />Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505 USA
| |
Collapse
|
68
|
Tidball AM, Bryan MR, Uhouse MA, Kumar KK, Aboud AA, Feist JE, Ess KC, Neely MD, Aschner M, Bowman AB. A novel manganese-dependent ATM-p53 signaling pathway is selectively impaired in patient-based neuroprogenitor and murine striatal models of Huntington's disease. Hum Mol Genet 2014; 24:1929-44. [PMID: 25489053 DOI: 10.1093/hmg/ddu609] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The essential micronutrient manganese is enriched in brain, especially in the basal ganglia. We sought to identify neuronal signaling pathways responsive to neurologically relevant manganese levels, as previous data suggested that alterations in striatal manganese handling occur in Huntington's disease (HD) models. We found that p53 phosphorylation at serine 15 is the most responsive cell signaling event to manganese exposure (of 18 tested) in human neuroprogenitors and a mouse striatal cell line. Manganese-dependent activation of p53 was severely diminished in HD cells. Inhibitors of ataxia telangiectasia mutated (ATM) kinase decreased manganese-dependent phosphorylation of p53. Likewise, analysis of ATM autophosphorylation and additional ATM kinase targets, H2AX and CHK2, support a role for ATM in the activation of p53 by manganese and that a defect in this process occurs in HD. Furthermore, the deficit in Mn-dependent activation of ATM kinase in HD neuroprogenitors was highly selective, as DNA damage and oxidative injury, canonical activators of ATM, did not show similar deficits. We assessed cellular manganese handling to test for correlations with the ATM-p53 pathway, and we observed reduced Mn accumulation in HD human neuroprogenitors and HD mouse striatal cells at manganese exposures associated with altered p53 activation. To determine if this phenotype contributes to the deficit in manganese-dependent ATM activation, we used pharmacological manipulation to equalize manganese levels between HD and control mouse striatal cells and rescued the ATM-p53 signaling deficit. Collectively, our data demonstrate selective alterations in manganese biology in cellular models of HD manifest in ATM-p53 signaling.
Collapse
Affiliation(s)
| | | | | | | | - Asad A Aboud
- Department of Neurology, Vanderbilt Brain Institute
| | | | - Kevin C Ess
- Department of Neurology, Vanderbilt Brain Institute, Department of Pediatrics, Vanderbilt Kennedy Center, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - M Diana Neely
- Department of Neurology, Vanderbilt Brain Institute, Vanderbilt Kennedy Center, Vanderbilt Center in Molecular Toxicology
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt Brain Institute, Department of Pediatrics, Vanderbilt Kennedy Center, Vanderbilt Center in Molecular Toxicology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
69
|
Kang MA, Kim MS, Kim JY, Shin YJ, Song JY, Jeong JH. A novel pyrido-thieno-pyrimidine derivative activates p53 through induction of phosphorylation and acetylation in colorectal cancer cells. Int J Oncol 2014; 46:342-50. [PMID: 25338966 DOI: 10.3892/ijo.2014.2720] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/03/2014] [Indexed: 11/06/2022] Open
Abstract
The tumor suppressor p53 plays a key role in regulation of the cell cycle, apoptosis and senescence in response to various stresses. We screened a library of 7920 chemical compounds for the p53 activator and identified N-[2-(dimethylamino)ethyl]-2,3-dimethyl-4-oxo-4H-pyrido[1,2-a]thieno[2,3-d]pyrimidine-9-carboxamide (PTP), which significantly increased p53-mediated reporter activity in colorectal cancer cells. PTP was found to induce p53 protein and activated transcription of downstream genes, such as p21 and PUMA, in HCT116 cells, leading to growth delay, G1-phase cell cycle arrest, cell senescence and cell death. Proximity ligation assay revealed that PTP weakened the interaction between p53 and murine double minute 2 (MDM2) in situ, thereby inhibiting MDM2-mediated p53 degradation. Although DNA damage has been known to promote phosphorylation of p53 and MDM2, thereby preventing their interaction and stabilizing p53, PTP did not cause DNA damage or activate any DNA damage response signaling. Instead, phosphorylation of p53 was mediated by Erk1/2 MAP kinase. In addition, PTP induced acetylation of p53 at Lys382 in a p300-dependent manner, but sirtuin (SIRT)1 and histone deacetylase (HDAC)1, a well-known p53-regulating deacetylase, were not involved. In the present study, the novel anticancer agent PTP was shown to cause the accumulation of p53 by inducing multiple post-translational modifications, as well as cell cycle arrest, senescence and cell death.
Collapse
Affiliation(s)
- Mi Ae Kang
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Mi-Sook Kim
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Ji Young Kim
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Young-Joo Shin
- Department of Radiation Oncology, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Jie-Young Song
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jae-Hoon Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| |
Collapse
|
70
|
Wu J, Song T, Liu S, Li X, Li G, Xu J. Icariside II inhibits cell proliferation and induces cell cycle arrest through the ROS-p38-p53 signaling pathway in A375 human melanoma cells. Mol Med Rep 2014; 11:410-6. [PMID: 25333296 DOI: 10.3892/mmr.2014.2701] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 07/23/2014] [Indexed: 11/05/2022] Open
Abstract
Icariside II (IS) is a metabolite of icariin, which is derived from Herba Epimedii. In the present study, the antiproliferative effects of IS on A375 human melanoma cells were examined in vitro and a possible mechanism through the ROS-p38-p53 pathway is discussed. A cell WST-8 assay revealed that treatment with IS markedly reduced cell viability from 77 to 21% (25 and 100 µM, respectively), and cell counting demonstrated that IS treatment reduced cell proliferation. IS treatment also induced cell cycle arrest of A375 cells at the G0/G1 and G2/M transitions and inhibited the expression of cell-cycle related proteins, including cyclin E, cyclin-dependent kinase 2 (CDK2), cyclin B1 and phosphorylated cyclin-dependent kinase 1 (P-CDK1). In this study, it was determined that IS inhibits cell proliferation and induces cell cycle arrest through the generation of reactive oxygen species and activation of p38 and p53. These findings were further supported by the evidence that pretreatment with N-acetyl-L-cysteine, SB203580 or pifithrin-α significantly blocked IS-induced reduction of cell viability, increase of cell death and cell cycle arrest. In conclusion, IS inhibits cell proliferation and induces cell cycle arrest. Crucially, it was confirmed that these effects were mediated at least in part by activating the ROS-p38-p53 pathway.
