1
|
Oren M, Prives C. p53: A tale of complexity and context. Cell 2024; 187:1569-1573. [PMID: 38552605 DOI: 10.1016/j.cell.2024.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 04/02/2024]
Abstract
The story of p53 is illuminating. Despite widespread attention, the tumor-suppressive functions of wild-type p53 or the oncogenic activities of its cancer-associated mutants are still not fully understood, and our discoveries have not yet led to major therapeutic breakthroughs. There is still much to learn about this fascinating protein.
Collapse
Affiliation(s)
- Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Otmani K, Rouas R, Berehab M, Lewalle P. The regulatory mechanisms of oncomiRs in cancer. Biomed Pharmacother 2024; 171:116165. [PMID: 38237348 DOI: 10.1016/j.biopha.2024.116165] [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: 10/26/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Cancer development is a complex process that primarily results from the combination of genetic alterations and the dysregulation of major signalling pathways due to interference with the epigenetic machinery. As major epigenetic regulators, miRNAs are central players in the control of many key tumour development factors. These miRNAs have been classified as oncogenic miRNAs (oncomiRs) when they target tumour suppressor genes and tumour suppressor miRNAs (TS miRNAs) when they inhibit oncogene protein expression. Most of the mechanisms that modulate oncomiR expression are linked to transcriptional or posttranscriptional regulation. However, non-transcriptional processes, such as gene amplification, have been described as alternative processes that are responsible for increasing oncomiR expression. The current review summarises the different mechanisms controlling the upregulation of oncomiR expression in cancer cells and the tumour microenvironment (TME). Detailed knowledge of the mechanism underlying the regulation of oncomiR expression in cancer may pave the way for understanding the critical role of oncomiRs in cancer development and progression.
Collapse
Affiliation(s)
- Khalid Otmani
- Hematology Laboratory, Hematology Department, Hôpital Universitaire de Bruxelles (H.U.B.) Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Redouane Rouas
- Hematology Laboratory, Hematology Department, Hôpital Universitaire de Bruxelles (H.U.B.) Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mimoune Berehab
- Hematology Laboratory, Hematology Department, Hôpital Universitaire de Bruxelles (H.U.B.) Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Lewalle
- Hematology Laboratory, Hematology Department, Hôpital Universitaire de Bruxelles (H.U.B.) Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
3
|
Shin DY. TP53 Mutation in Acute Myeloid Leukemia: An Old Foe Revisited. Cancers (Basel) 2023; 15:4816. [PMID: 37835510 PMCID: PMC10571655 DOI: 10.3390/cancers15194816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
INTRODUCTION TP53 is the most commonly mutated gene in human cancers and was the first tumor suppressor gene to be discovered in the history of medical science. Mutations in the TP53 gene occur at various genetic locations and exhibit significant heterogeneity among patients. Mutations occurring primarily within the DNA-binding domain of TP53 result in the loss of the p53 protein's DNA-binding capability. However, a complex phenotypic landscape often combines gain-of-function, dominant negative, or altered specificity features. This complexity poses a significant challenge in developing an effective treatment strategy, which eradicates TP53-mutated cancer clones. This review summarizes the current understanding of TP53 mutations in AML and their implications. TP53 mutation in AML: In patients with acute myeloid leukemia (AML), six hotspot mutations (R175H, G245S, R248Q/W, R249S, R273H/S, and R282W) within the DNA-binding domain are common. TP53 mutations are frequently associated with a complex karyotype and subgroups of therapy-related or secondary AML. The presence of TP53 mutation is considered as a poor prognostic factor. TP53-mutated AML is even classified as a distinct subgroup of AML by itself, as TP53-mutated AML exhibits a significantly distinct landscape in terms of co-mutation and gene expression profiles compared with wildtype (WT)-TP53 AML. CLINICAL IMPLICATIONS To better predict the prognosis in cancer patients with different TP53 mutations, several predictive scoring systems have been proposed based on screening experiments, to assess the aggressiveness of TP53-mutated cancer cells. Among those scoring systems, a relative fitness score (RFS) could be applied to AML patients with TP53 mutations in terms of overall survival (OS) and event-free survival (EFS). The current standard treatment, which includes cytotoxic chemotherapy and allogeneic hematopoietic stem cell transplantation, is largely ineffective for patients with TP53-mutated AML. Consequently, most patients with TP53-mutated AML succumb to leukemia within several months, despite active anticancer treatment. Decitabine, a hypomethylating agent, is known to be relatively effective in patients with AML. Numerous trials are ongoing to investigate the effects of novel drugs combined with hypomethylating agents, TP53-targeting agents or immunologic agents. CONCLUSIONS Developing an effective treatment strategy for TP53-mutated AML through innovative and multidisciplinary research is an urgent task. Directly targeting mutated TP53 holds promise as an approach to combating TP53-mutated AML, and recent developments in immunologic agents for AML offer hope in this field.
Collapse
Affiliation(s)
- Dong-Yeop Shin
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; ; Tel.: +82-2-2072-7209; Fax: +82-2-762-9662
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
4
|
Otmani K, Lewalle P. Tumor Suppressor miRNA in Cancer Cells and the Tumor Microenvironment: Mechanism of Deregulation and Clinical Implications. Front Oncol 2021; 11:708765. [PMID: 34722255 PMCID: PMC8554338 DOI: 10.3389/fonc.2021.708765] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/27/2021] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that have been identified as important posttranscriptional regulators of gene expression. miRNAs production is controlled at multiple levels, including transcriptional and posttranscriptional regulation. Extensive profiling studies have shown that the regulation of mature miRNAs expression plays a causal role in cancer development and progression. miRNAs have been identified to act as tumor suppressors (TS) or as oncogenes based on their modulating effect on the expression of their target genes. Upregulation of oncogenic miRNAs blocks TS genes and leads to tumor formation. In contrast, downregulation of miRNAs with TS function increases the translation of oncogenes. Several miRNAs exhibiting TS properties have been studied. In this review we focus on recent studies on the role of TS miRNAs in cancer cells and the tumor microenvironment (TME). Furthermore, we discuss how TS miRNA impacts the aggressiveness of cancer cells, with focus of the mechanism that regulate its expression. The study of the mechanisms of miRNA regulation in cancer cells and the TME may paved the way to understand its critical role in the development and progression of cancer and is likely to have important clinical implications in a near future. Finally, the potential roles of miRNAs as specific biomarkers for the diagnosis and the prognosis of cancer and the replacement of tumor suppressive miRNAs using miRNA mimics could be promising approaches for cancer therapy.
