1
|
The balance between p53 isoforms modulates the efficiency of HIV-1 infection in macrophages. J Virol 2021; 95:e0118821. [PMID: 34379507 DOI: 10.1128/jvi.01188-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several host factors influence HIV-1 infection and replication. The p53-mediated antiviral role in monocytes-derived macrophages (MDMs) was previously highlighted. Indeed, an increase in p53 level results in a stronger restriction against HIV-1 early replication steps through SAMHD1 activity. In this study, we investigated the potential role of some p53 isoforms in HIV-1 infection. Transfection of isoform-specific siRNA induces distinctive effects on the virus life cycle. For example, in contrast to a siRNA targeting all isoforms, a knockdown of Δ133p53 transcripts reduces virus replication in MDMs that is correlated with a decrease in phosphorylated inactive SAMHD1. Combination of Δ133p53 knockdown and Nutlin-3, a pharmacological inhibitor of MDM2 that stabilizes p53, further reduces susceptibility of MDMs to HIV-1 infection, thus suggesting an inhibitory role of Δ133p53 towards p53 antiviral activity. In contrast, p53β knockdown in MDMs increases the viral production independently of SAMHD1. Moreover, experiments with a Nef-deficient virus show that this viral protein plays a protective role against the antiviral environment mediated by p53. Finally, HIV-1 infection affects the expression pattern of p53 isoforms by increasing p53β and p53γ mRNA levels while stabilizing the protein level of p53α and some isoforms from the p53β subclass. The balance between the various p53 isoforms is therefore an important factor in the overall susceptibility of macrophages to HIV-1 infection, fine-tuning the p53 response against HIV-1. This study brings a new understanding of the complex role of p53 in virus replication processes in myeloid cells. Importance As of today, HIV-1 is still considered as a global pandemic without a functional cure, partly because of the presence of stable viral reservoirs. Macrophages constitute one of these cell reservoirs, contributing to the viral persistence. Studies investigating the host factors involved in cell susceptibility to HIV-1 infection might lead to a better understanding of the reservoir formation and will eventually allow the development of an efficient cure. Our team previously showed the antiviral role of p53 in macrophages, which acts by compromising the early steps of HIV-1 replication. In this study, we demonstrate the involvement of p53 isoforms, which regulates p53 activity and define the cellular environment influencing viral replication. In addition, the results concerning the potential role of p53 in antiviral innate immunity could be transposed to other fields of virology and suggest that knowledge in oncology can be applied to HIV-1 research.
Collapse
|
2
|
Melo Dos Santos N, de Oliveira GAP, Ramos Rocha M, Pedrote MM, Diniz da Silva Ferretti G, Pereira Rangel L, Morgado-Diaz JA, Silva JL, Rodrigues Pereira Gimba E. Loss of the p53 transactivation domain results in high amyloid aggregation of the Δ40p53 isoform in endometrial carcinoma cells. J Biol Chem 2019; 294:9430-9439. [PMID: 31028175 PMCID: PMC6579457 DOI: 10.1074/jbc.ra119.007566] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/17/2019] [Indexed: 01/18/2023] Open
Abstract
Dysfunctional p53 formation and activity can result from aberrant expression and subcellular localization of distinct p53 isoforms or aggregates. Endometrial carcinoma (EC) is a cancer type in which p53 status is correlated with prognosis, and TP53 mutations are a frequent genetic modification. Here we aimed to evaluate the expression patterns of different p53 isoforms and their contributions to the formation and subcellular localization of p53 amyloid aggregates in both EC and endometrial nontumor cell lines. We found that full-length (fl) p53 and a truncated p53 isoform, Δ40p53, resulting from alternative splicing of exon 2 or alternative initiation of translation at ATG-40, are the predominantly expressed p53 variants in EC cells. However, Δ40p53 was the major p53 isoform in endometrial nontumor cells. Immunofluorescence assays revealed that Δ40p53 is mainly localized to cytoplasmic punctate structures of EC cells, resembling solid-phase structures similar to those found in neurodegenerative pathologies. Using light-scattering kinetics, CD, and transmission EM, we noted that the p53 N-terminal transactivation domain significantly reduces aggregation of the WT p53 DNA-binding domain, confirming the higher aggregation tendency of Δ40p53, which lacks this domain. This is the first report of cytoplasmic Δ40p53 in EC cells being a major component of amyloid aggregates. The differential aggregation properties of p53 isoforms in EC cells may open up new avenues in the development of therapeutic strategies that preferentially target specific p53 isoforms to prevent p53 amyloid aggregate formation.
