201
|
Targeting cyclooxygenase by indomethacin decelerates progression of acute lymphoblastic leukemia in a xenograft model. Blood Adv 2020; 3:3181-3190. [PMID: 31698450 DOI: 10.1182/bloodadvances.2019000473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/05/2019] [Indexed: 01/02/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) develops in the bone marrow in the vicinity of stromal cells known to promote tumor development and treatment resistance. We previously showed that the cyclooxygenase (COX) inhibitor indomethacin prevents the ability of stromal cells to diminish p53-mediated killing of cocultured ALL cells in vitro, possibly by blocking the production of prostaglandin E2 (PGE2). Here, we propose that PGE2 released by bone marrow stromal cells might be a target for improved treatment of pediatric ALL. We used a xenograft model of human primary ALL cells in nonobese diabetic-scid IL2rγnull mice to show that indomethacin delivered in the drinking water delayed the progression of ALL in vivo. The progression was monitored by noninvasive in vivo imaging of the engrafted leukemic cells, as well as by analyses of CD19+CD10+ leukemic blasts present in spleen or bone marrow at the termination of the experiments. The indomethacin treatment increased the level of p53 in the leukemic cells, implying that COX inhibition might reduce progression of ALL by attenuating protective paracrine PGE2 signaling from bone marrow stroma to leukemic cells.
Collapse
|
202
|
Napolitano R, De Matteis S, Carloni S, Bruno S, Abbati G, Capelli L, Ghetti M, Bochicchio MT, Liverani C, Mercatali L, Calistri D, Cuneo A, Menon K, Musuraca G, Martinelli G, Simonetti G. Kevetrin induces apoptosis in TP53 wild‑type and mutant acute myeloid leukemia cells. Oncol Rep 2020; 44:1561-1573. [PMID: 32945487 PMCID: PMC7448420 DOI: 10.3892/or.2020.7730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor protein p53 is a key regulator of several cellular pathways, including DNA repair, cell cycle and angiogenesis. Kevetrin exhibits p53-dependent as well as-independent activity in solid tumors, while its effects on leukemic cells remain unknown. The aim of the present study was to analyze the response of acute myeloid leukemia (AML) cell lines (TP53 wild-type: OCI-AML3 and MOLM-13; and TP53-mutant: KASUMI-1 and NOMO-1) to kevetrin at a concentration range of 85–340 µM. The cellular and molecular effects of the treatment were analyzed in terms of cell growth, viability [Annexin V-propidium iodide (PI) staining] and cell cycle alterations (PI staining). Gene expression profiling, western blotting and immunofluorescence were performed to elucidate the pathways underlying kevetrin activity. Pulsed exposure exerted no effect on the wild-type cells, but was effective on mutant cells. After continuous treatment, significant cell growth arrest and apoptosis were observed in all cell lines, with TP53-mutant models displaying a higher sensitivity and p53 induction. Kevetrin also displayed efficacy against TP53 wild-type and mutant primary AML, with a preferential cytotoxic activity against blast cells. Gene expression profiling revealed a common core transcriptional program altered by drug exposure and the downregulation of glycolysis, DNA repair and unfolded protein response signatures. These findings suggest that kevetrin may be a promising therapeutic option for patients with both wild-type and TP53-mutant AML.