Collapse
Affiliation(s)
- Khalid Otmani
- Experimental Hematology Laboratory, Jules Bordet Institute, Université libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
5
|
He YH, Yeh MH, Chen HF, Wang TS, Wong RH, Wei YL, Huynh TK, Hu DW, Cheng FJ, Chen JY, Hu SW, Huang CC, Chen Y, Yu J, Cheng WC, Shen PC, Liu LC, Huang CH, Chang YJ, Huang WC. ERα determines the chemo-resistant function of mutant p53 involving the switch between lincRNA-p21 and DDB2 expressions. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:536-553. [PMID: 34589276 PMCID: PMC8463322 DOI: 10.1016/j.omtn.2021.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/30/2021] [Indexed: 12/16/2022]
Abstract
Mutant p53 (mutp53) commonly loses its DNA binding affinity to p53 response elements (p53REs) and fails to induce apoptosis fully. However, the p53 mutation does not predict chemoresistance in all subtypes of breast cancers, and the critical determinants remain to be identified. In this study, mutp53 was found to mediate chemotherapy-induced long intergenic noncoding RNA-p21 (lincRNA-p21) expression by targeting the G-quadruplex structure rather than the p53RE on its promoter to promote chemosensitivity. However, estrogen receptor alpha (ERα) suppressed mutp53-mediated lincRNA-p21 expression by hijacking mutp53 to upregulate damaged DNA binding protein 2 (DDB2) transcription for subsequent DNA repair and chemoresistance. Levels of lincRNA-p21 positively correlated with the clinical responses of breast cancer patients to neoadjuvant chemotherapy and had an inverse correlation with the ER status and DDB2 level. In contrast, the carboplatin-induced DDB2 expression was higher in ER-positive breast tumor tissues. These results demonstrated that ER status determines the oncogenic function of mutp53 in chemoresistance by switching its target gene preference from lincRNA-p21 to DDB2 and suggest that induction of lincRNA-p21 and targeting DDB2 would be effective strategies to increase the chemosensitivity of mutp53 breast cancer patients.
Collapse
Affiliation(s)
- Yu-Hao He
- The PhD Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Ming-Hsin Yeh
- Department of Surgery, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Hsiao-Fan Chen
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Drug Development Center, China Medical University, Taichung 40402, Taiwan
| | - Tsu-Shing Wang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Ruey-Hong Wong
- Department of Public Health, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Occupational Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Ya-Ling Wei
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Thanh Kieu Huynh
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Dai-Wei Hu
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Fang-Ju Cheng
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Graduate Institute of Basic Medical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Jhen-Yu Chen
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Shu-Wei Hu
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Chia-Chen Huang
- Department of Public Health, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yeh Chen
- Drug Development Center, China Medical University, Taichung 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
| | - Jiaxin Yu
- AI Innovation Center, China Medical University Hospital, Taiwan 40402, Taiwan
| | - Wei-Chung Cheng
- The PhD Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
| | - Pei-Chun Shen
- Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
| | - Liang-Chih Liu
- Division of Breast Surgery, China Medical University Hospital, Taichung 40402, Taiwan
| | - Chih-Hao Huang
- Division of Breast Surgery, China Medical University Hospital, Taichung 40402, Taiwan
| | - Ya-Jen Chang
- The PhD Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Wei-Chien Huang
- The PhD Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Drug Development Center, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
6
|
Yamulla RJ, Nalubola S, Flesken-Nikitin A, Nikitin AY, Schimenti JC. Most Commonly Mutated Genes in High-Grade Serous Ovarian Carcinoma Are Nonessential for Ovarian Surface Epithelial Stem Cell Transformation. Cell Rep 2021; 32:108086. [PMID: 32877668 DOI: 10.1016/j.celrep.2020.108086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/07/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the fifth leading cause of cancer-related deaths of women in the United States. Disease-associated mutations have been identified by the Cancer Genome Atlas Research Network. However, aside from mutations in TP53 or the RB1 pathway that are common in HGSOC, the contributions of mutation combinations are unclear. Here, we report CRISPR mutagenesis of 20 putative HGSOC driver genes to identify combinatorial disruptions of genes that transform either ovarian surface epithelium stem cells (OSE-SCs) or non-stem cells (OSE-NSs). Our results support the OSE-SC theory of HGSOC initiation and suggest that most commonly mutated genes in HGSOC have no effect on OSE-SC transformation initiation. Our results indicate that disruption of TP53 and PTEN, combined with RB1 disruption, constitutes a core set of mutations driving efficient transformation in vitro. The combined data may contribute to more accurate modeling of HGSOC development.
Collapse
Affiliation(s)
- Robert Joseph Yamulla
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Shreya Nalubola
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA; New York Medical College, Valhalla, NY 10595, USA
| | - Andrea Flesken-Nikitin
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Alexander Yu Nikitin
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - John C Schimenti
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA.