Collapse
Affiliation(s)
- Nataly Melo Dos Santos
- From the Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Oncobiologia Celular e Molecular, Rio de Janeiro, Brazil.,the Universidade Federal Fluminense, Instituto de Humanidades e Saúde, Departamento de Ciências da Natureza, Rio de Janeiro 28895-532, Brazil, and
| | - Guilherme A P de Oliveira
- the Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, Brazil.,the Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia 22908
| | - Murilo Ramos Rocha
- From the Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Oncobiologia Celular e Molecular, Rio de Janeiro, Brazil
| | - Murilo M Pedrote
- the Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, Brazil
| | - Giulia Diniz da Silva Ferretti
- the Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, Brazil
| | - Luciana Pereira Rangel
- the Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, Brazil.,the Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-170, Brazil
| | - José A Morgado-Diaz
- From the Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Oncobiologia Celular e Molecular, Rio de Janeiro, Brazil
| | - Jerson L Silva
- the Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, Brazil,
| | - Etel Rodrigues Pereira Gimba
- From the Instituto Nacional de Câncer, Coordenação de Pesquisa, Programa de Oncobiologia Celular e Molecular, Rio de Janeiro, Brazil, .,the Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, Brazil.,the Universidade Federal Fluminense, Instituto de Humanidades e Saúde, Departamento de Ciências da Natureza, Rio de Janeiro 28895-532, Brazil, and
| |
Collapse
|
3
|
Xiong Y, Deng Y, Wang K, Zhou H, Zheng X, Si L, Fu Z. Profiles of alternative splicing in colorectal cancer and their clinical significance: A study based on large-scale sequencing data. EBioMedicine 2018; 36:183-195. [PMID: 30243491 PMCID: PMC6197784 DOI: 10.1016/j.ebiom.2018.09.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023] Open
Abstract
Background Alternative splicing (AS), as a potent and pervasive mechanism of transcriptional regulatory, expands the genome's coding capacity and involves in the initiation and progression of cancer. Systematic analysis of alternative splicing in colorectal cancer (CRC) is lacking and greatly needed. Methods RNA-Seq data and corresponding clinical information of CRC cohort were downloaded from the TCGA data portal. Then, a java application, known as SpliceSeq, was used to evaluate the RNA splicing patterns and calculate the Percent Spliced In (PSI) value. Differently expressed AS events (DEAS) were identified based on PSI value between paired CRC and adjacent tissues. DEAS and its splicing networks were further analyzed by bioinformatics methods. Kaplan-Meier, Cox proportional regression and unsupervised clustering analysis were used to evaluate the association between DEAS and patients' clinical features. Results After strict filtering, a total of 34,334 AS events were identified, among which 421 AS events were found expressed differently. Parent genes of these DEAS play a important role in regulating CRC-related processes such as protein kinase activity (FDR<0.0001), PI3K-Akt signaling pathway (FDR = 0.0024) and p53 signaling pathway (FDR = 0.0143). 37 DEAS events were found to be associated with OS, and 68 DEAS events were found to be associated with DFS. Stratifying patients according to the PSI value of AT in CXCL12 and RI in CSTF3 formed significant Kaplan-Meier curves in both OS and DFS survival analysis. Unsupervised clustering analysis using DEAS revealed four clusters with distinct survival patterns, and associated with consensus molecular subtypes. Conclusions Large differences of AS events in CRC appear to exist, and these differences are likely to be important determinants of both prognosis and biological regulation. Our identified CRC-related AS events and uncovered splicing networks are valuable in deciphering the underlying mechanisms of AS in CRC, and provide clues of therapeutic targets to further validations.