Collapse
Affiliation(s)
- Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, P.R. China
| | - Tao Song
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 25002, P.R. China
| | - Shuyong Liu
- Department of Radiology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xiaomei Li
- Cancer Center, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Gang Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, P.R. China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, P.R. China
| |
Collapse
|
71
|
Parthasarathy G, Philipp MT. The MEK/ERK pathway is the primary conduit for Borrelia burgdorferi-induced inflammation and P53-mediated apoptosis in oligodendrocytes. Apoptosis 2014; 19:76-89. [PMID: 24114360 DOI: 10.1007/s10495-013-0913-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lyme neuroborreliosis (LNB) affects both the central and peripheral nervous systems. In a rhesus macaque model of LNB we had previously shown that brains of rhesus macaques inoculated with Borrelia burgdorferi release inflammatory mediators, and undergo oligodendrocyte and neuronal cell death. In vitro analysis of this phenomenon indicated that while B. burgdorferi can induce inflammation and apoptosis of oligodendrocytes per se, microglia are required for neuronal apoptosis. We hypothesized that the inflammatory milieu elicited by the bacterium in microglia or oligodendrocytes contributes to the apoptosis of neurons and glial cells, respectively, and that downstream signaling events in NFkB and/or MAPK pathways play a role in these phenotypes. To test these hypotheses in oligodendrocytes, several pathway inhibitors were used to determine their effect on inflammation and apoptosis, as induced by B. burgdorferi. In a human oligodendrocyte cell line (MO3.13), inhibition of the ERK pathway in the presence of B. burgdorferi markedly reduced inflammation, followed by the JNK, p38 and NFkB pathway inhibition. In addition to eliciting inflammation, B. burgdorferi also increased total p53 protein levels, and suppression of the ERK pathway mitigated this effect. While inhibition of p53 had a minimal effect in reducing inflammation, suppression of the ERK pathway or p53 reduced apoptosis as measured by active caspase-3 activity and the TUNEL assay. A similar result was seen in primary human oligodendrocytes wherein suppression of ERK or p53 reduced apoptosis. It is possible that inflammation and apoptosis in oligodendrocytes are divergent arms of MAPK pathways, particularly the MEK/ERK pathway.
Collapse
Affiliation(s)
- Geetha Parthasarathy
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, 18703, Three Rivers Road, Covington, LA, 70433, USA
| | | |
Collapse
|
72
|
Yang J, Bill MA, Young GS, La Perle K, Landesman Y, Shacham S, Kauffman M, Senapedis W, Kashyap T, Saint-Martin JR, Kendra K, Lesinski GB. Novel small molecule XPO1/CRM1 inhibitors induce nuclear accumulation of TP53, phosphorylated MAPK and apoptosis in human melanoma cells. PLoS One 2014; 9:e102983. [PMID: 25057921 PMCID: PMC4109950 DOI: 10.1371/journal.pone.0102983] [Citation(s) in RCA: 38] [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: 01/27/2014] [Accepted: 06/25/2014] [Indexed: 11/26/2022] Open
Abstract
XPO1/CRM1 is a key nuclear exporter protein that mediates translocation of numerous cellular regulatory proteins. We investigated whether XPO1 is a potential therapeutic target in melanoma using novel selective inhibitors of nuclear export (SINE). In vitro effects of SINE on cell growth and apoptosis were measured by MTS assay and flow cytometry [Annexin V/propidium iodide (PI)], respectively in human metastatic melanoma cell lines. Immunoblot analysis was used to measure nuclear localization of key cellular proteins. The in vivo activity of oral SINE was evaluated in NOD/SCID mice bearing A375 or CHL-1 human melanoma xenografts. SINE compounds induced cytostatic and pro-apoptotic effects in both BRAF wild type and mutant (V600E) cell lines at nanomolar concentrations. The cytostatic and pro-apoptotic effects of XPO1 inhibition were associated with nuclear accumulation of TP53, and CDKN1A induction in the A375 cell line with wild type TP53, while pMAPK accumulated in the nucleus regardless of TP53 status. The orally bioavailable KPT-276 and KPT-330 compounds significantly inhibited growth of A375 (p<0.0001) and CHL-1 (p = 0.0087) human melanoma cell lines in vivo at well tolerated doses. Inhibition of XPO1 using SINE represents a potential therapeutic approach for melanoma across cells with diverse molecular phenotypes by promoting growth inhibition and apoptosis.
Collapse
Affiliation(s)
- Jennifer Yang
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew A. Bill
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Gregory S. Young
- Center for Biostatistics, The Ohio State University, Columbus, Ohio, United States of America
| | - Krista La Perle
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Yosef Landesman
- Karyopharm Therapeutics, Natick, Massachusetts, United States of America
| | - Sharon Shacham
- Karyopharm Therapeutics, Natick, Massachusetts, United States of America
| | - Michael Kauffman
- Karyopharm Therapeutics, Natick, Massachusetts, United States of America
| | - William Senapedis
- Karyopharm Therapeutics, Natick, Massachusetts, United States of America
| | - Trinayan Kashyap
- Karyopharm Therapeutics, Natick, Massachusetts, United States of America
| | | | - Kari Kendra
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Gregory B. Lesinski
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
73
|
Toutounchian JJ, Steinle JJ, Makena PS, Waters CM, Wilson MW, Haik BG, Miller DD, Yates CR. Modulation of radiation injury response in retinal endothelial cells by quinic acid derivative KZ-41 involves p38 MAPK. PLoS One 2014; 9:e100210. [PMID: 24956278 PMCID: PMC4067294 DOI: 10.1371/journal.pone.0100210] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/22/2014] [Indexed: 01/09/2023] Open
Abstract
Radiation-induced damage to the retina triggers leukostasis, retinal endothelial cell (REC) death, and subsequent hypoxia. Resultant ischemia leads to visual loss and compensatory retinal neovascularization (RNV). Using human RECs, we demonstrated that radiation induced leukocyte adhesion through mechanisms involving p38MAPK, p53, and ICAM-1 activation. Additional phenotypic changes included p38MAPK-dependent tyrosine phosphorylation of the focal adhesion scaffolding protein, paxillin (Tyr118). The quinic acid derivative KZ-41 lessened leukocyte adhesion and paxillin-dependent proliferation via inhibition of p38MAPK-p53-ICAM-1 signaling. Using the murine oxygen-induced retinopathy (OIR) model, we examined the effect of KZ-41 on pathologic RNV. Daily ocular application of a KZ-41-loaded nanoemulsion significantly reduced both the avascular and neovascular areas in harvested retinal flat mounts when compared to the contralateral eye receiving vehicle alone. Our data highlight the potential benefit of KZ-41 in reducing both the retinal ischemia and neovascularization provoked by genotoxic insults. Further research into how quinic acid derivatives target and mitigate inflammation is needed to fully appreciate their therapeutic potential for the treatment of inflammatory retinal vasculopathies.
Collapse
Affiliation(s)
- Jordan J. Toutounchian
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Jena J. Steinle
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Patrudu S. Makena
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Christopher M. Waters
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Matthew W. Wilson
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Barrett G. Haik
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Duane D. Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Charles R. Yates
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
74
|
Song Y, Li X, Li Y, Li N, Shi X, Ding H, Zhang Y, Li X, Liu G, Wang Z. Non-esterified fatty acids activate the ROS-p38-p53/Nrf2 signaling pathway to induce bovine hepatocyte apoptosis in vitro. Apoptosis 2014; 19:984-997. [PMID: 24699798 DOI: 10.1007/s10495-014-0982-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A high plasma concentration of non-esterified fatty acids (NEFAs) is an important pathogenic factor that leads to ketosis and fatty liver in dairy cows. NEFAs may be associated with oxidative stress in dairy cows with ketosis or fatty liver and the subsequent induction of hepatocyte damage. However, the molecular mechanism of NEFAs-induced oxidative stress and whether NEFAs cause apoptosis of hepatocytes are unclear. Therefore, the aim of this study was to investigate the molecular mechanism of NEFAs-induced oxidative liver damage in bovine hepatocytes. The results showed that NEFAs increased oxidative stress, resulting in p38 phosphorylation. High activated p38 increased the expression, nuclear localization and transcriptional activity of p53 and decreased the nuclear localization and transcriptional activity of Nrf2 in bovine hepatocytes treated with high concentrations of NEFAs. High concentrations of NEFAs also promoted the apoptosis of bovine hepatocytes. Both N-acetyl-L-cysteine (NAC) and glucose (GLU) could attenuate the NEFA-induced apoptotic damage. These results indicate that NEFAs activate the ROS-p38-p53/Nrf2 signaling pathway to induce apoptotic damage in bovine hepatocytes.