| |
Collapse
|
7
|
El-Mekkawy HI, Al-Kahtani MA, Shati AA, Alshehri MA, Al-Doaiss AA, Elmansi AA, Ahmed AE. Black tea and curcumin synergistically mitigate the hepatotoxicity and nephropathic changes induced by chronic exposure to aflatoxin-B1 in Sprague-Dawley rats. J Food Biochem 2020; 44:e13346. [PMID: 32602579 DOI: 10.1111/jfbc.13346] [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: 03/27/2020] [Revised: 05/25/2020] [Accepted: 05/28/2020] [Indexed: 01/18/2023]
Abstract
The study aimed to clarify the characteristics of black tea (BTE) and/or curcumin (CMN) against aflatoxin-B1 (AFB1). Forty eight adult male Sprague-Dawley rats were divided into eight groups. G1 was non-treated control. G2, G3, and G4 were olive oil, BTE, and CMN, respectively. G5 was olive oil-dissolved AFB1 (25 µg/kg b.w). G6, G7, and G8 were AFB1 along with BTE (2%), CMN (200 mg/kg b.w.), and BTE plus CMN, respectively. All treatments were orally given for consecutive 90 days. After treatment period, rats were sacrificed. Serobiochemical analysis and histopathology showed hepatorenal dysfunction in response to AFB1. Glutathione-antioxidants were significantly decreased versus increased lipid peroxides (p < .05-.001). AFB1 significantly increased the expression of the antitumor p53, but decreased that of antiapoptotic Bcl2 in liver or kidney tissue, either (p < .05). BTE or CMN ameliorated those changes induced by AFB1 in both liver and kidney with highly pronounced improvement when combined BTE/CMN was used. PRACTICAL APPLICATIONS: Black tea (BTE) and curcumin (CMN) were known for their antioxidant effects, and several studies reported their independent effects against different toxicities including aflatoxicosis. The current study clarifies the ameliorative characteristics of both agents; BTE and/or CMN, against the toxicity resulted from the chronic exposure to aflatoxin-B1 (AFB1) (25 µg/kg b.w. for consecutive 90 days). The dose of either agents, BTE or CMN, was 200 mg/kg b.w. along with AFB1. The pathologic changes, serobiochemical parameters, oxidative stress, histological changes, and the molecular disruption, induced by AFB1 in both liver and kidney were obviously and significantly ameliorated after BTE and/or CMN treatments in variable potencies where both agents showed the most effective antitoxic capacities.
Collapse
Affiliation(s)
- Haitham I El-Mekkawy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A Al-Kahtani
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A Alshehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Amin A Al-Doaiss
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Histology and Anatomy, Faculty of Medicine, Sana'a University, Sana'a, Republic of Yemen
| | - Ahmed A Elmansi
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Theriogenology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| |
Collapse
|
8
|
Sborchia M, Keun HC, Phillips DH, Arlt VM. The Impact of p53 on Aristolochic Acid I-Induced Gene Expression In Vivo. Int J Mol Sci 2019; 20:ijms20246155. [PMID: 31817608 PMCID: PMC6940885 DOI: 10.3390/ijms20246155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to aristolochic acid (AA) is linked to kidney disease and urothelial cancer in humans. The major carcinogenic component of the AA plant extract is aristolochic acid I (AAI). The tumour suppressor p53 is frequently mutated in AA-induced tumours. We previously showed that p53 protects from AAI-induced renal proximal tubular injury, but the underlying mechanism(s) involved remain to be further explored. In the present study, we investigated the impact of p53 on AAI-induced gene expression by treating Trp53(+/+), Trp53(+/-), and Trp53(-/-) mice with 3.5 mg/kg body weight (bw) AAI daily for six days. The Clariom™ S Assay microarray was used to elucidate gene expression profiles in mouse kidneys after AAI treatment. Analyses in Qlucore Omics Explorer showed that gene expression in AAI-exposed kidneys is treatment-dependent. However, gene expression profiles did not segregate in a clear-cut manner according to Trp53 genotype, hence further investigations were performed by pathway analysis with MetaCore™. Several pathways were significantly altered to varying degrees for AAI-exposed kidneys. Apoptotic pathways were modulated in Trp53(+/+) kidneys; whereas oncogenic and pro-survival pathways were significantly altered for Trp53(+/-) and Trp53(-/-) kidneys, respectively. Alterations of biological processes by AAI in mouse kidneys could explain the mechanisms by which p53 protects from or p53 loss drives AAI-induced renal injury in vivo.
Collapse
Affiliation(s)
- Mateja Sborchia
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
| | - Hector C. Keun
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London W12 0NN, UK;
| | - David H. Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
| | - Volker M. Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
- Correspondence:
| |
Collapse
|
9
|
Sborchia M, De Prez EG, Antoine MH, Bienfait L, Indra R, Valbuena G, Phillips DH, Nortier JL, Stiborová M, Keun HC, Arlt VM. The impact of p53 on aristolochic acid I-induced nephrotoxicity and DNA damage in vivo and in vitro. Arch Toxicol 2019; 93:3345-3366. [PMID: 31602497 PMCID: PMC6823306 DOI: 10.1007/s00204-019-02578-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022]
Abstract
Exposure to aristolochic acid (AA) is associated with human nephropathy and urothelial cancer. The tumour suppressor TP53 is a critical gene in carcinogenesis and frequently mutated in AA-induced urothelial tumours. We investigated the impact of p53 on AAI-induced nephrotoxicity and DNA damage in vivo by treating Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice with 3.5 mg/kg body weight (bw) AAI daily for 2 or 6 days. Renal histopathology showed a gradient of intensity in proximal tubular injury from Trp53(+/+) to Trp53(-/-) mice, especially after 6 days. The observed renal injury was supported by nuclear magnetic resonance (NMR)-based metabonomic measurements, where a consistent Trp53 genotype-dependent trend was observed for urinary metabolites that indicate aminoaciduria (i.e. alanine), lactic aciduria (i.e. lactate) and glycosuria (i.e. glucose). However, Trp53 genotype had no impact on AAI-DNA adduct levels, as measured by 32P-postlabelling, in either target (kidney and bladder) or non-target (liver) tissues, indicating that the underlying mechanisms of p53-related AAI-induced nephrotoxicity cannot be explained by differences in AAI genotoxicity. Performing gas chromatography-mass spectrometry (GC-MS) on kidney tissues showed metabolic pathways affected by AAI treatment, but again Trp53 status did not clearly impact on such metabolic profiles. We also cultured primary mouse embryonic fibroblasts (MEFs) derived from Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice and exposed them to AAI in vitro (50 µM for up to 48 h). We found that Trp53 genotype impacted on the expression of NAD(P)H:quinone oxidoreductase (Nqo1), a key enzyme involved in AAI bioactivation. Nqo1 induction was highest in Trp53(+/+) MEFs and lowest in Trp53(-/-) MEFs; and it correlated with AAI-DNA adduct formation, with lowest adduct levels being observed in AAI-exposed Trp53(-/-) MEFs. Overall, our results clearly demonstrate that p53 status impacts on AAI-induced renal injury, but the underlying mechanism(s) involved remain to be further explored. Despite the impact of p53 on AAI bioactivation and DNA damage in vitro, such effects were not observed in vivo.
