1
|
Cui Y, Zhao N, Wang C, Long J, Chen Y, Deng Z, Zhang Z, Zhao R, Sun J, Wang Z, Liu F, Xu K, Wang R, Li Y. Acute ammonia stress-induced oxidative and heat shock responses modulated by transcription factors in Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2022; 128:181-187. [PMID: 35917888 DOI: 10.1016/j.fsi.2022.07.060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/03/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
The present study aimed to examine the effects of short-term exposure to ammonia on stress and oxidative responses in shrimp (Litopenaeus vannamei) and to determine whether the antioxidant system related to the regulatory role of transcription factors and stress proteins was activated. Shrimp were exposed ammonia-N at four concentrations: 0 (control), 5, 10, and 15 mg/L, for 48 h. The hepatopancreas was sampled to measure the levels of glutathione (GSH), malondialdehyde (MDA), nitric oxide (NO); the activities of superoxide dismutase (SOD), catalase (CAT), nitric oxide synthase (NOS); and the expression levels of GSH-px (encoding glutathione peroxidase), GST (encoding glutathione-S-transferase), HSP70 (encoding heat shock protein 70), HSP90 (encoding heat shock protein 90), p53, RELISH, and AKIRIN. We observed that exposure to a high ammonia content increased the abundance of oxidative factors (MDA, CAT, SOD, NOS, and NO), reduced the levels of GSH, and upregulated the mRNA expression levels of antioxidant genes (GSH-px and GST), stress-related genes (HSP70 and HSP90), and transcription factor genes (p53, RELISH, and AKIRIN). These results indicated that ammonia induced oxidative stress and inflammation. Both enzymatic and nonenzymatic antioxidant defense systems are involved, which might be regulated by HSPs, as well as certain transcription factors, such as p53 and nuclear factor kappa B (NF-κB), thus mounting an adaptive response to help rebalance redox homoeostasis.
Collapse
Affiliation(s)
- Yanting Cui
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China.
| | - Nannan Zhao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Cong Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Jinnan Long
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Yigeng Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Zhitong Deng
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Zhihao Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Ruiyang Zhao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Jinfeng Sun
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Zhongkai Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Fei Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Kefeng Xu
- Marine Science Research Institute of Shandong Province (National Oceanographic Center, Qingdao), Qingdao, Shandong Province, 266104, China
| | - Renjie Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China
| | - Yuquan Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China.
| |
Collapse
|
2
|
Ronayne CT, Jonnalagadda SK, Jonnalagadda S, Nelson GL, Solano LN, Palle H, Mani C, Rumbley J, Holy J, Mereddy VR. Synthesis and biological evaluation of a novel anticancer agent CBISC that induces DNA damage response and diminishes levels of mutant-p53. Biochem Biophys Res Commun 2021; 562:127-132. [PMID: 34051576 DOI: 10.1016/j.bbrc.2021.05.062] [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: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 11/26/2022]
Abstract
A novel nitrogen mustard CBISC has been synthesized and evaluated as an anticancer agent. CBISC has been shown to exhibit enhanced cell proliferation inhibition properties against mutant p53 cell lines colorectal cancer WiDr, pancreatic cancer (MIAPaCa-2 and PANC-1), and triple negative breast cancer (MDA-MB-231 and MDA-MB-468). In vitro mechanism of action studies revealed perturbations in the p53 pathway and increased cell death as evidenced by western blotting, immunofluorescent microscopy and MTT assay. Further, in vivo studies revealed that CBISC is well tolerated in healthy mice and exhibited significant in vivo tumor growth inhibition properties in WiDr and MIAPaCa-2 xenograft models. These studies illustrate the potential utility of CBISC as an anticancer agent.