Collapse
Affiliation(s)
- Roberta Napolitano
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Serena De Matteis
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Silvia Carloni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Samantha Bruno
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and Institute of Hematology 'L. e A. Seràgnoli', I‑40138 Bologna, Italy
| | - Giulia Abbati
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Laura Capelli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Martina Ghetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Chiara Liverani
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Laura Mercatali
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Daniele Calistri
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Antonio Cuneo
- Department of Medical Sciences, University of Ferrara‑Arcispedale Sant'Anna, I‑44124 Ferrara, Italy
| | | | - Gerardo Musuraca
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| | - Giovanni Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna and Institute of Hematology 'L. e A. Seràgnoli', I‑40138 Bologna, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, I‑47014 Meldola, Italy
| |
Collapse
|
203
|
Aguayo F, Muñoz JP, Perez-Dominguez F, Carrillo-Beltrán D, Oliva C, Calaf GM, Blanco R, Nuñez-Acurio D. High-Risk Human Papillomavirus and Tobacco Smoke Interactions in Epithelial Carcinogenesis. Cancers (Basel) 2020; 12:E2201. [PMID: 32781676 PMCID: PMC7465661 DOI: 10.3390/cancers12082201] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
Cervical, anogenital, and some head and neck cancers (HNC) are etiologically associated with high-risk human papillomavirus (HR-HPV) infection, even though additional cofactors are necessary. Epidemiological studies have established that tobacco smoke (TS) is a cofactor for cervical carcinogenesis because women who smoke are more susceptible to cervical cancer when compared to non-smokers. Even though such a relationship has not been established in HPV-related HNC, a group of HPV positive patients with this malignancy are smokers. TS is a complex mixture of more than 4500 chemical compounds and approximately 60 of them show oncogenic properties such as benzo[α]pyrene (BaP) and nitrosamines, among others. Some of these compounds have been evaluated for carcinogenesis through experimental settings in collaboration with HR-HPV. Here, we conducted a comprehensive review of the suggested molecular mechanisms involved in cooperation with both HR-HPV and TS for epithelial carcinogenesis. Furthermore, we propose interaction models in which TS collaborates with HR-HPV to promote epithelial cancer initiation, promotion, and progression. More studies are warranted to clarify interactions between oncogenic viruses and chemical or physical environmental factors for epithelial carcinogenesis.
Collapse
Affiliation(s)
- Francisco Aguayo
- Universidad de Tarapacá, Arica 1000000, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, Santiago 8330024, Chile
| | - Juan P. Muñoz
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (J.P.M.); (G.M.C.)
| | - Francisco Perez-Dominguez
- Laboratorio Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (F.P.-D.); (D.C.-B.); (C.O.); (R.B.); (D.N.-A.)
| | - Diego Carrillo-Beltrán
- Laboratorio Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (F.P.-D.); (D.C.-B.); (C.O.); (R.B.); (D.N.-A.)
| | - Carolina Oliva
- Laboratorio Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (F.P.-D.); (D.C.-B.); (C.O.); (R.B.); (D.N.-A.)
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (J.P.M.); (G.M.C.)
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
| | - Rances Blanco
- Laboratorio Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (F.P.-D.); (D.C.-B.); (C.O.); (R.B.); (D.N.-A.)
| | - Daniela Nuñez-Acurio
- Laboratorio Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (F.P.-D.); (D.C.-B.); (C.O.); (R.B.); (D.N.-A.)
| |
Collapse
|
204
|
Benor G, Fuks G, Chin S, Rueda OM, Mukherjee S, Arandkar S, Aylon Y, Caldas C, Domany E, Oren M. Transcriptional profiling reveals a subset of human breast tumors that retain wt TP53 but display mutant p53-associated features. Mol Oncol 2020; 14:1640-1652. [PMID: 32484602 PMCID: PMC7400784 DOI: 10.1002/1878-0261.12736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 11/25/2022] Open
Abstract
TP53 gene mutations are very common in human cancer. While such mutations abrogate the tumor suppressive activities of the wild-type (wt) p53 protein, some of them also endow the mutant (mut) protein with oncogenic gain of function (GOF), facilitating cancer progression. Yet, p53 may acquire altered functionality even without being mutated; in particular, experiments with cultured cells revealed that wtp53 can be rewired to adopt mut-like features in response to growth factors or cancer-mimicking genetic manipulations. To assess whether such rewiring also occurs in human tumors, we interrogated gene expression profiles and pathway deregulation patterns in the METABRIC breast cancer (BC) dataset as a function of TP53 gene mutation status. Harnessing the power of machine learning, we optimized a gene expression classifier for ER+Her2- patients that distinguishes tumors carrying TP53 mutations from those retaining wt TP53. Interestingly, a small subset of wt TP53 tumors displayed gene expression and pathway deregulation patterns markedly similar to those of TP53-mutated tumors. Moreover, similar to TP53-mutated tumors, these 'pseudomutant' cases displayed a signature for enhanced proliferation and had worse prognosis than typical wtp53 tumors. Notably, these tumors revealed upregulation of genes which, in BC cell lines, were reported to be positively regulated by p53 GOF mutants. Thus, such tumors may benefit from mut p53-associated activities without having to accrue TP53 mutations.