Collapse
Affiliation(s)
- Mateja Sborchia
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK
| | - Eric G De Prez
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Marie-Hélène Antoine
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Lucie Bienfait
- Department of Pathology, Erasme University Hospital, 1070, Brussels, Belgium
| | - Radek Indra
- Department of Biochemistry, Faculty of Science, Charles University Prague, 128 40, Prague, Czech Republic
| | - Gabriel Valbuena
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK
| | - Joëlle L Nortier
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University Prague, 128 40, Prague, Czech Republic
| | - Hector C Keun
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK.
| |
Collapse
|
10
|
Cancer therapeutic targeting using mutant-p53-specific siRNAs. Oncogene 2019; 38:3415-3427. [PMID: 30643191 PMCID: PMC6756012 DOI: 10.1038/s41388-018-0652-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/29/2018] [Accepted: 12/08/2018] [Indexed: 12/28/2022]
Abstract
Mutations in Tp53 compromise therapeutic response, due either to the dominant-negative effect over the functional wild-type allele; or as a result of the survival advantage conferred by mutant p53 to which cancer cells become addicted. Thus, targeting mutant p53 represents an effective therapeutic strategy to treat over half of all cancers. We have therefore generated a series of small-interfering-RNAs, capable of targeting four p53 hot-spot mutants which represent ~20% of all p53 mutations. These mutant–p53-specific siRNAs (MupSi) are highly specific in silencing the expression of the intended mutants without affecting wild-type p53. Functionally, these MupSis induce cell death by abrogating both the addiction to mutant p53 and the dominant-negative effect; and retard tumor growth in xenografts when administered in a therapeutic setting. These data together demonstrate the possibility of targeting mutant p53 specifically to improve clinical outcome.
Collapse
|
11
|
Zhao P, Pang X, Jiang J, Wang L, Zhu X, Yin Y, Zhai Q, Xiang X, Feng F, Xu W. TIPE1 promotes cervical cancer progression by repression of p53 acetylation and is associated with poor cervical cancer outcome. Carcinogenesis 2018; 40:592-599. [PMID: 30445600 DOI: 10.1093/carcin/bgy163] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/19/2018] [Accepted: 11/13/2018] [Indexed: 01/25/2023] Open
Affiliation(s)
- Peiqing Zhao
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Xiaoming Pang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Jie Jiang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Lianqing Wang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Xiaolan Zhu
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yingchun Yin
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Qiaoli Zhai
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Xinxin Xiang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Fan Feng
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wenlin Xu
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
12
|
Kim J, Park SH, Do KH, Kim D, Moon Y. Interference with mutagenic aflatoxin B1-induced checkpoints through antagonistic action of ochratoxin A in intestinal cancer cells: a molecular explanation on potential risk of crosstalk between carcinogens. Oncotarget 2018; 7:39627-39639. [PMID: 27119350 PMCID: PMC5129958 DOI: 10.18632/oncotarget.8914] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/05/2016] [Indexed: 12/16/2022] Open
Abstract
Foodborne aflatoxin B1 (AFB1) and ochratoxin A (OTA) cause genotoxic injury and subsequent tumor formation. As a biomarker of oncogenic stimulation by genotoxic mycotoxins, p53-triggered Mdm2 was assessed in intestinal cancer cells. AFB1 increased Mdm2 reporter expression in a dose-dependent manner. However, this was strongly antagonized by OTA treatment. As a positive transcription factor of Mdm2 expression, p53 levels were also increased by AFB1 alone and reduced by OTA. With marginal cell death responses, AFB1 induced p53-mediated S phase arrest and cell cycle-regulating target genes, which was completely suppressed by OTA. Although enterocyte-dominant CYP3A5 counteracted AFB1-induced DNA damage, expression of CYP3A5 was decreased by OTA or AFB1. Instead, OTA enhanced expression of another metabolic inactivating enzyme CYP3A4, attenuation of formation of AFB1-DNA adduct and p53-mediated cell cycle checking responses to the mutagens. Finally, the growth of intestinal cancer cells exposed to the mycotoxin mixture significantly exceeded the expected growth calculated from that of cells treated with each mycotoxin. Although AFB1-induced mutagen formation was decreased by OTA, interference with checkpoints through antagonistic action of OTA may contribute to the survival of tumor cells with deleterious mutations by genotoxic mycotoxins, potently increasing the risk of carcinogenesis.
Collapse
Affiliation(s)
- Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea
| | - Kee Hun Do
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea
| | - Dongwook Kim
- National Institute of Animal Science, RDA, Wanju, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea.,Research Institute for Basic Sciences and Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan, South Korea
| |
Collapse
|
13
|
Sabapathy K, Lane DP. Therapeutic targeting of p53: all mutants are equal, but some mutants are more equal than others. Nat Rev Clin Oncol 2017; 15:13-30. [DOI: 10.1038/nrclinonc.2017.151] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
14
|
He C, Li L, Guan X, Xiong L, Miao X. Mutant p53 Gain of Function and Chemoresistance: The Role of Mutant p53 in Response to Clinical Chemotherapy. Chemotherapy 2016; 62:43-53. [PMID: 27322648 DOI: 10.1159/000446361] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 04/18/2016] [Indexed: 11/19/2022]
Abstract
PURPOSE To review mechanisms underlying mutant p53 (mutp53) gain of function (GOF) and mutp53-induced chemoresistance, and to investigate the role of mutp53 in response to clinical chemotherapy. METHODS We searched the PubMed database for clinical studies from the past decade, including data evaluating the impact of mutp53 in clinical chemotherapy response. RESULTS Interactions between mutp53 and transcriptional factors, proteins or DNA structures, as well as epigenetic regulation, contribute to mutp53 GOF. Major mechanisms of mutp53-induced chemoresistance include enhanced drug efflux and metabolism, promoting survival, inhibiting apoptosis, upregulating DNA repair, suppressing autophagy, elevating microenvironmental resistance and inducing a stem-like phenotype. Clinically, mutp53 predicted resistance to chemotherapy in diffuse large B-cell lymphoma, and esophageal and oropharyngeal cancers, but its impact on chronic lymphocytic leukemia was unclear. In bladder cancer, mutp53 did not predict resistance, whereas in some breast and ovarian cancers, it was associated with sensitivity to certain chemotherapeutic agents. CONCLUSION mutp53 has an intricate role in the response to clinical chemotherapy and should not be interpreted in isolation. Furthermore, when predicting tumor response to chemotherapy based on the p53 status, the drugs used should also be taken into consideration. These concepts require further investigation.
