1
|
Adeniyi OO, Lenstra JA, Mastrangelo S, Lühken G. Genome-wide comparative analyses for selection signatures indicate candidate genes for between-breed variability in copper accretion in sheep. Animal 2024; 18:101329. [PMID: 39378609 DOI: 10.1016/j.animal.2024.101329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
The problem of copper (Cu) intoxication and deficiency continues to impact economic gains and animal welfare in sheep husbandry. This study investigated the ovine genome for regions and potential genes under selection for Cu accretion between sheep breeds. For this, we compared ovine single nucleotide polymorphism (SNP) data of three Cu-susceptible breeds with three Cu-tolerant breeds. After merging SNP data of breeds and removal of related individuals, a total of 229 sheep and 45 640 autosomal SNPs were left. Then, we selected 14 individuals per breed into two datasets (datasets 1 and 2) for analysis of selection signatures using the Fixation index, cross-population extended haplotype homozygosity and haplotype-based FLK methods. Selection regions shared by both datasets detected by at least two methods revealed regions on OAR 4, 8 and 11 containing 54 candidate genes under selection for Cu accretion. Enrichment analysis revealed that 19 gene ontologies and 1 enriched Kyoto encyclopaedia of genes and genomes pathway terms were associated with the candidate genes under selection. Genes such as TP53, TNFSF13, TNFSF12, ALOX15, ALOX12, EIF5A and PREP are associated with the regulation of Cu homeostasis, programmed cell death or inflammatory response. We also found an enrichment of arachidonate 15-lipoxygenase activity, arachidonate 12-lipoxygenase activity and ferroptosis that influence cellular inflammation and cell death. These results shed light on ovine genomic regions under selection for Cu accretion and provide information on candidate genes for further studies on breed differences in ovine Cu accretion.
Collapse
Affiliation(s)
- O O Adeniyi
- Institute of Animal Breeding and Genetics, Justus Liebig University of Giessen, Ludwigstrasse 21,35390 Giessen, Germany.
| | - J A Lenstra
- Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584CM Utrecht, the Netherlands
| | - S Mastrangelo
- Department of Agricultural, Food and Forest Sciences, University of Palermo, Viale delle Scienze, 90128 Palermo (PA), Italy
| | - G Lühken
- Institute of Animal Breeding and Genetics, Justus Liebig University of Giessen, Ludwigstrasse 21,35390 Giessen, Germany
| |
Collapse
|
2
|
Yan X, Zhang M, Wang D. Interplay between posttranslational modifications and liquid‒liquid phase separation in tumors. Cancer Lett 2024; 584:216614. [PMID: 38246226 DOI: 10.1016/j.canlet.2024.216614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024]
Abstract
Liquid‒liquid phase separation (LLPS) is a general phenomenon recently recognized to be critically involved in the regulation of a variety of cellular biological processes, such as transcriptional regulation, heterochromatin formation and signal transduction, through the compartmentalization of proteins or nucleic acids into droplet-like condensates. These processes are directly or indirectly related to tumor initiation and treatment. Posttranslational modifications (PTMs), which represent a rapid and reversible mechanism involved in the functional regulation of proteins, have emerged as key events in modulating LLPS under physiological or pathophysiological conditions, including tumorigenesis and antitumor therapy. In this review, we introduce the biological functions participated in cancer-associated LLPS, discuss the potential roles of LLPS during tumor onset or therapy, and emphasize the mechanistic characteristics of LLPS regulated by PTMs and its effects on tumor progression. We then provide a perspective on further studies on LLPS and its regulation by PTMs in cancer research. This review aims to broaden the understanding of the functions of LLPS and its regulation by PTMs under normal or aberrant cellular conditions.
Collapse
Affiliation(s)
- Xiaojun Yan
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Meng Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Donglai Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
3
|
Okamura K, Sato M, Suzuki T, Nohara K. Arsenite exposure induces premature senescence and senescence-associated secretory phenotype (SASP) in human hepatocyte-derived cell line Huh-7. Environ Health Prev Med 2024; 29:74. [PMID: 39756915 DOI: 10.1265/ehpm.24-00139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Chronic arsenite exposure has been known to induce cancer in various organs; however, the underlying mechanisms remain elusive. The characteristic feature of carcinogenesis due to arsenic exposure is that the disease develops after a prolonged latent period, even after cessation of exposure. Our previous study revealed that arsenite exposure induces premature senescence in hepatic stellate cells and suggests that the senescence-associated secretory phenotype (SASP) factors from the senescent cells promote hepatic carcinogenesis. However, arsenite exposure in the liver occurs not only in hepatic stellate cells, but also in hepatocytes. Therefore, we examined whether arsenite exposure in hepatocytes also causes premature senescence and the enhancement of SASP factors. We also assessed whether those effects remained after cessation of arsenite exposure. METHODS Human hepatocyte-derived cell line Huh-7 was exposed to sodium arsenite for 72 hours to determine the concentration at which cell proliferation was inhibited. In the 5 µM of exposure, various cellular senescence markers and SASP factors were analyzed and compared with unexposed cells. We also examined whether those senescence markers and SASP factors were maintained after cessation of arsenite exposure. Finally, we explored whether the increased expression of SASP factor, which was upregulated in hepatocytes by arsenic exposure in this study, is related to the prognosis of human hepatocellular carcinoma. RESULTS After exposure to 5 µM of sodium arsenite for 72 hours, various senescent features, such as the induction of P21 mRNA, the reduction of LAMINB1 mRNA, morphological changes, phosphorylation of P53, and the presence of SA-β-gal positive cells were observed. Those changes were maintained after cessation of arsenite exposure. In addition, mRNA levels of SASP factors (MMP1, MMP3, MMP10, GDF15, PAI-1, and IL-6) were increased after arsenite exposure, and their high expression levels were maintained after cessation of arsenite exposure. Furthermore, by analyzing the TCGA database, we found that the increased expression levels of many SASP factors negatively correlated with prognosis. CONCLUSIONS Arsenite exposure induces premature senescence in hepatocyte-derived cells and increases SASP factors that are related to hepatic tumorigenesis. Once arsenite exposure induces premature senescence, the senescent cells remain even after cessation of exposure.
Collapse
Affiliation(s)
- Kazuyuki Okamura
- Health and Environmental Risk Division, National Institute for Environmental Studies
| | - Miyuki Sato
- Health and Environmental Risk Division, National Institute for Environmental Studies
| | - Takehiro Suzuki
- Health and Environmental Risk Division, National Institute for Environmental Studies
| | - Keiko Nohara
- Health and Environmental Risk Division, National Institute for Environmental Studies
| |
Collapse
|
4
|
Abuetabh Y, Wu HH, Chai C, Al Yousef H, Persad S, Sergi CM, Leng R. DNA damage response revisited: the p53 family and its regulators provide endless cancer therapy opportunities. Exp Mol Med 2022; 54:1658-1669. [PMID: 36207426 PMCID: PMC9636249 DOI: 10.1038/s12276-022-00863-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 12/29/2022] Open
Abstract
Antitumor therapeutic strategies that fundamentally rely on the induction of DNA damage to eradicate and inhibit the growth of cancer cells are integral approaches to cancer therapy. Although DNA-damaging therapies advance the battle with cancer, resistance, and recurrence following treatment are common. Thus, searching for vulnerabilities that facilitate the action of DNA-damaging agents by sensitizing cancer cells is an active research area. Therefore, it is crucial to decipher the detailed molecular events involved in DNA damage responses (DDRs) to DNA-damaging agents in cancer. The tumor suppressor p53 is active at the hub of the DDR. Researchers have identified an increasing number of genes regulated by p53 transcriptional functions that have been shown to be critical direct or indirect mediators of cell fate, cell cycle regulation, and DNA repair. Posttranslational modifications (PTMs) primarily orchestrate and direct the activity of p53 in response to DNA damage. Many molecules mediating PTMs on p53 have been identified. The anticancer potential realized by targeting these molecules has been shown through experiments and clinical trials to sensitize cancer cells to DNA-damaging agents. This review briefly acknowledges the complexity of DDR pathways/networks. We specifically focus on p53 regulators, protein kinases, and E3/E4 ubiquitin ligases and their anticancer potential.
Collapse
Affiliation(s)
- Yasser Abuetabh
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - H Helena Wu
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Chengsen Chai
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
- College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Habib Al Yousef
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Sujata Persad
- Department of Pediatrics, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Consolato M Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON, K1H 8L1, Canada
| | - Roger Leng
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada.
| |
Collapse
|
5
|
At the Crossroads of Life and Death: The Proteins That Influence Cell Fate Decisions. Cancers (Basel) 2022; 14:cancers14112745. [PMID: 35681725 PMCID: PMC9179324 DOI: 10.3390/cancers14112745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cellular senescence and apoptosis were historically thought of as two distinct cell fate pathways. However, many of the proteins involved are integral to both pathways. In particular, the ability of p53 to regulate both senescence and apoptosis meant it was seen as the decisive factor in these decisions, yet questions remain about its ability to select on its own the most appropriate cell fate according to each situation. Therefore, cell fates are no longer considered fixed endpoints but dynamic states that can be shifted given the right combination of activation and/or inhibitions of cofactors. Abstract When a cell is damaged, it must decide how to respond. As a consequence of a variety of stresses, cells can induce well-regulated programmes such as senescence, a persistent proliferative arrest that limits their replication. Alternatively, regulated programmed cell death can be induced to remove the irreversibly damaged cells in a controlled manner. These programmes are mainly triggered and controlled by the tumour suppressor protein p53 and its complex network of effectors, but how it decides between these wildly different responses is not fully understood. This review focuses on the key proteins involved both in the regulation and induction of apoptosis and senescence to examine the key events that determine cell fate following damage. Furthermore, we examine how the regulation and activity of these proteins are altered during the progression of many chronic diseases, including cancer.
Collapse
|
6
|
Beneficial Oxidative Stress-Related trans-Resveratrol Effects in the Treatment and Prevention of Breast Cancer. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112211041] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resveratrol is one of the most investigated polyphenols for its multiple biological activities and many beneficial effects. These are mainly related to its ability to scavenge free radicals and reduce oxidative stress. Resveratrol has also been shown to have the ability to stimulate the production of antioxidant enzymes, which interact with numerous signaling pathways involved in tumor development, and to possess side effects associated with the use of chemotherapy drugs. In this review article we summarized the main discoveries about the impact resveratrol can have in helping to prevent, as well as adjuvant treating, breast cancer. A brief overview of the primary sources of resveratrol as well as some approaches for improving its bioavailability have been also discussed.
Collapse
|
7
|
Huang Y, Hou Y, Qu P, Cai Y. Editorial: Combating Cancer With Natural Products: What Would Non-Coding RNAs Bring? Front Oncol 2021; 11:747586. [PMID: 34604091 PMCID: PMC8484867 DOI: 10.3389/fonc.2021.747586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/17/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
- Yongye Huang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yue Hou
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Peng Qu
- Cancer and Inflammation Program, National Cancer Institute (NCI), Frederick, MD, United States
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
8
|
Akter R, Rahman MH, Kaushik D, Mittal V, Uivarosan D, Nechifor AC, Behl T, Karthika C, Stoicescu M, Munteanu MA, Bustea C, Bungau S. Chemo-Preventive Action of Resveratrol: Suppression of p53-A Molecular Targeting Approach. Molecules 2021; 26:molecules26175325. [PMID: 34500758 PMCID: PMC8433711 DOI: 10.3390/molecules26175325] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
Extensive experimental, clinical, and epidemiological evidence has explained and proven that products of natural origin are significantly important in preventing and/or ameliorating various disorders, including different types of cancer that researchers are extremely focused on. Among these studies on natural active substances, one can distinguish the emphasis on resveratrol and its properties, especially the potential anticancer role. Resveratrol is a natural product proven for its therapeutic activity, with remarkable anti-inflammatory properties. Various other benefits/actions have also been reported, such as cardioprotective, anti-ageing, antioxidant, etc. and its rapid digestion/absorption as well. This review aims to collect and present the latest published studies on resveratrol and its impact on cancer prevention, molecular signals (especially p53 protein participation), and its therapeutic prospects. The most recent information regarding the healing action of resveratrol is presented and concentrated to create an updated database focused on this topic presented above.
Collapse
Affiliation(s)
- Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka 1100, Bangladesh;
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
| | - Md. Habibur Rahman
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
- Correspondence: (M.H.R.); (S.B.)
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India; (D.K.); (V.M.)