Collapse
Affiliation(s)
- Conor T Ronayne
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Sravan K Jonnalagadda
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Shirisha Jonnalagadda
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Grady L Nelson
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Lucas N Solano
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Hithardha Palle
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Chinnadurai Mani
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Jon Rumbley
- Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN, 55812, USA
| | - Jon Holy
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Venkatram R Mereddy
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA; Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN, 55812, USA; Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN, 55812, USA.
| |
Collapse
|
3
|
Tsai YH, Borini Etichetti CM, Cicetti S, Girardini JE, Spanevello RA, Suárez AG, Sarotti AM. Design, synthesis and evaluation of novel levoglucosenone derivatives as promising anticancer agents. Bioorg Med Chem Lett 2020; 30:127247. [PMID: 32527547 DOI: 10.1016/j.bmcl.2020.127247] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/25/2020] [Accepted: 05/02/2020] [Indexed: 11/28/2022]
Abstract
A series of levoglucosenone-derived 1,2,3-triazoles and isoxazoles featuring a flexible spacer between the heteroaromatic and anhydropyranose cores have been designed and synthesized following an hetero Michael // 1,3-dipolar cycloaddition path. The use of a design of experiments approach allowed the optimization of the oxa-Michael reaction with propargyl alcohol as nucleophile, a key step for the synthesis of the target compounds. All of the compounds were tested for their anticancer activity on MDA-MB-231 cells, featuring mutant p53. The results highlighted the importance of the introduction of the flexible spacer as well as the higher activity of oxa-Michael isoxazole-derivatives. The most prominent compounds also showed anti-proliferative activities against lung and colon cancer cell lines. The compounds showed enhanced cytotoxic effects in the presence of mutant p53, determined both by endogenous mutant p53 knock down (R280K) and by reintroducing p53 R280K in cells lacking p53 expression.
Collapse
Affiliation(s)
- Yi-Hsuan Tsai
- Instituto de Química Rosario (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Carla M Borini Etichetti
- Instituto Fisiología Experimental de Rosario (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Soledad Cicetti
- Instituto de Química Rosario (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Javier E Girardini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Rolando A Spanevello
- Instituto de Química Rosario (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Alejandra G Suárez
- Instituto de Química Rosario (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Ariel M Sarotti
- Instituto de Química Rosario (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina.
| |
Collapse
|
4
|
Purification of supercoiled p53-encoding plasmid using an arginine-modified macroporous support. J Chromatogr A 2020; 1618:460890. [DOI: 10.1016/j.chroma.2020.460890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/02/2020] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
|
5
|
Nelson GL, Ronayne CT, Solano LN, Jonnalagadda SK, Jonnalagadda S, Rumbley J, Holy J, Rose-Hellekant T, Drewes LR, Mereddy VR. Development of Novel Silyl Cyanocinnamic Acid Derivatives as Metabolic Plasticity Inhibitors for Cancer Treatment. Sci Rep 2019; 9:18266. [PMID: 31797891 PMCID: PMC6892925 DOI: 10.1038/s41598-019-54709-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022] Open
Abstract
Novel silyl cyanocinnamic acid derivatives have been synthesized and evaluated as potential anticancer agents. In vitro studies reveal that lead derivatives 2a and 2b have enhanced cancer cell proliferation inhibition properties when compared to the parent monocarboxylate transporter (MCT) inhibitor cyano-hydroxycinnamic acid (CHC). Further, candidate compounds exhibit several-fold more potent MCT1 inhibition properties as determined by lactate-uptake studies, and these studies are supported by MCT homology modeling and computational inhibitor-docking studies. In vitro effects on glycolysis and mitochondrial metabolism also illustrate that the lead derivatives 2a and 2b lead to significant effects on both metabolic pathways. In vivo systemic toxicity and efficacy studies in colorectal cancer cell WiDr tumor xenograft demonstrate that candidate compounds are well tolerated and exhibit good single agent anticancer efficacy properties.
Collapse
Affiliation(s)
- Grady L Nelson
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Conor T Ronayne
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Lucas N Solano
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Sravan K Jonnalagadda
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Shirisha Jonnalagadda
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA
| | - Jon Rumbley
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA.,Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN, 55812, USA
| | - Jon Holy
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA.,Department of Biomedical Sciences, Medical School Duluth, University of Minnesota, Duluth, MN, 55812, USA
| | - Teresa Rose-Hellekant
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA.,Department of Biomedical Sciences, Medical School Duluth, University of Minnesota, Duluth, MN, 55812, USA
| | - Lester R Drewes
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA.,Department of Biomedical Sciences, Medical School Duluth, University of Minnesota, Duluth, MN, 55812, USA
| | - Venkatram R Mereddy
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN, 55812, USA. .,Department of Chemistry and Biochemistry, University of Minnesota Duluth, Duluth, MN, 55812, USA. .,Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN, 55812, USA.