Collapse
Affiliation(s)
- Yongfu Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Deng
- Department of Cardiovascular, The First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - He Zhou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangru Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangyi Si
- Department of Cardiovascular, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Zhongxue Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Targeting p53 as a promising therapeutic option for cancer by re-activating the wt or mutant p53’s tumor suppression. Future Med Chem 2018; 10:755-777. [DOI: 10.4155/fmc-2017-0175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
p53 protein, a product of the TP53 tumor suppressor gene, controls the cellular genome’s integrity and is an important regulator of cell cycling, proliferation, apoptosis and metabolism. Mutations of TP53 or inactivation of its gene product are among the first events initiating malignant transformation. The consequent loss of control over the cell cycle, resulting in accelerated cell proliferation and facilitating metabolic reprogramming, gives the initiated (premalignant) cells numerous advantages over healthy cells. Interestingly, p53 status is not only an important marker in cancer diagnosis; it has also become a promising target of personalized therapy. Depending on the TP53 status different therapeutic options have been developed. (Re)-activation of p53 functionality in cancer cells offers promising new alternatives to existing oncological therapies.
Collapse
|
5
|
Jiang W, Liu L, Chen Y. Simultaneous Detection of Human C-Terminal p53 Isoforms by Single Template Molecularly Imprinted Polymers (MIPs) Coupled with Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Targeted Proteomics. Anal Chem 2018; 90:3058-3066. [DOI: 10.1021/acs.analchem.7b02890] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Wenting Jiang
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
| | - Liang Liu
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
- China State Key Laboratory of Reproductive Medicine, Nanjing, China 210029
| |
Collapse
|
6
|
Yan R, Zhang J, Zellmer L, Chen L, Wu D, Liu S, Xu N, Liao JD. Probably less than one-tenth of the genes produce only the wild type protein without at least one additional protein isoform in some human cancer cell lines. Oncotarget 2017; 8:82714-82727. [PMID: 29137297 PMCID: PMC5669923 DOI: 10.18632/oncotarget.20015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/30/2017] [Indexed: 11/25/2022] Open
Abstract
To estimate how many genes produce multiple protein isoforms, we electrophoresed proteins from MCF7 and MDA-MB231 (MB231) human breast cancer cells in SDS-PAGE and excised narrow stripes of the gel at the 48kD, 55kD and 72kD. Proteins in these stripes were identified using liquid chromatography and tandem mass spectrometry. A total of 765, 750 and 679 proteins from MB231 cells, as well as 470, 390 and 490 proteins from MCF7 cells, were identified from the 48kD, 55kD and 72kD stripes, respectively. We arbitrarily allowed a 10% technical variation from the proteins' theoretical molecular mass (TMM) and considered those proteins with their TMMs within the 43-53 kD, 49-61 kD and 65-79 kD ranges as the wild type (WT) expected from the corresponding stripe, whereas those with a TMM above or below this range as a smaller- or larger-group, respectively. Only 263 (34.4%), 269 (35.9%) and 151 (22.2%) proteins from MB231 cells and 117 (24.9%), 135 (34.6%) and 130 (26.5%) proteins from MCF7 cells from the 48kD, 55kD and 72kD stripes, respectively, belonged to the WT, while the remaining majority belonged to the smaller- or larger-groups. Only about 3-16%, on average about 10% regardless of the stripe and cell line, of the proteins appeared in only one stripe and within the WT range, while the remaining preponderance appeared also in additional stripe(s) or had a larger or smaller TMM. We conclude that few (fewer than 10%) of the human genes produce only the WT protein without additional isoform(s).