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India; (D.K.); (V.M.)
| | - Diana Uivarosan
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Aurelia Cristina Nechifor
- Analytical Chemistry and Environmental Engineering Department, Polytechnic University of Bucharest, 011061 Bucharest, Romania;
| | - Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (M.S.); (M.A.M.); (C.B.)
| | - Mihai Alexandru Munteanu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (M.S.); (M.A.M.); (C.B.)
| | - Cristiana Bustea
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (M.S.); (M.A.M.); (C.B.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
- Correspondence: (M.H.R.); (S.B.)
| |
Collapse
|
9
|
Sari AN, Elwakeel A, Dhanjal JK, Kumar V, Sundar D, Kaul SC, Wadhwa R. Identification and Characterization of Mortaparib Plus-A Novel Triazole Derivative That Targets Mortalin-p53 Interaction and Inhibits Cancer-Cell Proliferation by Wild-Type p53-Dependent and -Independent Mechanisms. Cancers (Basel) 2021; 13:cancers13040835. [PMID: 33671256 PMCID: PMC7921971 DOI: 10.3390/cancers13040835] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 12/24/2022] Open
Abstract
p53 has an essential role in suppressing the carcinogenesis process by inducing cell cycle arrest/apoptosis/senescence. Mortalin/GRP75 is a member of the Hsp70 protein family that binds to p53 causing its sequestration in the cell cytoplasm. Hence, p53 cannot translocate to the nucleus to execute its canonical tumour suppression function as a transcription factor. Abrogation of mortalin-p53 interaction and subsequent reactivation of p53's tumour suppression function has been anticipated as a possible approach in developing a novel cancer therapeutic drug candidate. A chemical library was screened in a high-content screening system to identify potential mortalin-p53 interaction disruptors. By four rounds of visual assays for mortalin and p53, we identified a novel synthetic small-molecule triazole derivative (4-[(1E)-2-(2-phenylindol-3-yl)-1-azavinyl]-1,2,4-triazole, henceforth named MortaparibPlus). Its activities were validated using multiple bioinformatics and experimental approaches in colorectal cancer cells possessing either wild-type (HCT116) or mutant (DLD-1) p53. Bioinformatics and computational analyses predicted the ability of MortaparibPlus to competitively prevent the interaction of mortalin with p53 as it interacted with the p53 binding site of mortalin. Immunoprecipitation analyses demonstrated the abrogation of mortalin-p53 complex formation in MortaparibPlus-treated cells that showed growth arrest and apoptosis mediated by activation of p21WAF1, or BAX and PUMA signalling, respectively. Furthermore, we demonstrate that MortaparibPlus-induced cytotoxicity to cancer cells is mediated by multiple mechanisms that included the inhibition of PARP1, up-regulation of p73, and also the down-regulation of mortalin and CARF proteins that play critical roles in carcinogenesis. MortaparibPlus is a novel multimodal candidate anticancer drug that warrants further experimental and clinical attention.
Collapse
Affiliation(s)
- Anissa Nofita Sari
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 5-41, Tsukuba 305-8565, Japan; (A.N.S.); (A.E.); (J.K.D.)
- School of Integrative & Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Ahmed Elwakeel
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 5-41, Tsukuba 305-8565, Japan; (A.N.S.); (A.E.); (J.K.D.)
- School of Integrative & Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Jaspreet Kaur Dhanjal
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 5-41, Tsukuba 305-8565, Japan; (A.N.S.); (A.E.); (J.K.D.)
| | - Vipul Kumar
- DAILAB, Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110-016, India; (V.K.); (D.S.)
| | - Durai Sundar
- DAILAB, Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110-016, India; (V.K.); (D.S.)
| | - Sunil C. Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 5-41, Tsukuba 305-8565, Japan; (A.N.S.); (A.E.); (J.K.D.)
- Correspondence: (S.C.K.); (R.W.)
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 5-41, Tsukuba 305-8565, Japan; (A.N.S.); (A.E.); (J.K.D.)
- School of Integrative & Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
- Correspondence: (S.C.K.); (R.W.)
| |
Collapse
|
10
|
Sun Z, Qiu Z, Ma B, Wang Z. Encorafenib enhances TRAIL-induced apoptosis of colorectal cancer cells dependent on p53/PUMA signaling. Cytotechnology 2020; 73:63-70. [PMID: 33505114 DOI: 10.1007/s10616-020-00442-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/06/2020] [Indexed: 01/18/2023] Open
Abstract
TRAIL has been demonstrated to play a critical role in the apoptosis of colorectal cancer (CRC) cells, but drug resistance markedly restricts its therapeutic effects. Objectives: This study aims to investigate whether encorafenib can enhance TRAIL-induced apoptosis of colorectal cancer cells and the underlying mechanism. TRAIL was first used to induce CRC cells. CCK-8 assays were conducted for detecting cell viability of TRAIL-induced CRC cells with encorafenib treatment. Flow cytometry was used to detect the cell apoptosis of CRC cells and western blot was used to measure the expressions of apoptosis-related proteins. The expressions of DR4, DR5, p53, and PUMA were then evaluated by qPCR and western blot. After transfecting the interference plasmid of p53 into CRC cells, the expressions of PUMA and DR5 were further explored. TRAIL reduced the cell viability of CRC cells, and the inhibition was further reinforced under co-treatment of TRAIL and encorafenib. Encorafenib also triggered the promotion of CRC cell apoptosis induced by TRAIL. It was also found that encorafenib exerted its promoting effects on cell apoptosis of CRC cells via the elevation of DR5. Besides, encorafenib administration promoted the expression levels of p53 and PUMA in TRAIL-induced CRC cells. Furthermore, p53 knockdown attenuated the expression of PUMA and DR5 in TRAIL-induced CRC cells treated with encorafenib. This study indicates that encorafenib stimulates TRAIL-induced apoptosis of CRC cells dependent on p53/PUMA signaling, which may provide instructions for the treatment of CRC.
Collapse
Affiliation(s)
- Zhenqing Sun
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, N0.59, Haier Road, Laoshan District, Qingdao, 266100 Shandong China
| | - Zhigang Qiu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, N0.59, Haier Road, Laoshan District, Qingdao, 266100 Shandong China
| | - Bin Ma
- Affiliated Hospital of Qingdao University, Qingdao, 266100 Shandong China
| | - Zhengkun Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, N0.59, Haier Road, Laoshan District, Qingdao, 266100 Shandong China
| |
Collapse
|
11
|
Kaplan P, Tatarkova Z, Sivonova MK, Racay P, Lehotsky J. Homocysteine and Mitochondria in Cardiovascular and Cerebrovascular Systems. Int J Mol Sci 2020; 21:ijms21207698. [PMID: 33080955 PMCID: PMC7589705 DOI: 10.3390/ijms21207698] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022] Open
Abstract
Elevated concentration of homocysteine (Hcy) in the blood plasma, hyperhomocysteinemia (HHcy), has been implicated in various disorders, including cardiovascular and neurodegenerative diseases. Accumulating evidence indicates that pathophysiology of these diseases is linked with mitochondrial dysfunction. In this review, we discuss the current knowledge concerning the effects of HHcy on mitochondrial homeostasis, including energy metabolism, mitochondrial apoptotic pathway, and mitochondrial dynamics. The recent studies suggest that the interaction between Hcy and mitochondria is complex, and reactive oxygen species (ROS) are possible mediators of Hcy effects. We focus on mechanisms contributing to HHcy-associated oxidative stress, such as sources of ROS generation and alterations in antioxidant defense resulting from altered gene expression and post-translational modifications of proteins. Moreover, we discuss some recent findings suggesting that HHcy may have beneficial effects on mitochondrial ROS homeostasis and antioxidant defense. A better understanding of complex mechanisms through which Hcy affects mitochondrial functions could contribute to the development of more specific therapeutic strategies targeted at HHcy-associated disorders.
Collapse
|
12
|
Wang Y, Zhang J, Su Y, Wang C, Zhang G, Liu X, Chen Q, Lv M, Chang Y, Peng J, Hou M, Huang X, Zhang X. miRNA-98-5p Targeting IGF2BP1 Induces Mesenchymal Stem Cell Apoptosis by Modulating PI3K/Akt and p53 in Immune Thrombocytopenia. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:764-776. [PMID: 32428701 PMCID: PMC7232042 DOI: 10.1016/j.omtn.2020.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/15/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Immune thrombocytopenia (ITP) is a common hematological autoimmune disease, in which defective mesenchymal stem cells (MSCs) are potentially involved. Our previous study suggested that MSCs in ITP patients displayed enhanced apoptosis. MicroRNAs (miRNAs) play important roles in ITP by affecting megakaryopoiesis, platelet production and immunoregulation, whereas the roles of miRNAs in ITP-MSCs remain unknown. In a previous study, we performed microarray analysis to obtain mRNA and miRNA profiles of ITP-MSCs. In the present study, we reanalyze the data and identify miR-98-5p as a candidate miRNA contributing to MSC deficiency in ITP. miR-98-5p acts through targeting insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), and the subsequent downregulation of insulin-like growth factor 2 (IGF-2) causes inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which is involved in the process of MSC deficiency. Furthermore, miR-98-5p upregulates p53 by inhibiting β-transducin repeat-containing protein (β-TrCP)-dependent p53 ubiquitination. Moreover, miR-98-5p overexpression impairs the therapeutic effect of MSCs in ITP mice. All-trans retinoic acid (ATRA) protects MSCs from apoptosis by downregulating miR-98-5p, thus providing a potential therapeutic approach for ITP. Our findings demonstrate that miR-98-5p is a critical regulator of ITP-MSCs, which will help us thoroughly understand the pathogenesis of ITP.
Collapse
Affiliation(s)
- Yanan Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Jiamin Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Yan Su
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Chencong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Gaochao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Xiao Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Meng Lv
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China; National Clinical Research Center for Hematologic Disease, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China.
| |
Collapse
|
13
|
Li Y, Li X, Cao M, Jiang Y, Yan J, Liu Z, Yang R, Chen X, Sun P, Xiang R, Wang L, Shi Y. Seryl tRNA synthetase cooperates with POT1 to regulate telomere length and cellular senescence. Signal Transduct Target Ther 2019; 4:50. [PMID: 31815007 PMCID: PMC6882858 DOI: 10.1038/s41392-019-0078-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/04/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Deregulated telomere length is a causative factor in many physiological and pathological processes, including aging and cancer. Many studies focusing on telomeres have revealed important roles for cooperation between the Shelterin protein complex and telomerase in maintaining telomere length. However, it remains largely unknown whether and how aging-related stresses, such as deregulated protein homeostasis, impact telomere length. Here, we explored the possible roles of aminoacyl tRNA synthetases (AARSs), key enzymes catalyzing the first reactions in protein synthesis, in regulating telomere length and aging. We selected seryl tRNA synthetase (SerRS) since our previous studies discovered expanded functions of SerRS in the nucleus in addition to its canonical cytoplasmic role in protein synthesis. In this study, we revealed that overexpression of SerRS promoted cellular senescence and inhibited the growth of cervical tumor xenografts in mice by triggering the senescence of tumor cells. In the nucleus, SerRS directly bound to telomeric DNA repeats and tethered more POT1 proteins to telomeres through a direct interaction between the UNE-S domain of SerRS and the OB1 domain of POT1. We further demonstrated that SerRS-induced enrichment of POT1 prevented the recruitment of telomerase to telomeres, resulting in progressive telomere shortening. Our data suggested a possible molecular link between protein synthesis and telomere length control, the deregulation of which may be associated with aging and cancer.
Collapse
Affiliation(s)
- Yingxi Li
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Xiyang Li
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Mei Cao
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Yuke Jiang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Jie Yan
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Ze Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Rongcun Yang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Xu Chen
- Tianjin Key Laboratory Human Development and Reproductive Regulation, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Rong Xiang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Longlong Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
- Tianjin Key Laboratory Human Development and Reproductive Regulation, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Yi Shi
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071 China
- Tianjin Key Laboratory Human Development and Reproductive Regulation, Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, China
| |
Collapse
|
14
|
Hua Z, Zhan Y, Zhang S, Dong Y, Jiang M, Tan F, Liu Z, Thiele CJ, Li Z. P53/PUMA are potential targets that mediate the protection of brain-derived neurotrophic factor (BDNF)/TrkB from etoposide-induced cell death in neuroblastoma (NB). Apoptosis 2019; 23:408-419. [PMID: 29959561 DOI: 10.1007/s10495-018-1467-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The over-expressions of brain-derived neurotrophic factor (BDNF) and its tyrosine kinase receptor TrkB have been reported to induce chemo-resistance in neuroblastoma (NB) cells. In this study, we investigated the roles of P53 and BCL2 family members in the protection of BDNF/TrkB from etoposide-induced NB cell death. TB3 and TB8, two tetracycline (TET)-regulated TrkB-expressing NB cell lines, were utilized. The expressions of P53 and BCL2 family members were detected by Western blot or RT-PCR. Transfection of siRNAs was used to knockdown P53 or PUMA. Activated lentiviral was used to over-express PUMA. Cell survival was performed by MTS assay, and the percentage of cell confluence was measured by IncuCyte ZOOM. Our results showed that etoposide treatment induced significant and time-dependent increase of P53, which could be blocked by pre-treatment with BDNF, and knockdown P53 by transfecting siRNA attenuated etoposide-induced TrkB-expressing NB cell death. PUMA was the most significantly changed BCL2 family member after treatment with etoposide, and pre-treatment with BDNF blocked the increased expression of PUMA. Transfection with siRNA inhibited etoposide-induced increased expression of PUMA, and attenuated etoposide-induced NB cell death. We also found that over-expression of PUMA by infection of activated lentiviral induced TrkB-expressing NB cell death in the absence of etoposide, and treatment of BDNF protected NB cells from PUMA-induced cell death. Our results suggested that P53 and PUMA may be potential targets that mediated the protection of BDNF/TrkB from etoposide-induced NB cell death.