| |
Collapse
|
6
|
Valente J, Sousa A, Gaspar V, Queiroz J, Sousa F. The biological performance of purified supercoiled p53 plasmid DNA in different cancer cell lines. Process Biochem 2018. [DOI: 10.1016/j.procbio.2018.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
7
|
Isolation of 4,4'-bond secalonic acid D from the marine-derived fungus Penicillium oxalicum with inhibitory property against hepatocellular carcinoma. J Antibiot (Tokyo) 2018; 72:34-44. [PMID: 30258223 DOI: 10.1038/s41429-018-0104-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022]
Abstract
4,4'-bond secalonic acid D (4,4'-SAD) is a known compound isolated from the marine-derived fungus Penicillium oxalicum. No study about the antitumor effect of this compound has been reported, except for a few focusing on its bactericidal properties. Herein, we performed an in vitro biology test and found that 4,4'-SAD stimulated the apoptosis of tumor cells in the human hepatocellular carcinoma cell lines PLC/PRF/5 and HuH-7 by activating caspase-3, caspase-8, caspase-9, PARP, p53, and cyclin B1, as well as by regulating the Bax/Bcl-2 ratio. In vivo studies showed that 4,4'-SAD had antitumor efficacy in H22 cell xenograft model. Immunohistochemical analysis revealed that 4,4'-SAD could regulate Bax expression, which is a biomarker of tumor growth. In summary, 4,4'-SAD significantly inhibited tumor growth both in vivo and in vitro.
Collapse
|
8
|
Tsai YH, Borini Etichetti CM, Di Benedetto C, Girardini JE, Martins FT, Spanevello RA, Suárez AG, Sarotti AM. Synthesis of Triazole Derivatives of Levoglucosenone As Promising Anticancer Agents: Effective Exploration of the Chemical Space through retro-aza-Michael//aza-Michael Isomerizations. J Org Chem 2018; 83:3516-3528. [DOI: 10.1021/acs.joc.7b03141] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Yi-hsuan Tsai
- Instituto de Química Rosario (IQUIR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Carla M. Borini Etichetti
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, Rosario 2000, Argentina
| | - Carolina Di Benedetto
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, Rosario 2000, Argentina
| | - Javier E. Girardini
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, Rosario 2000, Argentina
| | - Felipe Terra Martins
- Instituto de Química, Universidade Federal de Goiás, Campus Samambaia, Goiânia, GO CP 131, 74001-970, Brazil
| | - Rolando A. Spanevello
- Instituto de Química Rosario (IQUIR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Alejandra G. Suárez
- Instituto de Química Rosario (IQUIR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| | - Ariel M. Sarotti
- Instituto de Química Rosario (IQUIR, CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina
| |
Collapse
|
9
|
Nuñez-Hernandez DM, Felix-Portillo M, Peregrino-Uriarte AB, Yepiz-Plascencia G. Cell cycle regulation and apoptosis mediated by p53 in response to hypoxia in hepatopancreas of the white shrimp Litopenaeus vannamei. CHEMOSPHERE 2018; 190:253-259. [PMID: 28992477 DOI: 10.1016/j.chemosphere.2017.09.131] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/23/2017] [Accepted: 09/27/2017] [Indexed: 06/07/2023]
Abstract
Although hypoxic aquatic environments cause negative effects on shrimp, these animals can withstand somewhat hypoxia, but the cellular mechanisms underlying this capacity are still poorly understood. In humans, mild hypoxia causes the induction of many proteins to allow cell survival. In contrast, apoptosis is induced during severe hypoxia leading to cell death. p53 is a key transcription factor that determines cells fate towards cell cycle arrest or induction of apoptosis in humans. The aim of this work was to study the role of p53 in cell cycle regulation and apoptosis in response to hypoxia in hepatopancreas of the white shrimp Litopenaeus vannamei. p53 was silenced by RNAi and afterwards the shrimp were exposed to hypoxia. Cdk-2 was used as indicator of cell cycle progression while caspase-3 expression and caspase activity were analyzed as indicators of apoptosis. p53 levels in hepatopancreas were significantly higher at 48 h after hypoxic treatment. Increased expression levels of Cdk-2 were found in p53-silenced shrimp after 24 and 48 h in the normoxic treatments as well as 48 h after hypoxia, indicating a possible role of p53 in cell cycle regulation. In response to hypoxia, unsilenced shrimp showed an increase in caspase-3 expression levels, however an increase was also observed in caspase activity at 24 h of normoxic conditions in p53-silenced shrimps. Taken together these results indicate the involvement of p53 in regulation of cell cycle and apoptosis in the white shrimp in response to hypoxia.