Collapse
Affiliation(s)
- Chao He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | |
Collapse
|
15
|
Krais AM, Speksnijder EN, Melis JPM, Indra R, Moserova M, Godschalk RW, van Schooten FJ, Seidel A, Kopka K, Schmeiser HH, Stiborova M, Phillips DH, Luijten M, Arlt VM. The impact of p53 on DNA damage and metabolic activation of the environmental carcinogen benzo[a]pyrene: effects in Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice. Arch Toxicol 2016; 90:839-51. [PMID: 25995008 PMCID: PMC4785204 DOI: 10.1007/s00204-015-1531-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 05/05/2015] [Indexed: 12/22/2022]
Abstract
The tumour suppressor p53 is one of the most important cancer genes. Previous findings have shown that p53 expression can influence DNA adduct formation of the environmental carcinogen benzo[a]pyrene (BaP) in human cells, indicating a role for p53 in the cytochrome P450 (CYP) 1A1-mediated biotransformation of BaP in vitro. We investigated the potential role of p53 in xenobiotic metabolism in vivo by treating Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice with BaP. BaP-DNA adduct levels, as measured by (32)P-postlabelling analysis, were significantly higher in liver and kidney of Trp53(-/-) mice than of Trp53(+/+) mice. Complementarily, significantly higher amounts of BaP metabolites were also formed ex vivo in hepatic microsomes from BaP-pretreated Trp53(-/-) mice. Bypass of the need for metabolic activation by treating mice with BaP-7,8-dihydrodiol-9,10-epoxide resulted in similar adduct levels in liver and kidney in all mouse lines, confirming that the influence of p53 is on the biotransformation of the parent compound. Higher BaP-DNA adduct levels in the livers of Trp53(-/-) mice correlated with higher CYP1A protein levels and increased CYP1A enzyme activity in these animals. Our study demonstrates a role for p53 in the metabolism of BaP in vivo, confirming previous in vitro results on a novel role for p53 in CYP1A1-mediated BaP metabolism. However, our results also suggest that the mechanisms involved in the altered expression and activity of the CYP1A1 enzyme by p53 in vitro and in vivo are different.
Collapse
Affiliation(s)
- Annette M Krais
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment & Health, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Ewoud N Speksnijder
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), 3721 MA, Bilthoven, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Joost P M Melis
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), 3721 MA, Bilthoven, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Radek Indra
- Department of Biochemistry, Faculty of Science, Charles University, 12840, Prague 2, Czech Republic
| | - Michaela Moserova
- Department of Biochemistry, Faculty of Science, Charles University, 12840, Prague 2, Czech Republic
| | - Roger W Godschalk
- Department of Toxicology, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University Medical Centre, 6200 MD, Maastricht, The Netherlands
| | - Frederik-J van Schooten
- Department of Toxicology, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University Medical Centre, 6200 MD, Maastricht, The Netherlands
| | - Albrecht Seidel
- Biochemical Institute for Environmental Carcinogens, Prof. Dr. Gernot Grimmer-Foundation, 22927, Grosshansdorf, Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Heinz H Schmeiser
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, 12840, Prague 2, Czech Republic
| | - David H Phillips
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment & Health, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Mirjam Luijten
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), 3721 MA, Bilthoven, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment & Health, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
16
|
Karni-Schmidt O, Lokshin M, Prives C. The Roles of MDM2 and MDMX in Cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:617-44. [PMID: 27022975 DOI: 10.1146/annurev-pathol-012414-040349] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For more than 25 years, MDM2 and its homolog MDMX (also known as MDM4) have been shown to exert oncogenic activity. These two proteins are best understood as negative regulators of the p53 tumor suppressor, although they may have additional p53-independent roles. Understanding the dysregulation of MDM2 and MDMX in human cancers and how they function either together or separately in tumorigenesis may improve methods of diagnosis and for assessing prognosis. Targeting the proteins themselves, or their regulators, may be a promising therapeutic approach to treating some forms of cancer.
Collapse
Affiliation(s)
- Orit Karni-Schmidt
- Department of Biological Sciences, Columbia University, New York, NY 10027;
| | - Maria Lokshin
- Department of Biological Sciences, Columbia University, New York, NY 10027;
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, NY 10027;
| |
Collapse
|
17
|
Gurtner A, Falcone E, Garibaldi F, Piaggio G. Dysregulation of microRNA biogenesis in cancer: the impact of mutant p53 on Drosha complex activity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:45. [PMID: 26971015 PMCID: PMC4789259 DOI: 10.1186/s13046-016-0319-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 12/13/2022]
Abstract
A widespread decrease of mature microRNAs is often observed in human malignancies giving them potential to act as tumor suppressors. Thus, microRNAs may be potential targets for cancer therapy. The global miRNA deregulation is often the result of defects in the miRNA biogenesis pathway, such as genomic mutation or aberrant expression/localization of enzymes and cofactors responsible of miRNA maturation. Alterations in the miRNA biogenesis machinery impact on the establishment and development of cancer programs. Accumulation of pri-microRNAs and corresponding depletion of mature microRNAs occurs in human cancers compared to normal tissues, strongly indicating an impairment of crucial steps in microRNA biogenesis. In agreement, inhibition of microRNA biogenesis, by depletion of Dicer1 and Drosha, tends to enhance tumorigenesis in vivo. The p53 tumor suppressor gene, TP53, is mutated in half of human tumors resulting in an oncogene with Gain-Of-Function activities. In this review we discuss recent studies that have underlined a role of mutant p53 (mutp53) on the global regulation of miRNA biogenesis in cancer. In particular we describe how a new transcriptionally independent function of mutant p53 in miRNA maturation, through a mechanism by which this oncogene is able to interfere with the Drosha processing machinery, generally inhibits miRNA processing in cancer and consequently impacts on carcinogenesis.
