1
|
Guo Y, Wu H, Wiesmüller L, Chen M. Canonical and non-canonical functions of p53 isoforms: potentiating the complexity of tumor development and therapy resistance. Cell Death Dis 2024; 15:412. [PMID: 38866752 PMCID: PMC11169513 DOI: 10.1038/s41419-024-06783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
Full-length p53 (p53α) plays a pivotal role in maintaining genomic integrity and preventing tumor development. Over the years, p53 was found to exist in various isoforms, which are generated through alternative splicing, alternative initiation of translation, and internal ribosome entry site. p53 isoforms, either C-terminally altered or N-terminally truncated, exhibit distinct biological roles compared to p53α, and have significant implications for tumor development and therapy resistance. Due to a lack of part and/or complete C- or N-terminal domains, ectopic expression of some p53 isoforms failed to induce expression of canonical transcriptional targets of p53α like CDKN1A or MDM2, even though they may bind their promoters. Yet, p53 isoforms like Δ40p53α still activate subsets of targets including MDM2 and BAX. Furthermore, certain p53 isoforms transactivate even novel targets compared to p53α. More recently, non-canonical functions of p53α in DNA repair and of different isoforms in DNA replication unrelated to transcriptional activities were discovered, amplifying the potential of p53 as a master regulator of physiological and tumor suppressor functions in human cells. Both regarding canonical and non-canonical functions, alternative p53 isoforms frequently exert dominant negative effects on p53α and its partners, which is modified by the relative isoform levels. Underlying mechanisms include hetero-oligomerization, changes in subcellular localization, and aggregation. These processes ultimately influence the net activities of p53α and give rise to diverse cellular outcomes. Biological roles of p53 isoforms have implications for tumor development and cancer therapy resistance. Dysregulated expression of isoforms has been observed in various cancer types and is associated with different clinical outcomes. In conclusion, p53 isoforms have expanded our understanding of the complex regulatory network involving p53 in tumors. Unraveling the mechanisms underlying the biological roles of p53 isoforms provides new avenues for studies aiming at a better understanding of tumor development and developing therapeutic interventions to overcome resistance.
Collapse
Affiliation(s)
- Yitian Guo
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China.
| | - Hang Wu
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Ming Chen
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Ray Das S, Delahunt B, Lasham A, Li K, Wright D, Print C, Slatter T, Braithwaite A, Mehta S. Combining TP53 mutation and isoform has the potential to improve clinical practice. Pathology 2024; 56:473-483. [PMID: 38594116 DOI: 10.1016/j.pathol.2024.02.003] [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: 10/02/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 04/11/2024]
Abstract
The clinical importance of assessing and combining data on TP53 mutations and isoforms is discussed in this article. It gives a succinct overview of the structural makeup and key biological roles of the isoforms. It then provides a comprehensive summary of the roles that p53 isoforms play in cancer development, therapy response and resistance. The review provides a summary of studies demonstrating the role of p53 isoforms as potential prognostic indicators. It further provides evidence on how the presence of TP53 mutations may affect one or more of these activities and the association of p53 isoforms with clinicopathological data in various tumour types. The review gives insight into the present diagnostic hurdles for identifying TP53 isoforms and makes recommendations to improve their evaluation. In conclusion, this review offers suggestions for enhancing the identification and integration of TP53 isoforms in conjunction with mutation data within the clinical context.
Collapse
Affiliation(s)
- Sankalita Ray Das
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Brett Delahunt
- Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - Annette Lasham
- Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand; Te Aka Mātauranga Matepukupuku (Centre for Cancer Research), University of Auckland, Auckland, New Zealand
| | - Kunyu Li
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Deborah Wright
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Cristin Print
- Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand; Te Aka Mātauranga Matepukupuku (Centre for Cancer Research), University of Auckland, Auckland, New Zealand
| | - Tania Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Antony Braithwaite
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Sunali Mehta
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
3
|
Wiles AK, Mehta S, Millier M, Woolley AG, Li K, Parker K, Kazantseva M, Wilson M, Young K, Bowie S, Ray S, Slatter TL, Stamp LK, Hessian PA, Braithwaite AW. Activated CD90/Thy-1 fibroblasts co-express the Δ133p53β isoform and are associated with highly inflamed rheumatoid arthritis. Arthritis Res Ther 2023; 25:62. [PMID: 37060003 PMCID: PMC10105423 DOI: 10.1186/s13075-023-03040-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/29/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND The p53 isoform Δ133p53β is known to be associated with cancers driven by inflammation. Many of the features associated with the development of inflammation in rheumatoid arthritis (RA) parallel those evident in cancer progression. However, the role of this isoform in RA has not yet been explored. The aim of this study was to determine whether Δ133p53β is driving aggressive disease in RA. METHODS Using RA patient synovia, we carried out RT-qPCR and RNAScope-ISH to determine both protein and mRNA levels of Δ133p53 and p53. We also used IHC to determine the location and type of cells with elevated levels of Δ133p53β. Plasma cytokines were also measured using a BioPlex cytokine panel and data analysed by the Milliplex Analyst software. RESULTS Elevated levels of pro-inflammatory plasma cytokines were associated with synovia from RA patients displaying extensive tissue inflammation, increased immune cell infiltration and the highest levels of Δ133TP53 and TP53β mRNA. Located in perivascular regions of synovial sub-lining and surrounding ectopic lymphoid structures (ELS) were a subset of cells with high levels of CD90, a marker of 'activated fibroblasts' together with elevated levels of Δ133p53β. CONCLUSIONS Induction of Δ133p53β in CD90+ synovial fibroblasts leads to an increase in cytokine and chemokine expression and the recruitment of proinflammatory cells into the synovial joint, creating a persistently inflamed environment. Our results show that dysregulated expression of Δ133p53β could represent one of the early triggers in the immunopathogenesis of RA and actively perpetuates chronic synovial inflammation. Therefore, Δ133p53β could be used as a biomarker to identify RA patients more likely to develop aggressive disease who might benefit from targeted therapy to cytokines such as IL-6.
Collapse
Affiliation(s)
- Anna K Wiles
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
- Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Sunali Mehta
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
- Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Melanie Millier
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Adele G Woolley
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
- Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Kunyu Li
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
| | - Kim Parker
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
| | - Marina Kazantseva
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
- Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Michelle Wilson
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
| | - Katie Young
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
| | - Sarah Bowie
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
| | - Sankalita Ray
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
| | - Tania L Slatter
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand
- Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Paul A Hessian
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Antony W Braithwaite
- Department of Pathology, University of Otago, Hercus Building, 58 Hanover Street, Dunedin, New Zealand.
- Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand.
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, New Zealand.
| |
Collapse
|
4
|
Singh S, Rani H, Sharma N, Behl T, Zahoor I, Makeen HA, Albratty M, Alhazm HA, Aleya L. Targeting multifunctional magnetic nanowires for drug delivery in cancer cell death: an emerging paradigm. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:57219-57235. [PMID: 37010687 DOI: 10.1007/s11356-023-26650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/21/2023] [Indexed: 05/10/2023]
Abstract
Apoptosis, often known as programmed cell death is a mechanism used by numerous species to maintain tissue homeostasis. The process leading to cell death is complicated because it requires the stimulation of caspases. According to several studies, nanowires have important medical benefits, can kill cells by adhering to cancer cells, destroying them, and killing the entire cell using a triple attack that integrates vibration, heat, and drug delivery to trigger apoptosis. The sewage effluents and industrial, fertilizer and organic wastes decomposition can produce elevated levels of chemicals in the environment which may interrupt the cell cycle and activate apoptosis. The purpose of this review is to give a thorough summary of the evidence that is currently available on apoptosis. Current review discussed topics like the morphological and biochemical alterations that occur during apoptosis, as well as the various mechanisms that cause cell death, including the intrinsic (or mitochondrial), extrinsic (or death receptor), and intrinsic endoplasmic reticulum pathway. The apoptosis reduction in cancer development is mediated by (i) an imbalance between pro- and anti-apoptotic proteins, such as members of the B-cell lymphoma-2 (BCL2) family of proteins, tumour protein 53 and inhibitor of apoptosis proteins, (ii) a reduction in caspase activity, and (iii) impaired death receptor signalling. This review does an excellent task of outlining the function of nanowires in both apoptosis induction and targeted drug delivery for cancer cells. A comprehensive summary of the relevance of nanowires synthesised for the purpose of inducing apoptosis in cancer cells has been compiled collectively.
Collapse
Affiliation(s)
- Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Hema Rani
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana, 141104, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India.
| | - Tapan Behl
- School of Health Sciences &Technology, University of Petroleum and Energy Studies, Bidholi, Uttarakhand, 248007, Dehradun, India
| | - Ishrat Zahoor
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazm
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| |
Collapse
|
5
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
6
|
Alterations in the p53 isoform ratio govern breast cancer cell fate in response to DNA damage. Cell Death Dis 2022; 13:907. [PMID: 36307393 PMCID: PMC9616954 DOI: 10.1038/s41419-022-05349-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022]
Abstract
Our previous studies have shown that p53 isoform expression is altered in breast cancer and related to prognosis. In particular, a high ∆40p53:p53α ratio is associated with worse disease-free survival. In this manuscript, the influence of altered Δ40p53 and p53α levels on the response to standard of care DNA-damaging agents used in breast cancer treatment was investigated in vitro. Our results revealed that a high Δ40p53:p53α ratio causes cells to respond differently to doxorubicin and cisplatin treatments. Δ40p53 overexpression significantly impairs the cells' sensitivity to doxorubicin through reducing apoptosis and DNA damage, whereas Δ40p53 knockdown has the opposite effect. Further, a high Δ40p53:p53α ratio inhibited the differential expression of several genes following doxorubicin and promoted DNA repair, impairing the cells' canonical response. Overall, our results suggest that the response of breast cancer cells to standard of care DNA-damaging therapies is dependent on the expression of p53 isoforms, which may contribute to outcomes in breast cancer.
Collapse
|
7
|
Mehta S, Campbell H, Drummond CJ, Li K, Murray K, Slatter T, Bourdon JC, Braithwaite AW. Adaptive homeostasis and the p53 isoform network. EMBO Rep 2021; 22:e53085. [PMID: 34779563 PMCID: PMC8647153 DOI: 10.15252/embr.202153085] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/12/2021] [Accepted: 10/28/2021] [Indexed: 12/25/2022] Open
Abstract
All living organisms have developed processes to sense and address environmental changes to maintain a stable internal state (homeostasis). When activated, the p53 tumour suppressor maintains cell and organ integrity and functions in response to homeostasis disruptors (stresses) such as infection, metabolic alterations and cellular damage. Thus, p53 plays a fundamental physiological role in maintaining organismal homeostasis. The TP53 gene encodes a network of proteins (p53 isoforms) with similar and distinct biochemical functions. The p53 network carries out multiple biological activities enabling cooperation between individual cells required for long‐term survival of multicellular organisms (animals) in response to an ever‐changing environment caused by mutation, infection, metabolic alteration or damage. In this review, we suggest that the p53 network has evolved as an adaptive response to pathogen infections and other environmental selection pressures.