Collapse
Affiliation(s)
- Dahlia M Nuñez-Hernandez
- Centro de Investigación en Alimentación y Desarrollo, A.C. P.O. Box 1735. Carretera a Ejido La Victoria Km. 0.6 Hermosillo, Sonora, 83304, Mexico
| | - Monserrath Felix-Portillo
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua, Periférico Francisco R. Almada, Km 1, Chihuahua, Chihuahua, 33820, Mexico
| | - Alma B Peregrino-Uriarte
- Centro de Investigación en Alimentación y Desarrollo, A.C. P.O. Box 1735. Carretera a Ejido La Victoria Km. 0.6 Hermosillo, Sonora, 83304, Mexico
| | - Gloria Yepiz-Plascencia
- Centro de Investigación en Alimentación y Desarrollo, A.C. P.O. Box 1735. Carretera a Ejido La Victoria Km. 0.6 Hermosillo, Sonora, 83304, Mexico.
| |
Collapse
|
10
|
Mantovani F, Walerych D, Sal GD. Targeting mutant p53 in cancer: a long road to precision therapy. FEBS J 2016; 284:837-850. [PMID: 27808469 DOI: 10.1111/febs.13948] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/05/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
Abstract
The TP53 tumor suppressor is the most frequently mutated gene in human cancers. In recent years, a blooming of research efforts based on both cell lines and mouse models have highlighted how deeply mutant p53 proteins affect fundamental cellular pathways with cancer-promoting outcomes. Neomorphic mutant p53 activities spread over multiple levels, impinging on chromatin structure, transcriptional regulation and microRNA maturation, shaping the proteome and the cell's metabolic pathways, and also exerting cytoplasmic functions and displaying cell-extrinsic effects. These tumorigenic activities are inextricably linked with the blend of highly corrupted processes that characterize the tumor context. Recent studies indicate that successful strategies to extract core aspects of mutant p53 oncogenic potential and to identify unique tumor dependencies entail the superimposition of large-scale analyses performed in multiple experimental systems, together with a mindful use of animal models. This will hopefully soon lead to the long-awaited inclusion of mutant p53 as an actionable target of clinical antitumor therapies.
Collapse
Affiliation(s)
- Fiamma Mantovani
- Laboratorio Nazionale CIB (LNCIB), Trieste, Italy.,Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Italy
| | | | - Giannino Del Sal
- Laboratorio Nazionale CIB (LNCIB), Trieste, Italy.,Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Italy
| |
Collapse
|
11
|
Soares J, Raimundo L, Pereira NAL, Monteiro Â, Gomes S, Bessa C, Pereira C, Queiroz G, Bisio A, Fernandes J, Gomes C, Reis F, Gonçalves J, Inga A, Santos MMM, Saraiva L. Reactivation of wild-type and mutant p53 by tryptophanolderived oxazoloisoindolinone SLMP53-1, a novel anticancer small-molecule. Oncotarget 2016; 7:4326-43. [PMID: 26735173 PMCID: PMC4826208 DOI: 10.18632/oncotarget.6775] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/28/2015] [Indexed: 12/28/2022] Open
Abstract
Restoration of the p53 pathway, namely by reactivation of mutant (mut) p53, represents a valuable anticancer strategy. Herein, we report the identification of the enantiopure tryptophanol-derived oxazoloisoindolinone SLMP53-1 as a novel reactivator of wild-type (wt) and mut p53, using a yeast-based screening strategy. SLMP53-1 has a p53-dependent anti-proliferative activity in human wt and mut p53R280K-expressing tumor cells. Additionally, SLMP53-1 enhances p53 transcriptional activity and restores wt-like DNA binding ability to mut p53R280K. In wt/mut p53-expressing tumor cells, SLMP53-1 triggers p53 transcription-dependent and mitochondrial apoptotic pathways involving BAX, and wt/mut p53 mitochondrial translocation. SLMP53-1 inhibits the migration of wt/mut p53-expressing tumor cells, and it shows promising p53-dependent synergistic effects with conventional chemotherapeutics. In xenograft mice models, SLMP53-1 inhibits the growth of wt/mut p53-expressing tumors, but not of p53-null tumors, without apparent toxicity. Collectively, besides the potential use of SLMP53-1 as anticancer drug, the tryptophanol-derived oxazoloisoindolinone scaffold represents a promissing starting point for the development of effective p53-reactivating drugs.