Collapse
Affiliation(s)
- Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, 130062, Jilin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Zhu W, Cromie MM, Cai Q, Lv T, Singh K, Gao W. Curcumin and vitamin E protect against adverse effects of benzo[a]pyrene in lung epithelial cells. PLoS One 2014; 9:e92992. [PMID: 24664296 PMCID: PMC3963982 DOI: 10.1371/journal.pone.0092992] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/27/2014] [Indexed: 11/18/2022] Open
Abstract
Benzo[a]pyrene (BaP), a well-known environmental carcinogen, promotes oxidative stress and DNA damage. Curcumin and vitamin E (VE) have potent antioxidative activity that protects cells from oxidative stress and cellular damage. The objectives of the present study were to investigate the adverse effects of BaP on normal human lung epithelial cells (BEAS-2B), the potential protective effects of curcumin and VE against BaP-induced cellular damage, and the molecular mechanisms of action. MTT assay, flow cytometry, fluorescence microplate assay, HPLC, qRT-PCR, and western blot were performed to analyze cytotoxicity, cell cycle, reactive oxygen species (ROS), BaP diol-epoxidation (BPDE)-DNA adducts, gene expression, and protein expression, respectively. Curcumin or VE prevented cells from BaP-induced cell cycle arrest and growth inhibition, significantly suppressed BaP-induced ROS levels, and decreased BPDE-DNA adducts. While CYP1A1 and 1B1 were induced by BaP, these inductions were not significantly reduced by curcumin or VE. Moreover, the level of activated p53 and PARP-1 were significantly induced by BaP, whereas this induction was markedly reduced after curcumin and VE co-treatment. Survivin was significantly down-regulated by BaP, and curcumin significantly restored survivin expression in BaP-exposed cells. The ratio of Bax/Bcl-2 was also significantly increased in cells exposed to BaP and this increase was reversed by VE co-treatment. Taken together, BaP-induced cytotoxicity occurs through DNA damage, cell cycle arrest, ROS production, modulation of metabolizing enzymes, and the expression/activation of p53, PARP-1, survivin, and Bax/Bcl-2. Curcumin and VE could reverse some of these BaP-mediated alterations and therefore be effective natural compounds against the adverse effects of BaP in lung cells.
Collapse
Affiliation(s)
- Wenbin Zhu
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, Texas, United States of America
| | - Meghan M. Cromie
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, Texas, United States of America
| | - Qingsong Cai
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, Texas, United States of America
| | - Tangfeng Lv
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, Texas, United States of America
| | - Kamaleshwar Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, Texas, United States of America
| | - Weimin Gao
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
76
|
Akhter R, Sanphui P, Biswas SC. The essential role of p53-up-regulated modulator of apoptosis (Puma) and its regulation by FoxO3a transcription factor in β-amyloid-induced neuron death. J Biol Chem 2014; 289:10812-10822. [PMID: 24567336 DOI: 10.1074/jbc.m113.519355] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Neurodegeneration underlies the pathology of Alzheimer disease (AD). The molecules responsible for such neurodegeneration in AD brain are mostly unknown. Recent findings indicate that the BH3-only proteins of the Bcl-2 family play an essential role in various cell death paradigms, including neurodegeneration. Here we report that Puma (p53-up-regulated modulator of apoptosis), an important member of the BH3-only protein family, is up-regulated in neurons upon toxic β-amyloid 1-42 (Aβ(1-42)) exposure both in vitro and in vivo. Down-regulation of Puma by specific siRNA provides significant protection against neuron death induced by Aβ(1-42). We further demonstrate that the activation of p53 and inhibition of PI3K/Akt pathways induce Puma. The transcription factor FoxO3a, which is activated when PI3K/Akt signaling is inhibited, directly binds with the Puma gene and induces its expression upon exposure of neurons to oligomeric Aβ(1-42). Moreover, Puma cooperates with another BH3-only protein, Bim, which is already implicated in AD. Our results thus suggest that Puma is activated by both p53 and PI3K/Akt/FoxO3a pathways and cooperates with Bim to induce neuron death in response to Aβ(1-42).
Collapse
Affiliation(s)
- Rumana Akhter
- Cell Biology and Physiology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India
| | - Priyankar Sanphui
- Cell Biology and Physiology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India
| | - Subhas Chandra Biswas
- Cell Biology and Physiology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700 032, India.
| |
Collapse
|
77
|
Tang WH, Stitham J, Jin Y, Liu R, Lee SH, Du J, Atteya G, Gleim S, Spollett G, Martin K, Hwa J. Aldose reductase-mediated phosphorylation of p53 leads to mitochondrial dysfunction and damage in diabetic platelets. Circulation 2014; 129:1598-609. [PMID: 24474649 DOI: 10.1161/circulationaha.113.005224] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Platelet abnormalities are well-recognized complications of diabetes mellitus. Mitochondria play a central role in platelet metabolism and activation. Mitochondrial dysfunction is evident in diabetes mellitus. The molecular pathway for hyperglycemia-induced mitochondrial dysfunction in platelets in diabetes mellitus is unknown. METHODS AND RESULTS Using both human and humanized mouse models, we report that hyperglycemia-induced aldose reductase activation and subsequent reactive oxygen species production lead to increased p53 phosphorylation (Ser15), which promotes mitochondrial dysfunction, damage, and rupture by sequestration of the antiapoptotic protein Bcl-xL. In a glucose dose-dependent manner, severe mitochondrial damage leads to loss of mitochondrial membrane potential and platelet apoptosis (cytochrome c release, caspase 3 activation, and phosphatidylserine exposure). Although platelet hyperactivation, mitochondrial dysfunction, aldose reductase activation, reactive oxygen species production, and p53 phosphorylation are all induced by hyperglycemia, we demonstrate that platelet apoptosis and hyperactivation are 2 distinct states that depend on the severity of the hyperglycemia and mitochondrial damage. Combined, both lead to increased thrombus formation in a mouse blood stasis model. CONCLUSIONS Aldose reductase contributes to diabetes-mediated mitochondrial dysfunction and damage through the activation of p53. The degree of mitochondrial dysfunction and damage determines whether hyperactivity (mild damage) or apoptosis (severe damage) will ensue. These signaling components provide novel therapeutic targets for thrombotic complications in diabetes mellitus.