Collapse
Affiliation(s)
- Zhongyan Hua
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Yue Zhan
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Simeng Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Yudi Dong
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Min Jiang
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Fei Tan
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhihui Liu
- Cellular & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carol J Thiele
- Cellular & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhijie Li
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
15
|
Camici M, Garcia-Gil M, Pesi R, Allegrini S, Tozzi MG. Purine-Metabolising Enzymes and Apoptosis in Cancer. Cancers (Basel) 2019; 11:cancers11091354. [PMID: 31547393 PMCID: PMC6769685 DOI: 10.3390/cancers11091354] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/03/2019] [Accepted: 09/07/2019] [Indexed: 12/17/2022] Open
Abstract
The enzymes of both de novo and salvage pathways for purine nucleotide synthesis are regulated to meet the demand of nucleic acid precursors during proliferation. Among them, the salvage pathway enzymes seem to play the key role in replenishing the purine pool in dividing and tumour cells that require a greater amount of nucleotides. An imbalance in the purine pools is fundamental not only for preventing cell proliferation, but also, in many cases, to promote apoptosis. It is known that tumour cells harbour several mutations that might lead to defective apoptosis-inducing pathways, and this is probably at the basis of the initial expansion of the population of neoplastic cells. Therefore, knowledge of the molecular mechanisms that lead to apoptosis of tumoural cells is key to predicting the possible success of a drug treatment and planning more effective and focused therapies. In this review, we describe how the modulation of enzymes involved in purine metabolism in tumour cells may affect the apoptotic programme. The enzymes discussed are: ectosolic and cytosolic 5'-nucleotidases, purine nucleoside phosphorylase, adenosine deaminase, hypoxanthine-guanine phosphoribosyltransferase, and inosine-5'-monophosphate dehydrogenase, as well as recently described enzymes particularly expressed in tumour cells, such as deoxynucleoside triphosphate triphosphohydrolase and 7,8-dihydro-8-oxoguanine triphosphatase.
Collapse
Affiliation(s)
- Marcella Camici
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy.
| | - Mercedes Garcia-Gil
- Dipartimento di Biologia, Unità di Fisiologia Generale, Via S. Zeno 31, 56127 Pisa, Italy
| | - Rossana Pesi
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy
| | - Simone Allegrini
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy
| | - Maria Grazia Tozzi
- Dipartimento di Biologia, Unità di Biochimica, Via S. Zeno 51, 56127 Pisa, Italy
| |
Collapse
|
16
|
MDM2-mediated degradation of WRN promotes cellular senescence in a p53-independent manner. Oncogene 2018; 38:2501-2515. [PMID: 30532073 DOI: 10.1038/s41388-018-0605-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/27/2018] [Accepted: 11/13/2018] [Indexed: 01/12/2023]
Abstract
MDM2 (Murine double minute 2) acts as a key repressor for p53-mediated tumor-suppressor functions, which includes cellular senescence. We found that MDM2 can promote cellular senescence by modulating WRN stability. Werner syndrome (WS), caused by mutations of the WRN gene, is an autosomal recessive disease, which is characterized by premature aging. Loss of WRN function induces cellular senescence in human cancer cells. Here, we found that MDM2 acts as an E3 ligase for WRN protein. MDM2 interacts with WRN both in vivo and in vitro. MDM2 induces ubiquitination of WRN and dramatically downregulates the levels of WRN protein in human cells. During DNA damage response, WRN is translocated to the nucleoplasm to facilitate its DNA repair functions; however, it is degraded by the MDM2-mediated ubiquitination pathway. Moreover, the senescent phenotype induced by DNA damage reagents, such as Etoposide, is at least in part mediated by MDM2-dependent WRN degradation as it can be significantly attenuated by ectopic expression of WRN. These results show that MDM2 is critically involved in regulating WRN function via ubiquitin-dependent degradation and reveal an unexpected role of MDM2 in promoting cellular senescence through a p53-independent manner.
Collapse
|
17
|
Wang Y, Ding Q, Lu YC, Cao SY, Liu QX, Zhang L. Interferon-stimulated gene 15 enters posttranslational modifications of p53. J Cell Physiol 2018; 234:5507-5518. [PMID: 30317575 DOI: 10.1002/jcp.27347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Abstract
The tumor suppressor protein p53 is a central governor of various cellular signals. It is well accepted that ubiquitination as well as ubiquitin-like (UBL) modifications of p53 protein is critical in the control of its activity. Interferon-stimulated gene 15 (ISG15) is a well-known UBL protein with pleiotropic functions, serving both as a free intracellular molecule and as a modifier by conjugating to target proteins. Initially, attentions have historically focused on the antiviral effects of ISG15 pathway. Remarkably, a significant role in the processes of autophagy, DNA repair, and protein translation provided considerable insight into the new functions of ISG15 pathway. Despite the deterministic revelation of the relation between ISG15 and p53, the functional consequence of p53 ISGylation appears somewhat confused. More important, more recent studies have hinted p53 ubiquitination or other UBL modifications that might interconnect with its ISGylation. Here, we aim to summarize the current knowledge of p53 ISGylation and the differences in other significant modifications, which would be beneficial for the development of p53-based cancer therapy.
Collapse
Affiliation(s)
- Yang Wang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Qi Ding
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Yu-Chen Lu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Shi-Yang Cao
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Qing-Xue Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,The Key Laboratory of Major Autoimmune Disease, School of Pharmacy, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| |
Collapse
|
18
|
Geelvink M, Babmorad A, Maurer A, Stöhr R, Grimm T, Bach C, Knuechel R, Rose M, Gaisa NT. Diagnostic and Prognostic Implications of FGFR3 high/Ki67 high Papillary Bladder Cancers. Int J Mol Sci 2018; 19:E2548. [PMID: 30154342 PMCID: PMC6163244 DOI: 10.3390/ijms19092548] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/19/2018] [Accepted: 08/24/2018] [Indexed: 01/22/2023] Open
Abstract
Prognostic/therapeutic stratification of papillary urothelial cancers is solely based upon histology, despite activated FGFR3-signaling was found to be associated with low grade tumors and favorable outcome. However, there are FGFR3-overexpressing tumors showing high proliferation-a paradox of coexisting favorable and adverse features. Therefore, our study aimed to decipher the relevance of FGFR3-overexpression/proliferation for histopathological grading and risk stratification. N = 142 (n = 82 pTa, n = 42 pT1, n = 18 pT2-4) morphologically G1⁻G3 tumors were analyzed for immunohistochemical expression of FGFR3 and Ki67. Mutation analysis of FGFR3 and TP53 and FISH for FGFR3 amplification and rearrangement was performed. SPSS 23.0 was used for statistical analysis. Overall FGFR3high/Ki67high status (n = 58) resulted in a reduced ∆mean progression-free survival (PFS) (p < 0.01) of 63.92 months, and shorter progression-free survival (p < 0.01; mean PFS: 55.89 months) in pTa tumors (n = 50). FGFR3mut/TP53mut double mutations led to a reduced ∆mean PFS (p < 0.01) of 80.30 months in all tumors, and FGFR3mut/TP53mut pTa tumors presented a dramatically reduced PFS (p < 0.001; mean PFS: 5.00 months). Our results identified FGFR3high/Ki67high papillary pTa tumors as a subgroup with poor prognosis and encourage histological grading as high grade tumors. Tumor grading should possibly be augmented by immunohistochemical stainings and suitable clinical surveillance by endoscopy should be performed.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers, Tumor
- Carcinoma, Papillary/diagnosis
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/mortality
- Female
- Humans
- Immunohistochemistry
- Ki-67 Antigen/genetics
- Ki-67 Antigen/metabolism
- Male
- Middle Aged
- Mutation
- Neoplasm Grading
- Neoplasm Staging
- Prognosis
- Proportional Hazards Models
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Signal Transduction
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Urinary Bladder Neoplasms/diagnosis
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/mortality
Collapse
Affiliation(s)
- Mirja Geelvink
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Armin Babmorad
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Angela Maurer
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Robert Stöhr
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany.
| | - Tobias Grimm
- Department of Urology, Ludwig Maximilian University Munich, 81377 Munich, Germany.
| | - Christian Bach
- Department of Urology, RWTH Aachen University, 52074 Aachen, Germany.
| | - Ruth Knuechel
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Michael Rose
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Nadine T Gaisa
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
| |
Collapse
|
19
|
Silva G, Marins M, Chaichanasak N, Yoon Y, Fachin AL, Pinhanelli VC, Regasini LO, dos Santos MB, Ayusso GM, Marques BDC, Wu WW, Phue JN, Shen RF, Baek SJ. Trans-chalcone increases p53 activity via DNAJB1/HSP40 induction and CRM1 inhibition. PLoS One 2018; 13:e0202263. [PMID: 30118500 PMCID: PMC6097677 DOI: 10.1371/journal.pone.0202263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/31/2018] [Indexed: 11/19/2022] Open
Abstract
Naturally-occurring chalcones and synthetic chalcone analogues have been demonstrated to have many biological effects, including anti-inflammatory, anti-malarial, anti-fungal, and anti-oxidant/anti-cancerous activities. Compared to other chalcones, trans-chalcone exhibits superior inhibitory activity in cancer cell growth as shown via in vitro assays, and exerts anti-cancerous effects via the activation of the p53 tumor suppressor protein. Thus, characterization of the specific mechanisms, by which trans-chalcone activates p53, can aid development of new chemotherapeutic drugs that can be used individually or synergistically with other drugs. In this report, we found that trans-chalcone modulates many p53 target genes, HSP40 being the most induced gene in the RNA-Seq data using trans-chalcone-treated cells. CRM1 is also inhibited by trans-chalcone, resulting in the accumulation of p53 and other tumor suppressor proteins in the nucleus. Similar effects were seen using trans-chalcone derivatives. Overall, trans-chalcone could provide a strong foundation for the development of chalcone-based anti-cancer drugs.
Collapse
Affiliation(s)
- Gabriel Silva
- Biotechnology Unit, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Mozart Marins
- Biotechnology Unit, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
- Medicine School, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Nadda Chaichanasak
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Yongdae Yoon
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Ana Lúcia Fachin
- Biotechnology Unit, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
- Medicine School, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | | | - Luis Octávio Regasini
- Department of Chemistry and Environmental Chemistry, São Paulo State University (UNESP), São Paulo, Brazil
| | - Mariana Bastos dos Santos
- Department of Chemistry and Environmental Chemistry, São Paulo State University (UNESP), São Paulo, Brazil
| | - Gabriela Miranda Ayusso
- Department of Chemistry and Environmental Chemistry, São Paulo State University (UNESP), São Paulo, Brazil
| | | | - Wells W. Wu
- Facility for Biotechnology Resources, CBER, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Je-Nie Phue
- Facility for Biotechnology Resources, CBER, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Rong-Fong Shen
- Facility for Biotechnology Resources, CBER, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Seung Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
20
|
XZ-1 regulates cell apoptosis of gastric epithelial dysplasia via NF-κB/p53/Ki67 signaling pathway. Biosci Rep 2018; 38:BSR20171529. [PMID: 29588340 PMCID: PMC5997797 DOI: 10.1042/bsr20171529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/31/2018] [Accepted: 03/26/2018] [Indexed: 11/17/2022] Open
Abstract
We aimed to determine the effect of ‘Xiaozeng No. 1’ (XZ-1) on cellular apoptosis changes of gastric epithelial dysplasia (GED) and to explore the underlying mechanism. Specimens taken from the pyloric area of the stomachs from rats in each group were subjected to Hematoxylin and Eosin (H&E) staining for pathological examination, TUNEL staining for apoptosis detection, and Western blot analysis for apoptosis-related proteins. The results showed that XZ-1 decreased GED incidence and enhanced gastric epithelial apoptosis. Furthermore, XZ-1 up-regulated the proapoptotic proteins including cleaved caspases (cysteine-dependent aspartate-specific protease) (-3, -8, and -9), Fas, Bax, and Bid, and facilitated the release of cytochrome c from mitochondria to the cytoplasm. Interestingly, XZ-1 enhanced protein expression of NF-κB p65, Ki67, and p53. Moreover, inhibition of NF-κB pathway suppressed the XZ-induced p53 expression, whereas inhibition of NF-κB or p53 pathway suppressed the XZ-induced Ki67. More importantly, inhibition of NF-κB or p53 pathway attenuated the XZ-1-mediated induction of gastric epithelial apoptosis and decline of GED incidence. Collectively, our results demonstrated that XZ-1, almost equivalent effect exerted by the positive control Retin-A, dramatically decreased GED incidence and enhanced gastric epithelial apoptosis. Meanwhile, XZ-1 activated the NF-κB/p53/Ki67-apoptosis signaling pathway, which might be one of the mechanisms whereby XZ-1 reversed GED.