Collapse
Affiliation(s)
- Joana Soares
- UCIBIO/REQUIMTE, Universidade do Porto, Porto, Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Liliana Raimundo
- UCIBIO/REQUIMTE, Universidade do Porto, Porto, Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Nuno A L Pereira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Ângelo Monteiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Gomes
- UCIBIO/REQUIMTE, Universidade do Porto, Porto, Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Cláudia Bessa
- UCIBIO/REQUIMTE, Universidade do Porto, Porto, Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Clara Pereira
- UCIBIO/REQUIMTE, Universidade do Porto, Porto, Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Glória Queiroz
- Laboratório de Farmacologia, Departamento de Ciências do Medicamento, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Alessandra Bisio
- CIBIO, Centre for Integrative Biology, Laboratory of Transcriptional Networks, University of Trento, Trento, Italy
| | - João Fernandes
- Laboratório de Farmacologia e Terapêutica Experimental, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | - Célia Gomes
- Laboratório de Farmacologia e Terapêutica Experimental, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | - Flávio Reis
- Laboratório de Farmacologia e Terapêutica Experimental, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | - Jorge Gonçalves
- Laboratório de Farmacologia, Departamento de Ciências do Medicamento, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Alberto Inga
- CIBIO, Centre for Integrative Biology, Laboratory of Transcriptional Networks, University of Trento, Trento, Italy
| | - Maria M M Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Lucília Saraiva
- UCIBIO/REQUIMTE, Universidade do Porto, Porto, Portugal.,Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
Walerych D, Lisek K, Sommaggio R, Piazza S, Ciani Y, Dalla E, Rajkowska K, Gaweda-Walerych K, Ingallina E, Tonelli C, Morelli MJ, Amato A, Eterno V, Zambelli A, Rosato A, Amati B, Wiśniewski JR, Del Sal G. Proteasome machinery is instrumental in a common gain-of-function program of the p53 missense mutants in cancer. Nat Cell Biol 2016; 18:897-909. [PMID: 27347849 DOI: 10.1038/ncb3380] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 05/25/2016] [Indexed: 12/17/2022]
Abstract
In cancer, the tumour suppressor gene TP53 undergoes frequent missense mutations that endow mutant p53 proteins with oncogenic properties. Until now, a universal mutant p53 gain-of-function program has not been defined. By means of multi-omics: proteome, DNA interactome (chromatin immunoprecipitation followed by sequencing) and transcriptome (RNA sequencing/microarray) analyses, we identified the proteasome machinery as a common target of p53 missense mutants. The mutant p53-proteasome axis globally affects protein homeostasis, inhibiting multiple tumour-suppressive pathways, including the anti-oncogenic KSRP-microRNA pathway. In cancer cells, p53 missense mutants cooperate with Nrf2 (NFE2L2) to activate proteasome gene transcription, resulting in resistance to the proteasome inhibitor carfilzomib. Combining the mutant p53-inactivating agent APR-246 (PRIMA-1MET) with the proteasome inhibitor carfilzomib is effective in overcoming chemoresistance in triple-negative breast cancer cells, creating a therapeutic opportunity for treatment of solid tumours and metastasis with mutant p53.