Collapse
Affiliation(s)
- Sunali Mehta
- Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Hamish Campbell
- Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Catherine J Drummond
- Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Kunyu Li
- Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Kaisha Murray
- Dundee Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Tania Slatter
- Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Jean-Christophe Bourdon
- Dundee Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Antony W Braithwaite
- Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Biodiscovery, University of Otago, Dunedin, New Zealand
| |
Collapse
|
8
|
Zhang X, Groen K, Morten BC, Steffens Reinhardt L, Campbell HG, Braithwaite AW, Bourdon JC, Avery-Kiejda KA. Effect of p53 and its N-terminally truncated isoform, Δ40p53, on breast cancer migration and invasion. Mol Oncol 2021; 16:447-465. [PMID: 34657382 PMCID: PMC8763661 DOI: 10.1002/1878-0261.13118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/27/2021] [Accepted: 10/14/2021] [Indexed: 01/07/2023] Open
Abstract
Breast cancer is the most diagnosed malignancy in women, with over half a million women dying from this disease each year. In our previous studies, ∆40p53, an N‐terminally truncated p53 isoform, was found to be upregulated in breast cancers, and a high ∆40p53 : p53α ratio was linked with worse disease‐free survival. Although p53α inhibits cancer migration and invasion, little is known about the role of ∆40p53 in regulating these metastasis‐related processes and its role in contributing to worse prognosis. The aim of this study was to assess the role of ∆40p53 in breast cancer migration and invasion. A relationship between Δ40p53 and gene expression profiles was identified in oestrogen‐receptor‐positive breast cancer specimens. To further evaluate the role of Δ40p53 in oestrogen‐receptor‐positive breast cancer, MCF‐7 and ZR75‐1 cell lines were transduced to knockdown p53α or Δ40p53 and overexpress Δ40p53. Proliferation, migration and invasion were assessed in the transduced sublines, and gene expression was assessed through RNA‐sequencing and validated by reverse‐transcription quantitative PCR. Knockdown of both p53α and ∆40p53 resulted in increased proliferation, whereas overexpression of ∆40p53 reduced proliferation rates. p53α knockdown was also associated with increased cell mobility. ∆40p53 overexpression reduced both migratory and invasive properties of the transduced cells. Phenotypic findings are supported by gene expression data, including differential expression of LRG1, HYOU1, UBE2QL1, SERPINA5 and PCDH7. Taken together, these results suggest that, at the basal level, ∆40p53 works similarly to p53α in suppressing cellular mobility and proliferation, although the role of Δ40p53 may be cell context‐specific.
Collapse
Affiliation(s)
- Xiajie Zhang
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia
| | - Kira Groen
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia
| | - Brianna C Morten
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia
| | - Luiza Steffens Reinhardt
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia
| | - Hamish G Campbell
- Children's Medical Research Institute, University of Sydney, NSW, Australia
| | - Antony W Braithwaite
- Children's Medical Research Institute, University of Sydney, NSW, Australia.,Department of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand
| | | | - Kelly A Avery-Kiejda
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia
| |
Collapse
|
9
|
Δ133p53β isoform pro-invasive activity is regulated through an aggregation-dependent mechanism in cancer cells. Nat Commun 2021; 12:5463. [PMID: 34526502 PMCID: PMC8443592 DOI: 10.1038/s41467-021-25550-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/04/2021] [Indexed: 11/09/2022] Open
Abstract
The p53 isoform, Δ133p53β, is critical in promoting cancer. Here we report that Δ133p53β activity is regulated through an aggregation-dependent mechanism. Δ133p53β aggregates were observed in cancer cells and tumour biopsies. The Δ133p53β aggregation depends on association with interacting partners including p63 family members or the CCT chaperone complex. Depletion of the CCT complex promotes accumulation of Δ133p53β aggregates and loss of Δ133p53β dependent cancer cell invasion. In contrast, association with p63 family members recruits Δ133p53β from aggregates increasing its intracellular mobility. Our study reveals novel mechanisms of cancer progression for p53 isoforms which are regulated through sequestration in aggregates and recruitment upon association with specific partners like p63 isoforms or CCT chaperone complex, that critically influence cancer cell features like EMT, migration and invasion.
Collapse
|
10
|
Żydowicz-Machtel P, Dutkiewicz M, Swiatkowska A, Gurda-Woźna D, Ciesiołka J. Translation of human Δ133p53 mRNA and its targeting by antisense oligonucleotides complementary to the 5'-terminal region of this mRNA. PLoS One 2021; 16:e0256938. [PMID: 34492050 PMCID: PMC8423303 DOI: 10.1371/journal.pone.0256938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/18/2021] [Indexed: 11/18/2022] Open
Abstract
The p53 protein is expressed as at least twelve protein isoforms. Within intron 4 of the human TP53 gene, a P2 transcription initiation site is located and this transcript encodes two p53 isoforms: Δ133p53 and Δ160p53. Here, the secondary structure of the 5'-terminal region of P2-initiated mRNA was characterized by means of the SHAPE and Pb2+-induced cleavage methods and for the first time, a secondary structure model of this region was proposed. Surprisingly, only Δ133p53 isoform was synthetized in vitro from the P2-initiated p53 mRNA while translation from both initiation codons occurred after the transfection of vector-encoded model mRNA to HCT116 cells. Interestingly, translation performed in the presence of the cap analogue suggested that the cap-independent process contributes to the translation of P2-initiated p53 mRNA. Subsequently, several antisense oligonucleotides targeting the 5'-terminal region of P2-initiated p53 mRNA were designed. The selected oligomers were applied in in vitro translation assays as well as in cell lines and their impact on the Δ133p53 synthesis and on cell viability was investigated. The results show that these oligomers are attractive tools in the modulation of the translation of P2-initiated p53 mRNA through attacking the 5' terminus of the transcript. Since cell proliferation is also reduced by antisense oligomers that lower the level of Δ133p53, this demonstrates an involvement of this isoform in tumorigenesis.
Collapse
Affiliation(s)
| | - Mariola Dutkiewicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Agata Swiatkowska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Dorota Gurda-Woźna
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Jerzy Ciesiołka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
- * E-mail:
| |
Collapse
|
11
|
Legscha KJ, Antunes Ferreira E, Chamoun A, Lang A, Awwad MHS, Ton GNHQ, Galetzka D, Guezguez B, Hundemer M, Bourdon JC, Munder M, Theobald M, Echchannaoui H. Δ133p53α enhances metabolic and cellular fitness of TCR-engineered T cells and promotes superior antitumor immunity. J Immunother Cancer 2021; 9:jitc-2020-001846. [PMID: 34112738 PMCID: PMC8194333 DOI: 10.1136/jitc-2020-001846] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2021] [Indexed: 12/13/2022] Open
Abstract
Background Tumor microenvironment-associated T cell senescence is a key limiting factor for durable effective cancer immunotherapy. A few studies have demonstrated the critical role of the tumor suppressor TP53-derived p53 isoforms in cellular senescence process of non-immune cells. However, their role in lymphocytes, in particular tumor-antigen (TA) specific T cells remain largely unexplored. Methods Human T cells from peripheral blood were retrovirally engineered to coexpress a TA-specific T cell receptor and the Δ133p53α-isoform, and characterized for their cellular phenotype, metabolic profile and effector functions. Results Phenotypic analysis of Δ133p53α-modified T cells revealed a marked reduction of the T-cell inhibitory molecules (ie, CD160 and TIGIT), a lower frequency of senescent-like CD57+ and CD160+ CD8+ T cell populations, and an increased number of less differentiated CD28+ T cells. Consistently, we demonstrated changes in the cellular metabolic program toward a quiescent T cell state. On a functional level, Δ133p53α-expressing T cells acquired a long-term proliferative capacity, showed superior cytokine secretion and enhanced tumor-specific killing in vitro and in mouse tumor model. Finally, we demonstrated the capacity of Δ133p53α to restore the antitumor response of senescent T cells isolated from multiple myeloma patients. Conclusion This study uncovered a broad effect of Δ133p53α isoform in regulating T lymphocyte function. Enhancing fitness and effector functions of senescent T cells by modulation of p53 isoforms could be exploited for future translational research to improve cancer immunotherapy and immunosenescence-related diseases.
Collapse
Affiliation(s)
- Kevin Jan Legscha
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Edite Antunes Ferreira
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Antonios Chamoun
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander Lang
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | - Danuta Galetzka
- Department of Radiation Oncology and Radiotherapy, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Borhane Guezguez
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Partner Site, Mainz, Germany
| | - Michael Hundemer
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | - Markus Munder
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Partner Site, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hakim Echchannaoui
- Department of Hematology, Oncology and Pneumology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany .,German Cancer Consortium (DKTK), Partner Site, Mainz, Germany
| |
Collapse
|
12
|
p53/p73 Protein Network in Colorectal Cancer and Other Human Malignancies. Cancers (Basel) 2021; 13:cancers13122885. [PMID: 34207603 PMCID: PMC8227208 DOI: 10.3390/cancers13122885] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The p53 family of proteins comprises p53, p63, and p73, which share high structural and functional similarity. The two distinct promoters of each locus, the alternative splicing, and the alternative translation initiation sites enable the generation of numerous isoforms with different protein-interacting domains and distinct activities. The co-expressed p53/p73 isoforms have significant but distinct roles in carcinogenesis. Their activity is frequently impaired in human tumors including colorectal carcinoma due to dysregulated expression and a dominant-negative effect accomplished by some isoforms and p53 mutants. The interactions between isoforms are particularly important to understand the onset of tumor formation, progression, and therapeutic response. The understanding of the p53/p73 network can contribute to the development of new targeted therapies. Abstract The p53 tumor suppressor protein is crucial for cell growth control and the maintenance of genomic stability. Later discovered, p63 and p73 share structural and functional similarity with p53. To understand the p53 pathways more profoundly, all family members should be considered. Each family member possesses two promoters and alternative translation initiation sites, and they undergo alternative splicing, generating multiple isoforms. The resulting isoforms have important roles in carcinogenesis, while their expression is dysregulated in several human tumors including colorectal carcinoma, which makes them potential targets in cancer treatment. Their activities arise, at least in part, from the ability to form tetramers that bind to specific DNA sequences and activate the transcription of target genes. In this review, we summarize the current understanding of the biological activities and regulation of the p53/p73 isoforms, highlighting their role in colorectal tumorigenesis. The analysis of the expression patterns of the p53/p73 isoforms in human cancers provides an important step in the improvement of cancer therapy. Furthermore, the interactions among the p53 family members which could modulate normal functions of the canonical p53 in tumor tissue are described. Lastly, we emphasize the importance of clinical studies to assess the significance of combining the deregulation of different members of the p53 family to define the outcome of the disease.