Collapse
Affiliation(s)
- Wai Ho Tang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (W.H.T., J.S., Y.J., R.L., S.H.L., J.D., G.A., S.G., K.M., J.H.) and Section of Endocrinology and Metabolism, Department of Internal Medicine (G.S.), Yale University School of Medicine, New Haven, CT
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Karabulut S, Demiroğlu-Zergeroğlu A, Yılmaz E, Sağır F, Delikara N. p53 and mitogen-activated protein kinase pathway protein profiles in fresh and frozen spermatozoa. Andrologia 2013; 46:1113-7. [DOI: 10.1111/and.12200] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2013] [Indexed: 11/28/2022] Open
Affiliation(s)
- S. Karabulut
- Department of Molecular Biology and Genetics; Gebze Institute of Technology; Kocaeli Turkey
- Florence Nightingale Hospital; Center for Reproductive Medicine; Istanbul Turkey
| | | | - E. Yılmaz
- Florence Nightingale Hospital; Center for Reproductive Medicine; Istanbul Turkey
| | - F. Sağır
- Department of Molecular Biology and Genetics; Gebze Institute of Technology; Kocaeli Turkey
| | - N. Delikara
- Florence Nightingale Hospital; Center for Reproductive Medicine; Istanbul Turkey
| |
Collapse
|
79
|
Wentilactone A as a novel potential antitumor agent induces apoptosis and G2/M arrest of human lung carcinoma cells, and is mediated by HRas-GTP accumulation to excessively activate the Ras/Raf/ERK/p53-p21 pathway. Cell Death Dis 2013; 4:e952. [PMID: 24309939 PMCID: PMC3877555 DOI: 10.1038/cddis.2013.484] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/30/2013] [Accepted: 11/04/2013] [Indexed: 02/06/2023]
Abstract
Chemotherapy remains the common therapeutic for patients with lung cancer. Novel, selective antitumor agents are pressingly needed. This study is the first to investigate a different, however, effective antitumor drug candidate Wentilactone A (WA) for its development as a novel agent. In NCI-H460 and NCI-H446 cell lines, WA triggered G2/M phase arrest and mitochondrial-related apoptosis, accompanying the accumulation of reactive oxygen species (ROS). It also induced activation of mitogen-activated protein kinase and p53 and increased expression of p21. When we pre-treated cells with ERK, JNK, p38, p53 inhibitor or NAC followed by WA treatment, only ERK and p53 inhibitors blocked WA-induced apoptosis and G2/M arrest. We further observed Ras (HRas, KRas and NRas) and Raf activation, and found that WA treatment increased HRas–Raf activation. Knockdown of HRas by using small interfering RNA (siRNA) abolished WA-induced apoptosis and G2/M arrest. HRas siRNA also halted Raf, ERK, p53 activation and p21 accumulation. Molecular docking analysis suggested that WA could bind to HRas-GTP, causing accumulation of Ras-GTP and excessive activation of Raf/ERK/p53-p21. The direct binding affinity was confirmed by surface plasmon resonance (SPR). In vivo, WA suppressed tumor growth without adverse toxicity and presented the same mechanism as that in vitro. Taken together, these findings suggest WA as a promising novel, potent and selective antitumor drug candidate for lung cancer.
Collapse
|
80
|
Saleem A, Carter HN, Hood DA. p53 is necessary for the adaptive changes in cellular milieu subsequent to an acute bout of endurance exercise. Am J Physiol Cell Physiol 2013; 306:C241-9. [PMID: 24284795 DOI: 10.1152/ajpcell.00270.2013] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An acute bout of exercise activates downstream signaling cascades that ultimately result in mitochondrial biogenesis. In addition to inducing mitochondrial synthesis, exercise triggers the removal of damaged cellular material via autophagy and of dysfunctional mitochondria through mitophagy. Here, we investigated the necessity of p53 to the changes that transpire within the muscle upon an imposed metabolic and physiological challenge, such as a bout of endurance exercise. We randomly assigned wild-type (WT) and p53 knockout (KO) mice to control, acute exercise (AE; 90 min at 15 m/min), and AE + 3 h recovery (AER) groups and measured downstream alterations in markers of mitochondrial biogenesis, autophagy, and mitophagy. In the absence of p53, activation of p38 MAPK upon exercise was abolished, whereas CaMKII and AMP-activated protein kinase only displayed an attenuated enhancement in the AER group compared with WT mice. The translocation of peroxisome proliferator-activated receptor-γ coactivator-1 α to the nucleus was diminished and only observed in the AER group, and the subsequent increase in messenger RNA transcripts related to mitochondrial biogenesis with exercise and recovery was absent in the p53 KO animals. Whole-muscle autophagic and lysosomal markers did not respond to exercise, irrespective of the genotype of the exercised mice, with the exception of increased ubiquitination observed in KO mice with exercise. Markers of mitophagy were elevated in response to AE and AER conditions in both WT and p53 KO runners. The data suggest that p53 is important for the exercise-induced activation of mitochondrial synthesis and is integral in regulating autophagy during control conditions but not in response to exercise.
Collapse
Affiliation(s)
- Ayesha Saleem
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | | | | |
Collapse
|
81
|
MacNeil AJ, Jiao SC, McEachern LA, Yang YJ, Dennis A, Yu H, Xu Z, Marshall JS, Lin TJ. MAPK kinase 3 is a tumor suppressor with reduced copy number in breast cancer. Cancer Res 2013; 74:162-72. [PMID: 24233520 DOI: 10.1158/0008-5472.can-13-1310] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancers are initiated as a result of changes that occur in the genome. Identification of gains and losses in the structure and expression of tumor-suppressor genes and oncogenes lies at the root of the understanding of cancer cell biology. Here, we show that the mitogen-activated protein kinase (MAPK) MKK3 suppresses the growth of breast cancer, in which it varies in copy number. A pervasive loss of MKK3 gene copy number in patients with breast cancer is associated with an impairment of MKK3 expression and protein level in malignant tissues. To assess the functional role of MKK3 in breast cancer, we showed in an animal model that MKK3 activity is required for suppression of tumor growth. Active MKK3 enhanced expression of the cyclin-dependent kinase inhibitors p21(Cip1/Waf1) and p27(Kip1), leading to increased cell-cycle arrest in G1 phase of the cell cycle. Our results reveal the functional significance of MKK3 as a tumor suppressor and improve understanding of the dynamic role of the MAPK pathway in tumor progression.
Collapse
Affiliation(s)
- Adam J MacNeil
- Authors' Affiliations: Departments of Microbiology and Immunology, Pediatrics, Physiology and Biophysics, and Pathology, Dalhousie University; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada; Department of Medical Oncology, General Hospital of the People's Liberation Army, Beijing; and Institute of Zoonosis, College of Animal Sciences and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Liu H, Hwang J, Li W, Choi TW, Liu K, Huang Z, Jang JH, Thimmegowda NR, Lee KW, Ryoo IJ, Ahn JS, Bode AM, Zhou X, Yang Y, Erikson RL, Kim BY, Dong Z. A derivative of chrysin suppresses two-stage skin carcinogenesis by inhibiting mitogen- and stress-activated kinase 1. Cancer Prev Res (Phila) 2013; 7:74-85. [PMID: 24169959 DOI: 10.1158/1940-6207.capr-13-0133] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mitogen- and stress-activated kinase 1 (MSK1) is a nuclear serine/threonine protein kinase that acts downstream of both extracellular signal-regulated kinases and p38 mitogen-activated protein kinase in response to stress or mitogenic extracellular stimuli. Increasing evidence has shown that MSK1 is closely associated with malignant transformation and cancer development. MSK1 should be an effective target for cancer chemoprevention and chemotherapy. However, very few MSK1 inhibitors, especially natural compounds, have been reported. We used virtual screening of a natural products database and the active conformation of the C-terminal kinase domain of MSK1 (PDB id 3KN) as the receptor structure to identify chrysin and its derivative, compound 69407, as inhibitors of MSK1. Compared with chrysin, compound 69407 more strongly inhibited proliferation and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced neoplastic transformation of JB6 P+ cells with lower cytotoxicity. Western blot data demonstrated that compound 69407 suppressed phosphorylation of the MSK1 downstream effector histone H3 in intact cells. Knocking down the expression of MSK1 effectively reduced the sensitivity of JB6 P+ cells to compound 69407. Moreover, topical treatment with compound 69407 before TPA application significantly reduced papilloma development in terms of number and size in a two-stage mouse skin carcinogenesis model. The reduction in papilloma development was accompanied by the inhibition of histone H3 phosphorylation at Ser10 in tumors extracted from mouse skin. The results indicated that compound 69407 exerts inhibitory effects on skin tumorigenesis by directly binding with MSK1 and attenuates the MSK1/histone H3 signaling pathway, which makes it an ideal chemopreventive agent against skin cancer.