Collapse
Affiliation(s)
- Dawid Walerych
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy
| | - Kamil Lisek
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Dipartimento di Scienze della Vita-Università degli Studi di Trieste, Trieste 34127, Italy
| | - Roberta Sommaggio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova 35128, Italy
| | - Silvano Piazza
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy
| | - Yari Ciani
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy
| | - Emiliano Dalla
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy
| | - Katarzyna Rajkowska
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy
| | - Katarzyna Gaweda-Walerych
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Laboratory of Neurogenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw 02106, Poland
| | - Eleonora Ingallina
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Dipartimento di Scienze della Vita-Università degli Studi di Trieste, Trieste 34127, Italy
| | - Claudia Tonelli
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan 20141, Italy
| | - Marco J Morelli
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan 20139, Italy
| | - Angela Amato
- Laboratory of Experimental Oncology and Pharmacogenomics, IRCCS 'Salvatore Maugeri' Foundation, Pavia 27100, Italy
| | - Vincenzo Eterno
- Laboratory of Experimental Oncology and Pharmacogenomics, IRCCS 'Salvatore Maugeri' Foundation, Pavia 27100, Italy
| | - Alberto Zambelli
- Laboratory of Experimental Oncology and Pharmacogenomics, IRCCS 'Salvatore Maugeri' Foundation, Pavia 27100, Italy.,Unit of Medical Oncology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo 24127, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova 35128, Italy.,Istituto Oncologico Veneto IOV-IRCCS, Padova 35128, Italy
| | - Bruno Amati
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan 20141, Italy.,Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan 20139, Italy
| | - Jacek R Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried D82152, Germany
| | - Giannino Del Sal
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Dipartimento di Scienze della Vita-Università degli Studi di Trieste, Trieste 34127, Italy
| |
Collapse
|
13
|
Walerych D, Lisek K, Del Sal G. Mutant p53: One, No One, and One Hundred Thousand. Front Oncol 2015; 5:289. [PMID: 26734571 PMCID: PMC4685664 DOI: 10.3389/fonc.2015.00289] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/07/2015] [Indexed: 11/16/2022] Open
Abstract
Encoded by the mutated variants of the TP53 tumor suppressor gene, mutant p53 proteins are getting an increased experimental support as active oncoproteins promoting tumor growth and metastasis. p53 missense mutant proteins are losing their wild-type tumor suppressor activity and acquire oncogenic potential, possessing diverse transforming abilities in cell and mouse models. Whether various mutant p53s differ in their oncogenic potential has been a matter of debate. Recent discoveries are starting to uncover the existence of mutant p53 downstream programs that are common to different mutant p53 variants. In this review, we discuss a number of studies on mutant p53, underlining the advantages and disadvantages of alternative experimental approaches that have been used to describe the numerous mutant p53 gain-of-function activities. Therapeutic possibilities are also discussed, taking into account targeting either individual or multiple mutant p53 proteins in human cancer.
Collapse
Affiliation(s)
- Dawid Walerych
- Laboratorio Nazionale CIB, Area Science Park Padriciano , Trieste , Italy
| | - Kamil Lisek
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Giannino Del Sal
- Laboratorio Nazionale CIB, Area Science Park Padriciano, Trieste, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| |
Collapse
|
14
|
Di Agostino S, Sorrentino G, Ingallina E, Valenti F, Ferraiuolo M, Bicciato S, Piazza S, Strano S, Del Sal G, Blandino G. YAP enhances the pro-proliferative transcriptional activity of mutant p53 proteins. EMBO Rep 2015; 17:188-201. [PMID: 26691213 DOI: 10.15252/embr.201540488] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/19/2015] [Indexed: 01/02/2023] Open
Abstract
Mutant p53 proteins are present in more than half of human cancers. Yes-associated protein (YAP) is a key transcriptional regulator controlling organ growth, tissue homeostasis, and cancer. Here, we report that these two determinants of human malignancy share common transcriptional signatures. YAP physically interacts with mutant p53 proteins in breast cancer cells and potentiates their pro-proliferative transcriptional activity. We found YAP as well as mutant p53 and the transcription factor NF-Y onto the regulatory regions of cyclin A, cyclin B, and CDK1 genes. Either mutant p53 or YAP depletion down-regulates cyclin A, cyclin B, and CDK1 gene expression and markedly slows the growth of diverse breast cancer cell lines. Pharmacologically induced cytoplasmic re-localization of YAP reduces the expression levels of cyclin A, cyclin B, and CDK1 genes both in vitro and in vivo. Interestingly, primary breast cancers carrying p53 mutations and displaying high YAP activity exhibit higher expression levels of cyclin A, cyclin B, and CDK1 genes when compared to wt-p53 tumors.