Collapse
|
13
|
Genetic Drivers of Head and Neck Squamous Cell Carcinoma: Aberrant Splicing Events, Mutational Burden, HPV Infection and Future Targets. Genes (Basel) 2021; 12:genes12030422. [PMID: 33804181 PMCID: PMC7998272 DOI: 10.3390/genes12030422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/16/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
Head and neck cancers include cancers that originate from a variety of locations. These include the mouth, nasal cavity, throat, sinuses, and salivary glands. These cancers are the sixth most diagnosed cancers worldwide. Due to the tissues they arise from, they are collectively named head and neck squamous cell carcinomas (HNSCC). The most important risk factors for head and neck cancers are infection with human papillomavirus (HPV), tobacco use and alcohol consumption. The genetic basis behind the development and progression of HNSCC includes aberrant non-coding RNA levels. However, one of the most important differences between healthy tissue and HNSCC tissue is changes in the alternative splicing of genes that play a vital role in processes that can be described as the hallmarks of cancer. These changes in the expression profile of alternately spliced mRNA give rise to various protein isoforms. These protein isoforms, alternate methylation of proteins, and changes in the transcription of non-coding RNAs (ncRNA) can be used as diagnostic or prognostic markers and as targets for the development of new therapeutic agents. This review aims to describe changes in alternative splicing and ncRNA patterns that contribute to the development and progression of HNSCC. It will also review the use of the changes in gene expression as biomarkers or as the basis for the development of new therapies.
Collapse
|
14
|
Niu G, Hellmuth I, Flisikowska T, Pausch H, Rieblinger B, Carrapeiro A, Schade B, Böhm B, Kappe E, Fischer K, Klinger B, Steiger K, Burgkart R, Bourdon JC, Saur D, Kind A, Schnieke A, Flisikowski K. Porcine model elucidates function of p53 isoform in carcinogenesis and reveals novel circTP53 RNA. Oncogene 2021; 40:1896-1908. [PMID: 33603167 PMCID: PMC7946636 DOI: 10.1038/s41388-021-01686-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 01/31/2023]
Abstract
Recent years have seen an increasing number of genetically engineered pig models of human diseases including cancer. We previously generated pigs with a modified TP53 allele that carries a Cre-removable transcriptional stop signal in intron 1, and an oncogenic mutation TP53R167H (orthologous to human TP53R175H) in exon 5. Pigs with the unrecombined mutant allele (flTP53R167H) develop mainly osteosarcoma but also nephroblastomas and lymphomas. This observation suggested that TP53 gene dysfunction is itself the key initiator of bone tumorigenesis, but raises the question which aspects of the TP53 regulation lead to the development of such a narrow tumour spectrum. Molecular analysis of p53 revealed the presence of two internal TP53 promoters (Pint and P2) equivalent to those found in human. Consequently, both pig and human express TP53 isoforms. Data presented here strongly suggest that P2-driven expression of the mutant R167H-Δ152p53 isoform (equivalent to the human R175H-Δ160p53 isoform) and its circular counterpart circTP53 determine the tumour spectrum and play a critical role in the malignant transformation in flTP53R167H pigs. The detection of Δ152p53 isoform mRNA in serum is indicative of tumorigenesis. Furthermore, we showed a tissue-specific p53-dependent deregulation of the p63 and p73 isoforms in these tumours. This study highlights important species-specific differences in the transcriptional regulation of TP53. Considering the similarities of TP53 regulation between pig and human, these observations provide useful pointers for further investigation into isoform function including the novel circTP53 in both the pig model and human patients.
Collapse
Affiliation(s)
- Guanglin Niu
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Isabel Hellmuth
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | | | - Beate Rieblinger
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Alexander Carrapeiro
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Benjamin Schade
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - Brigitte Böhm
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - Eva Kappe
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - Konrad Fischer
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Bernhard Klinger
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Katja Steiger
- School of Medicine, Institute of Pathology, Technische Universität München, Munich, Germany
| | - Reiner Burgkart
- Klinik und Poliklinik für Orthopädie und Sportorthopädie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, Technische Universität München, Munich, Germany
| | | |
Collapse
|
15
|
Heissig B, Salama Y, Osada T, Okumura K, Hattori K. The Multifaceted Role of Plasminogen in Cancer. Int J Mol Sci 2021; 22:ijms22052304. [PMID: 33669052 PMCID: PMC7956603 DOI: 10.3390/ijms22052304] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrinolytic factors like plasminogen, tissue-type plasminogen activator (tPA), and urokinase plasminogen activator (uPA) dissolve clots. Though mere extracellular-matrix-degrading enzymes, fibrinolytic factors interfere with many processes during primary cancer growth and metastasis. Their many receptors give them access to cellular functions that tumor cells have widely exploited to promote tumor cell survival, growth, and metastatic abilities. They give cancer cells tools to ensure their own survival by interfering with the signaling pathways involved in senescence, anoikis, and autophagy. They can also directly promote primary tumor growth and metastasis, and endow tumor cells with mechanisms to evade myelosuppression, thus acquiring drug resistance. In this review, recent studies on the role fibrinolytic factors play in metastasis and controlling cell-death-associated processes are presented, along with studies that describe how cancer cells have exploited plasminogen receptors to escape myelosuppression.
Collapse
Affiliation(s)
- Beate Heissig
- Immunological Diagnosis, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan;
- Correspondence: ; Tel.: +81-3-3813-3111
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine;
| | - Taro Osada
- Department of Gastroenterology Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, Chiba 279-0021, Japan;
| | - Ko Okumura
- Immunological Diagnosis, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan;
| | - Koichi Hattori
- Center for Genomic & Regenerative Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan;
| |
Collapse
|
16
|
The Δ133p53 Isoforms, Tuners of the p53 Pathway. Cancers (Basel) 2020; 12:cancers12113422. [PMID: 33218139 PMCID: PMC7698932 DOI: 10.3390/cancers12113422] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary TP53, the most frequently mutated gene in human cancers, has a key role in the maintenance of the genetic stability and, thus, in preventing tumor development. The p53-dependent responses were long thought to be solely driven by canonical p53α. However, it is now known that TP53 physiologically expresses at least 12 p53 isoforms including Δ133p53α, Δ133p53β and Δ133p53γ. The Δ133p53 isoforms are potent modulators of the p53 pathway that regulate critical functions in cancer, physiological and premature aging, neurodegenerative diseases, immunity and inflammation, and tissue repair. This review aims to summarize the current knowledge on the Δ133p53 isoforms and how they contribute to multiple physiological and pathological mechanisms. Critically, further characterization of p53 isoforms may identify novel regulatory modes of p53 pathway functions that contribute to disease progression and facilitate the development of new therapeutic strategies. Abstract The TP53 gene is a critical tumor suppressor and key determinant of cell fate which regulates numerous cellular functions including DNA repair, cell cycle arrest, cellular senescence, apoptosis, autophagy and metabolism. In the last 15 years, the p53 pathway has grown in complexity through the discovery that TP53 differentially expresses twelve p53 protein isoforms in human cells with both overlapping and unique biologic activities. Here, we summarize the current knowledge on the Δ133p53 isoforms (Δ133p53α, Δ133p53β and Δ133p53γ), which are evolutionary derived and found only in human and higher order primates. All three isoforms lack both of the transactivation domains and the beginning of the DNA-binding domain. Despite the absence of these canonical domains, the Δ133p53 isoforms maintain critical functions in cancer, physiological and premature aging, neurodegenerative diseases, immunity and inflammation, and tissue repair. The ability of the Δ133p53 isoforms to modulate the p53 pathway functions underscores the need to include these p53 isoforms in our understanding of how the p53 pathway contributes to multiple physiological and pathological mechanisms. Critically, further characterization of p53 isoforms may identify novel regulatory modes of p53 pathway functions that contribute to disease progression and facilitate the development of new therapeutic strategies.
Collapse
|
17
|
Silencing of Testin expression is a frequent event in spontaneous lymphomas from Trp53-mutant mice. Sci Rep 2020; 10:16255. [PMID: 33004921 PMCID: PMC7530732 DOI: 10.1038/s41598-020-73229-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 09/09/2020] [Indexed: 02/03/2023] Open
Abstract
The tumour suppressor gene, TES, is frequently methylated in many human tumours. Previously, we demonstrated that TES promoter methylation and transcriptional silencing was the most common molecular abnormality detected in childhood acute lymphoblastic leukaemia (ALL). Trp53-mutant mouse models predominantly develop B- and T-cell lymphomas, which are widely considered equivalent to childhood T and B ALL. In this study, we examined expression of Tes transcript and Testin protein in spontaneous tumours obtained from three Trp53-mutant mouse models. Using immunohistochemistry, we report that 47% of lymphomas lacked Testin protein compared to only 7% of non-lymphoid tumours. Further examination of the lymphomas from Trp53-null and Trp53-mΔpro homozygous mutant mice revealed that 63% and 69% respectively of the isolated lymphomas were Testin negative, which is similar to reported rates in childhood T-ALL. Surprisingly, lymphomas from Trp53-Δ122 mice were frequently Testin positive (> 60%), suggesting that the presence of the Trp53-Δ122 protein appeared to mitigate the requirement for Tes silencing in lymphomagenesis. Quantitative RT-PCR results confirmed that this lack of Testin protein was due to Tes transcriptional silencing, although bisulfite sequencing demonstrated that this was not due to promoter methylation. These results are consistent with the Testin protein having lymphoid tumour suppressor activity in both mice and humans.
Collapse
|
18
|
The Emerging Landscape of p53 Isoforms in Physiology, Cancer and Degenerative Diseases. Int J Mol Sci 2019; 20:ijms20246257. [PMID: 31835844 PMCID: PMC6941119 DOI: 10.3390/ijms20246257] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022] Open
Abstract
p53, first described four decades ago, is now established as a master regulator of cellular stress response, the “guardian of the genome”. p53 contributes to biological robustness by behaving in a cellular-context dependent manner, influenced by several factors (e.g., cell type, active signalling pathways, the type, extent and intensity of cellular damage, cell cycle stage, nutrient availability, immune function). The p53 isoforms regulate gene transcription and protein expression in response to the stimuli so that the cell response is precisely tuned to the cell signals and cell context. Twelve isoforms of p53 have been described in humans. In this review, we explore the interactions between p53 isoforms and other proteins contributing to their established cellular functions, which can be both tumour-suppressive and oncogenic in nature. Evidence of p53 isoform in human cancers is largely based on RT-qPCR expression studies, usually investigating a particular type of isoform. Beyond p53 isoform functions in cancer, it is implicated in neurodegeneration, embryological development, progeroid phenotype, inflammatory pathology, infections and tissue regeneration, which are described in this review.