Collapse
Affiliation(s)
- Haidan Liu
- University of Minnesota, 801 16th Avenue NE, Austin, MN 55912. Phone: 507-437-9600; Fax: 507-437-9606; ; and Bo-Yeon Kim, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 363-883, Republic of Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Nakata Y, Nishi K, Nishimoto S, Sugahara T. Phenylhydroquinone induces loss of thymocytes through cell cycle arrest and apoptosis elevation in p53-dependent pathway. J Toxicol Sci 2013; 38:325-35. [PMID: 23665931 DOI: 10.2131/jts.38.325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
ortho-Phenylphenol has been employed in post-harvest treatment of citrus fruits. Although o-phenylphenol has been reported to cause carcinomas in the urinary tract in rats, toxicity to the immune organs is still unknown. Herein, we report that administration of o-phenylphenol induces thymic atrophy and loss of thymocytes in female BALB/c mice. The influence seems to result from inhibition of the thymocyte development, because increased and decreased populations of the CD4⁻ CD8⁻ double-negative and CD4⁺ CD8⁺ double-positive thymocytes were observed in the o-phenylphenol-administered mice, respectively. ortho-Phenylphenol is metabolized to phenylhydroquinone by cytochrome P450 monooxygenases. Phenylhydroquinone made cell cycle of thymocytes to be arrested through reduced expression of the genes associated with G₂/M phase and through phosphorylation of p53 at Ser15. Phosphorylation of p53 at Ser15 was upregulated by activation of not only ATR but also Erk1/2 and p38, leading to increase of apoptosis. Gene expression of cytochrome P450 1A1 (CYP1A1) was promoted in thymocytes from the o-phenylphenol-administered mice. Overall, our results suggest that o-phenylphenol induces CYP1A1 expression and is metabolized into phenylhydroquinone by the expressed CYP1A1 in thymocytes. The produced phenylhydroquinone in turn induces inhibition of thymocyte development through cell cycle arrest and apoptosis in the p53-dependent pathway.
Collapse
|
84
|
Choi SH, Jung SY, Yoo SY, Yoo SM, Kim DY, Kang S, Baek SH, Kwon SM. Regulation of ROS-independent ERK signaling rescues replicative cellular senescence in ex vivo expanded human c-kit-positive cardiac progenitor cells. Int J Cardiol 2013; 169:73-82. [DOI: 10.1016/j.ijcard.2013.08.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 06/14/2013] [Accepted: 08/28/2013] [Indexed: 11/16/2022]
|
85
|
ATM kinase enables the functional axis of YAP, PML and p53 to ameliorate loss of Werner protein-mediated oncogenic senescence. Cell Death Differ 2013; 20:1498-509. [PMID: 23933816 DOI: 10.1038/cdd.2013.101] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 06/11/2013] [Accepted: 07/02/2013] [Indexed: 01/01/2023] Open
Abstract
Werner syndrome (WS) results from dysfunction of the WRN protein, and is associated with premature aging and early death. Here we report that loss of WRN function elicits accumulation of the Yes-associated protein (YAP protein), a major effector of the Hippo tumor suppressor pathway, both experimentally and in WS-derived fibroblasts. YAP upregulation correlates with slower cell proliferation and accelerated senescence, which are partially mediated by the formation of a complex between YAP and the PML protein, whose activity promotes p53 activation. The ATM kinase is necessary for YAP and PML accumulation in WRN-depleted cells. Notably, the depletion of either YAP or PML partially impairs the induction of senescence following WRN loss. Altogether, our findings reveal that loss of WRN activity triggers the activation of an ATM-YAP-PML-p53 axis, thereby accelerating cellular senescence. The latter has features of SASP (senescence-associated secretory phenotype), whose protumorigenic properties are potentiated by YAP, PML and p53 depletion.
Collapse
|
86
|
Jeong JB, Yang X, Clark R, Choi J, Baek SJ, Lee SH. A mechanistic study of the proapoptotic effect of tolfenamic acid: involvement of NF-κB activation. Carcinogenesis 2013; 34:2350-60. [PMID: 23784084 DOI: 10.1093/carcin/bgt224] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent studies demonstrate that tolfenamic acid (TA) induces apoptosis and suppresses the development and progression of several types of cancers. However, the underlying mechanisms are complex and remain to be fully elucidated. Nuclear factor-kappaB (NF-κB) plays a critical role in inflammation, cancer development and progression. Although non-steroidal anti-inflammatory drugs modulate NF-κB signaling pathway in different ways, the link between NF-κB and TA-induced apoptosis of colorectal cancer cells has yet to be thoroughly investigated. In this study, we examined the effects of TA on the NF-κB pathway and apoptosis. TA activated NF-κB transcriptional activity and binding affinity of NF-κB to DNA. TA-induced NF-κB activation was mediated by an increased phosphorylation and proteosomal degradation of IκB-α and subsequent p65 nuclear translocation. We also observed that TA stabilized p65 and increased nuclear accumulation via an increase of p65 phosphorylation at Ser276 residue, which was mediated by p38 mitogen-activated protein kinase and extracellular signal-regulated kinase. The knockout of p53 blocked TA-induced transcriptional activation of NF-κB, but not the p65 nuclear accumulation. TA increased transcriptional activity of p53 and the binding affinity of p53 with p65, which are mediated by p38 mitogen-activated protein kinase and extracellular signal-regulated kinase-stimulated Ser276 phosphorylation. TA-induced apoptosis was ameliorated by the knockout of p65 and p53 and the point mutation of p65 at Ser276 residue. We demonstrate a novel molecular mechanism by which TA induced the NF-κB and apoptosis in human colorectal cancer cells.