Collapse
Affiliation(s)
- Silvia Di Agostino
- Translational Oncogenomic Unit, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Sorrentino
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Eleonora Ingallina
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Fabio Valenti
- Translational Oncogenomic Unit, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Ferraiuolo
- Translational Oncogenomic Unit, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy Molecular Chemoprevention Group, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvano Piazza
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy
| | - Sabrina Strano
- Molecular Chemoprevention Group, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| | - Giannino Del Sal
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Giovanni Blandino
- Translational Oncogenomic Unit, Molecular Medicine Area Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
15
|
Braicu O, Pileczki V, Braicu C, Achimas-Cadariu P, Irimie A, Berindan-Neagoe I. p53 siRNA - a therapeutic tool with significant implication in the modulation of apoptosis and angiogenic pathways. ACTA ACUST UNITED AC 2015; 88:333-7. [PMID: 26609266 PMCID: PMC4632892 DOI: 10.15386/cjmed-434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/04/2015] [Accepted: 05/09/2015] [Indexed: 12/28/2022]
Abstract
Background and aims siRNAs represent an encouraging novel alternative in cancer therapy as a result of targeting the mutated tumour suppressor genes or activated oncogenes. Targeting oncogenic signals, as the mutated p53 gene that gains oncogenic role, we observed inhibition of migration, a downregulation of specific genes involved in apoptosis but also in angiogenesis, connected with a reduction in invasion rate in the case of p53siRNA therapy. Methods The study was designed to assess the role of p53 by using RNAi (RNA interference) in Hela in vitro cell culture model. Therefore cell migration rate was assessed by using xCELLigence Systems, gene expression for a panel of genes involved in apoptosis and angiogenesis, and validation of gene expression data at protein level. Results On the selected in vitro model p53 siRNA therapy was correlated with the reduction of cell migration. The downregulation of p53, PTEN, TNFα, NFkB, BCL-2, ICAM-2, VEGF, and FGFb was evidenced as response to p53 inhibition. Conclusion RNAi may be a valuable technology in order to restore the normal cellular phenotype. The results in the current research may also have an important significance outside the context of cervical cancer, by using specific inhibitors for p53 for increasing the therapeutic response in a wide range of tumoral pathology.
Collapse
Affiliation(s)
- Ovidiu Braicu
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Surgical Oncology, The Oncological Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Romania
| | - Valentina Pileczki
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patriciu Achimas-Cadariu
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Surgical Oncology, The Oncological Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Romania
| | - Alexandru Irimie
- Department of Surgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Surgical Oncology, The Oncological Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Department of Functional Genomics and Experimental Pathology, The Oncological Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Romania
| |
Collapse
|
16
|
Distinct novel quinazolinone exhibits selective inhibition in MGC-803 cancer cells by dictating mutant p53 function. Eur J Med Chem 2015; 95:377-87. [DOI: 10.1016/j.ejmech.2015.03.053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/18/2015] [Accepted: 03/23/2015] [Indexed: 01/15/2023]
|
17
|
A tryptophanol-derived oxazolopiperidone lactam is cytotoxic against tumors via inhibition of p53 interaction with murine double minute proteins. Pharmacol Res 2015; 95-96:42-52. [PMID: 25814188 DOI: 10.1016/j.phrs.2015.03.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 03/12/2015] [Accepted: 03/12/2015] [Indexed: 12/25/2022]
Abstract