Collapse
|
19
|
Functional interplay between p53 and Δ133p53 in adaptive stress response. Cell Death Differ 2019; 27:1618-1632. [PMID: 31659281 DOI: 10.1038/s41418-019-0445-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 10/10/2019] [Accepted: 10/15/2019] [Indexed: 01/20/2023] Open
Abstract
Apart from its well-known prodeath activity, p53 is also implicated in promoting cell survival. How p53 can mediate such seemingly opposing effects is largely unclear. We report here a novel mechanism in which p53-mediated proapoptosis is switched to antiapoptosis via its interaction with a p53 isoform, Δ133p53. We show that the expression of Δ133p53 is induced by mild or a moderate level of stress via an HIF1-dependent mechanism. Increased Δ133p53 levels contribute to the adaptive response by shifting the p53 binding at the Bcl2 promoter from suppressive responsive elements (RE) to activating REs, resulting in induction of Bcl2. In accordance with this mode of action, pretreatment of mice with mild stress induces Δ133p53 and Bcl2, which is associated with protection of animals from toxicity caused by high doses of DNA damage agents. Collectively, our work uncovers a novel functional interplay between p53 and Δ133p53 determining cell fate; survival or death in response to stress.
Collapse
|
20
|
The Δ133p53β isoform promotes an immunosuppressive environment leading to aggressive prostate cancer. Cell Death Dis 2019; 10:631. [PMID: 31431617 PMCID: PMC6702175 DOI: 10.1038/s41419-019-1861-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 07/25/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022]
Abstract
Prostate cancer is the second most common cancer in men, for which there are no reliable biomarkers or targeted therapies. Here we demonstrate that elevated levels of Δ133TP53β isoform characterize prostate cancers with immune cell infiltration, particularly T cells and CD163+ macrophages. These cancers are associated with shorter progression-free survival, Gleason scores ≥ 7, and an immunosuppressive environment defined by a higher proportion of PD-1, PD-L1 and colony-stimulating factor 1 receptor (CSF1R) positive cells. Consistent with this, RNA-seq of tumours showed enrichment for pathways associated with immune signalling and cell migration. We further show a role for hypoxia and wild-type p53 in upregulating Δ133TP53 levels. Finally, AUC analysis showed that Δ133TP53β expression level alone predicted aggressive disease with 88% accuracy. Our data identify Δ133TP53β as a highly accurate prognostic factor for aggressive prostate cancer.
Collapse
|
21
|
Loss of p53 Causes Stochastic Aberrant X-Chromosome Inactivation and Female-Specific Neural Tube Defects. Cell Rep 2019; 27:442-454.e5. [DOI: 10.1016/j.celrep.2019.03.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 02/11/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022] Open
|
22
|
Lei J, Qi R, Tang Y, Wang W, Wei G, Nussinov R, Ma B. Conformational stability and dynamics of the cancer-associated isoform Δ133p53β are modulated by p53 peptides and p53-specific DNA. FASEB J 2019; 33:4225-4235. [PMID: 30540922 PMCID: PMC6404584 DOI: 10.1096/fj.201801973r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/12/2018] [Indexed: 01/01/2023]
Abstract
p53 is a tumor suppressor protein that maintains genome stability, but its Δ133p53β and Δ160p53β isoforms promote breast cancer cell invasion. The sequence truncations in the p53 core domain raise key questions related to their physicochemical properties, including structural stabilities, interaction mechanisms, and DNA-binding abilities. Herein, we investigated the conformational dynamics of Δ133p53β and Δ160p53β with and without binding to p53-specific DNA by using molecular dynamics simulations. We observed that the core domains of the 2 truncated isoforms are much less stable than wild-type (wt) p53β, and the increased solvent exposure of their aggregation-triggering segment indicates their higher aggregation propensities than wt p53. We also found that Δ133p53β stability is modulable by peptide or DNA interactions. Adding a p53 peptide (derived from truncated p53 sequence 107-129) may help stabilize Δ133p53. Most importantly, our simulations of p53 isomer-DNA complexes indicate that Δ133p53β dimer, but not Δ160p53β dimer, could form a stable complex with p53-specific DNA, which is consistent with recent experiments. This study provides physicochemical insight into Δ133p53β, Δ133p53β-DNA complexes, Δ133p53β's pathologic mechanism, and peptide-based inhibitor design against p53-related cancers.-Lei, J., Qi, R., Tang, Y., Wang, W., Wei, G., Nussinov, R., Ma, B. Conformational stability and dynamics of the cancer-associated isoform Δ133p53β are modulated by p53 peptides and p53-specific DNA.
Collapse
Affiliation(s)
- Jiangtao Lei
- State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Sciences–Ministry of Education, Department of Physics, Fudan University, Shanghai, China
| | - Ruxi Qi
- State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Sciences–Ministry of Education, Department of Physics, Fudan University, Shanghai, China
| | - Yegen Tang
- Department of Chemistry, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wenning Wang
- Department of Chemistry, Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Guanghong Wei
- State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Sciences–Ministry of Education, Department of Physics, Fudan University, Shanghai, China
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland, USA; and
- Department of Human Genetics and Molecular Medicine, Sackler Institute of Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland, USA; and
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Childhood blastomas, unlike adult cancers, originate from developing organs in which molecular and cellular features exhibit differentiation arrest and embryonic characteristics. Conventional cancer therapies, which rely on the generalized cytotoxic effect on rapidly dividing cells, may damage delicate organs in young children, leading to multiple late effects. Deep understanding of the biology of embryonal cancers is crucial in reshaping the cancer treatment paradigm for children. RECENT FINDINGS p53 plays a major physiological role in embryonic development, by controlling cell proliferation, differentiation and responses to cellular stress. Tumor suppressor function of p53 is commonly lost in adult cancers through genetic alterations. However, both somatic and germline p53 mutations are rare in childhood blastomas, suggesting that in these cancers, p53 may be inactivated through other mechanisms than mutation. In this review, we summarize current knowledge about p53 pathway inactivation in childhood blastomas (specifically neuroblastoma, retinoblastoma and Wilms' tumor) through various upstream mechanisms. Laboratory evidence and clinical trials of targeted therapies specific to exploiting p53 upstream regulators are discussed. SUMMARY Despite the low rate of inherent TP53 mutations, p53 pathway inactivation is a common denominator in childhood blastomas. Exploiting p53 and its regulators is likely to translate into more effective targeted therapies with minimal late effects for children. (see Video Abstract, Supplemental Digital Content 1, http://links.lww.com/COON/A23).
Collapse
Affiliation(s)
- Lixian Oh
- Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia
| | - Hind Hafsi
- Institute of Advanced Biosciences, University of Grenoble-Alpes, La Tronche, France
| | - Pierre Hainaut
- Institute of Advanced Biosciences, University of Grenoble-Alpes, La Tronche, France
| | - Hany Ariffin
- Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
Kazantseva M, Eiholzer RA, Mehta S, Taha A, Bowie S, Roth I, Zhou J, Joruiz SM, Royds JA, Hung NA, Slatter TL, Braithwaite AW. Elevation of the TP53 isoform Δ133p53β in glioblastomas: an alternative to mutant p53 in promoting tumor development. J Pathol 2018; 246:77-88. [PMID: 29888503 PMCID: PMC6120556 DOI: 10.1002/path.5111] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 05/05/2018] [Accepted: 06/05/2018] [Indexed: 01/22/2023]
Abstract
As tumor protein 53 (p53) isoforms have tumor‐promoting, migration, and inflammatory properties, this study investigated whether p53 isoforms contributed to glioblastoma progression. The expression levels of full‐length TP53α (TAp53α) and six TP53 isoforms were quantitated by RT‐qPCR in 89 glioblastomas and correlated with TP53 mutation status, tumor‐associated macrophage content, and various immune cell markers. Elevated levels of Δ133p53β mRNA characterised glioblastomas with increased CD163‐positive macrophages and wild‐type TP53. In situ‐based analyses found Δ133p53β expression localised to malignant cells in areas with increased hypoxia, and in cells with the monocyte chemoattractant protein C‐C motif chemokine ligand 2 (CCL2) expressed. Tumors with increased Δ133p53β had increased numbers of cells positive for macrophage colony‐stimulating factor 1 receptor (CSF1R) and programmed death ligand 1 (PDL1). In addition, cells expressing a murine ‘mimic’ of Δ133p53 (Δ122p53) were resistant to temozolomide treatment and oxidative stress. Our findings suggest that elevated Δ133p53β is an alternative pathway to TP53 mutation in glioblastoma that aids tumor progression by promoting an immunosuppressive and chemoresistant environment. Adding Δ133p53β to a TP53 signature along with TP53 mutation status will better predict treatment resistance in glioblastoma. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Marina Kazantseva
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Ramona A Eiholzer
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sunali Mehta
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Ahmad Taha
- Department of Neurosurgery, Southern District Heath Board, New Zealand
| | - Sara Bowie
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Imogen Roth
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Jean Zhou
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Department of Radiology, Southern District Health Board, New Zealand
| | - Sebastien M Joruiz
- Jacqui Wood Cancer Centre, Division of Cancer Research, University of Dundee, UK
| | - Janice A Royds
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Noelyn A Hung
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Tania L Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Antony W Braithwaite
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| |
Collapse
|
25
|
A mouse model of the Δ133p53 isoform: roles in cancer progression and inflammation. Mamm Genome 2018; 29:831-842. [PMID: 29992419 DOI: 10.1007/s00335-018-9758-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 07/05/2018] [Indexed: 01/19/2023]
Abstract
This review paper outlines studies on the Δ122p53 mouse, a model of the human Δ133p53 isoform, together with studies in other model organisms, cell culture, and where available, clinical investigations. In general, these studies imply that, in contrast to the canonical p53 tumor suppressor, Δ133p53 family members have oncogenic capability. Δ122p53 is multi-functional, conferring survival and proliferative advantages on cells, promoting invasion, metastasis and vascularization, as does Δ133p53. Cancers with high levels of Δ133p53 often have poor prognosis. Δ122p53 mediates its effects through the JAK-STAT and RhoA-ROCK signaling pathways. We propose that Δ133p53 isoforms have evolved as inflammatory signaling molecules to deal with the consequent tissue damage of p53 activation. However, if sustained expression of the isoforms occur, pathologies may result.