Collapse
Affiliation(s)
- Rui Yan
- Nephrology Department, Guizhou Medical University Hospital, Guiyang, P.R. China
| | - Ju Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Lucas Zellmer
- Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Lichan Chen
- Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Di Wu
- Beijing Protein Innovation Co., Ltd, Beijing, P.R. China
| | - Siqi Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Joshua D Liao
- Department of Pathology, Guizhou Medical University Hospital, Guiyang, P.R. China
| |
Collapse
|
7
|
p53 Proteoforms and Intrinsic Disorder: An Illustration of the Protein Structure-Function Continuum Concept. Int J Mol Sci 2016; 17:ijms17111874. [PMID: 27834926 PMCID: PMC5133874 DOI: 10.3390/ijms17111874] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/27/2016] [Accepted: 11/03/2016] [Indexed: 01/10/2023] Open
Abstract
Although it is one of the most studied proteins, p53 continues to be an enigma. This protein has numerous biological functions, possesses intrinsically disordered regions crucial for its functionality, can form both homo-tetramers and isoform-based hetero-tetramers, and is able to interact with many binding partners. It contains numerous posttranslational modifications, has several isoforms generated by alternative splicing, alternative promoter usage or alternative initiation of translation, and is commonly mutated in different cancers. Therefore, p53 serves as an important illustration of the protein structure–function continuum concept, where the generation of multiple proteoforms by various mechanisms defines the ability of this protein to have a multitude of structurally and functionally different states. Considering p53 in the light of a proteoform-based structure–function continuum represents a non-canonical and conceptually new contemplation of structure, regulation, and functionality of this important protein.
Collapse
|
8
|
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
|
9
|
Gadea G, Arsic N, Fernandes K, Diot A, Joruiz SM, Abdallah S, Meuray V, Vinot S, Anguille C, Remenyi J, Khoury MP, Quinlan PR, Purdie CA, Jordan LB, Fuller-Pace FV, de Toledo M, Cren M, Thompson AM, Bourdon JC, Roux P. TP53 drives invasion through expression of its Δ133p53β variant. eLife 2016; 5. [PMID: 27630122 PMCID: PMC5067115 DOI: 10.7554/elife.14734] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 09/13/2016] [Indexed: 12/28/2022] Open
Abstract
TP53 is conventionally thought to prevent cancer formation and progression to metastasis, while mutant TP53 has transforming activities. However, in the clinic, TP53 mutation status does not accurately predict cancer progression. Here we report, based on clinical analysis corroborated with experimental data, that the p53 isoform Δ133p53β promotes cancer cell invasion, regardless of TP53 mutation status. Δ133p53β increases risk of cancer recurrence and death in breast cancer patients. Furthermore Δ133p53β is critical to define invasiveness in a panel of breast and colon cell lines, expressing WT or mutant TP53. Endogenous mutant Δ133p53β depletion prevents invasiveness without affecting mutant full-length p53 protein expression. Mechanistically WT and mutant Δ133p53β induces EMT. Our findings provide explanations to 2 long-lasting and important clinical conundrums: how WT TP53 can promote cancer cell invasion and reciprocally why mutant TP53 gene does not systematically induce cancer progression. DOI:http://dx.doi.org/10.7554/eLife.14734.001 Most cancers are caused by a build-up of mutations that are acquired throughout life. One gene in particular, called TP53, is the most commonly mutated gene in many types of human cancers. This suggests that TP53 mutations play an important role in cancer development. It is widely considered that the TP53 gene normally stops tumors from forming, while mutant forms of the gene somehow promote cancer growth. Evidence from patients with cancer has shown, however, that the relationship between TP53 mutations and cancer is not that simple. Some very aggressive cancers that resist treatment and spread have a normal TP53 gene. Some cancers with a mutated gene do not spread and respond well to cancer treatments. Recent studies have shown that the normal TP53 gene produces many different versions of its protein, and that some of these naturally occurring forms are found more often in tumors that others. However, it was not clear if certain versions of TP53’s proteins contributed to the development of cancer. Now, Gadea, Arsic, Fernandes et al. show that Δ133p53β, one version of the protein produced by the TP53 gene in human cells, helps tumor cells to spread to other organs. Tests of 273 tumors taken from patients with breast cancer revealed that tumors with the Δ133p53β protein were more likely to spread. Patients with these Δ133p53β-containing tumors were also more likely to develop secondary tumors at other sites in the body and to die within five years. Next, a series of experiments showed that removing Δ133p53β from breast cancer cells grown in the laboratory made them less likely to invade, while adding it back had the opposite effect. The same thing happened in colon cancer cells grown in the laboratory. The experiments showed that Δ133p53β causes tumor cells with the normal TP53 gene or a mutated TP53 gene to spread to other organs. Together the new findings help explain why some aggressive cancers develop even with a normal version of the tumor-suppressing TP53 gene. They also help explain why not all cancers with a mutant version of the TP53 gene go on to spread. Future studies will be needed to determine whether drugs that prevent the production of the Δ133p53β protein can help to treat aggressive cancers. DOI:http://dx.doi.org/10.7554/eLife.14734.002