Collapse
Affiliation(s)
- Gal Benor
- Department of Physics of Complex SystemsThe Weizmann Institute of ScienceRehovotIsrael
| | - Garold Fuks
- Department of Physics of Complex SystemsThe Weizmann Institute of ScienceRehovotIsrael
| | - Suet‐Feung Chin
- Cancer Research UK Cambridge Institute and Department of OncologyLi Ka Shing CentreUniversity of CambridgeCambridgeUK
| | - Oscar M. Rueda
- Cancer Research UK Cambridge Institute and Department of OncologyLi Ka Shing CentreUniversity of CambridgeCambridgeUK
| | - Saptaparna Mukherjee
- Department of Molecular Cell BiologyThe Weizmann Institute of ScienceRehovotIsrael
| | - Sharathchandra Arandkar
- Department of Molecular Cell BiologyThe Weizmann Institute of ScienceRehovotIsrael
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC)Tata Memorial CentreKhargharIndia
| | - Yael Aylon
- Department of Molecular Cell BiologyThe Weizmann Institute of ScienceRehovotIsrael
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of OncologyLi Ka Shing CentreUniversity of CambridgeCambridgeUK
| | - Eytan Domany
- Department of Physics of Complex SystemsThe Weizmann Institute of ScienceRehovotIsrael
| | - Moshe Oren
- Department of Molecular Cell BiologyThe Weizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
205
|
Bennani-Baiti B, Pinker K, Zimmermann M, Helbich TH, Baltzer PA, Clauser P, Kapetas P, Bago-Horvath Z, Stadlbauer A. Non-Invasive Assessment of Hypoxia and Neovascularization with MRI for Identification of Aggressive Breast Cancer. Cancers (Basel) 2020; 12:cancers12082024. [PMID: 32721996 PMCID: PMC7464174 DOI: 10.3390/cancers12082024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to investigate the potential of magnetic resonance imaging (MRI) for a non-invasive synergistic assessment of tumor microenvironment (TME) hypoxia and induced neovascularization for the identification of aggressive breast cancer. Fifty-three female patients with breast cancer underwent multiparametric breast MRI including quantitative blood-oxygen-level-dependent (qBOLD) imaging for hypoxia and vascular architecture mapping for neovascularization. Quantitative MRI biomarker maps of oxygen extraction fraction (OEF), metabolic rate of oxygen (MRO2), mitochondrial oxygen tension (mitoPO2), microvessel radius (VSI), microvessel density (MVD), and microvessel type indicator (MTI) were calculated. Histopathology was the standard of reference. Histopathological markers (vascular endothelial growth factor receptor 1 (FLT1), podoplanin, hypoxia-inducible factor 1-alpha (HIF-1alpha), carbonic anhydrase 9 (CA IX), vascular endothelial growth factor C (VEGF-C)) were used to confirm imaging biomarker findings. Univariate and multivariate regression analyses were performed to differentiate less aggressive luminal from aggressive non-luminal (HER2-positive, triple negative) malignancies and assess the interplay between hypoxia and neoangiogenesis markers. Aggressive non-luminal cancers (n = 40) presented with significantly higher MRO2 (i.e., oxygen consumption), lower mitoPO2 values (i.e., hypoxia), lower MTI, and higher MVD than less aggressive cancers (n = 13). Data suggest that a model derived from OEF, mitoPO2, and MVD can predict tumor proliferation rate. This novel MRI approach, which can be easily implemented in routine breast MRI exams, aids in the non-invasive identification of aggressive breast cancer.
Collapse
Affiliation(s)
- Barbara Bennani-Baiti
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, 1090 Vienna, Austria; (B.B.-B.); (K.P.); (P.A.B.); (P.C.); (P.K.)
| | - Katja Pinker
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, 1090 Vienna, Austria; (B.B.-B.); (K.P.); (P.A.B.); (P.C.); (P.K.)
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Max Zimmermann
- Department of Neurosurgery, University of Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (A.S.)
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas H. Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, 1090 Vienna, Austria; (B.B.-B.); (K.P.); (P.A.B.); (P.C.); (P.K.)