Collapse
|
26
|
Mehta SY, Morten BC, Antony J, Henderson L, Lasham A, Campbell H, Cunliffe H, Horsfield JA, Reddel RR, Avery-Kiejda KA, Print CG, Braithwaite AW. Regulation of the interferon-gamma (IFN-γ) pathway by p63 and Δ133p53 isoform in different breast cancer subtypes. Oncotarget 2018; 9:29146-29161. [PMID: 30018742 PMCID: PMC6044385 DOI: 10.18632/oncotarget.25635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
The TP53 family consists of three sets of transcription factor genes, TP53, TP63 and TP73, each of which expresses multiple RNA variants and protein isoforms. Of these, TP53 is mutated in 25-30% of breast cancers. How TP53 mutations affect the interaction of TP53 family members and their isoforms in breast cancer is unknown. To investigate this, 3 independent breast cancer cohorts were stratified into 4 groups based on oestrogen receptor (ER) and TP53 mutation status. Using bioinformatic methodologies, principal signalling pathways associated with the expression of TP53 family members were identified. Results show an enrichment of IFN-γ signalling associated with TP63 RNA in wild type TP53 (wtTP53), ER negative (ER-) tumours and with Δ133TP53 RNA in mutant TP53 (mTP53) ER positive (ER+) tumours. Moreover, tumours with low IFN-γ signalling were associated with significantly poorer patient outcome. The predicted changes in expression of a subset of RNAs involved in IFN-γ signalling were confirmed in vitro. Our data show that different members of the TP53 family can drive transcription of genes involved in IFN-γ signalling in different breast cancer subgroups.
Collapse
Affiliation(s)
- Sunali Y Mehta
- Pathology Department, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Brianna C Morten
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Calvary Mater Hospital, Waratah NSW, Australia
| | - Jisha Antony
- Pathology Department, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Luke Henderson
- Pathology Department, University of Otago, Dunedin, New Zealand
| | - Annette Lasham
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Hamish Campbell
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | | | - Julia A Horsfield
- Pathology Department, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Roger R Reddel
- Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Kelly A Avery-Kiejda
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Calvary Mater Hospital, Waratah NSW, Australia
| | - Cristin G Print
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Antony W Braithwaite
- Pathology Department, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
27
|
∆133p53 isoform promotes tumour invasion and metastasis via interleukin-6 activation of JAK-STAT and RhoA-ROCK signalling. Nat Commun 2018; 9:254. [PMID: 29343721 PMCID: PMC5772473 DOI: 10.1038/s41467-017-02408-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 11/29/2017] [Indexed: 02/06/2023] Open
Abstract
∆122p53 mice (a model of ∆133p53 isoform) are tumour-prone, have extensive inflammation and elevated serum IL-6. To investigate the role of IL-6 we crossed ∆122p53 mice with IL-6 null mice. Here we show that loss of IL-6 reduced JAK-STAT signalling, tumour incidence and metastasis. We also show that ∆122p53 activates RhoA-ROCK signalling leading to tumour cell invasion, which is IL-6-dependent and can be reduced by inhibition of JAK-STAT and RhoA-ROCK pathways. Similarly, we show that Δ133p53 activates these pathways, resulting in invasive and migratory phenotypes in colorectal cancer cells. Gene expression analysis of colorectal tumours showed enrichment of GPCR signalling associated with ∆133TP53 mRNA. Patients with elevated ∆133TP53 mRNA levels had a shorter disease-free survival. Our results suggest that ∆133p53 promotes tumour invasion by activation of the JAK-STAT and RhoA-ROCK pathways, and that patients whose tumours have high ∆133TP53 may benefit from therapies targeting these pathways. Aberrant expression of the Δ133p53 isoform is linked to many cancers. Here, the authors utilise a model of the Δ133p53 isoform that is prone to tumours and inflammation, showing that Δ133p53 promotes tumour cell invasion by activation of the JAK-STAT and RhoA-ROCK pathways in an IL-6 dependent manner.
Collapse
|
28
|
Leroy B, Ballinger ML, Baran-Marszak F, Bond GL, Braithwaite A, Concin N, Donehower LA, El-Deiry WS, Fenaux P, Gaidano G, Langerød A, Hellstrom-Lindberg E, Iggo R, Lehmann-Che J, Mai PL, Malkin D, Moll UM, Myers JN, Nichols KE, Pospisilova S, Ashton-Prolla P, Rossi D, Savage SA, Strong LC, Tonin PN, Zeillinger R, Zenz T, Fraumeni JF, Taschner PEM, Hainaut P, Soussi T. Recommended Guidelines for Validation, Quality Control, and Reporting of TP53 Variants in Clinical Practice. Cancer Res 2017; 77:1250-1260. [PMID: 28254861 DOI: 10.1158/0008-5472.can-16-2179] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/12/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
Accurate assessment of TP53 gene status in sporadic tumors and in the germline of individuals at high risk of cancer due to Li-Fraumeni Syndrome (LFS) has important clinical implications for diagnosis, surveillance, and therapy. Genomic data from more than 20,000 cancer genomes provide a wealth of information on cancer gene alterations and have confirmed TP53 as the most commonly mutated gene in human cancer. Analysis of a database of 70,000 TP53 variants reveals that the two newly discovered exons of the gene, exons 9β and 9γ, generated by alternative splicing, are the targets of inactivating mutation events in breast, liver, and head and neck tumors. Furthermore, germline rearrange-ments in intron 1 of TP53 are associated with LFS and are frequently observed in sporadic osteosarcoma. In this context of constantly growing genomic data, we discuss how screening strategies must be improved when assessing TP53 status in clinical samples. Finally, we discuss how TP53 alterations should be described by using accurate nomenclature to avoid confusion in scientific and clinical reports. Cancer Res; 77(6); 1250-60. ©2017 AACR.
Collapse
Affiliation(s)
- Bernard Leroy
- Sorbonne Université, UPMC Univ Paris 06, Paris, France
| | - Mandy L Ballinger
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Fanny Baran-Marszak
- Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'H ematologie Biologique, France
| | - Gareth L Bond
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford, United Kingdom
| | - Antony Braithwaite
- Dept of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand.,Children's Medical Research Institute, University of Sydney, Westmead NSW, Australia
| | - Nicole Concin
- Department of Gynecology and Obstetrics, Innsbruck Medical University, Innsbruck, Austria
| | | | - Wafik S El-Deiry
- Department of Hematology/Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Pierre Fenaux
- Service d'hématologie séniors, Hôpital St Louis/Université Paris 7, 1 avenue Claude Vellefaux, Paris, France
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Anita Langerød
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Eva Hellstrom-Lindberg
- Karolinska Institute, Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Richard Iggo
- Bergonié Cancer Institute University of Bordeaux 229 cours de l'Argonne 33076 Bordeaux, France
| | | | - Phuong L Mai
- Cancer Genetics Program, Magee Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David Malkin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, New York
| | - Jeffrey N Myers
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kim E Nichols
- Department of Oncology, Division of Cancer Predisposition, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sarka Pospisilova
- Masaryk University, CEITEC - Molecular Medicine and University Hospital Brno, Department of Internal Medicine - Hematology and Oncology, Brno, Czech Republic
| | - Patricia Ashton-Prolla
- Universidade Federal do Rio Grande do Sul (UFRGS) e Serviço deGenética Médica-HCPA, Rua Ramiro Barcelos, Porto Alegre, Brasil
| | - Davide Rossi
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Louise C Strong
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia N Tonin
- Departments of Medicine and Human Genetics, McGill University and Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Robert Zeillinger
- Molecular Oncology Group, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Thorsten Zenz
- University of Heidelberg, Department of Medicine V, Heidelberg, Germany; Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Joseph F Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Peter E M Taschner
- Generade Centre of Expertise Genomics and University of Applied Sciences Leiden, Leiden, the Netherlands
| | - Pierre Hainaut
- Institut Albert Bonniot, Inserm 823, Université Grenoble Alpes, Rond Point de la Chantourne, La Tronche, France
| | - Thierry Soussi
- Sorbonne Université, UPMC Univ Paris 06, Paris, France. .,Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden.,INSERM, U1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
29
|
Nasef NA, Mehta S, Ferguson LR. Susceptibility to chronic inflammation: an update. Arch Toxicol 2017; 91:1131-1141. [PMID: 28130581 DOI: 10.1007/s00204-016-1914-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 12/13/2016] [Indexed: 12/25/2022]
Abstract
Chronic inflammation is defined by the persistence of inflammatory processes beyond their physiological function, resulting in tissue destruction. Chronic inflammation is implicated in the progression of many chronic diseases and plays a central role in chronic inflammatory and autoimmune disease. As such, this review aims to collate some of the latest research in relation to genetic and environmental susceptibilities to chronic inflammation. In the genetic section, we discuss some of the updates in cytokine research and current treatments that are being developed. We also discuss newly identified canonical and non-canonical genes associated with chronic inflammation. In the environmental section, we highlight some of the latest updates and evidence in relation to the role that infection, diet and stress play in promoting inflammation. The aim of this review is to provide an overview of the latest research to build on our current understanding of chronic inflammation. It highlights the complexity associated with chronic inflammation, as well as provides insights into potential new targets for therapies that could be used to treat chronic inflammation and consequently prevent disease progression.
Collapse
Affiliation(s)
- Noha Ahmed Nasef
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sunali Mehta
- Department of Pathology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Lynnette R Ferguson
- Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
30
|
Mehta S, Tsai P, Lasham A, Campbell H, Reddel R, Braithwaite A, Print C. A Study of TP53 RNA Splicing Illustrates Pitfalls of RNA-seq Methodology. Cancer Res 2016; 76:7151-7159. [PMID: 27913434 DOI: 10.1158/0008-5472.can-16-1624] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 09/02/2016] [Accepted: 09/27/2016] [Indexed: 11/16/2022]
Abstract
TP53 undergoes multiple RNA-splicing events, resulting in at least nine mRNA transcripts encoding at least 12 functionally different protein isoforms. Antibodies specific to p53 protein isoforms have proven difficult to develop, thus researchers must rely on the transcript information to infer isoform abundance. In this study, we used deep RNA-seq, droplet digital PCR (ddPCR), and real-time quantitative reverse transcriptase PCR (RT-qPCR) from nine human cell lines and RNA-seq data available for tumors in The Cancer Genome Atlas to analyze TP53 splice variant expression. All three methods detected expression of the FL/40TP53α_T1 variant in most human tumors and cell lines. However, other less abundant variants were only detected with PCR-based methods. Using RNA-seq simulation analysis, we determined why RNA-seq is unable to detect less abundant TP53 transcripts and discuss the implications of these findings for the general interpretation of RNA-seq data. Cancer Res; 76(24); 7151-9. ©2016 AACR.