Collapse
Affiliation(s)
- Gilles Gadea
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - Nikola Arsic
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - Kenneth Fernandes
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Alexandra Diot
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Sébastien M Joruiz
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Samer Abdallah
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - Valerie Meuray
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Stéphanie Vinot
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - Christelle Anguille
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - Judit Remenyi
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Marie P Khoury
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Philip R Quinlan
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Colin A Purdie
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Lee B Jordan
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Frances V Fuller-Pace
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Marion de Toledo
- Université Montpellier, Montpellier, France.,CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Maïlys Cren
- Université Montpellier, Montpellier, France.,IRB, Institut de Recherche en Biothérapie, Montpellier, France
| | - Alastair M Thompson
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom.,Department of Surgical Oncology, MD Anderson Cancer Centre, Houston, United States
| | - Jean-Christophe Bourdon
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Pierre Roux
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| |
Collapse
|
10
|
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
|
11
|
Abstract
p53 is a master regulatory protein that is involved in diverse cellular metabolic processes such as apoptosis, DNA repair, and cell cycle arrest. The protective function of p53 (in its homotetrameric form) as a tumor suppressor is lost in more than 50% of human cancers.Despite considerable experimental evidence suggesting the presence of multiple p53 states, it has been difficult to correlate the status of p53 with cancer response to treatments and clinical outcomes, which suggest the importance of complex but essential p53 regulatory pathways.Recent studies have indicated that the expression pattern of p53 isoforms may play a crucial role in regulating normal and cancer cell fates in response to diverse stresses. The human TP53 gene encodes at least 12 p53 isoforms, which are produced in normal tissue through alternative initiation of translation, usage of alternative promoters, and alternative splicing. Furthermore, some researchers have suggested that the formation of mutant p53 aggregates may be associated with cancer pathogenesis due to loss-of function (LoF), dominant-negative (DN), and gain-of function (GoF) effects.As different isoforms or the aggregation state of p53 may influence tumorigenesis, this review aims to examine the correlation of p53 isoforms and aggregation with cancer.
Collapse
Affiliation(s)
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
- Correspondence: Seong Soo A. An, Department of Bionano Techonology, Gachon University65 San Bokjung-dong, Sujung-gu, Seongnam-si, Gyeonggi-do 461-701, Republic of Korea (e-mail: )
| |
Collapse
|
12
|
Targeted Proteomics Enables Simultaneous Quantification of Folate Receptor Isoforms and Potential Isoform-based Diagnosis in Breast Cancer. Sci Rep 2015; 5:16733. [PMID: 26573433 PMCID: PMC4648081 DOI: 10.1038/srep16733] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023] Open
Abstract
The distinct roles of protein isoforms in cancer are becoming increasingly evident. FRα and FRβ, two major isoforms of the folate receptor family, generally have different cellular distribution and tissue specificity. However, the presence of FRβ in breast tumors, where FRα is normally expressed, complicates this situation. Prior to applying any FR isoform-based diagnosis and therapeutics, it is essential to monitor the expression profile of FR isoforms in a more accurate manner. An LC-MS/MS-based targeted proteomics assay was developed and validated in this study because of the lack of suitable methodology for the simultaneous and specific measurement of highly homologous isoforms occurring at low concentrations. FRα and FRβ monitoring was achieved by measuring their surrogate isoform-specific peptides. Five human breast cell lines, isolated macrophages and 60 matched pairs of breast tissue samples were subjected to the analysis. The results indicated that FRβ was overexpressed in tumor-associated macrophages (TAMs) but not epithelial cells, in addition to an enhanced level of FRα in breast cancer cells and tissue samples. Moreover, the levels of the FR isoforms were evaluated according to the histology, histopathological features and molecular subtypes of breast cancer. Several positive associations with PR/ER and HER2 status and metastasis were revealed.