Collapse
Affiliation(s)
- Jin Boo Jeong
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD 20742, USA and
| | | | | | | | | | | |
Collapse
|
87
|
Niero ELDO, Machado-Santelli GM. Cinnamic acid induces apoptotic cell death and cytoskeleton disruption in human melanoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:31. [PMID: 23701745 PMCID: PMC3667113 DOI: 10.1186/1756-9966-32-31] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/17/2013] [Indexed: 01/05/2023]
Abstract
Anticancer activities of cinnamic acid derivatives include induction of apoptosis by irreversible DNA damage leading to cell death. The present work aimed to compare the cytotoxic and genotoxic potential of cinnamic acid in human melanoma cell line (HT-144) and human melanocyte cell line derived from blue nevus (NGM). Viability assay showed that the IC50 for HT-144 cells was 2.4 mM, while NGM cells were more resistant to the treatment. The growth inhibition was probably associated with DNA damage leading to DNA synthesis inhibition, as shown by BrdU incorporation assay, induction of nuclear aberrations and then apoptosis. The frequency of cell death caused by cinnamic acid was higher in HT-144 cells. Activated-caspase 3 staining showed apoptosis after 24 hours of treatment with cinnamic acid 3.2 mM in HT-144 cells, but not in NGM. We observed microtubules disorganization after cinnamic acid exposure, but this event and cell death seem to be independent according to M30 and tubulin labeling. The frequency of micronucleated HT-144 cells was higher after treatment with cinnamic acid (0.4 and 3.2 mM) when compared to the controls. Cinnamic acid 3.2 mM also increased the frequency of micronucleated NGM cells indicating genotoxic activity of the compound, but the effects were milder. Binucleation and multinucleation counting showed similar results. We conclude that cinnamic acid has effective antiproliferative activity against melanoma cells. However, the increased frequency of micronucleation in NGM cells warrants the possibility of genotoxicity and needs further investigation.
Collapse
Affiliation(s)
- Evandro Luís de Oliveira Niero
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Av, Prof, Lineu Prestes, 1524, Cidade Universitária, 05508-000 São Paulo, SP, Brazil
| | | |
Collapse
|
88
|
Saleem A, Hood DA. Acute exercise induces tumour suppressor protein p53 translocation to the mitochondria and promotes a p53-Tfam-mitochondrial DNA complex in skeletal muscle. J Physiol 2013; 591:3625-36. [PMID: 23690562 DOI: 10.1113/jphysiol.2013.252791] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The major tumour suppressor protein p53 plays an important role in maintaining mitochondrial content and function in skeletal muscle. p53 has been shown to reside in the mitochondria complexed with mitochondrial DNA (mtDNA); however, the physiological repercussions of mitochondrial p53 remain unknown. We endeavoured to elucidate whether an acute bout of endurance exercise could mediate an increase in mitochondrial p53 levels. C57Bl6 mice (n = 6 per group) were randomly assigned to sedentary, acute exercise (AE, 15 m min(-1) for 90 min) or acute exercise + 3 h recovery (AER) groups. Exercise concomitantly increased the mRNA content of nuclear-encoded (PGC-1α, Tfam, NRF-1, COX-IV, citrate synthase) and mtDNA-encoded (COX-I) genes in the AE group, and further by ∼5-fold in the AER group. Nuclear p53 protein levels were reduced in the AE and AER groups, while in contrast, the abundance of p53 was drastically enhanced by ∼2.4-fold and ∼3.9-fold in subsarcolemmal and intermyofibrillar mitochondria, respectively, in the AER conditions. Within the mitochondria, the interaction of p53 with mtDNA at the D-loop and with Tfam was elevated by ∼4.6-fold and ∼3.6-fold, respectively, in the AER group. In the absence of p53, the enhanced COX-I mRNA content observed with AE and AER was abrogated. This study is the first to indicate that endurance exercise can signal to localize p53 to the mitochondria where it may serve to positively modulate the activity of the mitochondrial transcription factor Tfam. Our findings help us understand the mechanisms underlying the effects of exercise as a therapeutic intervention designed to trigger the pro-metabolic functions of p53.
Collapse
Affiliation(s)
- Ayesha Saleem
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | | |
Collapse
|
89
|
Flores-López LA, Díaz-Flores M, García-Macedo R, Ávalos-Rodríguez A, Vergara-Onofre M, Cruz M, Contreras-Ramos A, Konigsberg M, Ortega-Camarillo C. High glucose induces mitochondrial p53 phosphorylation by p38 MAPK in pancreatic RINm5F cells. Mol Biol Rep 2013; 40:4947-58. [PMID: 23657598 DOI: 10.1007/s11033-013-2595-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 04/29/2013] [Indexed: 01/09/2023]
Abstract
Pancreatic β-cell death in type 2 diabetes has been related to p53 subcellular localisation and phosphorylation. However, the mechanisms by which p53 is phosphorylated and its activation in response to oxidative stress remain poorly understood. Therefore, the aim of this study was to investigate mitochondrial p53 phosphorylation, its subcellular localisation and its relationship with apoptotic induction in RINm5F cells cultured under high glucose conditions. Our results show that p53 phosphorylation in the mitochondrial fraction was greater at ser392 than at ser15. This increased phosphorylation correlated with an increase in reactive oxygen species, a decrease in the Bcl-2/Bax ratio, a release of cytochrome c and an increase in the rate of apoptosis. We also observed a decline in ERK 1/2 phosphorylation over time, which is an indicator of cell proliferation. To identify the kinase responsible for phosphorylating p53, p38 mitogen-activated protein kinase (MAPK) activation was analysed. We found that high glucose induced an increase in p38 MAPK phosphorylation in the mitochondria after 24-72 h. Moreover, the phosphorylation of p53 (ser392) by p38 MAPK in mitochondria was confirmed by colocalisation studies with confocal microscopy. The addition of a specific p38 MAPK inhibitor (SB203580) to the culture medium during high glucose treatment blocked p53 mobilisation to the mitochondria and phosphorylation; thus, the release of cytochrome c and the apoptosis rate in RINm5F cells decreased. These results suggest that mitochondrial p53 phosphorylation by p38 MAPK plays an important role in RINm5F cell death under high glucose conditions.
Collapse
Affiliation(s)
- Luis A Flores-López
- Unidad de Investigación Médica en Bioquímica, HE, Centro Médico Nacional Siglo XXI. IMSS., Av. Cuauhtémoc 330, Col Doctores, Del. Cuauhtémoc, México, DF, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Taylor CA, Zheng Q, Liu Z, Thompson JE. Role of p38 and JNK MAPK signaling pathways and tumor suppressor p53 on induction of apoptosis in response to Ad-eIF5A1 in A549 lung cancer cells. Mol Cancer 2013; 12:35. [PMID: 23638878 PMCID: PMC3660295 DOI: 10.1186/1476-4598-12-35] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/17/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The eukaryotic translation initiation factor 5A1 (eIF5A1) is a highly conserved protein involved in many cellular processes including cell division, translation, apoptosis, and inflammation. Induction of apoptosis is the only function of eIF5A1 that is known to be independent of post-translational hypusine modification. In the present study, we investigated the involvement of mitogen- and stress-activated protein kinases during apoptosis of A549 lung cancer cells infected with adenovirus expressing eIF5A1 or a mutant of eIF5A1 that cannot be hypusinated (eIF5A1K50A). METHODS Using adenoviral-mediated transfection of human A549 lung cancer cells to over-express eIF5A1 and eIF5A1K50A, the mechanism by which unhypusinated eIF5A1 induces apoptosis was investigated by Western blotting, flow cytometry, and use of MAPK and p53 inhibitors. RESULTS Phosphorylation of ERK, p38 MAPK, and JNK was observed in response to adenovirus-mediated over-expression of eIF5A1 or eIF5A1K50A, along with phosphorylation and stabilization of the p53 tumor suppressor protein. Synthetic inhibitors of p38 and JNK kinase activity, but not inhibitors of ERK1/2 or p53 activity, significantly inhibited apoptosis induced by Ad-eIF5A1. Importantly, normal lung cells were more resistant to apoptosis induced by eIF5A1 and eIF5A1K50A than A549 lung cancer cells. CONCLUSIONS Collectively these data indicate that p38 and JNK MAP kinase signaling are important for eIF5A1-induced cell death and that induction of apoptosis was not dependent on p53 activity.