Collapse
Affiliation(s)
- Sunali Mehta
- Department of Pathology, University of Otago, Dunedin, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Otago, Dunedin, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, Faculty of Medicine, University of Auckland, Auckland, New Zealand.,Bioinformatics Institute, University of Auckland, Auckland, New Zealand
| | - Annette Lasham
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Otago, Dunedin, New Zealand.,Department of Molecular Medicine and Pathology, Faculty of Medicine, University of Auckland, Auckland, New Zealand
| | - Hamish Campbell
- Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Roger Reddel
- Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Antony Braithwaite
- Department of Pathology, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Otago, Dunedin, New Zealand.,Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Cristin Print
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Otago, Dunedin, New Zealand.,Department of Molecular Medicine and Pathology, Faculty of Medicine, University of Auckland, Auckland, New Zealand.,Bioinformatics Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Abstract
It is poorly understood how a single protein, p53, can be responsive to so many stress signals and orchestrates very diverse cell responses to maintain/restore cell/tissue functions. The uncovering that TP53 gene physiologically expresses, in a tissue-dependent manner, several p53 splice variants (isoforms) provides an explanation to its pleiotropic biological activities. Here, we summarize a decade of research on p53 isoforms. The clinical studies and the diverse cellular and animal models of p53 isoforms (zebrafish, Drosophila, and mouse) lead us to realize that a p53-mediated cell response is, in fact, the sum of the intrinsic activities of the coexpressed p53 isoforms and that unbalancing expression of different p53 isoforms leads to cancer, premature aging, (neuro)degenerative diseases, inflammation, embryo malformations, or defects in tissue regeneration. Cracking the p53 isoforms' code is, thus, a necessary step to improve cancer treatment. It also opens new exciting perspectives in tissue regeneration.
Collapse
Affiliation(s)
- Sebastien M Joruiz
- Dundee Cancer Centre, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - Jean-Christophe Bourdon
- Dundee Cancer Centre, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| |
Collapse
|
32
|
Banerjee S, Uppal T, Strahan R, Dabral P, Verma SC. The Modulation of Apoptotic Pathways by Gammaherpesviruses. Front Microbiol 2016; 7:585. [PMID: 27199919 PMCID: PMC4847483 DOI: 10.3389/fmicb.2016.00585] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/11/2016] [Indexed: 12/11/2022] Open
Abstract
Apoptosis or programmed cell death is a tightly regulated process fundamental for cellular development and elimination of damaged or infected cells during the maintenance of cellular homeostasis. It is also an important cellular defense mechanism against viral invasion. In many instances, abnormal regulation of apoptosis has been associated with a number of diseases, including cancer development. Following infection of host cells, persistent and oncogenic viruses such as the members of the Gammaherpesvirus family employ a number of different mechanisms to avoid the host cell’s “burglar” alarm and to alter the extrinsic and intrinsic apoptotic pathways by either deregulating the expressions of cellular signaling genes or by encoding the viral homologs of cellular genes. In this review, we summarize the recent findings on how gammaherpesviruses inhibit cellular apoptosis via virus-encoded proteins by mediating modification of numerous signal transduction pathways. We also list the key viral anti-apoptotic proteins that could be exploited as effective targets for novel antiviral therapies in order to stimulate apoptosis in different types of cancer cells.
Collapse
Affiliation(s)
- Shuvomoy Banerjee
- Amity Institute of Virology and Immunology, Amity University Noida, India
| | - Timsy Uppal
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Roxanne Strahan
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Prerna Dabral
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Subhash C Verma
- Department of Microbiology and Immunology, Center for Molecular Medicine, School of Medicine, University of Nevada, Reno Reno, NV, USA
| |
Collapse
|
33
|
The Δ133p53 isoform and its mouse analogue Δ122p53 promote invasion and metastasis involving pro-inflammatory molecules interleukin-6 and CCL2. Oncogene 2016; 35:4981-9. [PMID: 26996665 DOI: 10.1038/onc.2016.45] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/17/2016] [Accepted: 02/08/2016] [Indexed: 12/12/2022]
Abstract
A number of naturally occurring isoforms of the tumour suppressor protein p53 have been discovered, which appear to have differing roles in tumour prevention or promotion. We are investigating the tumour-promoting activities of the Δ133p53 isoform using our mouse model of Δ133p53 (Δ122p53). Here, we report that tumours from Δ122p53 homozygous mice show evidence of invasion and metastasis and that Δ122p53 promotes migration though a 3-dimensional collagen matrix. We also show that Δ122p53 and Δ133p53 promote cell migration in scratch wound and Transwell assays, similar to the 'gain-of-function' phenotypes seen with mutant p53. Using the well-defined B16 mouse melanoma metastatic model, we show that Δ122p53 leads to faster generation of lung metastases. The increased migratory phenotypes are dependent on secreted factors, including the cytokine interleukin-6 and the chemokine CCL2. We propose that Δ122p53 (and Δ133p53) acts in a similar manner to 'gain-of-function' mutant p53 proteins to promote migration, invasion and metastasis, which may contribute to poor survival in patients with Δ133p53-expressing tumours.
Collapse
|
34
|
Blackburn J, Roden DL, Ng R, Wu J, Bosman A, Epstein RJ. Damage-inducible intragenic demethylation of the human TP53 tumor suppressor gene is associated with transcription from an alternative intronic promoter. Mol Carcinog 2015; 55:1940-1951. [PMID: 26676339 PMCID: PMC5111752 DOI: 10.1002/mc.22441] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 10/29/2015] [Accepted: 11/17/2015] [Indexed: 12/23/2022]
Abstract
Wild-type TP53 exons 5-8 contain CpG dinucleotides that are prone to methylation-dependent mutation during carcinogenesis, but the regulatory effects of methylation affecting these CpG sites are unclear. To clarify this, we first assessed site-specific TP53 CpG methylation in normal and transformed cells. Both DNA damage and cell ageing were associated with site-specific CpG demethylation in exon 5 accompanied by induction of a truncated TP53 isoform regulated by an adjacent intronic promoter (P2). We then synthesized novel synonymous TP53 alleles with divergent CpG content but stable encodement of the wild-type polypeptide. Expression of CpG-enriched TP53 constructs selectively reduced production of the full-length transcript (P1), consistent with a causal relationship between intragenic demethylation and transcription. 450K methylation comparison of normal (TP53-wildtype) and cancerous (TP53-mutant) human cells and tissues revealed focal cancer-associated declines in CpG methylation near the P1 transcription start site, accompanied by rises near the alternate exon 5 start site. These data confirm that site-specific changes of intragenic TP53 CpG methylation are extrinsically inducible, and suggest that human cancer progression is mediated in part by dysregulation of damage-inducible intragenic CpG demethylation that alters TP53 P1/P2 isoform expression. © 2015 The Authors. Molecular Carcinogenesis Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- James Blackburn
- Laboratory of Genome Evolution, The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney, Australia.,UNSW Medicine, St. Vincent's Clinical School, Darlinghurst, Sydney, Australia
| | - Daniel L Roden
- Laboratory of Cancer Biology, The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney, Australia
| | - Robert Ng
- Laboratory of Genome Evolution, The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney, Australia.,UNSW Medicine, St. Vincent's Clinical School, Darlinghurst, Sydney, Australia
| | - Jianmin Wu
- Laboratory of Cancer Bioinformatics, The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney, Australia
| | - Alexis Bosman
- Laboratory of Developmental and Stem Cell Biology, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Richard J Epstein
- Laboratory of Genome Evolution, The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney, Australia.,UNSW Medicine, St. Vincent's Clinical School, Darlinghurst, Sydney, Australia.,Clinical Informatics & Research Centre, Department of Oncology, St. Vincent's Hospital, Sydney, Australia
| |
Collapse
|
35
|
Dong Y, Wu G. Azadircta indica as a modulator of membrane stability parameters and surface changes during 1,2 dimethylhydrazine-induced colorectal carcinogenesis. ACTA ACUST UNITED AC 2015; 63:204-9. [PMID: 26271242 DOI: 10.1016/j.patbio.2015.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 06/25/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The aim of the present study was to study the modulatory potential of Azadirta indica on colonic surface abnormalities and membrane fluidity changes following 1,2 dimethylhydrazine-induced [DMH] colon carcinogenesis. MATERIALS AND METHODS Brush border membranes [BBM] were isolated from the colon of rats and the viscosity as well as fluidity parameters were assessed by using the membrane extrinsic fluorophore pyrene. RESULTS DMH treatment resulted in a significant increase in lipid peroxidation [LPO]. Reduced glutathione levels [GSH] and the activities of glutathione reductase [GR], glutathione transferase [GST], superoxide dismutase [SOD], catalase [CAT] and glutathione peroxidase [GPx] were found to be significantly decreased following DMH treatment. On the other hand, supplementation with AI, DMH-treated rats resulted in a significant decrease in the levels of lipid peroxidation but caused a significant increase in the levels of GSH as well in the activities of GR, GST, SOD, CAT and GPx. The results further demonstrated a marked decrease in membrane microviscosity following DMH treatment. On the other hand, a significant increase was observed in the excimer/monomer ratio and fluidity parameter of DMH-treated rats when compared to normal control rats. However, the alterations in membrane microviscosity and the fluidity parameters were significantly restored following A. indica treatment. Further, histological as well as colon surface alterations were also observed following DMH treatment, which however were greatly prevented upon AI co-administration. CONCLUSIONS The study, therefore, concludes that A. indica proves to be useful in modulating the colonic surface abnormalities and membrane stability following DMH-induced colon carcinogenesis.