Collapse
|
21
|
Zhou R, Wu J, Tang X, Wei X, Ju C, Zhang F, Sun J, Shuai D, Zhang Z, Liu Q, Lv XB. Histone deacetylase inhibitor AR-42 inhibits breast cancer cell growth and demonstrates a synergistic effect in combination with 5-FU. Oncol Lett 2018; 16:1967-1974. [PMID: 30008890 DOI: 10.3892/ol.2018.8854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/06/2018] [Indexed: 12/30/2022] Open
Abstract
AR-42 is a member of a novelly discovered class of phenylbutyrate-derived histone deacetylase inhibitors, and has a number of antitumor effects in a variety of tumor types; however, the role of AR-42 and its possible mechanisms have not been reported in the treatment of breast cancer. The aim of the present study was to investigate the antitumor effects of AR-42 and its associated mechanisms in breast cancer. MTT assays and colony formation assays were conducted to measure the proliferation of MCF-7 cells, and flow cytometry was used to analyze cell apoptosis. The results revealed that AR-42 induced cell apoptosis and suppressed cell growth in a dose- and time-dependent manner. Mechanistically, AR-42 treatment increased the acetylation of the p53 protein and prolonged the half-life of the p53 protein; furthermore, AR-42 treatment upregulated p21 and PUMA expression. Notably, AR-42 had a synergistic effect on MCF-7 cells in combination with fluorouracil, which is one of the most commonly used chemotherapeutic agents. In conclusion, the results indicated that AR-42 inhibits breast cancer cell proliferation and induces apoptosis, indicating that AR-42 is a potential therapeutic agent.
Collapse
Affiliation(s)
- Ruihao Zhou
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China.,First Clinical Department, Medical School of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Juan Wu
- Guangzhou Key Laboratory of Translational Medicine on Malignant Tumor Treatment, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Xiaofeng Tang
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Xin Wei
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Cheng Ju
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Feifei Zhang
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Jun Sun
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Deyong Shuai
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Zhiping Zhang
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Qiong Liu
- Department of Cardiovascular Medicine, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiao-Bin Lv
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| |
Collapse
|
22
|
Hasche D, Vinzón SE, Rösl F. Cutaneous Papillomaviruses and Non-melanoma Skin Cancer: Causal Agents or Innocent Bystanders? Front Microbiol 2018; 9:874. [PMID: 29770129 PMCID: PMC5942179 DOI: 10.3389/fmicb.2018.00874] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
There is still controversy in the scientific field about whether certain types of cutaneous human papillomaviruses (HPVs) are causally involved in the development of non-melanoma skin cancer (NMSC). Deciphering the etiological role of cutaneous HPVs requires - besides tissue culture systems - appropriate preclinical models to match the obtained results with clinical data from affected patients. Clear scientific evidence about the etiology and underlying mechanisms involved in NMSC development is fundamental to provide reasonable arguments for public health institutions to classify at least certain cutaneous HPVs as group 1 carcinogens. This in turn would have implications on fundraising institutions and health care decision makers to force - similarly as for anogenital cancer - the implementation of a broad vaccination program against "high-risk" cutaneous HPVs to prevent NMSC as the most frequent cancer worldwide. Precise knowledge of the multi-step progression from normal cells to cancer is a prerequisite to understand the functional and clinical impact of cofactors that affect the individual outcome and the personalized treatment of a disease. This overview summarizes not only recent arguments that favor the acceptance of a viral etiology in NMSC development but also reflects aspects of causality in medicine, the use of empirically meaningful model systems and strategies for prevention.
Collapse
Affiliation(s)
- Daniel Hasche
- Division of Viral Transformation Mechanisms, Research Program "Infection, Inflammation and Cancer", German Cancer Research Center, Heidelberg, Germany
| | - Sabrina E Vinzón
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| | - Frank Rösl
- Division of Viral Transformation Mechanisms, Research Program "Infection, Inflammation and Cancer", German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
23
|
Tiys ES, Ivanisenko TV, Demenkov PS, Ivanisenko VA. FunGeneNet: a web tool to estimate enrichment of functional interactions in experimental gene sets. BMC Genomics 2018; 19:76. [PMID: 29504895 PMCID: PMC5836822 DOI: 10.1186/s12864-018-4474-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Estimation of functional connectivity in gene sets derived from genome-wide or other biological experiments is one of the essential tasks of bioinformatics. A promising approach for solving this problem is to compare gene networks built using experimental gene sets with random networks. One of the resources that make such an analysis possible is CrossTalkZ, which uses the FunCoup database. However, existing methods, including CrossTalkZ, do not take into account individual types of interactions, such as protein/protein interactions, expression regulation, transport regulation, catalytic reactions, etc., but rather work with generalized types characterizing the existence of any connection between network members. Results We developed the online tool FunGeneNet, which utilizes the ANDSystem and STRING to reconstruct gene networks using experimental gene sets and to estimate their difference from random networks. To compare the reconstructed networks with random ones, the node permutation algorithm implemented in CrossTalkZ was taken as a basis. To study the FunGeneNet applicability, the functional connectivity analysis of networks constructed for gene sets involved in the Gene Ontology biological processes was conducted. We showed that the method sensitivity exceeds 0.8 at a specificity of 0.95. We found that the significance level of the difference between gene networks of biological processes and random networks is determined by the type of connections considered between objects. At the same time, the highest reliability is achieved for the generalized form of connections that takes into account all the individual types of connections. By taking examples of the thyroid cancer networks and the apoptosis network, it is demonstrated that key participants in these processes are involved in the interactions of those types by which these networks differ from random ones. Conclusions FunGeneNet is a web tool aimed at proving the functionality of networks in a wide range of sizes of experimental gene sets, both for different global networks and for different types of interactions. Using examples of thyroid cancer and apoptosis networks, we have shown that the links over-represented in the analyzed network in comparison with the random ones make possible a biological interpretation of the original gene/protein sets. The FunGeneNet web tool for assessment of the functional enrichment of networks is available at http://www-bionet.sscc.ru/fungenenet/. Electronic supplementary material The online version of this article (10.1186/s12864-018-4474-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Evgeny S Tiys
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia. .,Laboratory of Computer Genomics, Novosibirsk State University, Pirogova Str. 2, 630090, Novosibirsk, Russia.
| | - Timofey V Ivanisenko
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia.,Laboratory of Computer Genomics, Novosibirsk State University, Pirogova Str. 2, 630090, Novosibirsk, Russia
| | - Pavel S Demenkov
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia
| | - Vladimir A Ivanisenko
- The Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Prospekt Lavrentyeva 10, 630090, Novosibirsk, Russia
| |
Collapse
|
24
|
Hasche D, Stephan S, Braspenning-Wesch I, Mikulec J, Niebler M, Gröne HJ, Flechtenmacher C, Akgül B, Rösl F, Vinzón SE. The interplay of UV and cutaneous papillomavirus infection in skin cancer development. PLoS Pathog 2017; 13:e1006723. [PMID: 29190285 PMCID: PMC5708609 DOI: 10.1371/journal.ppat.1006723] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Cutaneous human papillomaviruses (HPVs) are considered as cofactors for non-melanoma skin cancer (NMSC) development, especially in association with UVB. Extensively studied transgenic mouse models failed to mimic all aspects of virus-host interactions starting from primary infection to the appearance of a tumor. Using the natural model Mastomys coucha, which reflects the human situation in many aspects, we provide the first evidence that only UVB and Mastomys natalensis papillomavirus (MnPV) infection strongly promote NMSC formation. Using UVB exposures that correspond to UV indices of different geographical regions, irradiated animals developed either well-differentiated keratinizing squamous cell carcinomas (SCCs), still supporting productive infections with high viral loads and transcriptional activity, or poorly differentiated non-keratinizing SCCs almost lacking MnPV DNA and in turn, early and late viral transcription. Intriguingly, animals with the latter phenotype, however, still showed strong seropositivity, clearly verifying a preceding MnPV infection. Of note, the mere presence of MnPV could induce γH2AX foci, indicating that viral infection without prior UVB exposure can already perturb genome stability of the host cell. Moreover, as shown both under in vitro and in vivo conditions, MnPV E6/E7 expression also attenuates the excision repair of cyclobutane pyrimidine dimers upon UVB irradiation, suggesting a viral impact on the DNA damage response. While mutations of Ras family members (e.g. Hras, Kras, and Nras) were absent, the majority of SCCs harbored-like in humans-Trp53 mutations especially at two hot-spots in the DNA-binding domain, resulting in a loss of function that favored tumor dedifferentiation, counter-selective for viral maintenance. Such a constellation provides a reasonable explanation for making continuous viral presence dispensable during skin carcinogenesis as observed in patients with NMSC.
Collapse
Affiliation(s)
- Daniel Hasche
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sonja Stephan
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ilona Braspenning-Wesch
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julita Mikulec
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Virus-associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Niebler
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann-Josef Gröne
- Division of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Baki Akgül
- Institute of Virology, University of Cologne, Cologne, Germany
| | - Frank Rösl
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabrina E. Vinzón
- Division of Viral Transformation Mechanisms, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
25
|
Chang YL, Hsu YJ, Chen Y, Wang YW, Huang SM. Theophylline exhibits anti-cancer activity via suppressing SRSF3 in cervical and breast cancer cell lines. Oncotarget 2017; 8:101461-101474. [PMID: 29254178 PMCID: PMC5731888 DOI: 10.18632/oncotarget.21464] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/18/2017] [Indexed: 11/25/2022] Open
Abstract
Caffeine, theophylline, and theobromine are the most well-known members of methylxanthines. Caffeine-induced serine/arginine-rich splicing factor 2, SRSF2, and SRSF3 are required for the alternative splicing of a subset of cancer-associated genes. However, it remains to be investigated whether and how theophylline and theobromine as well as caffeine exert their antitumor effects through mediating the alternative splicing process. Here, we reveal that theophylline down-regulated SRSF3 expression and switched p53 from alpha into a beta isoform as caffeine did in HeLa and MCF-7 cells via the reverse-transcriptase polymerase chain reaction and Western blot analysis. Further functional studies show that theophylline induced cellular apoptosis, senescence, and decreased colony formation. Interestingly, theophylline had a suppressive effect on cellular proliferation, whereas caffeine enhanced cellular proliferation rates via the 5-bromo-2-deoxyuridine analysis. Theophylline and caffeine had no effect on MCF-10A cells, which is a normal breast cell line. Our results provide an insight that theophylline as well as caffeine could be repurposed as antitumor leading compounds via the downregulation of splicing factor SRSF3 and its target genes.
Collapse
Affiliation(s)
- Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yu-Juei Hsu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Ying Chen
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yi-Wen Wang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
26
|
Huang C, Li R, Zhang Y, Gong J. Amarogentin Induces Apoptosis of Liver Cancer Cells via Upregulation of p53 and Downregulation of Human Telomerase Reverse Transcriptase in Mice. Technol Cancer Res Treat 2017; 16:546-558. [PMID: 27402632 PMCID: PMC5665146 DOI: 10.1177/1533034616657976] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 04/27/2016] [Accepted: 06/06/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Amarogentin has been reported to have a preventive effect on liver cancer via inducing cancer cell apoptosis. We attempted to elucidate the roles of p53-associated apoptosis pathways in the chemopreventive mechanism of amarogentin. The findings of this study will facilitate the development of a novel supplementary strategy for the treatment of liver cancer. MATERIALS AND METHODS The purity of amarogentin was assessed by high-performance liquid chromatography. The inhibitory ratios of the liver cell lines were determined using a Cell Counting Kit-8 following treatment with a gradient concentration of amarogentin. Cell apoptosis was detected by flow cytometry using annexin V-fluorescein isothiocyanate/propidium iodide kits. The gene and protein expression of p53-associated molecules, such as Akt, human telomerase reverse transcriptase, RelA, and p38, was detected by real-time quantitative polymerase chain reaction, Western blotting, and immunohistochemical staining in liver cancer cells and mouse tumor tissues after treatment with amarogentin. RESULTS The inhibitory effect of amarogentin on cell proliferation was more obvious in liver cancer cells, and amarogentin was more likely to induce the apoptosis of liver cancer cells than that of normal liver cells. The gene and protein expression levels of Akt, RelA, and human telomerase reverse transcriptase were markedly higher in the control group than in the preventive group and treatment groups. Only the expression of human telomerase reverse transcriptase was downregulated, accompanied by the upregulation of p53. CONCLUSION The results of our study suggest that amarogentin promotes apoptosis of liver cancer cells by the upregulation of p53 and downregulation of human telomerase reverse transcriptase and prevents the malignant transformation of these cells.