Collapse
Affiliation(s)
- Aymone Gurtner
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Emmanuela Falcone
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Francesca Garibaldi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144, Rome, Italy.
| | - Giulia Piaggio
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144, Rome, Italy
| |
Collapse
|
18
|
Krais AM, Speksnijder EN, Melis JP, Singh R, Caldwell A, Gamboa da Costa G, Luijten M, Phillips DH, Arlt VM. Metabolic activation of 2-amino-1-methyl-6-phenylimidazo [4,5-b]pyridine and DNA adduct formation depends on p53: Studies in Trp53(+/+),Trp53(+/-) and Trp53(-/-) mice. Int J Cancer 2016; 138:976-82. [PMID: 26335255 PMCID: PMC4832306 DOI: 10.1002/ijc.29836] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/20/2015] [Indexed: 11/07/2022]
Abstract
The expression of the tumor suppressor p53 can influence the bioactivation of, and DNA damage induced by, the environmental carcinogen benzo[a]pyrene, indicating a role for p53 in its cytochrome P450 (CYP)-mediated biotransformation. The carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), which is formed during the cooking of food, is also metabolically activated by CYP enzymes, particularly CYP1A2. We investigated the potential role of p53 in PhIP metabolism in vivo by treating Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice with a single oral dose of 50 mg/kg body weight PhIP. N-(Deoxyguanosin-8-yl)-2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP-C8-dG) levels in DNA, measured by liquid chromatography-tandem mass spectrometry, were significantly lower in liver, colon, forestomach and glandular stomach of Trp53(-/-) mice compared to Trp53(+/+) mice. Lower PhIP-DNA adduct levels in the livers of Trp53(-/-) mice correlated with lower Cyp1a2 enzyme activity (measured by methoxyresorufin-O-demethylase activity) in these animals. Interestingly, PhIP-DNA adduct levels were significantly higher in kidney and bladder of Trp53(-/-) mice compared to Trp53(+/+) mice, which was accompanied by higher sulfotransferase (Sult) 1a1 protein levels and increased Sult1a1 enzyme activity (measured by 2-naphthylsulfate formation from 2-naphthol) in kidneys of these animals. Our study demonstrates a role for p53 in the metabolism of PhIP in vivo, extending previous results on a novel role for p53 in xenobiotic metabolism. Our results also indicate that the impact of p53 on PhIP biotransformation is tissue-dependent and that in addition to Cyp1a enzymes, Sult1a1 can contribute to PhIP-DNA adduct formation.
Collapse
Affiliation(s)
- Annette M. Krais
- Analytical and Environmental Sciences Division, MRC‐PHE Centre for Environment and HealthKing's College LondonLondonSE1 9NHUnited Kingdom
- Annette M. Krais current address is: Division of Occupational and Environmental MedicineLund University221 85LundSweden
| | - Ewoud N. Speksnijder
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM)BilthovenMA3721The Netherlands
- Department of Human GeneticsLeiden University Medical CenterLeiden2300The NetherlandsRC
| | - Joost P.M. Melis
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM)BilthovenMA3721The Netherlands
- Department of Human GeneticsLeiden University Medical CenterLeiden2300The NetherlandsRC
| | - Rajinder Singh
- Department of Cancer Studies and Molecular MedicineUniversity of LeicesterLeicesterLE2 7LXUnited Kingdom
| | - Anna Caldwell
- Mass Spectrometry Facility, King's College LondonLondonSE1 9NHUnited Kingdom
| | - Gonçalo Gamboa da Costa
- Division of Biochemical ToxicologyNational Center for Toxicological ResearchJeffersonAR72079
| | - Mirjam Luijten
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM)BilthovenMA3721The Netherlands
- Department of Human GeneticsLeiden University Medical CenterLeiden2300The NetherlandsRC
| | - David H. Phillips
- Analytical and Environmental Sciences Division, MRC‐PHE Centre for Environment and HealthKing's College LondonLondonSE1 9NHUnited Kingdom
| | - Volker M. Arlt
- Analytical and Environmental Sciences Division, MRC‐PHE Centre for Environment and HealthKing's College LondonLondonSE1 9NHUnited Kingdom
| |
Collapse
|
19
|
Sabapathy K. The Contrived Mutant p53 Oncogene - Beyond Loss of Functions. Front Oncol 2015; 5:276. [PMID: 26697411 PMCID: PMC4674554 DOI: 10.3389/fonc.2015.00276] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/24/2015] [Indexed: 12/22/2022] Open
Abstract
Mutations in p53 are almost synonymous with cancer – be it susceptibility to the disease or response to treatment – and therefore, are a critical determinant of overall survival. As most of these mutations occur in the DNA-binding domain of p53, many of the clinical correlations with mutant p53 have been initially relegated to the loss of its transcription-dependent activities as a tumor suppressor. However, significant efforts over the last two decades have led to the vast knowledge on the potential functions of the mutated p53 protein, which have been attributed to the physical presence of the mutant protein rather than the loss of its wild-type (WT) functions. Beyond the inhibitory effects of mutant p53 on the remaining WT protein that leads to the dominant-negative effect in the heterozygous state, mutant p53’s presence has also been significantly attributed to novel gain-of-functions that lead to addiction of cancer cells to its presence for survival, as well as for their ability to invade and metastasize, elevating it to a contrived oncogene that drives the cancer cells forward. This review will summarize the functional consequences of the presence of mutant p53 protein on cellular and organismal physiology.