Collapse
Affiliation(s)
- Y Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China; Department of Oncology, Xiangyang Hospital Affiliated to Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - G Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China.
| |
Collapse
|
36
|
Δ122p53, a mouse model of Δ133p53α, enhances the tumor-suppressor activities of an attenuated p53 mutant. Cell Death Dis 2015; 6:e1783. [PMID: 26068791 PMCID: PMC4669831 DOI: 10.1038/cddis.2015.149] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 04/09/2015] [Accepted: 05/06/2015] [Indexed: 01/29/2023]
Abstract
Growing evidence suggests the Δ133p53α isoform may function as an oncogene. It is overexpressed in many tumors, stimulates pathways involved in tumor progression, and inhibits some activities of wild-type p53, including transactivation and apoptosis. We hypothesized that Δ133p53α would have an even more profound effect on p53 variants with weaker tumor-suppressor capability. We tested this using a mouse model heterozygous for a Δ133p53α-like isoform (Δ122p53) and a p53 mutant with weak tumor-suppressor function (mΔpro). The Δ122p53/mΔpro mice showed a unique survival curve with a wide range of survival times (92-495 days) which was much greater than mΔpro/- mice (range 120-250 days) and mice heterozygous for the Δ122p53 and p53 null alleles (Δ122p53/-, range 78-150 days), suggesting Δ122p53 increased the tumor-suppressor activity of mΔpro. Moreover, some of the mice that survived longest only developed benign tumors. In vitro analyses to investigate why some Δ122p53/mΔpro mice were protected from aggressive tumors revealed that Δ122p53 stabilized mΔpro and prolonged the response to DNA damage. Similar effects of Δ122p53 and Δ133p53α were observed on wild-type of full-length p53, but these did not result in improved biological responses. The data suggest that Δ122p53 (and Δ133p53α) could offer some protection against tumors by enhancing the p53 response to stress.
Collapse
|
37
|
Sawhney S, Hood K, Shaw A, Braithwaite AW, Stubbs R, Hung NA, Royds JA, Slatter TL. Alpha-enolase is upregulated on the cell surface and responds to plasminogen activation in mice expressing a ∆133p53α mimic. PLoS One 2015; 10:e0116270. [PMID: 25643152 PMCID: PMC4313950 DOI: 10.1371/journal.pone.0116270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 12/05/2014] [Indexed: 01/11/2023] Open
Abstract
The p53 protein is a master regulator of the stress response. It acts as a tumor suppressor by inducing transcriptional activation of p53 target genes, with roles in apoptosis, cell cycle arrest and metabolism. The discovery of at least 12 isoforms of p53, some of which have tumor-promoting properties, has opened new avenues of research. Our previous work studied tumor phenotypes in four mouse models with different p53 backgrounds: wild-type p53, p53 null, mutant p53 lacking the proline domain (mΔpro), and a mimic for the human Δ133p53α p53 isoform (Δ122p53). To identify the major proteins affected by p53 function early in the response to DNA damage, the current study investigated the entire proteome of bone marrow, thymus, and lung in the four p53 models. Protein extracts from untreated controls and those treated with amsacrine were analyzed using two-dimensional fluorescence difference gel electrophoresis. In the bone marrow, reactive proteins were universally decreased by wild-type p53, including α-enolase. Further analysis of α-enolase in the p53 models revealed that it was instead increased in Δ122p53 hematopoietic and tumor cell cytosol and on the cell surface. Alpha-enolase on the surface of Δ122p53 cells acted as a plasminogen receptor, with tumor necrosis factor alpha induced upon plasminogen stimulation. Taken together, these data identified new proteins associated with p53 function. One of these proteins, α-enolase, is regulated differently by wild-type p53 and Δ122p53 cells, with reduced abundance as part of a wild-type p53 response and increased abundance with Δ122p53 function. Increased cell surface α-enolase on Δ122p53 cells provides a possible explanation for the model’s pro-inflammatory features and suggests that p53 isoforms may direct an inflammatory response by increasing the amount of α-enolase on the cell surface.
Collapse
Affiliation(s)
- Sonal Sawhney
- Wakefield Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Kylie Hood
- Wakefield Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Alisha Shaw
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Antony W. Braithwaite
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Children’s Medical Research Institute, University of Sydney, Westmead, Australia
| | - Richard Stubbs
- Wakefield Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Noelyn A. Hung
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Janice A. Royds
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Tania L. Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- * E-mail:
| |
Collapse
|
38
|
Engelmann D, Pützer BM. Emerging from the shade of p53 mutants: N-terminally truncated variants of the p53 family in EMT signaling and cancer progression. Sci Signal 2014; 7:re9. [PMID: 25270260 DOI: 10.1126/scisignal.2005699] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prevailing view has been that N-terminally truncated p53 family isoforms (ΔNp53, ΔNp63, and DNp73) predominantly counteract cell cycle arrest and apoptosis. Recent progress in the field extend these well-known functions and place these isoforms in the center of a comprehensive regulatory network controlling major epithelial-to-mesenchymal transition (EMT)-relevant signaling pathways [such as transforming growth factor-β (TGF-β), wingless-int (WNT), insulin-like growth factor (IGF), and signal transducer and activator of transcription (STAT)], microRNAs, and EMT-associated transcription factors that promote invasion, loss of tumor cell polarity, and metastatic behavior in conjunction with a chemoresistant phenotype. These observations add new weight to the concept that currently underappreciated truncated forms of this tumor suppressor family play an equally important role in promoting cancer aggressiveness as do mutant p53 proteins, and illustrate how the consequences of ΔN/DN expression depend on cellular contexts. The tumor microenvironment contributes to the emergence of these variants, thereby linking inflammation to the activation of the mesenchymal program. In addition, molecular connections between ΔN/DN forms and self-renewal have arisen, suggesting their potential function in the generation of cancer stem cells (CSCs) from bulk tumor cells. These intriguing insights provoke a new understanding of the acquisition of aggressive traits by carcinoma cells in the absence of p53 mutations, and may help direct the development of new therapies for a broad range of cancers.
Collapse
Affiliation(s)
- David Engelmann
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany
| | - Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany.
| |
Collapse
|
39
|
Cooks T, Harris CC, Oren M. Caught in the cross fire: p53 in inflammation. Carcinogenesis 2014; 35:1680-90. [PMID: 24942866 PMCID: PMC4123652 DOI: 10.1093/carcin/bgu134] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 06/06/2014] [Accepted: 06/10/2014] [Indexed: 12/17/2022] Open
Abstract
The p53 transcription factor is a major tumor suppressor, whose diverse activities serve to ensure genome stability and inhibit neoplastic processes. In recent years, it is becoming increasingly clear that p53 also plays a broader role in maintaining cellular homeostasis, as well as contributing to tissue homeostasis in a non-cell-autonomous fashion. Chronic inflammation is a potential cancer-promoting condition, and as such is also within the radar of p53, which mounts a multifaceted attempt to prevent the escalation of chronic tissue imbalance into neoplasia. Recent understanding of the p53 pathway and other family members reveals a broad interaction with inflammatory elements such as reactive oxygen and nitrogen species, cytokines, infectious agents and major immune-regulatory pathways like nuclear factor-kappaB. This complex cross talk is highly dependent on p53 status, as different p53 isoforms and p53 mutants can mediate different responses and even promote chronic inflammation and associated cancer, acting in the tumor cells as well as in the stromal and immune compartments.
Collapse
Affiliation(s)
- Tomer Cooks
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD 20892-4258, USA and Molecular Cell Biology, Weizmann Institute for Science, Rehovot 76100, Israel
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD 20892-4258, USA and Molecular Cell Biology, Weizmann Institute for Science, Rehovot 76100, Israel
| | - Moshe Oren
- Molecular Cell Biology, Weizmann Institute for Science, Rehovot 76100, Israel
| |
Collapse
|
40
|
|
41
|
Surget S, Khoury MP, Bourdon JC. Uncovering the role of p53 splice variants in human malignancy: a clinical perspective. Onco Targets Ther 2013; 7:57-68. [PMID: 24379683 PMCID: PMC3872270 DOI: 10.2147/ott.s53876] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Thirty-five years of research on p53 gave rise to more than 68,000 articles and reviews, but did not allow the uncovering of all the mysteries that this major tumor suppressor holds. How p53 handles the different signals to decide the appropriate cell fate in response to a stress and its implication in tumorigenesis and cancer progression remains unclear. Nevertheless, the uncovering of p53 isoforms has opened new perspectives in the cancer research field. Indeed, the human TP53 gene encodes not only one but at least twelve p53 protein isoforms, which are produced in normal tissues through alternative initiation of translation, usage of alternative promoters, and alternative splicing. In recent years, it became obvious that the different p53 isoforms play an important role in regulating cell fate in response to different stresses in normal cells by differentially regulating gene expression. In cancer cells, abnormal expression of p53 isoforms contributes actively to cancer formation and progression, regardless of TP53 mutation status. They can also be associated with response to treatment, depending on the cell context. The determination of p53 isoform expression and p53 mutation status helps to define different subtypes within a particular cancer type, which would have different responses to treatment. Thus, the understanding of the regulation of p53 isoform expression and their biological activities in relation to the cellular context would constitute an important step toward the improvement of the diagnostic, prognostic, and predictive values of p53 in cancer treatment. This review aims to summarize the involvement of p53 isoforms in cancer and to highlight novel potential therapeutic targets.
Collapse
Affiliation(s)
- Sylvanie Surget
- Dundee Cancer Centre, University of Dundee, Dundee, UK ; Jacqui Wood Cancer Centre, Ninewells Hospital, University of Dundee, Dundee, UK
| | - Marie P Khoury
- Dundee Cancer Centre, University of Dundee, Dundee, UK ; Jacqui Wood Cancer Centre, Ninewells Hospital, University of Dundee, Dundee, UK
| | - Jean-Christophe Bourdon
- Dundee Cancer Centre, University of Dundee, Dundee, UK ; Jacqui Wood Cancer Centre, Ninewells Hospital, University of Dundee, Dundee, UK
| |
Collapse
|
42
|
Avery-Kiejda KA, Morten B, Wong-Brown MW, Mathe A, Scott RJ. The relative mRNA expression of p53 isoforms in breast cancer is associated with clinical features and outcome. Carcinogenesis 2013; 35:586-96. [PMID: 24336193 DOI: 10.1093/carcin/bgt411] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mutation of p53 is a common feature of cancer. Breast cancer is the most common malignancy that develops in women; however, somatic mutation of p53 is rare, suggesting that p53 becomes inactivated by other mechanisms. p53 is expressed as smaller isoforms, some of which inhibit wild-type p53. There are no studies that have examined the relative expression of all isoforms in this disease. We have analysed the relative messenger RNA expression of the p53 isoforms, Δ40, Δ133, β and γ in a panel of 6 breast cancer cell lines, 148 breast cancers specimens and 31 matched normal adjacent tissues by semi-quantitative real-time reverse transcription-PCR and analysed their relationship to clinical features and outcome. We have identified several important clinical associations, particularly with Δ40p53, which was expressed at levels that were ~50-fold higher than the least expressed isoform p53γ. Δ40p53 was significantly upregulated in tumour tissue when compared with the normal breast and was significantly associated with an aggressive breast cancer subtype-triple negative. Additionally, p53β expression was significantly negatively associated with tumour size and positively associated with disease-free survival, where high levels of p53β were protective, particularly in patients with a mutation in p53, suggesting p53β may counteract the damage inflicted by mutant p53. In conclusion, the relative expression of p53 isoforms is related to clinical features of breast cancer and outcome. These results have implications for the stratification of breast cancer based on p53 function and may provide an alternate explanation for deregulated p53 signalling in breast cancer.