Collapse
Affiliation(s)
- Chun Huang
- Chongqing Key Laboratory of Hepatobiliary Surgery, Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Division of Basic Medical Science, Chongqing Three Gorges Medical College, Chongqing, Wanzhou, People’s Republic of China
| | - Runqin Li
- Division of Basic Medical Science, Chongqing Three Gorges Medical College, Chongqing, Wanzhou, People’s Republic of China
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yinglin Zhang
- Department of Hepatobiliary Surgery, The Third Hospital of Mianyang, Mianyang, Sichuan, People’s Republic of China
| | - Jianping Gong
- Chongqing Key Laboratory of Hepatobiliary Surgery, Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
27
|
Jiang C, Liu G, Luckhardt T, Antony V, Zhou Y, Carter AB, Thannickal VJ, Liu RM. Serpine 1 induces alveolar type II cell senescence through activating p53-p21-Rb pathway in fibrotic lung disease. Aging Cell 2017; 16:1114-1124. [PMID: 28722352 PMCID: PMC5595683 DOI: 10.1111/acel.12643] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 12/23/2022] Open
Abstract
Senescence of alveolar type 2 (ATII) cells, progenitors of the alveolar epithelium, is implicated in the pathogeneses of idiopathic pulmonary fibrosis (IPF), an aging‐related progressive fatal lung disorder with unknown etiology. The mechanism underlying ATII cell senescence in fibrotic lung diseases, however, remains poorly understood. In this study, we report that ATII cells in IPF lungs express higher levels of serpine 1, also known as plasminogen activator inhibitor 1 (PAI‐1), and cell senescence markers p21 and p16, compared to ATII cells in control lungs. Silencing PAI‐1 or inhibition of PAI‐1 activity in cultured rat ATII (L2) cells leads to decreases in p53 serine 18 phosphorylation (p53S18P), p53 and p21 protein expressions; an increase in retinoblastoma protein phosphorylation (ppRb); and a reduction in the sensitivity to bleomycin‐ and doxorubicin‐induced senescence. Silencing p53, on the other hand, abrogates PAI‐1 protein‐stimulated p21 expression and cell senescence. In vivo studies, using ATII cell‐specific PAI‐1 conditional knockout mouse model generated recently in this laboratory, further support the role of PAI‐1 in the activation of p53‐p21‐Rb cell cycle repression pathway, ATII cell senescence, and lung fibrosis induced by bleomycin. This study reveals a novel function of PAI‐1 in regulation of cell cycle and suggests that elevation of PAI‐1 contributes importantly to ATII cell senescence in fibrotic lung diseases.
Collapse
Affiliation(s)
- Chunsun Jiang
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - Tracy Luckhardt
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - Veena Antony
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - Yong Zhou
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - A. Brent Carter
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - Victor J. Thannickal
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care; Department of Medicine; School of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| |
Collapse
|
28
|
Carter CA, Oronsky BT, Roswarski J, Oronsky AL, Oronsky N, Scicinski J, Lybeck H, Kim MM, Lybeck M, Reid TR. No patient left behind: The promise of immune priming with epigenetic agents. Oncoimmunology 2017; 6:e1315486. [PMID: 29123948 PMCID: PMC5665084 DOI: 10.1080/2162402x.2017.1315486] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023] Open
Abstract
Checkpoint inhibitors, monoclonal antibodies that inhibit PD-1 or CTLA-4, have revolutionized the treatment of multiple cancers. Despite the enthusiasm for the clinical successes of checkpoint inhibitors, and immunotherapy, in general, only a minority of patients with specific tumor types actually benefit from treatment. Emerging evidence implicates epigenetic alterations as a mechanism of clinical resistance to immunotherapy. This review presents evidence for that association, summarizes the epi-based mechanisms by which tumors evade immunogenic cell death, discusses epigenetic modulation as a component of an integrated strategy to boost anticancer T cell effector function in relation to a tumor immunosuppression cycle and, finally, makes the case that the success of this no-patient-left-behind strategy critically depends on the toxicity profile of the epigenetic agent(s).
Collapse
Affiliation(s)
- Corey A Carter
- Walter Reed National Military Medical Center, Murtha Cancer Center, Bethesda, MD, USA
| | | | - Joseph Roswarski
- Walter Reed National Military Medical Center, Murtha Cancer Center, Bethesda, MD, USA
| | | | | | | | - Harry Lybeck
- University of Helsinki, Department of Physiology, Helsinki, Finland
| | - Michelle M Kim
- University of Michigan, Department of Radiation Oncology, Ann Arbor, MI, USA
| | | | - Tony R Reid
- University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
29
|
Hsieh HH, Chen YC, Jhan JR, Lin JJ. The serine protease inhibitor serpinB2 binds and stabilizes p21 in senescent cells. J Cell Sci 2017; 130:3272-3281. [PMID: 28794016 DOI: 10.1242/jcs.204974] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/03/2017] [Indexed: 01/18/2023] Open
Abstract
SerpinB2 is a serine protease inhibitor also known as plasminogen activator inhibitor type 2 (PAI-2). It has been well documented that serpinB2 is an inhibitor of urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA). Interestingly, serpinB2 levels are increased in senescent cells and serpinB2 is thus considered a senescence biomarker. In this study, by mimicking the elevated levels of serpinB2 in senescent cells, proliferating human fibroblasts were induced into senescence. Senescence induced by serpinB2 did not relate to its extracellular function, as inhibition of serpinB2 secretion, exogenous introduced serpinB2, or a serpinB2 mutant that failed to bind to its extracellular target uPA did not affect senescence. We also showed that serpinB2 is a direct downstream target of p53 that is activated by the DNA damage response pathway. Significantly, serpinB2 bound to and stabilized p21 to mediate senescence in a proteasome-independent manner, indicating that serpinB2 has a direct role in senescence. Thus, this study reveals a unique mechanism by which serpinB2 maintains senescence through stabilization of p21 protein levels.
Collapse
Affiliation(s)
- Hsi-Hsien Hsieh
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan 112
| | - Ying-Chieh Chen
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan 112
| | - Jing-Ru Jhan
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan 112
| | - Jing-Jer Lin
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan 112 .,Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan 100
| |
Collapse
|
30
|
Lopes-Aguiar L, Costa EFD, Nogueira GAS, Lima TRP, Visacri MB, Pincinato EC, Calonga L, Mariano FV, de Almeida Milani Altemani AM, Altemani JMC, Coutinho-Camillo CM, Ribeiro Alves MAVF, Moriel P, Ramos CD, Chone CT, Lima CSP. XPD c.934G>A polymorphism of nucleotide excision repair pathway in outcome of head and neck squamous cell carcinoma patients treated with cisplatin chemoradiation. Oncotarget 2017; 8:16190-16201. [PMID: 26918827 PMCID: PMC5369956 DOI: 10.18632/oncotarget.7668] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/16/2016] [Indexed: 11/25/2022] Open
Abstract
This study aimed to investigate the associations of XPC c.2815A>C, XPD c.934G>A and c.2251A>C, XPF c.2505T>C and ERCC1 c.354C>T single nucleotide polymorphisms (SNPs) of nucleotide excision repair pathway in outcome of head and neck squamous cell carcinoma (HNSCC) patients treated with cisplatin (CDDP) chemoradiation. Patients with XPC c.2815AC or CC and XPD c.934GA or AA genotypes had 0.20 and 0.38 less chances of presenting moderate/severe ototoxicity and nausea, respectively. Patients with XPD c.934AA and c.2251AC or CC genotypes had 8.64, 12.29 and 3.55 more chances of achieving complete response (CR), consistent ototoxicity and nephrotoxicity, respectively. AA haplotype of XPD and ACT haplotype of XPD and ERCC1 SNPs were associated with 9.30 and 3.41 more chances of achieving CR and consistent nephrotoxicity, respectively. At 24 months of follow-up, patients with XPD c.934AA genotype presented lower progression-free survival and overall survival in Kaplan-Meier estimates, and differences between groups remained the same in univariate Cox analysis. Patients with XPD c.934AA genotype had 2.13 and 2.04 more risks of presenting tumor progression and death than others in multivariate Cox analysis. Our data present preliminary evidence that XPC c.2815A>C, XPD c.934G>A and c.2251A>C, and ERCC1 c.354C>T SNPs alter outcome of HNSCC patients treated with CDDP chemoradiation.
Collapse
Affiliation(s)
- Leisa Lopes-Aguiar
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | - Tathiane Regine Penna Lima
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Marília Berlofa Visacri
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Eder Carvalho Pincinato
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Luciane Calonga
- Department of Ophthalmology and Otorhinolaryngology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Fernanda Viviane Mariano
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | | | | | - Patrícia Moriel
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Celso Dario Ramos
- Department of Radiology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Carlos Takahiro Chone
- Department of Ophthalmology and Otorhinolaryngology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Carmen Silvia Passos Lima
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
31
|
Seo M, Kim MS, Jang A, Chung HJ, Noh Y, Kim DH, Lee J, Ko K, Myung SC. Epigenetic suppression of the anti-aging gene KLOTHO in human prostate cancer cell lines. Anim Cells Syst (Seoul) 2017; 21:223-232. [PMID: 30460073 PMCID: PMC6138301 DOI: 10.1080/19768354.2017.1336112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 04/30/2017] [Accepted: 05/04/2017] [Indexed: 12/23/2022] Open
Abstract
KLOTHO was originally identified as an aging-suppressor gene that causes a human aging-like phenotype when tested in KLOTHO-deficient-mice. Recent evidence suggests that KLOTHO functions as a tumor suppressor by inhibiting Wnt signaling. KLOTHO gene silencing, including DNA methylation, has been observed in some human cancers. Aberrant activation of Wnt signaling plays a significant role in aging, and its silencing may be related to prostate cancer and other types of cancers. Thus, we investigated whether the expression of the anti-aging gene KLOTHO was associated with epigenetic changes in prostate cancer cell lines. KLOTHO mRNA was detected in the 22Rv1 cell line while it was not detected in DU145 and PC-3 cell lines. The restoration of KLOTHO mRNA in the DU145 and PC-3 cell lines was induced with a DNA methyltransferase inhibitor. Methylation-specific PCR was performed to determine the specific CpG sites in the KLOTHO promoter responsible for expression. In addition, the level of methylation was assessed in each CpG by performing bisulfite sequencing and quantitative pyrosequencing analysis. The results suggested a remarkable inverse relationship between KLOTHO expression and promoter methylation in prostate cancer cell lines.
Collapse
Affiliation(s)
- Minkyu Seo
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Min Su Kim
- Department of Urology, Seoul Medical Center, Seoul, Republic of Korea
| | - Ara Jang
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Republic of Korea
| | - Hyun Joo Chung
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Republic of Korea
| | - Yoohun Noh
- Department of Anatomy and Cell Biology and Neurology, College of Medicine, Chung-Ang University, Seoul, Korea.,Famenity Biomedical Research Center, Famenity, Inc., Gyeonggi, Korea
| | - Do-Hee Kim
- Natural Pharmaceutical R&D Center, Naturesense, Inc., Gyeonggi, Korea
| | - Jaehyouk Lee
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Republic of Korea
| | - Kisung Ko
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Soon Chul Myung
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Advanced Urogenital Diseases Research Center, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
32
|
Abstract
Increasing epidemiological and experimental evidence has demonstrated an inverse relationship between the consumption of plant foods and the incidence of chronic diseases, including cancer. Microcomponents that are naturally present in such foods, especially polyphenols, are responsible for the benefits to human health. Resveratrol is a diet-derived cancer chemopreventive agent with high therapeutic potential, as demonstrated by different authors. The aim of this review is to collect and present recent evidence from the literature regarding resveratrol and its effects on cancer prevention, molecular signaling (especially regarding the involvement of p53 protein), and therapeutic perspectives with an emphasis on clinical trial results to date.