Collapse
Affiliation(s)
- Kanaga Sabapathy
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Humphrey Oei Institute of Cancer Research , Singapore ; Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School , Singapore ; Institute of Molecular and Cellular Biology , Singapore ; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| |
Collapse
|
20
|
Bello SM, Smith CL, Eppig JT. Allele, phenotype and disease data at Mouse Genome Informatics: improving access and analysis. Mamm Genome 2015; 26:285-94. [PMID: 26162703 PMCID: PMC4534497 DOI: 10.1007/s00335-015-9582-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/23/2015] [Indexed: 11/16/2022]
Abstract
A core part of the Mouse Genome Informatics (MGI) resource is the collection of mouse mutations and the annotation phenotypes and diseases displayed by mice carrying these mutations. These data are integrated with the rest of data in MGI and exported to numerous other resources. The use of mouse phenotype data to drive translational research into human disease has expanded rapidly with the improvements in sequencing technology. MGI has implemented many improvements in allele and phenotype data annotation, search, and display to facilitate access to these data through multiple avenues. For example, the description of alleles has been modified to include more detailed categories of allele attributes. This allows improved discrimination between mutation types. Further, connections have been created between mutations involving multiple genes and each of the genes overlapping the mutation. This allows users to readily find all mutations affecting a gene and see all genes affected by a mutation. In a similar manner, the genes expressed by transgenic or knock-in alleles are now connected to these alleles. The advanced search forms and public reports have been updated to take advantage of these improvements. These search forms and reports are used by an expanding number of researchers to identify novel human disease genes and mouse models of human disease.
Collapse
Affiliation(s)
- Susan M Bello
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, 04609, USA,
| | | | | |
Collapse
|
21
|
Mantovani F, Zannini A, Rustighi A, Del Sal G. Interaction of p53 with prolyl isomerases: Healthy and unhealthy relationships. Biochim Biophys Acta Gen Subj 2015; 1850:2048-60. [PMID: 25641576 DOI: 10.1016/j.bbagen.2015.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/17/2015] [Accepted: 01/19/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND The p53 protein family, comprising p53, p63 and p73, is primarily involved in preserving genome integrity and preventing tumor onset, and also affects a range of physiological processes. Signal-dependent modifications of its members and of other pathway components provide cells with a sophisticated code to transduce a variety of stress signaling into appropriate responses. TP53 mutations are highly frequent in cancer and lead to the expression of mutant p53 proteins that are endowed with oncogenic activities and sensitive to stress signaling. SCOPE OF REVIEW p53 family proteins have unique structural and functional plasticity, and here we discuss the relevance of prolyl-isomerization to actively shape these features. MAJOR CONCLUSIONS The anti-proliferative functions of the p53 family are carefully activated upon severe stress and this involves the interaction with prolyl-isomerases. In particular, stress-induced stabilization of p53, activation of its transcriptional control over arrest- and cell death-related target genes and of its mitochondrial apoptotic function, as well as certain p63 and p73 functions, all require phosphorylation of specific S/T-P motifs and their subsequent isomerization by the prolyl-isomerase Pin1. While these functions of p53 counteract tumorigenesis, under some circumstances their activation by prolyl-isomerases may have negative repercussions (e.g. tissue damage induced by anticancer therapies and ischemia-reperfusion, neurodegeneration). Moreover, elevated Pin1 levels in tumor cells may transduce deregulated phosphorylation signaling into activation of mutant p53 oncogenic functions. GENERAL SIGNIFICANCE The complex repertoire of biological outcomes induced by p53 finds mechanistic explanations, at least in part, in the association between prolyl-isomerases and the p53 pathway. This article is part of a Special Issue entitled Proline-directed foldases: Cell signaling catalysts and drug targets.
Collapse
Affiliation(s)
- Fiamma Mantovani
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Alessandro Zannini
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Alessandra Rustighi
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, 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.
| |
Collapse
|
22
|
Soussi T. The TP53 gene network in a postgenomic era. Hum Mutat 2014; 35:641-2. [PMID: 24753184 DOI: 10.1002/humu.22562] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Abstract
Inactivation of TP53 pathways are the most common defects observed in human cancer. Although missense mutations remain the most frequent genetic event, it is now evident that dysfunction of several members of this network such as MDM2, MDM4 (mdmX), or miR-125b can substitute for TP53 mutations. This special issue on TP53 brings the TP53 gene into the post-genomic era. Several fundamental features of wild type and mutant proteins and their modifications are reviewed, as well as animal models and clinical aspects such as recommendations for patient care. The complex structure of this gene warrants innovative strategies to infer a more accurate status of human tumors. Recommendations and guidelines for reporting and annotating TP53 variants are also provided, to help researchers generate standardized data that are easy to understand, analyze, and exchange across various cancer variant databases.
Collapse
|
23
|
Leroy B, Anderson M, Soussi T. TP53 mutations in human cancer: database reassessment and prospects for the next decade. Hum Mutat 2014; 35:672-88. [PMID: 24665023 DOI: 10.1002/humu.22552] [Citation(s) in RCA: 269] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 03/04/2014] [Indexed: 12/18/2022]
Abstract
More than 50% of human tumors carry TP53 gene mutations and in consequence more than 45,000 somatic and germline mutations have been gathered in the UMD TP53 database (http://p53.fr). Analyses of these mutations have been invaluable for bettering our knowledge on the structure-function relationships within the TP53 protein and the high degree of heterogeneity of the various TP53 mutants in human cancer. In this review, we discuss how with the release of the sequences of thousands of tumor genomes issued from high-throughput sequencing, the description of novel TP53 mutants is now reaching a plateau indicating that we are close to the full set of mutants that target the elusive tumor-suppressive activity of this protein. We performed an extensive and thorough analysis of the TP53 mutation database, focusing particularly on specific sets of mutations that were overlooked in the past because of their low frequencies, for example, synonymous mutations, splice mutations, or mutations-targeting residues subject to posttranslational modifications. We also discuss the evolution of the statistical methods used to differentiate TP53 passenger mutations and artifactual data from true mutations, a process vital to the release of an accurate TP53 mutation database that will in turn be an invaluable tool for both clinicians and researchers.