Collapse
Affiliation(s)
- Catherine A Taylor
- Department of Biology, University of Waterloo, 200 University Ave. W., Waterloo, ON N2L 3G1, Canada
| | - Qifa Zheng
- Department of Biology, University of Waterloo, 200 University Ave. W., Waterloo, ON N2L 3G1, Canada
| | - Zhongda Liu
- Department of Biology, University of Waterloo, 200 University Ave. W., Waterloo, ON N2L 3G1, Canada
| | - John E Thompson
- Department of Biology, University of Waterloo, 200 University Ave. W., Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
91
|
Fukuda R, Suico MA, Koyama K, Omachi K, Kai Y, Matsuyama S, Mitsutake K, Taura M, Morino-Koga S, Shuto T, Kai H. Mild electrical stimulation at 0.1-ms pulse width induces p53 protein phosphorylation and G2 arrest in human epithelial cells. J Biol Chem 2013; 288:16117-26. [PMID: 23599430 DOI: 10.1074/jbc.m112.442442] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Exogenous low-intensity electrical stimulation has been used for treatment of various intractable diseases despite the dearth of information on the molecular underpinnings of its effects. Our work and that of others have demonstrated that applied electrical stimulation at physiological strength or mild electrical stimulation (MES) activates the PI3K-Akt pathway, but whether MES activates other molecules remains unknown. Considering that MES is a form of physiological stress, we hypothesized that it can activate the tumor suppressor p53, which is a key modulator of the cell cycle and apoptosis in response to cell stresses. The potential response of p53 to an applied electrical current of low intensity has not been investigated. Here, we show that p53 was transiently phosphorylated at Ser-15 in epithelial cells treated with an imperceptible voltage (1 V/cm) and a 0.1-ms pulse width. MES-induced p53 phosphorylation was inhibited by pretreatment with a p38 MAPK inhibitor and transfection of dominant-negative mutants of p38, MKK3b, and MKK6b, implying the involvement of the p38 MAPK signaling pathway. Furthermore, MES treatment enhanced p53 transcriptional function and increased the expression of p53 target genes p21, BAX, PUMA, NOXA, and IRF9. Importantly, MES treatment triggered G2 cell cycle arrest, but not cell apoptosis. MES treatment had no effect on the cell cycle in HCT116 p53(-/-) cells, suggesting a dependence on p53. These findings identify some molecular targets of electrical stimulation and incorporate the p38-p53 signaling pathway among the transduction pathways that MES affects.
Collapse
Affiliation(s)
- Ryosuke Fukuda
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
SUN KEKANG, ZHONG NING, YANG YANG, ZHAO LIN, JIAO YANG. Enhanced radiosensitivity of NSCLC cells by transducer of erbB2.1 (TOB1) through modulation of the MAPK/ERK pathway. Oncol Rep 2013; 29:2385-91. [DOI: 10.3892/or.2013.2403] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/26/2013] [Indexed: 11/06/2022] Open
|
93
|
Banh S, Hales BF. Hydroxyurea exposure triggers tissue-specific activation of p38 mitogen-activated protein kinase signaling and the DNA damage response in organogenesis-stage mouse embryos. Toxicol Sci 2013; 133:298-308. [PMID: 23492809 PMCID: PMC3663560 DOI: 10.1093/toxsci/kft069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hydroxyurea (HU) is commonly used to treat myeloproliferative diseases and sickle cell anemia. The administration of HU to gestation day 9 CD1 mice causes predominantly hindlimb, tail, and neural tube defects. HU induces oxidative stress and p38 mitogen-activated protein kinase (MAPK) signaling in embryos. HU also inactivates ribonucleotide reductase, leading to DNA replication stress and DNA damage response signaling. We hypothesize that HU exposure induces p38 MAPK activation and DNA damage response signaling during organogenesis preferentially in malformation-sensitive tissues. HU treatment (400 or 600mg/kg) induced the activation of MEK3/6, upstream MAP2K3 kinases, within 30min; phospho-MEK3/6 immunoreactivity was increased throughout the embryo. Activation of the downstream p38 MAPK peaked 3h post-HU treatment. At this time, phospho-p38 MAPK immunoreactivity was enhanced in the cytoplasm and nucleus of cells in the rostral and caudal neuroepithelium and neural tube; significant increases in p38 MAPK signaling were not observed in the somites or heart. Interestingly, the DNA damage response, as assessed by the formation of γH2AX foci, was increased at 3h in HU-exposed embryos in all tissues examined, including the somites and heart. Increases in pyknotic nuclei and cell fragmentation were observed in all tissues except the heart, an organ that is relatively resistant to HU-induced malformations. Thus, although HU induces a widespread DNA damage response, the activation of p38 MAPK is localized to the rostral and caudal neuroepithelium and neural tube, suggesting that p38 MAPK pathways may play a role in mediating the specific malformations observed after HU exposure.
Collapse
Affiliation(s)
- Serena Banh
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| | | |
Collapse
|
94
|
Picone P, Nuzzo D, Di Carlo M. Ferulic acid: a natural antioxidant against oxidative stress induced by oligomeric A-beta on sea urchin embryo. THE BIOLOGICAL BULLETIN 2013; 224:18-28. [PMID: 23493505 DOI: 10.1086/bblv224n1p18] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder, characterized by loss of memory and impairment of multiple cognitive functions. Amyloid beta peptide (Aβ) is the main component of amyloid plaques observed in the brain of individuals affected by AD. Oxidative stress and mitochondrial dysfunction, induced by Aβ, are among the earliest events in AD, triggering neuronal degeneration and cell death. Use of natural molecules with antioxidant properties could be a suitable strategy for inhibiting the cell death cascade. Here, by employing the sea urchin Paracentrotus lividus as a model system, and Aβ oligomers, we tested the effectiveness of ferulic acid (FA), a natural antioxidant, as a putative AD neuroprotective compound. By microscopic inspection we observed that FA is able to reverse morphological defects induced by Aβ oligomers in P. lividus embryos. In addition, FA is able to neutralize reactive oxygen species (ROS), recover mitochondrial membrane potential, and block apoptotic pathways. Moreover, this model system has allowed us to obtain information about down- or up-regulation of some key molecules--Foxo3a, ERK, and p53--involved in the antioxidant mechanism.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto di Biomedicina ed Immunologia Molecolare (IBIM)-CNR, via Ugo La Malfa 153, 90146, Palermo, Italy
| | | | | |
Collapse
|
95
|
Santoro R, Mori F, Marani M, Grasso G, Cambria MA, Blandino G, Muti P, Strano S. Blockage of melatonin receptors impairs p53-mediated prevention of DNA damage accumulation. Carcinogenesis 2013; 34:1051-61. [PMID: 23354312 DOI: 10.1093/carcin/bgt025] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Melatonin has been known to be a chemopreventive agent since its levels inversely correlate with the risk of developing cancer. We have recently shown that melatonin induces p38-dependent phosphorylation of both p53 and histone H2AX. This is associated with a p53-mediated increase in repair of both endogenous and chemotherapy-induced DNA damage. In addition, the inhibition of p38 activities impairs melatonin's capability to induce a p53-dependent DNA damage response and thus its ability to maintain genome integrity. Since melatonin-induced p53 phosphorylation requires an intact p38 phosphorylation cascade and p38 can be activated by G proteins, we supposed that melatonin's activities could be mediated by its G-protein-coupled membrane receptors, MT1 and MT2. Here, we show that the activation of the p53-dependent DNA damage response by melatonin is indeed mediated by MT1 and MT2. As a result, the absence of either receptor impairs melatonin's ability to reduce both cell proliferation and clonogenic potential of cancer cells. In addition, this causes an impairment of the p53-dependent DNA damage response. By providing molecular insight, our findings might have translational impact, suggesting the involvement of melatonin receptors in tumorigenesis.