Collapse
|
33
|
Hao L, Leng J, Xiao R, Kingsley T, Li X, Tu Z, Yang X, Deng X, Xiong M, Xiong J, Zhang Q. Bioinformatics analysis of the prognostic value of Tripartite Motif 28 in breast cancer. Oncol Lett 2017; 13:2670-2678. [PMID: 28454449 PMCID: PMC5403292 DOI: 10.3892/ol.2017.5764] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/06/2016] [Indexed: 02/06/2023] Open
Abstract
Tripartite motif containing 28 (TRIM28) is a transcriptional regulator acting as an essential corepressor for Krüppel-associated box zinc finger domain-containing proteins in multiple tissue and cell types. An increasing number of studies have investigated the function of TRIM28; however, its prognostic value in breast cancer (BC) remains unclear. In the present study, the expression of TRIM28 was identified to be significantly higher in cancerous compared with healthy tissue samples. Furthermore, it was demonstrated that TRIM28 expression was significantly correlated with several clinicopathological characteristics of patients with BC, such as p53 mutation, tumor recurrence and Elston grade of the tumor. In addition, a protein-protein interaction network was created to illustrate the interactions of TRIM28 with other proteins. The prognostic value of TRIM28 in patients with BC was investigated using the Kaplan-Meier Plotter database, which revealed that high expression of TRIM28 is a predictor of poor prognosis in patients with BC. In conclusion, the results of the present study indicate that TRIM28 provides a survival advantage to patients with BC and is a novel prognostic biomarker, in addition to being a therapeutic target for the treatment of BC.
Collapse
Affiliation(s)
- Ling Hao
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jun Leng
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ruijing Xiao
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Tembo Kingsley
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xinran Li
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhenbo Tu
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xiangyong Yang
- College of Engineering Technology, Hubei University of Technology, Wuhan, Hubei 430068, P.R. China
| | - Xinzhou Deng
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Meng Xiong
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qiuping Zhang
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
34
|
Lang PY, Nanjangud GJ, Sokolsky-Papkov M, Shaw C, Hwang D, Parker JS, Kabanov AV, Gershon TR. ATR maintains chromosomal integrity during postnatal cerebellar neurogenesis and is required for medulloblastoma formation. Development 2016; 143:4038-4052. [PMID: 27803059 PMCID: PMC5117143 DOI: 10.1242/dev.139022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/16/2016] [Indexed: 02/01/2023]
Abstract
Microcephaly and medulloblastoma may both result from mutations that compromise genomic stability. We report that ATR, which is mutated in the microcephalic disorder Seckel syndrome, sustains cerebellar growth by maintaining chromosomal integrity during postnatal neurogenesis. Atr deletion in cerebellar granule neuron progenitors (CGNPs) induced proliferation-associated DNA damage, p53 activation, apoptosis and cerebellar hypoplasia in mice. Co-deletions of either p53 or Bax and Bak prevented apoptosis in Atr-deleted CGNPs, but failed to fully rescue cerebellar growth. ATR-deficient CGNPs had impaired cell cycle checkpoint function and continued to proliferate, accumulating chromosomal abnormalities. RNA-Seq demonstrated that the transcriptional response to ATR-deficient proliferation was highly p53 dependent and markedly attenuated by p53 co-deletion. Acute ATR inhibition in vivo by nanoparticle-formulated VE-822 reproduced the developmental disruptions seen with Atr deletion. Genetic deletion of Atr blocked tumorigenesis in medulloblastoma-prone SmoM2 mice. Our data show that p53-driven apoptosis and cell cycle arrest - and, in the absence of p53, non-apoptotic cell death - redundantly limit growth in ATR-deficient progenitors. These mechanisms may be exploited for treatment of CGNP-derived medulloblastoma using ATR inhibition.
Collapse
Affiliation(s)
- Patrick Y Lang
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gouri J Nanjangud
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Research Center, New York, NY 10021, USA
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Christine Shaw
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Research Center, New York, NY 10021, USA
| | - Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joel S Parker
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Timothy R Gershon
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
35
|
Lee YY, Wang CT, Huang SKH, Wu WJ, Huang CN, Li CC, Chan TC, Liang PI, Hsing CH, Li CF. Downregulation of RNF128 Predicts Progression and Poor Prognosis in Patients with Urothelial Carcinoma of the Upper Tract and Urinary Bladder. J Cancer 2016; 7:2187-2196. [PMID: 27994654 PMCID: PMC5166527 DOI: 10.7150/jca.16798] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/04/2016] [Indexed: 01/10/2023] Open
Abstract
Background: The TP53 tumor suppressor gene plays a crucial role in the carcinogenesis of many malignancies, including urothelial carcinoma (UC). Overexpression of p53 is associated with poor prognosis in UC. Recently, RING finger protein 128 (RNF128) was shown to be involved in p53-induced apoptosis, forming a negative feedback loop. However, the significance of RNF128 in patients with UC remains unknown. In this study, our aim was to evaluate the expression of RNF128 in UC and to assess its predictive and prognostic value in a well-established cohort. Methods: Through data mining from a published transcriptome (GSE31684), RNF128 was identified as the most differentially expressed gene in UC among those associated with negative regulation of the cytokine biosynthetic process (GO:0042036). Its immunoexpression was further evaluated using the H-scores of 340 patients with upper urinary tract UC (UTUC) and 295 with urinary bladder UC (UBUC). The scores were correlated with clinicopathological features, disease-specific survival (DSS) and metastasis-free survival (MeFS). We also used Western blot analysis to evaluate RNF128 protein expression in human urothelial cell (HUC) lines. Results: Downregulation of RNF128 expression was significantly associated with advanced pT stage (p<0.001), high histological grade (UTUC, p<0.001; UBUC, p=0.035), nodal metastasis (UTUC, p<0.001; UBUC, p=0.001), vascular invasion (UTUC, p<0.001; UBUC, p=0.008) and high mitotic rate (UTUC, p=0.003; UBUC, p=0.023). Low expression of RNF128 was an adverse prognosticator for DSS (UTUC, p<0.0001; UBUC, p<0.0001) and MeFS (UTUC, p<0.0001; UBUC, p=0.0002). Moreover, low expression was predictive of poor DSS (UTUC, p=0.006; UBUC, p=0.003) and MeFS (UTUC, p=0.009; UBUC, p=0.036) in multivariate comparisons. Western blot analysis showed that the RNF128 protein was downregulated in invasive urothelial cancer cell lines. Conclusion: Our findings showed that downregulation of RNF128 was correlated with cancer invasiveness and metastasis as well as reduced survival in patients with UTUC and UBUC, identifying RNF128 as a prognostic factor in UC.
Collapse
Affiliation(s)
- Yi-Ying Lee
- Department of Pathology, Chi Mei Medical Center, Liouying, Taiwan
- Institute of Biomedical Science National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chieh-Tien Wang
- Department of Pathology, Chi Mei Medical Center, Liouying, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology
| | | | - Wen-Jeng Wu
- Department of Urology, Faculty of Medicine, Kaohsiung Medical University
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital
| | - Chun-Nung Huang
- Department of Urology, Faculty of Medicine, Kaohsiung Medical University
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
| | - Ching-Chia Li
- Department of Urology, Faculty of Medicine, Kaohsiung Medical University
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital
| | - Ti-Chun Chan
- Institute of Biomedical Science National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Feng Li
- Division of Clinical Pathology, Chi Mei Medical Center, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
36
|
Pioglitazone induces cell growth arrest and activates mitochondrial apoptosis in human uterine leiomyosarcoma cells by a peroxisome proliferator-activated receptor γ-independent mechanism. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:37-48. [PMID: 27664035 DOI: 10.1007/s00210-016-1291-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) agonists, thiazolidinediones, including pioglitazone (PIO) exhibit anti-tumour activities in cancer cells. The present study investigates the effects of PIO on cell proliferation and apoptosis in SK-UT-1 cells, a human uterine leiomyosarcoma cell line, and human uterine smooth muscle cells (HUtSMC). The proliferation and viability of SK-UT-1 cells treated with vehicle or PIO were assessed by cell counting and WST-1 assay. The activity of MEK/ERK and p38 MAPK signalling pathways and the expression of p53, the cyclin-dependent kinase inhibitor, p21, Bax, Bad and Bim proteins and cleaved caspase-3 were analysed by Western blotting. Quiescent SK-UT-1 cells intensively proliferate and display high levels of phosphorylated, activated MEK1/2, ERK1/2 and p38 MAPK. PIO (10 or 25 μM) induced time- and dose-dependently cell-growth arrest, reduced the cell numbers and effectively suppressed the over-activated MEK/ERK and p38 MAPK signalling pathways as evidenced by the abolished levels of phosphorylated MEK1/2, ERK1/2 and p38 MAPK. PIO activated the intrinsic apoptotic pathway, i.e. up-regulated the p53, p21, Bax and Bad proteins and cleaved caspase-3. PIO also reduced cell numbers of highly proliferative SK-UT-1 cells cultured in growth medium. The anti-proliferative and pro-apoptotic actions of PIO were not PPARγ dependent and exclusive for SK-UT-1 cells as PIO did not interfere with the proliferation of HUtSMC. The pronounced anti-tumorigenic effects of PIO in SK-UT-1 cells address an important issue about the relevance of the PPARγ agonist in the treatment of the human uterine leiomyosarcoma.
Collapse
|
37
|
miR-100 antagonism triggers apoptosis by inhibiting ubiquitination-mediated p53 degradation. Oncogene 2016; 36:1023-1037. [PMID: 27524417 DOI: 10.1038/onc.2016.270] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 06/13/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022]
Abstract
During tumourigenesis, p53 functions as 'the guardian of the genome' because p53-dependent apoptosis strongly regulates the fate of cancer cells. Therefore, p53 regulation must be sensitive and accurate. p53 activity is regulated through its ubiquitination and deubiquitination. However, the role of microRNA in ubiquitin-mediated p53 degradation has not been previously studied. Our previous studies indicated that miR-100 is required for apoptosis. In the current study, the mechanism of p53 protein ubiquitination mediated by miR-100 was characterized. An analysis of primary tumour samples from gastric cancer patients showed a significant correlation between miR-100 upregulation and primary human gastric tumourigenesis and progression. The in vivo and in vitro data indicated that miR-100 antagonism specifically induced the apoptosis of poorly differentiated gastric cancer cells but not non-cancerous gastric cells, indicating that miR-100 has a crucial role in regulating the progression of gastric tumours. In the regulation of p53-dependent apoptosis, miR-100 antagonism inhibited ubiquitin-mediated p53 protein degradation by activating RNF144B, an E3 ubiquitination ligase. Consequently, the miR-100-RNF144B-pirh2-p53-dependent pathway was initiated. Our findings highlight a novel mechanism of ubiquitin-mediated p53 protein degradation in apoptosis.
Collapse
|
38
|
Zhang JM, Feng FE, Wang QM, Zhu XL, Fu HX, Xu LP, Liu KY, Huang XJ, Zhang XH. Platelet-Derived Growth Factor-BB Protects Mesenchymal Stem Cells (MSCs) Derived From Immune Thrombocytopenia Patients Against Apoptosis and Senescence and Maintains MSC-Mediated Immunosuppression. Stem Cells Transl Med 2016; 5:1631-1643. [PMID: 27471307 DOI: 10.5966/sctm.2015-0360] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 05/13/2016] [Indexed: 12/12/2022] Open
Abstract
: Immune thrombocytopenia (ITP) is characterized by platelet destruction and megakaryocyte dysfunction. Mesenchymal stem cells (MSCs) from ITP patients (MSC-ITP) do not exhibit conventional proliferative abilities and thus exhibit defects in immunoregulation, suggesting that MSC impairment might be a mechanism involved in ITP. Platelet-derived growth factor (PDGF) improves growth and survival in various cell types. Moreover, PDGF promotes MSC proliferation. The aim of the present study was to analyze the effects of PDGF-BB on MSC-ITP. We showed that MSC-ITP expanded more slowly and appeared flattened and larger. MSC-ITP exhibited increased apoptosis and senescence compared with controls. Both the intrinsic and extrinsic pathways account for the enhanced apoptosis. P53 and p21 expression were upregulated in MSC-ITP, but inhibition of p53 with pifithrin-α markedly inhibited apoptosis and senescence. Furthermore, MSCs from ITP patients showed a lower capacity for inhibiting the proliferation of activated T cells inducing regulatory T cells (Tregs) and suppressing the synthesis of anti-glycoprotein (GP)IIb-IIIa antibodies. PDGF-BB treatment significantly decreased the expression of p53 and p21 and increased survivin expression in MSC-ITP. In addition, the apoptotic rate and number of senescent cells in ITP MSCs were reduced. Their impaired ability for inhibiting activated T cells, inducing Tregs, and suppressing the synthesis of anti-GPIIb-IIIa antibodies was restored after PDGF-BB treatment. In conclusion, we have demonstrated that PDGF-BB protects MSCs derived from ITP patients against apoptosis, senescence, and immunomodulatory defects. This protective effect of PDGF-BB is likely mediated via the p53/p21 pathway, thus potentially providing a new therapeutic approach for ITP. SIGNIFICANCE Immune thrombocytopenia (ITP) is characterized by platelet destruction and megakaryocyte dysfunction. Platelet-derived growth factor (PDGF) improves growth and survival in various cell types and promotes mesenchymal stem cell (MSC) proliferation. PDGF-BB protects MSCs derived from ITP patients against apoptosis, senescence, and immunomodulatory defects. This protective effect of PDGF-BB is likely mediated via the p53/p21 pathway, thus potentially providing a new therapeutic approach for ITP.