Collapse
|
13
|
Xu F, Yang T, Sheng Y, Zhong T, Yang M, Chen Y. Simultaneous Quantification of Protein Phosphorylation Sites using Liquid Chromatography–Tandem Mass Spectrometry-Based Targeted Proteomics: A Linear Algebra Approach for Isobaric Phosphopeptides. J Proteome Res 2014; 13:5452-60. [DOI: 10.1021/pr500339u] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Feifei Xu
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ting Yang
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuan Sheng
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ting Zhong
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Mi Yang
- Nanjing Gulou Hospital, Nanjing 210008, China
| | - Yun Chen
- School
of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
14
|
Leroy B, Girard L, Hollestelle A, Minna JD, Gazdar AF, Soussi T. Analysis of TP53 mutation status in human cancer cell lines: a reassessment. Hum Mutat 2014; 35:756-65. [PMID: 24700732 DOI: 10.1002/humu.22556] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 03/20/2014] [Indexed: 12/14/2022]
Abstract
Tumor-derived cell lines play an important role in the investigation of tumor biology and genetics. Across a wide array of studies, they have been tools of choice for the discovery of important genes involved in cancer and for the analysis of the cellular pathways that are impaired by diverse oncogenic events. They are also invaluable for screening novel anticancer drugs. The TP53 protein is a major component of multiple pathways that regulate cellular response to various types of stress. Therefore, TP53 status affects the phenotype of tumor cell lines profoundly and must be carefully ascertained for any experimental project. In the present review, we use the 2014 release of the UMD TP53 database to show that TP53 status is still controversial for numerous cell lines, including some widely used lines from the NCI-60 panel. Our analysis clearly confirms that, despite numerous warnings, the misidentification of cell lines is still present as a silent and neglected issue, and that extreme care must be taken when determining the status of p53, because errors may lead to disastrous experimental interpretations. A novel compendium gathering the TP53 status of 2,500 cell lines has been made available (http://p53.fr). A stand-alone application can be used to browse the database and extract pertinent information on cell lines and associated TP53 mutations. It will be updated regularly to minimize any scientific issues associated with the use of misidentified cell lines (http://p53.fr).