Collapse
Affiliation(s)
- Bernard Leroy
- Université Pierre et Marie Curie-Paris 6, Paris, 75005, France
| | | | | |
Collapse
|
24
|
Whole-genome sequencing analysis of phenotypic heterogeneity and anticipation in Li-Fraumeni cancer predisposition syndrome. Proc Natl Acad Sci U S A 2014; 111:15497-501. [PMID: 25313051 DOI: 10.1073/pnas.1417322111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Li-Fraumeni syndrome (LFS) and its variant form (LFL) is a familial predisposition to multiple forms of childhood, adolescent, and adult cancers associated with germ-line mutation in the TP53 tumor suppressor gene. Individual disparities in tumor patterns are compounded by acceleration of cancer onset with successive generations. It has been suggested that this apparent anticipation pattern may result from germ-line genomic instability in TP53 mutation carriers, causing increased DNA copy-number variations (CNVs) with successive generations. To address the genetic basis of phenotypic disparities of LFS/LFL, we performed whole-genome sequencing (WGS) of 13 subjects from two generations of an LFS kindred. Neither de novo CNV nor significant difference in total CNV was detected in relation with successive generations or with age at cancer onset. These observations were consistent with an experimental mouse model system showing that trp53 deficiency in the germ line of father or mother did not increase CNV occurrence in the offspring. On the other hand, individual records on 1,771 TP53 mutation carriers from 294 pedigrees were compiled to assess genetic anticipation patterns (International Agency for Research on Cancer TP53 database). No strictly defined anticipation pattern was observed. Rather, in multigeneration families, cancer onset was delayed in older compared with recent generations. These observations support an alternative model for apparent anticipation in which rare variants from noncarrier parents may attenuate constitutive resistance to tumorigenesis in the offspring of TP53 mutation carriers with late cancer onset.
Collapse
|
25
|
Soussi T, Leroy B, Taschner PEM. Recommendations for analyzing and reporting TP53 gene variants in the high-throughput sequencing era. Hum Mutat 2014; 35:766-78. [PMID: 24729566 DOI: 10.1002/humu.22561] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/02/2014] [Indexed: 12/27/2022]
Abstract
The architecture of TP53, the most frequently mutated gene in human cancer, is more complex than previously thought. Using TP53 variants as clinical biomarkers to predict response to treatment or patient outcome requires an unequivocal and standardized procedure toward a definitive strategy for the clinical evaluation of variants to provide maximum diagnostic sensitivity and specificity. An intronic promoter and two novel exons have been identified resulting in the expression of multiple transcripts and protein isoforms. These regions are additional targets for mutation events impairing the tumor suppressive activity of TP53. Reassessment of variants located in these regions is needed to refine their prognostic value in many malignancies. We recommend using the stable Locus Reference Genomic reference sequence for detailed and unequivocal reports and annotations of germ line and somatic alterations on all TP53 transcripts and protein isoforms according to the recommendations of the Human Genome Variation Society. This novel and comprehensive description framework will generate standardized data that are easy to understand, analyze, and exchange across various cancer variant databases. Based on the statistical analysis of more than 45,000 variants in the latest version of the UMD TP53 database, we also provide a classification of their functional effects ("pathogenicity").
Collapse
Affiliation(s)
- Thierry Soussi
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institute, Stockholm, Sweden; Université Pierre et Marie Curie-Paris 6, Paris, 75005, France
| | | | | |
Collapse
|
26
|
Bisio A, Ciribilli Y, Fronza G, Inga A, Monti P. TP53 Mutants in the Tower of Babel of Cancer Progression. Hum Mutat 2014; 35:689-701. [DOI: 10.1002/humu.22514] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 01/06/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Alessandra Bisio
- Laboratory of Transcriptional Networks; Centre for Integrative Biology (CIBIO); University of Trento; Trento Italy
| | - Yari Ciribilli
- Laboratory of Transcriptional Networks; Centre for Integrative Biology (CIBIO); University of Trento; Trento Italy
| | - Gilberto Fronza
- Mutagenesis Unit; IRCSS Azienda Ospedaliera Universitaria San Martino-IST-Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
| | - Alberto Inga
- Laboratory of Transcriptional Networks; Centre for Integrative Biology (CIBIO); University of Trento; Trento Italy
| | - Paola Monti
- Mutagenesis Unit; IRCSS Azienda Ospedaliera Universitaria San Martino-IST-Istituto Nazionale per la Ricerca sul Cancro; Genoa Italy
| |
Collapse
|
27
|
Nguyen TA, Menendez D, Resnick MA, Anderson CW. Mutant TP53 posttranslational modifications: challenges and opportunities. Hum Mutat 2014; 35:738-55. [PMID: 24395704 DOI: 10.1002/humu.22506] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022]
Abstract
The wild-type (WT) human p53 (TP53) tumor suppressor can be posttranslationally modified at over 60 of its 393 residues. These modifications contribute to changes in TP53 stability and in its activity as a transcription factor in response to a wide variety of intrinsic and extrinsic stresses in part through regulation of protein-protein and protein-DNA interactions. The TP53 gene frequently is mutated in cancers, and in contrast to most other tumor suppressors, the mutations are mostly missense often resulting in the accumulation of mutant (MUT) protein, which may have novel or altered functions. Most MUT TP53s can be posttranslationally modified at the same residues as in WT TP53. Strikingly, however, codons for modified residues are rarely mutated in human tumors, suggesting that TP53 modifications are not essential for tumor suppression activity. Nevertheless, these modifications might alter MUT TP53 activity and contribute to a gain-of-function leading to increased metastasis and tumor progression. Furthermore, many of the signal transduction pathways that result in TP53 modifications are altered or disrupted in cancers. Understanding the signaling pathways that result in TP53 modification and the functions of these modifications in both WT TP53 and its many MUT forms may contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Thuy-Ai Nguyen
- Chromosome Stability Section, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | |
Collapse
|