- Correspondence: ; Tel.: +43-1-40400-48980
| | - Pascal A. Baltzer
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, 1090 Vienna, Austria; (B.B.-B.); (K.P.); (P.A.B.); (P.C.); (P.K.)
| | - Paola Clauser
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, 1090 Vienna, Austria; (B.B.-B.); (K.P.); (P.A.B.); (P.C.); (P.K.)
| | - Panagiotis Kapetas
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, 1090 Vienna, Austria; (B.B.-B.); (K.P.); (P.A.B.); (P.C.); (P.K.)
| | | | - Andreas Stadlbauer
- Department of Neurosurgery, University of Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (A.S.)
- Institute of Medical Radiology, University Clinic of St. Pölten, 3100 St. Pölten, Austria
| |
Collapse
|
206
|
Shan L, Yu J, He Z, Chen S, Liu M, Ding H, Xu L, Zhao J, Yang A, Jiang H. Defining relative mutational difficulty to understand cancer formation. Cell Discov 2020; 6:48. [PMID: 32704382 PMCID: PMC7371891 DOI: 10.1038/s41421-020-0177-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Most mutations in human cancer are low-frequency missense mutations, whose functional status remains hard to predict. Here, we show that depending on the type of nucleotide change and the surrounding sequences, the tendency to generate each type of nucleotide mutations varies greatly, even by several hundred folds. Therefore, a cancer-promoting mutation may appear only in a small number of cancer cases, if the underlying nucleotide change is too difficult to generate. We propose a method that integrates both the original mutation counts and their relative mutational difficulty. Using this method, we can accurately predict the functionality of hundreds of low-frequency missense mutations in p53, PTEN, and INK4A. Many loss-of-function p53 mutations with dominant negative effects were identified, and the functional importance of several regions in p53 structure were highlighted by this analysis. Our study not only established relative mutational difficulties for different types of mutations in human cancer, but also showed that by incorporating such a parameter, we can bring new angles to understanding cancer formation.
Collapse
Affiliation(s)
- Lin Shan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jiao Yu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhengjin He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Shishuang Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Mingxian Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hongyu Ding
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Liang Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jie Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ailing Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hai Jiang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
207
|
Lozano G. Restoring p53 in cancer: the promises and the challenges. J Mol Cell Biol 2020; 11:615-619. [PMID: 31283825 PMCID: PMC6736346 DOI: 10.1093/jmcb/mjz063] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Guillermina Lozano
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence to: Guillermina Lozano, E-mail:
| |
Collapse
|
208
|
Eriksson SE, Ceder S, Bykov VJN, Wiman KG. p53 as a hub in cellular redox regulation and therapeutic target in cancer. J Mol Cell Biol 2020; 11:330-341. [PMID: 30892598 PMCID: PMC6734141 DOI: 10.1093/jmcb/mjz005] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/21/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022] Open
Abstract
The TP53 tumor suppressor gene encodes a DNA-binding transcription factor that regulates multiple cellular processes including cell growth and cell death. The ability of p53 to bind to DNA and activate transcription is tightly regulated by post-translational modifications and is dependent on a reducing cellular environment. Some p53 transcriptional target genes are involved in regulation of the cellular redox homeostasis, e.g. TIGAR and GLS2. A large fraction of human tumors carry TP53 mutations, most commonly missense mutations that lead to single amino acid substitutions in the core domain. Mutant p53 proteins can acquire so called gain-of-function activities and influence the cellular redox balance in various ways, for instance by binding of the Nrf2 transcription factor, a major regulator of cellular redox state. The DNA-binding core domain of p53 has 10 cysteine residues, three of which participate in holding a zinc atom that is critical for p53 structure and function. Several novel compounds that refold and reactivate missense mutant p53 bind to specific p53 cysteine residues. These compounds can also react with other thiols and target components of the cellular redox system, such as glutathione. Dual targeting of mutant p53 and redox homeostasis may allow more efficient treatment of cancer.