Collapse
Affiliation(s)
- Kelly A Avery-Kiejda
- Centre for Information Based Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | | | | | | | | |
Collapse
|
43
|
Lee YJ, Park IS, Lee YJ, Shim JH, Cho MK, Nam HS, Park JW, Oh MH, Lee SH. Resveratrol contributes to chemosensitivity of malignant mesothelioma cells with activation of p53. Food Chem Toxicol 2013; 63:153-60. [PMID: 24239893 DOI: 10.1016/j.fct.2013.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/22/2013] [Accepted: 11/05/2013] [Indexed: 12/18/2022]
Abstract
Resveratrol is a naturally occurring polyphenolic phytoalexin with chemopreventive properties. We previously reported a synergistic anti-proliferative effect of resveratrol and clofarabine against malignant mesothelioma (MM) cells. Here, we further investigated molecular mechanisms involved in the synergistic interaction of these compounds in MM MSTO-211H cells. Resveratrol, in combination with clofarabine, time-dependently induced a strong cytotoxic effect with the nuclear accumulation of phospho-p53 (p-p53) in MSTO-211H cells, but not in normal mesothelial MeT-5A cells. Combination treatment up-regulated the levels of p-p53, cleaved caspase-3, and cleaved PARP proteins. Gene silencing with p53-targeting siRNA attenuated the sensitivity of cells to the combined treatment of two compounds. Analyses of p53 DNA binding assay, p53 reporter gene assay, and RTP-CR toward p53-regulated genes, including Bax, PUMA, Noxa and p21, demonstrated that induced p-p53 is transcriptionally active. These results were further confirmed by the siRNA-mediated knockdown of p53 gene. Combination treatment significantly caused the accumulation of cells at G1 phase with the increases in the sub-G0/G1 peak, DNA ladder, nuclear fragmentation, and caspase-3/7 activity. Taken together, these results demonstrate that resveratrol and clofarabine synergistically elicit apoptotic signal via a p53-dependent pathway, and provide a scientific rationale for clinical evaluation of resveratrol as a promising chemopotentiator in MM.
Collapse
Affiliation(s)
- Yoon-Jin Lee
- Soonchunhyung Environmental Health Center for Asbestos, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 330-930, Republic of Korea; Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Ihl-Sung Park
- Department of Pediatrics, Soonchunhyang University Cheonan Hospital, Cheonan 330-721, Republic of Korea
| | - Yong-Jin Lee
- Soonchunhyung Environmental Health Center for Asbestos, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 330-930, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Moon-Kyun Cho
- Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Hae-Seon Nam
- Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Ji Woong Park
- Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Myung-Ho Oh
- Department of Pediatrics, Soonchunhyang University Cheonan Hospital, Cheonan 330-721, Republic of Korea
| | - Sang-Han Lee
- Soonchunhyung Environmental Health Center for Asbestos, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 330-930, Republic of Korea; Department of Pediatrics, Soonchunhyang University Cheonan Hospital, Cheonan 330-721, Republic of Korea.
| |
Collapse
|
44
|
|
45
|
Tang X, Asano M, O'Reilly A, Farquhar A, Yang Y, Amar S. p53 is an important regulator of CCL2 gene expression. Curr Mol Med 2013; 12:929-43. [PMID: 22804246 DOI: 10.2174/156652412802480844] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 12/11/2022]
Abstract
The p53 protein is a sequence-specific DNA-binding factor that regulates inflammatory genes such as CCL2/MCP-1 that may play a role in various diseases. A recent study has indicated that the knockdown of human p53 leads to a strong negative regulation of CCL2 induction. We are therefore interested in how p53 regulates CCL2 gene expression. In the following study, our findings indicate that UV-induced p53 accumulation in mouse macrophages significantly decreases LPS-induced CCL2 production, and that p53 binds to CCL2 5'UTR in the region (16-35). We also found that a p53 domain (p53pep170) mimics full length p53 to down-regulate CCL2 promoter activity. Treatment of p53-deficient mouse primary macrophages with synthetic p53pep170 was found to decrease LPS-induced production of CCL2 without association with cellular endogenous p53. CCL2 production induced by lentiCLG in human monocytes or mouse primary macrophages was blocked in the presence of p53pep170. Overall, these results demonstrate that p53 or its derived peptide (p53pep170) is an important regulator of CCL2 gene expression via its binding activity, and acts as a novel model for future studies linking p53 and its short peptide to pave the way to possible pharmaceutical intervention of CCL2-mediated inflammatory and cancer diseases.
Collapse
Affiliation(s)
- X Tang
- Center for Anti- Inflammatory Therapeutics, Boston University, 650 Albany Street, X- 343, Boston, MA 02118, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Pathogenic bacterium Helicobacter pylori alters the expression profile of p53 protein isoforms and p53 response to cellular stresses. Proc Natl Acad Sci U S A 2012; 109:E2543-50. [PMID: 22927405 DOI: 10.1073/pnas.1205664109] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The p53 protein plays a central role in the prevention of tumorigenesis. Cellular stresses, such as DNA damage and aberrant oncogene activation, trigger induction of p53 that halts cellular proliferation and allows cells to be repaired. If cellular damage is beyond the capability of the repair mechanisms, p53 induces apoptosis or cell cycle arrest, preventing damaged cells from becoming cancerous. However, emerging evidence suggests that the function of p53 needs to be considered as isoform-specific. Here, we report that the expression profile of p53 can be shifted toward inhibitory p53 isoforms by the pathogenic bacterium Helicobacter pylori, which is known for its strong association with gastric cancer and gastric mucosa-associated lymphoid tissue lymphoma. We found that interaction of H. pylori with gastric epithelial cells, mediated via the cag pathogenicity island, induces N-terminally truncated Δ133p53 and Δ160p53 isoforms in human cells. Induction of an orthologous p53 isoform, Δ153p53, was also found in H. pylori-infected Mongolian gerbils. The p53 isoforms inhibit p53 and p73 activities, induce NF-κB, and increase survival of infected cells. Expression of Δ133p53, in response to H. pylori infection, is regulated by phosphorylation of c-Jun and activation of activator protein-1-dependent transcription. Together, these results provide unique insights into the regulation of p53 protein and may contribute to the understanding of tumorigenesis associated with H. pylori.
Collapse
|
47
|
Selenium as a modulator of membrane stability parameters and surface changes during the initiation phase of 1,2-dimethylhydrazine induced colorectal carcinogenesis. Mol Cell Biochem 2012; 369:119-26. [PMID: 22752389 DOI: 10.1007/s11010-012-1374-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/20/2012] [Indexed: 01/02/2023]
|
48
|
p53 Family: Role of Protein Isoforms in Human Cancer. J Nucleic Acids 2011; 2012:687359. [PMID: 22007292 PMCID: PMC3191818 DOI: 10.1155/2012/687359] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 07/04/2011] [Indexed: 01/07/2023] Open
Abstract
TP53, TP63, and TP73 genes comprise the p53 family. Each gene produces protein isoforms through multiple mechanisms including extensive alternative mRNA splicing. Accumulating evidence shows that these isoforms play a critical role in the regulation of many biological processes in normal cells. Their abnormal expression contributes to tumorigenesis and has a profound effect on tumor response to curative therapy. This paper is an overview of isoform diversity in the p53 family and its role in cancer.
Collapse
|
49
|
Wong RSY. Apoptosis in cancer: from pathogenesis to treatment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2011; 30:87. [PMID: 21943236 PMCID: PMC3197541 DOI: 10.1186/1756-9966-30-87] [Citation(s) in RCA: 1785] [Impact Index Per Article: 137.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 09/26/2011] [Indexed: 01/10/2023]
Abstract
Apoptosis is an ordered and orchestrated cellular process that occurs in physiological and pathological conditions. It is also one of the most studied topics among cell biologists. An understanding of the underlying mechanism of apoptosis is important as it plays a pivotal role in the pathogenesis of many diseases. In some, the problem is due to too much apoptosis, such as in the case of degenerative diseases while in others, too little apoptosis is the culprit. Cancer is one of the scenarios where too little apoptosis occurs, resulting in malignant cells that will not die. The mechanism of apoptosis is complex and involves many pathways. Defects can occur at any point along these pathways, leading to malignant transformation of the affected cells, tumour metastasis and resistance to anticancer drugs. Despite being the cause of problem, apoptosis plays an important role in the treatment of cancer as it is a popular target of many treatment strategies. The abundance of literature suggests that targeting apoptosis in cancer is feasible. However, many troubling questions arise with the use of new drugs or treatment strategies that are designed to enhance apoptosis and critical tests must be passed before they can be used safely in human subjects.
Collapse
Affiliation(s)
- Rebecca S Y Wong
- Division of Human Biology, School of Medical and Health Sciences, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000 Kuala Lumpur, Malaysia.
| |
Collapse
|
50
|
Marcel V, Dichtel-Danjoy ML, Sagne C, Hafsi H, Ma D, Ortiz-Cuaran S, Olivier M, Hall J, Mollereau B, Hainaut P, Bourdon JC. Biological functions of p53 isoforms through evolution: lessons from animal and cellular models. Cell Death Differ 2011; 18:1815-24. [PMID: 21941372 DOI: 10.1038/cdd.2011.120] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The TP53 tumour-suppressor gene is expressed as several protein isoforms generated by different mechanisms, including use of alternative promoters, splicing sites and translational initiation sites, that are conserved through evolution and within the TP53 homologues, TP63 and TP73. Although first described in the eighties, the importance of p53 isoforms in regulating the suppressive functions of p53 has only become evident in the last 10 years, by analogy with observations that p63 and p73 isoforms appeared indispensable to fully understand the biological functions of TP63 and TP73. This review summarizes recent advances in the field of 'p53 isoforms', including new data on p63 and p73 isoforms. Details of the alternative mechanisms that produce p53 isoforms and cis- and trans-regulators identified are provided. The main focus is on their biological functions (apoptosis, cell cycle, aging and so on) in cellular and animal models, including mouse, zebrafish and Drosophila. Finally, the deregulation of p53 isoform expression in human cancers is reviewed. Based on these latest results, several developments are expected in the future: the identification of drugs modulating p53 isoform expression; the generation of animal models and the evaluation of the use of p53 isoform as biomarkers in human cancers.
Collapse
Affiliation(s)
- V Marcel
- Centre for Oncology and Molecular Medicine, INSERM-European Associated Laboratory, University of Dundee, Ninewells Hospital, Dundee, Scotland, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|