Collapse
Affiliation(s)
- Jia-Min Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, People's Republic of China
| | - Fei-Er Feng
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
| | - Qian-Ming Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
| | - Xiao-Lu Zhu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
| | - Hai-Xia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, People's Republic of China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, People's Republic of China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, People's Republic of China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, People's Republic of China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, People's Republic of China
| |
Collapse
|
39
|
Banerjee A, Thyagarajan K, Chatterjee S, Chakraborty P, Kesarwani P, Soloshchenko M, Al-Hommrani M, Andrijauskaite K, Moxley K, Janakiraman H, Scheffel MJ, Helke K, Armenson K, Palanisamy V, Rubinstein MP, Mayer EG, Cole DJ, Paulos CM, Christina-Voelkel-Johnson, Nishimura MI, Mehrotra S. Lack of p53 Augments Antitumor Functions in Cytolytic T Cells. Cancer Res 2016; 76:5229-5240. [PMID: 27466285 DOI: 10.1158/0008-5472.can-15-1798] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Repetitive stimulation of T-cell receptor (TCR) with cognate antigen results in robust proliferation and expansion of the T cells, and also imprints them with replicative senescence signatures. Our previous studies have shown that life-span and antitumor function of T cells can be enhanced by inhibiting reactive oxygen species (ROS) or intervening with ROS-dependent JNK activation that leads to its activation-induced cell death. Because tumor suppressor protein p53 is also a redox active transcription factor that regulates cellular ROS generation that triggers downstream factor-mediating apoptosis, we determined if p53 levels could influence persistence and function of tumor-reactive T cells. Using h3T TCR transgenic mice, with human tyrosinase epitope-reactive T cells developed on p53 knockout (KO) background, we determined its role in regulating antitumor T-cell function. Our data show that as compared with h3T cells, h3T-p53 KO T cells exhibited enhanced glycolytic commitment that correlated with increased proliferation, IFNγ secretion, cytolytic capacity, expression of stemness gene signature, and decreased TGF-β signaling. This increased effector function correlated to the improved control of subcutaneously established murine melanoma after adoptive transfer of p53-KO T cells. Pharmacological inhibition of human TCR-transduced T cells using a combination of p53 inhibitors also potentiated the T-cell effector function and improved persistence. Thus, our data highlight the key role of p53 in regulating the tumor-reactive T-cell response and that targeting this pathway could have potential translational significance in adoptive T-cell therapy. Cancer Res; 76(18); 5229-40. ©2016 AACR.
Collapse
Affiliation(s)
- Anirban Banerjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | | | - Shilpak Chatterjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Pravin Kesarwani
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | | | - Mazen Al-Hommrani
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | | | - Kelly Moxley
- Department of Surgery, Oncology Institute, Loyola University, Maywood, IL 60153
| | | | - Matthew J Scheffel
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Kristi Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Kent Armenson
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425
| | - Viswanathan Palanisamy
- Department of Oral Health Research, Medical University of South Carolina, Charleston, SC 29425
| | - Mark P Rubinstein
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Elizabeth-Garrett Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425
| | - David J Cole
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Chrystal M Paulos
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
| | | | - Michael I Nishimura
- Department of Surgery, Oncology Institute, Loyola University, Maywood, IL 60153
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
40
|
Abstract
Reactive oxygen species (ROS) and oxidative stress have long been linked to aging and diseases prominent in the elderly such as hypertension, atherosclerosis, diabetes and atrial fibrillation (AF). NADPH oxidases (Nox) are a major source of ROS in the vasculature and are key players in mediating redox signalling under physiological and pathophysiological conditions. In this review, we focus on the Nox-mediated ROS signalling pathways involved in the regulation of 'longevity genes' and recapitulate their role in age-associated vascular changes and in the development of age-related cardiovascular diseases (CVDs). This review is predicated on burgeoning knowledge that Nox-derived ROS propagate tightly regulated yet varied signalling pathways, which, at the cellular level, may lead to diminished repair, the aging process and predisposition to CVDs. In addition, we briefly describe emerging Nox therapies and their potential in improving the health of the elderly population.
Collapse
|
41
|
Liu QJ, Shen HL, Lin J, Xu XH, Ji ZG, Han X, Shang DH, Yang PQ. Synergistic roles of p53 and HIF1α in human renal cell carcinoma-cell apoptosis responding to the inhibition of mTOR and MDM2 signaling pathways. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:745-55. [PMID: 26937175 PMCID: PMC4762585 DOI: 10.2147/dddt.s88779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Introduction mTOR and MDM2 signaling pathways are frequently deregulated in cancer development, and inhibition of mTOR or MDM2 independently enhances carcinoma-cell apoptosis. However, responses to mTOR and MDM2 antagonists in renal cell carcinoma (RCC) remain unknown. Materials and methods A498 cells treated with MDM2 antagonist MI-319 and/or mTOR inhibitor rapamycin were employed in the present study. Cell apoptosis and Western blot analysis were performed. Results and conclusion We found that the MDM2 inhibitor MI-319 induced RCC cell apoptosis mainly dependent on p53 overexpression, while the mTOR antagonist rapamycin promoted RCC cell apoptosis primarily through upregulation of HIF1α expression. Importantly, strong synergistic effects of MI-319 and rapamycin combinations at relatively low concentrations on RCC cell apoptosis were observed. Depletion of p53 or HIF1α impaired both antagonist-elicited apoptoses to differential extents, corresponding to their expression changes responding to chemical treatments, and double knockdown of p53 and HIF1α remarkably hindered MI-319- or rapamycin-induced apoptosis, suggesting that both p53 and HIF1α are involved in MDM2 or mTOR antagonist-induced apoptosis. Collectively, we propose that concurrent activation of p53 and HIF1α may effectively result in cancer-cell apoptosis, and that combined MDM2 antagonists and mTOR inhibitors may be useful in RCC therapy.
Collapse
Affiliation(s)
- Qing-jun Liu
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hong-liang Shen
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jun Lin
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiu-hong Xu
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zheng-guo Ji
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiao Han
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Dong-hao Shang
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Pei-qian Yang
- Department of Urology Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
42
|
Su Z, Yang Z, Xu Y, Chen Y, Yu Q. MicroRNAs in apoptosis, autophagy and necroptosis. Oncotarget 2016; 6:8474-90. [PMID: 25893379 PMCID: PMC4496162 DOI: 10.18632/oncotarget.3523] [Citation(s) in RCA: 278] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/10/2015] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are endogenous 22 nt non-coding RNAs that target mRNAs for cleavage or translational repression. Numerous miRNAs regulate programmed cell death including apoptosis, autophagy and necroptosis. We summarize how miRNAs regulate apoptotic, autophagic and necroptotic pathways and cancer progression. We also discuss how miRNAs link different types of cell death.
Collapse
Affiliation(s)
- Zhenyi Su
- Department of Biochemistry and Molecular Biology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China.,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, China.,Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan 650118, China
| | - Yongqing Xu
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan 650118, China
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Qiang Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
43
|
Smonskey M, Lasorsa E, Rosario S, Kirk JS, Hernandez-Ilizaliturri FJ, Ellis L. EZH2 inhibition re-sensitizes multidrug resistant B-cell lymphomas to etoposide mediated apoptosis. Oncoscience 2016; 3:21-30. [PMID: 26973857 PMCID: PMC4751913 DOI: 10.18632/oncoscience.288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/22/2016] [Indexed: 01/02/2023] Open
Abstract
Reactivation of apoptotic pathways is an attractive strategy for patients with treatment-resistant B-cell lymphoma. The tumor suppressor, p53 is central for apoptotic response to multiple DNA damaging agents used to treat aggressive B-cell lymphomas, including etoposide. It has been demonstrated that etoposide induced DNA damage and therapeutic efficacy is enhanced by combination with inhibitors of the histone methyltransferase, enhancer of zeste homolog 2 (EZH2). Further, EZH2 was identified to regulate cell fate decisions in response to DNA damage. Using B-cell lymphoma cell lines resistant to etoposide induced cell death; we show that p53 is dramatically down regulated and MDMX, a negative regulator of p53, is significantly up regulated. However, these cell lines remain responsive to etoposide mediated DNA damage and exhibit cell cycle inhibition and induction of senescence. Furthermore, chemical inhibition of EZH2 directs DNA damage to a predominant p53 dependent apoptotic response associated with loss of MDMX and BCL-XL. These data provide confirmation of EZH2 in determining cell fate following DNA damage and propose a novel therapeutic strategy for patients with aggressive treatment-resistant B-cell lymphoma.
Collapse
Affiliation(s)
- Matthew Smonskey
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Elena Lasorsa
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Spencer Rosario
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA; Department of Molecular Pharmacology and Cancer Therapeutics, State University of New York at Buffalo, NY, USA
| | - Jason S Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Leigh Ellis
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA; Department of Molecular Pharmacology and Cancer Therapeutics, State University of New York at Buffalo, NY, USA
| |
Collapse
|
44
|
Tsai CW, Kao YT, Chiang IN, Wang JH, Young TH. Chitosan Treatment Delays the Induction of Senescence in Human Foreskin Fibroblast Strains. PLoS One 2015; 10:e0140747. [PMID: 26465338 PMCID: PMC4605659 DOI: 10.1371/journal.pone.0140747] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/30/2015] [Indexed: 12/25/2022] Open
Abstract
Fibroblasts have been extensively used as a model to study cellular senescence. The purpose of this study was to investigate whether the human foreskin fibroblast aging process could be regulated by using the biomaterial chitosan. Fibroblasts cultured on commercial tissue culture polystyrene (TCPS) entered senescence after 55–60 population doublings (PDs), and were accompanied by larger cell shape, higher senescence-associated β-galactosidase (SA β-gal) activity, lower proliferation capacity, and upregulation of senescence-associated molecular markers p21, p53, retinoblastoma (pRB), and p16. Before senescence was reached, PD48 cells were collected from TCPS and seeded on chitosan for three days (PD48-Cd3) to form multicellular spheroids. The protein expression of senescence-associated secretory phenotypes (SASPs) and senescence-associated molecular markers of these cells in PD48-Cd3 spheroids were downregulated significantly. Following chitosan treatment, fibroblasts reseeded on TCPS showed lower SA β-gal activity, increased cellular motility, and a higher proliferation ability of 70–75 PDs. These phenotypic changes were not accompanied by colonies forming in soft agar and a continuous decrease in the senescence-associated proteins p53 and pRB which act as a barrier to tumorigenesis. These results demonstrate that chitosan treatment could delay the induction of senescence which may be useful and safe for future tissue engineering applications.
Collapse
Affiliation(s)
- Ching-Wen Tsai
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan, No.1, Sec. 1, Jen - Ai Rd., Taipei 100, Taiwan
| | - Yu-Ting Kao
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan, No.1, Sec. 1, Jen - Ai Rd., Taipei 100, Taiwan
| | - I-Ni Chiang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan, No.1, Sec. 1, Jen - Ai Rd., Taipei 100, Taiwan
- Department of Urology, National Taiwan University Hospital, Taipei 100, Taiwan, No.7, Chung-Shan S. Rd., Taipei 100, Taiwan
| | - Jyh-Horng Wang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei 100, Taiwan, No.7, Chung-Shan S. Rd., Taipei 100, Taiwan
- * E-mail: (JHW); (THY)
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan, No.1, Sec. 1, Jen - Ai Rd., Taipei 100, Taiwan
- * E-mail: (JHW); (THY)
| |
Collapse
|
45
|
Tower J. Programmed cell death in aging. Ageing Res Rev 2015; 23:90-100. [PMID: 25862945 DOI: 10.1016/j.arr.2015.04.002] [Citation(s) in RCA: 273] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/15/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
Abstract
Programmed cell death (PCD) pathways, including apoptosis and regulated necrosis, are required for normal cell turnover and tissue homeostasis. Mis-regulation of PCD is increasingly implicated in aging and aging-related disease. During aging the cell turnover rate declines for several highly-mitotic tissues. Aging-associated disruptions in systemic and inter-cell signaling combined with cell-autonomous damage and mitochondrial malfunction result in increased PCD in some cell types, and decreased PCD in other cell types. Increased PCD during aging is implicated in immune system decline, skeletal muscle wasting (sarcopenia), loss of cells in the heart, and neurodegenerative disease. In contrast, cancer cells and senescent cells are resistant to PCD, enabling them to increase in abundance during aging. PCD pathways limit life span in fungi, but whether PCD pathways normally limit adult metazoan life span is not yet clear. PCD is regulated by a balance of negative and positive factors, including the mitochondria, which are particularly subject to aging-associated malfunction.