Collapse
Affiliation(s)
- Raffaela Santoro
- Molecular Chemoprevention Group, Molecular Medicine Area, Regina Elena National Cancer Institute, Rome 00144, Italy
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Mir Mohammadrezaei F, Mohseni kouchesfehani H, Montazeri H, Gharghabi M, Ostad SN, Ghahremani MH. Signaling crosstalk of FHIT, CHK2 and p38 in etoposide induced growth inhibition in MCF-7 cells. Cell Signal 2013; 25:126-32. [DOI: 10.1016/j.cellsig.2012.09.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 09/13/2012] [Indexed: 10/27/2022]
|
97
|
Role of JNK and p38 MAPK in Taiwanin A-induced cell death. Life Sci 2012; 91:1358-65. [PMID: 23123629 DOI: 10.1016/j.lfs.2012.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 10/15/2012] [Accepted: 10/18/2012] [Indexed: 11/23/2022]
Abstract
AIM The lignan compound Taiwanin A is cytotoxic for human cancer cells. Taiwanin A has been previously shown to damage microtubules, induce mitotic arrest and cause apoptosis in cancer cells. The goal of the current study is to identify intracellular signaling pathways that are involved in Taiwanin A-mediated apoptosis. MAIN METHODS We examined the activation of three mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinase (ERK), p38 and c-Jun N-terminal kinase (JNK), in HepG2 cells after Taiwanin A treatment. The role of MAPK activation in Taiwanin A-induced apoptosis was examined using Western blotting, caspase activity assays combined with specific MAPK inhibitors and shRNA treatment to knockdown JNK. KEY FINDINGS Taiwanin A activated all three MAPKs (ERK, p38 and JNK). Cytotoxicity was blocked by the p38 MAPK inhibitor SB203580 and the JNK inhibitor SP600125 but not by the ERK inhibitor PD98059. A combined treatment of SB203580 and SP600125 showed increased effects on the inhibition of Taiwanin A cytotoxicity, suggesting that both JNK and p38 play a role in Taiwanin A-induced apoptosis. Inhibition of p38 activity reduced Taiwanin A-induced p53 phosphorylation on Ser15. Direct interaction of Taiwanin A-activated p38 and p53 was demonstrated by immunoprecipitation. In addition, inhibition of JNK by SP600125 or silencing of the JNK scaffold protein JIP2 reduced phosphorylation of Bcl-2, which may help to promote anti-apoptotic pathways. SIGNIFICANCE We demonstrated for the first time that two distinct apoptotic pathways, the p38-p53 and JNK-Bcl-2 pathways, were triggered by the anti-microtubule compound Taiwanin A.
Collapse
|
98
|
Matsuoka M, Igisu H. Effects of heavy metals on mitogen-activated protein kinase pathways. Environ Health Prev Med 2012; 6:210-7. [PMID: 21432337 DOI: 10.1007/bf02897972] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2001] [Accepted: 10/22/2001] [Indexed: 01/07/2023] Open
Abstract
The signaling pathways leading to cellular protection or cell death following exposure to heavy metals have not been fully clarified. Mitogen-activated protein kinases (MAPKs), i.e., extracellular signal-regulated protein kinase (ERK), c-Jun NH(2)-terminal kinase (JNK) and p38 MAPK transmit extracellular signals into the nucleus, and have been shown to participate in a diverse array of cellular functions such as cell growth, differentiation and apoptosis. Treatment with cadmium, inorganic mercury or tributyltin can activate ERK, JNK and p38 MAPK, and induces the expression of c-fos and c-jun genes prior to the development of apoptosis. However, the members of the MAPK family appear to be differentially activated depending on the heavy metal and the cell type exposed. Consequently, various cellular responses may be caused by the distinct pattern of MAPKs activation. MAPKs may be one of the important cellular signal transduction pathways affected by various environmental pollutants, including heavy metals.
Collapse
Affiliation(s)
- Masato Matsuoka
- Department of Environmental Toxicology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, 807-8555, Kitakyushu, Japan,
| | | |
Collapse
|
99
|
Wang L, Xiang S, Williams KA, Dong H, Bai W, Nicosia SV, Khochbin S, Bepler G, Zhang X. Depletion of HDAC6 enhances cisplatin-induced DNA damage and apoptosis in non-small cell lung cancer cells. PLoS One 2012; 7:e44265. [PMID: 22957056 PMCID: PMC3434198 DOI: 10.1371/journal.pone.0044265] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 07/31/2012] [Indexed: 11/18/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) are promising therapeutic agents which are currently used in combination with chemotherapeutic agents in clinical trials for cancer treatment including non-small cell lung cancer (NSCLC). However, the mechanisms underlying their anti-tumor activities remain elusive. Previous studies showed that inhibition of HDAC6 induces DNA damage and sensitizes transformed cells to anti-tumor agents such as etoposide and doxorubicin. Here, we showed that depletion of HDAC6 in two NSCLC cell lines, H292 and A549, sensitized cells to cisplatin, one of the first-line chemotherapeutic agents used to treat NSCLC. We suggested that depletion of HDAC6 increased cisplatin-induced cytotoxicity was due to the enhancement of apoptosis via activating ATR/Chk1 pathway. Furthermore, we showed that HDAC6 protein levels were positively correlated with cisplatin IC(50) in 15 NSCLC cell lines. Lastly, depletion of HDAC6 in H292 xenografts rendered decreased tumor weight and volume and exhibited increased basal apoptosis compared with the controls in a xenograft mouse model. In summary, our findings suggest that HDAC6 is positively associated with cisplatin resistance in NSCLC and reveal HDAC6 as a potential novel therapeutic target for platinum refractory NSCLC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
| | - Shengyan Xiang
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
| | - Kendra A. Williams
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
| | - Huiqin Dong
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
| | - Wenlong Bai
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
- Program of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Santo V. Nicosia
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
- Experimental Therapeutics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Saadi Khochbin
- French National Institute of Health and Medical Research, The Albert Bonniot Institute, Grenoble, France
| | - Gerold Bepler
- Department of Oncology, Karmanos Cancer Institute, Detroit, Michigan, United States of America
- * E-mail: (GB); (XZ)
| | - Xiaohong Zhang
- Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, Florida, United States of America
- Program of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- * E-mail: (GB); (XZ)
| |
Collapse
|
100
|
Katsuyama M, Iwata K, Ibi M, Matsuno K, Matsumoto M, Yabe-Nishimura C. Clioquinol induces DNA double-strand breaks, activation of ATM, and subsequent activation of p53 signaling. Toxicology 2012; 299:55-9. [DOI: 10.1016/j.tox.2012.05.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 05/11/2012] [Accepted: 05/14/2012] [Indexed: 11/30/2022]
|