Collapse
Affiliation(s)
- Bernard Leroy
- Université Pierre et Marie Curie-Paris 6, Paris, 75005, France
| | | | | | | | | | | |
Collapse
|
15
|
Fant X, Durieu E, Chicanne G, Payrastre B, Sbrissa D, Shisheva A, Limanton E, Carreaux F, Bazureau JP, Meijer L. cdc-like/dual-specificity tyrosine phosphorylation-regulated kinases inhibitor leucettine L41 induces mTOR-dependent autophagy: implication for Alzheimer's disease. Mol Pharmacol 2014; 85:441-50. [PMID: 24366666 PMCID: PMC6067634 DOI: 10.1124/mol.113.090837] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/23/2013] [Indexed: 12/26/2022] Open
Abstract
Leucettines, a family of pharmacological inhibitors of dual-specificity tyrosine phosphorylation regulated kinases and cdc-like kinases (CLKs), are currently under investigation for their potential therapeutic application to Down syndrome and Alzheimer's disease. We here report that leucettine L41 triggers bona fide autophagy in osteosarcoma U-2 OS cells and immortalized mouse hippocampal HT22 cells, characterized by microtubule-associated protein light chain 3 membrane translocation and foci formation. Leucettine L41-triggered autophagy requires the Unc-51-like kinase and is sensitive to the phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and 3-methyladenine, suggesting that it acts through the mammalian target of rapamycin (mTOR)/PI3K-dependent pathway. Leucettine L41 does not act by modifying the autophagic flux of vesicles. Leucettine L41-induced autophagy correlates best with inhibition of CLKs. Leucettine L41 modestly inhibited phosphatidylinositol-3-phosphate 5-kinase, FYVE domain-containing activity as tested both in vitro and in vivo, which may also contribute to autophagy induction. Altogether these results demonstrate that leucettines can activate the autophagic mTOR/PI3K pathway, a characteristic that may turn advantageous in the context of Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Xavier Fant
- Centre National de la Recherche Scientifique (CNRS), USR3151, "Protein Phosphorylation and Human Disease," Station Biologique, Roscoff cedex, France (X.F., E.D.); Institut National de la Santé et de la Recherche Médicale/Université Paul Sabatier Unité Mixte de Recherche (UMR) 1048, "Production et fonctions plaquettaires: signalisation et phosphoinositides" group, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse cedex, France (G.C., B.P.); Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan (D.S., A.S.); Laboratoire Sciences Chimiques de Rennes, UMR CNRS 6226, Groupe Ingénierie Chimique et Molécules pour le Vivant (ICMV), Université de Rennes, Campus de Beaulieu, Rennes cedex, France (E.L., F.C., J.-P.B.); and ManRos Therapeutics, Perharidy Research Center, Roscoff, Bretagne, France (L.M.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mondal AM, Horikawa I, Pine SR, Fujita K, Morgan KM, Vera E, Mazur SJ, Appella E, Vojtesek B, Blasco MA, Lane DP, Harris CC. p53 isoforms regulate aging- and tumor-associated replicative senescence in T lymphocytes. J Clin Invest 2013; 123:5247-57. [PMID: 24231352 PMCID: PMC3859419 DOI: 10.1172/jci70355] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/10/2013] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence contributes to aging and decline in tissue function. p53 isoform switching regulates replicative senescence in cultured fibroblasts and is associated with tumor progression. Here, we found that the endogenous p53 isoforms Δ133p53 and p53β are physiological regulators of proliferation and senescence in human T lymphocytes in vivo. Peripheral blood CD8+ T lymphocytes collected from healthy donors displayed an age-dependent accumulation of senescent cells (CD28-CD57+) with decreased Δ133p53 and increased p53β expression. Human lung tumor-associated CD8+ T lymphocytes also harbored senescent cells. Cultured CD8+ blood T lymphocytes underwent replicative senescence that was associated with loss of CD28 and Δ133p53 protein. In poorly proliferative, Δ133p53-low CD8+CD28- cells, reconstituted expression of either Δ133p53 or CD28 upregulated endogenous expression of each other, which restored cell proliferation, extended replicative lifespan and rescued senescence phenotypes. Conversely, Δ133p53 knockdown or p53β overexpression in CD8+CD28+ cells inhibited cell proliferation and induced senescence. This study establishes a role for Δ133p53 and p53β in regulation of cellular proliferation and senescence in vivo. Furthermore, Δ133p53-induced restoration of cellular replicative potential may lead to a new therapeutic paradigm for treating immunosenescence disorders, including those associated with aging, cancer, autoimmune diseases, and HIV infection.
Collapse
Affiliation(s)
- Abdul M. Mondal
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sharon R. Pine
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kaori Fujita
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Katherine M. Morgan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Elsa Vera
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sharlyn J. Mazur
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ettore Appella
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Borivoj Vojtesek
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Maria A. Blasco
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - David P. Lane
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Department of Medicine, UMDNJ/Robert Wood Johnson Medical School, The Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
Telomeres and Telomerase Group/Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, C/Melchor Fernández Almagro, Madrid, Spain.
Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic.
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|