Collapse
|
46
|
Haupt S, Buckley D, Pang JMB, Panimaya J, Paul PJ, Gamell C, Takano EA, Lee YY, Hiddingh S, Rogers TM, Teunisse AFAS, Herold MJ, Marine JC, Fox SB, Jochemsen A, Haupt Y. Targeting Mdmx to treat breast cancers with wild-type p53. Cell Death Dis 2015; 6:e1821. [PMID: 26181202 PMCID: PMC4650725 DOI: 10.1038/cddis.2015.173] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/15/2015] [Accepted: 05/22/2015] [Indexed: 01/07/2023]
Abstract
The function of the tumor suppressor p53 is universally compromised in cancers. It is the most frequently mutated gene in human cancers (reviewed). In cases where p53 is not mutated, alternative regulatory pathways inactivate its tumor suppressive functions. This is primarily achieved through elevation in the expression of the key inhibitors of p53: Mdm2 or Mdmx (also called Mdm4) (reviewed). In breast cancer (BrCa), the frequency of p53 mutations varies markedly between the different subtypes, with basal-like BrCas bearing a high frequency of p53 mutations, whereas luminal BrCas generally express wild-type (wt) p53. Here we show that Mdmx is unexpectedly highly expressed in normal breast epithelial cells and its expression is further elevated in most luminal BrCas, whereas p53 expression is generally low, consistent with wt p53 status. Inducible knockdown (KD) of Mdmx in luminal BrCa MCF-7 cells impedes the growth of these cells in culture, in a p53-dependent manner. Importantly, KD of Mdmx in orthotopic xenograft transplants resulted in growth inhibition associated with prolonged survival, both in a preventative model and also in a treatment model. Growth impediment in response to Mdmx KD was associated with cellular senescence. The growth inhibitory capacity of Mdmx KD was recapitulated in an additional luminal BrCa cell line MPE600, which expresses wt p53. Further, the growth inhibitory capacity of Mdmx KD was also demonstrated in the wt p53 basal-like cell line SKBR7 line. These results identify Mdmx growth dependency in wt p53 expressing BrCas, across a range of subtypes. Based on our findings, we propose that Mdmx targeting is an attractive strategy for treating BrCas harboring wt p53.
Collapse
Affiliation(s)
- S Haupt
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - D Buckley
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J-M B Pang
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J Panimaya
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - P J Paul
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - C Gamell
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - E A Takano
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Y Ying Lee
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - S Hiddingh
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - T-M Rogers
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - A F A S Teunisse
- Department of Molecular Cell Biology, University Medical Centre, Leiden, The Netherlands
| | - M J Herold
- 1] Department of Molecular Genetics of Cancer, The Walter and Eliza Hall Institute, Parkville, Victoria, Australia [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - J-C Marine
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - S B Fox
- 1] Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - A Jochemsen
- Department of Molecular Cell Biology, University Medical Centre, Leiden, The Netherlands
| | - Y Haupt
- 1] Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia [3] Department of Pathology, University of Melbourne, Parkville, Victoria, Australia [4] Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
47
|
Escobar D, Hepp MI, Farkas C, Campos T, Sodir NM, Morales M, Álvarez CI, Swigart L, Evan GI, Gutiérrez JL, Nishinakamura R, Castro AF, Pincheira R. Sall2 is required for proapoptotic Noxa expression and genotoxic stress-induced apoptosis by doxorubicin. Cell Death Dis 2015; 6:e1816. [PMID: 26181197 PMCID: PMC4650718 DOI: 10.1038/cddis.2015.165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/13/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
The Sall2 transcription factor is deregulated in several cancers; however, little is known about its cellular functions, including its target genes. Recently, we demonstrated that p53 directly regulates Sall2 expression under genotoxic stress. Here, we investigated the role of Sall2 in the context of cellular response to genotoxic stress. In addition, we further examined the Sall2-p53 relationship during genotoxic stress in primary mouse embryo fibroblasts (MEFs), which are derived from Sall2 knockout mice separately, or in combination with the p53ERTAM knock-in mice. We found that the levels of Sall2 mRNA and protein are dynamically modulated in response to doxorubicin. At early times of stress, Sall2 is downregulated, but increases under extension of the stress in a p53-independent manner. Based on caspase-3/7 activities, expression of cleaved poly (ADP-ribose) polymerase, expression of cleaved caspase-3 and induction of proapoptotic proteins, Sall2 expression was correlated with cellular apoptosis. Consequently, Sall2-/- MEFs have decreased apoptosis, which relates with increased cell viability in response to doxorubicin. Importantly, Sall2 was required for apoptosis even in the presence of fully activated p53. Searching for putative Sall2 targets that could mediate its role in apoptosis, we identified proapoptotic NOXA/PMAIP1 (phorbol-12-myristate-13-acetate-induced protein 1). We demonstrated that Sall2 positively regulates Noxa promoter activity. Conserved putative Sall2-binding sites at the NOXA promoter were validated in vitro by electrophoretic mobility shift assay and in vivo by ChIP experiments, identifying NOXA as a novel Sall2 target. In agreement, induction of Noxa protein and mRNA in response to doxorubicin was significantly decreased in Sall2-/- MEFs. In addition, studies in leukemia Jurkat T cells support the existence of the Sall2/Noxa axis, and the significance of this axis on the apoptotic response to doxorubicin in cancer cells. Our study highlights the relevance of Sall2 in the apoptotic response to extended genotoxic stress, which is important for understanding its role in normal physiology and disease.
Collapse
Affiliation(s)
- D Escobar
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - M I Hepp
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - C Farkas
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - T Campos
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - N M Sodir
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - M Morales
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - C I Álvarez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - L Swigart
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94115, USA
| | - G I Evan
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - J L Gutiérrez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - A F Castro
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Pincheira
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
48
|
Yoshioka KI, Atsumi Y, Nakagama H, Teraoka H. Development of cancer-initiating cells and immortalized cells with genomic instability. World J Stem Cells 2015; 7:483-489. [PMID: 25815132 PMCID: PMC4369504 DOI: 10.4252/wjsc.v7.i2.483] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/29/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023] Open
Abstract
Cancers that develop after middle age usually exhibit genomic instability and multiple mutations. This is in direct contrast to pediatric tumors that usually develop as a result of specific chromosomal translocations and epigenetic aberrations. The development of genomic instability is associated with mutations that contribute to cellular immortalization and transformation. Cancer occurs when cancer-initiating cells (CICs), also called cancer stem cells, develop as a result of these mutations. In this paper, we explore how CICs develop as a result of genomic instability, including looking at which cancer suppression mechanisms are abrogated. A recent in vitro study revealed the existence of a CIC induction pathway in differentiating stem cells. Under aberrant differentiation conditions, cells become senescent and develop genomic instabilities that lead to the development of CICs. The resulting CICs contain a mutation in the alternative reading frame of CDKN2A (ARF)/p53 module, i.e., in either ARF or p53. We summarize recently established knowledge of CIC development and cellular immortality, explore the role of the ARF/p53 module in protecting cells from transformation, and describe a risk factor for genomic destabilization that increases during the process of normal cell growth and differentiation and is associated with the downregulation of histone H2AX to levels representative of growth arrest in normal cells.
Collapse
|
49
|
Wilking MJ, Singh C, Nihal M, Zhong W, Ahmad N. SIRT1 deacetylase is overexpressed in human melanoma and its small molecule inhibition imparts anti-proliferative response via p53 activation. Arch Biochem Biophys 2014; 563:94-100. [PMID: 24751483 DOI: 10.1016/j.abb.2014.04.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/03/2014] [Accepted: 04/05/2014] [Indexed: 11/30/2022]
Abstract
Melanoma causes more deaths than any other skin cancer, and its incidence in the US continues to rise. Current medical therapies are insufficient to control this deadly neoplasm, necessitating the development of new target-based approaches. The objective of this study was to determine the role and functional significance of the class III histone deacetylase SIRT1 in melanoma. We have found that SIRT1 is overexpressed in clinical human melanoma tissues and human melanoma cell lines (Sk-Mel-2, WM35, G361, A375, and Hs294T) compared to normal skin and normal melanocytes, respectively. In addition, treatment of melanoma cell lines A375, Hs294T, and G361 with Tenovin-1, a small molecule SIRT1 inhibitor, resulted in a significant decrease in cell growth and cell viability. Further, Tenovin-1 treatment also resulted in a marked decrease in the clonogenic survival of melanoma cells. Further experiments showed that the anti-proliferative response of Tenovin-1 was accompanied by an increase in the protein as well as activity of the tumor suppressor p53. This increase in p53 activity was substantiated by an increase in the protein level of its downstream target p21. Overall, these data suggest that small molecule inhibition of SIRT1 causes anti-proliferative effects in melanoma cells. SIRT1 appears to be acting through the activity of the tumor suppressor p53, which is not mutated in the majority of melanomas. However, future detailed studies are needed to further explore the role and mechanism of SIRT1 in melanoma development and progression and its usefulness in melanoma treatment.
Collapse
Affiliation(s)
- Melissa J Wilking
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | - Chandra Singh
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | - Minakshi Nihal
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA
| | - Weixiong Zhong
- Department of Pathology, University of Wisconsin, Madison, WI 53705, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA.
| |
Collapse
|
50
|
Sasaki H, Takeuchi I, Okada M, Sawada R, Kanie K, Kiyota Y, Honda H, Kato R. Label-free morphology-based prediction of multiple differentiation potentials of human mesenchymal stem cells for early evaluation of intact cells. PLoS One 2014; 9:e93952. [PMID: 24705458 PMCID: PMC3976343 DOI: 10.1371/journal.pone.0093952] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 03/11/2014] [Indexed: 12/11/2022] Open
Abstract
Precise quantification of cellular potential of stem cells, such as human bone marrow-derived mesenchymal stem cells (hBMSCs), is important for achieving stable and effective outcomes in clinical stem cell therapy. Here, we report a method for image-based prediction of the multiple differentiation potentials of hBMSCs. This method has four major advantages: (1) the cells used for potential prediction are fully intact, and therefore directly usable for clinical applications; (2) predictions of potentials are generated before differentiation cultures are initiated; (3) prediction of multiple potentials can be provided simultaneously for each sample; and (4) predictions of potentials yield quantitative values that correlate strongly with the experimental data. Our results show that the collapse of hBMSC differentiation potentials, triggered by in vitro expansion, can be quantitatively predicted far in advance by predicting multiple potentials, multi-lineage differentiation potentials (osteogenic, adipogenic, and chondrogenic) and population doubling potential using morphological features apparent during the first 4 days of expansion culture. In order to understand how such morphological features can be effective for advance predictions, we measured gene-expression profiles of the same early undifferentiated cells. Both senescence-related genes (p16 and p21) and cytoskeleton-related genes (PTK2, CD146, and CD49) already correlated to the decrease of potentials at this stage. To objectively compare the performance of morphology and gene expression for such early prediction, we tested a range of models using various combinations of features. Such comparison of predictive performances revealed that morphological features performed better overall than gene-expression profiles, balancing the predictive accuracy with the effort required for model construction. This benchmark list of various prediction models not only identifies the best morphological feature conversion method for objective potential prediction, but should also allow clinicians to choose the most practical morphology-based prediction method for their own purposes.
Collapse
Affiliation(s)
- Hiroto Sasaki
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| | - Ichiro Takeuchi
- Department of Computer Science/Scientific and Engineering Simulation, Graduate School of Engineering, Nagoya Institute of Technology, Nagoya, Aichi, Japan
| | - Mai Okada
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi, Japan
| | - Rumi Sawada
- Division of Medical Devices, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Kei Kanie
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi, Japan
| | | | - Hiroyuki Honda
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
| | - Ryuji Kato
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi, Japan
- * E-mail:
| |
Collapse
|