Inactivation of the p53 tumor suppressor protein by interaction with murine double minute (MDM) proteins, MDM2 and MDMX, is a common event in human tumors expressing wild-type p53. In these tumors, the simultaneous inhibition of these interactions with MDMs, for a full p53 reactivation, represents a promising anticancer strategy. Herein, we report the identification of a dual inhibitor of the p53 interaction with MDM2 and MDMX, the (S)-tryptophanol derivative OXAZ-1, from the screening of a small library of enantiopure tryptophanol-derived oxazolopiperidone lactams, using a yeast-based assay. With human colon adenocarcinoma HCT116 cell lines expressing wild-type p53 (HCT116 p53(+/+)) and its p53-null isogenic derivative (HCT116 p53(-/-)), it was shown that OXAZ-1 induced a p53-dependent tumor growth-inhibitory effect. In fact, OXAZ-1 induced p53 stabilization, up-regulated p53 transcription targets, such as MDM2, MDMX, p21, Puma and Bax, and led to PARP cleavage, in p53(+/+), but not in p53(-/-), HCT116 cells. In addition, similar tumor cytotoxic effects were observed for OXAZ-1 against MDMX-overexpressing breast adenocarcinoma MCF-7 tumor cells, commonly described as highly resistant to MDM2-only inhibitors. In HCT116 p53(+/+) cells, the disruption of the p53 interaction with MDMs by OXAZ-1 was further confirmed by co-immunoprecipitation. It was also shown that OXAZ-1 potently triggered a p53-dependent mitochondria-mediated apoptosis, characterized by reactive oxygen species generation, mitochondrial membrane potential dissipation, Bax translocation to mitochondria, and cytochrome c release, and exhibited a p53-dependent synergistic effect with conventional chemotherapeutic drugs. Collectively, in this work, a novel selective activator of the p53 pathway is reported with promising antitumor properties to be explored either alone or combined with conventional chemotherapeutic drugs. Moreover, OXAZ-1 may represent a promising starting scaffold to search for new dual inhibitors of the p53-MDMs interaction.
Collapse
|
18
|
Wang JZ, Liu BG, Zhang Y. Pin1-based diagnostic and therapeutic strategies for breast cancer. Pharmacol Res 2014; 93:28-35. [PMID: 25553719 DOI: 10.1016/j.phrs.2014.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 01/12/2023]
Abstract
Pin1 is the only known cis-to-trans isomerase that recognizes the phosphorylated pThr/pSer-Pro motifs in many signaling molecules, playing unique roles in the pathogenesis of breast cancer. First, Pin1 is prevalently over-expressed in kinds of breast cancer cell lines and tissues, such as MDA-MB-231 cell, MCF-7 cell, Her2+, ERα+, and basal-like breast cancer subtypes. Second, Pin1 amplifies many oncogenic signaling pathways, inhibits multiple tumor suppressors, promotes the angiogenesis and metastasis of breast cancer cells, and enhances the resistance of breast cancer cells to anti-tumor medicines. Third, inhibiting Pin1 blocks most of these detrimental effects in a great number of breast cancer cell lines. These findings suggest Pin1 as a promising diagnostic biomarker as well as an efficient therapeutic target for breast cancer. It is strongly expected that a Pin1-positive subtype of breast cancers should be extremely concerned and that the therapeutic efficacy of Pin1 inhibitors on breast cancer patients should be evaluated as soon as possible. Nonetheless, Pin1-based therapeutic strategies for breast cancer still deserve some debates. Hence, we give the predictions of several important issues, such as application precondition, side effects, and personalized medication, when Pin1 inhibitors are used in the breast cancer therapy. These proposals are meaningful for the further development of Pin1-based diagnostic and therapeutic strategies in order to conquer breast cancer.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan 056002, PR China.
| | - Bao-Guo Liu
- Department of Medical Technology, Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| | - Yong Zhang
- Department of Medical Technology, Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| |
Collapse
|
19
|
IL-1Ra selectively protects intestinal crypt epithelial cells, but not tumor cells, from chemotoxicity via p53-mediated upregulation of p21WAF1 and p27KIP1. Pharmacol Res 2014; 82:21-33. [DOI: 10.1016/j.phrs.2014.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 12/27/2022]
|