Collapse
Affiliation(s)
- Sofi E Eriksson
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| | - Sophia Ceder
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| | - Vladimir J N Bykov
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| | - Klas G Wiman
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| |
Collapse
|
209
|
Stein Y, Aloni-Grinstein R, Rotter V. Mutant p53-a potential player in shaping the tumor-stroma crosstalk. J Mol Cell Biol 2020; 11:600-604. [PMID: 31318969 PMCID: PMC6736352 DOI: 10.1093/jmcb/mjz071] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 06/26/2019] [Indexed: 12/28/2022] Open
Abstract
A plethora of studies suggest that the non-transformed cellular and non-cellular components of the tumor, collectively known as the tumor microenvironment, have a significant impact on the tumorigenic process. It was suggested that the microenvironment, which initially restricts tumor development, is recruited by the tumor and maintains a crosstalk that further promotes cancer progression. Indeed, many of the molecules that participate in the tumor–stroma crosstalk have been characterized. However, the crucial factors that are responsible for the initiation of this crosstalk or the ‘recruitment’ process remain poorly understood. We propose that oncogenes themselves may influence the ‘recruitment’ of the stromal cells, while focusing on mutant p53. Apart from losing its tumor-suppressing properties, mutant p53 gains novel oncogenic functions, a phenomenon dubbed mutant p53 gain of function (GOF). Here, we discuss possible ways in which mutant p53 may modulate the microenvironment in order to promote tumorigenesis. We thus propose that mutant p53 may serve as a key player in the modulation of the tumor–stroma crosstalk in a way that benefits the tumor. Further elucidation of these ‘recruitment’ processes, dictated by mutant p53, may be utilized for tailoring personalized therapeutic approaches for patients with tumors that harbor p53 mutation.
Collapse
Affiliation(s)
- Yan Stein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Box 19, Ness Ziona 7410001, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
210
|
Nakayama M, Oshima M. Mutant p53 in colon cancer. J Mol Cell Biol 2020; 11:267-276. [PMID: 30496442 PMCID: PMC6487790 DOI: 10.1093/jmcb/mjy075] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 11/13/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
The accumulation of genetic alterations in driver genes is responsible for the development and malignant progression of colorectal cancer. Comprehensive genome analyses have revealed the driver genes, including APC, KRAS, TGFBR2, and TP53, whose mutations are frequently found in human colorectal cancers. Among them, the p53 mutation is found in ~60% of colorectal cancers, and a majority of mutations are missense-type at ‘hot spots’, suggesting an oncogenic role of mutant p53 by ‘gain-of-function’ mechanisms. Mouse model studies have shown that one of these missense-type mutations, p53 R270H (corresponding to human R273H), causes submucosal invasion of intestinal tumors, while the loss of wild-type p53 has a limited effect on the invasion process. Furthermore, the same mutant p53 promotes metastasis when combined with Kras activation and TGF-β suppression. Importantly, either missense-type p53 mutation or loss of wild-type p53 induces NF-κB activation by a variety of mechanisms, such as increasing promoter accessibility by chromatin remodeling, which may contribute to progression to epithelial–mesenchymal transition. These results indicate that missense-type p53 mutations together with loss of wild-type p53 accelerate the late stage of colorectal cancer progression through the activation of both oncogenic and inflammatory pathways. Accordingly, the suppression of the mutant p53 function via the inhibition of nuclear accumulation is expected to be an effective strategy against malignant progression of colorectal cancer.
Collapse
Affiliation(s)
- Mizuho Nakayama
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,WPI-Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,WPI-Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
211
|
Koifman G, Aloni-Grinstein R, Rotter V. p53 balances between tissue hierarchy and anarchy. J Mol Cell Biol 2020; 11:553-563. [PMID: 30925590 PMCID: PMC6735948 DOI: 10.1093/jmcb/mjz022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/17/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023] Open
Abstract
Normal tissues are organized in a hierarchical model, whereas at the apex of these hierarchies reside stem cells (SCs) capable of self-renewal and of producing differentiated cellular progenies, leading to normal development and homeostasis. Alike, tumors are organized in a hierarchical manner, with cancer SCs residing at the apex, contributing to the development and nourishment of tumors. p53, the well-known ‘guardian of the genome’, possesses various roles in embryonic development as well as in adult SC life and serves as the ‘guardian of tissue hierarchy’. Moreover, p53 serves as a barrier for dedifferentiation and reprogramming by constraining the cells to a somatic state and preventing their conversion to SCs. On the contrary, the mutant forms of p53 that lost their tumor suppressor activity and gain oncogenic functions serve as ‘inducers of tissue anarchy’ and promote cancer development. In this review, we discuss these two sides of the p53 token that sentence a tissue either to an ordered hierarchy and life or to anarchy and death. A better understanding of these processes may open new horizons for the development of new cancer therapies.
Collapse
Affiliation(s)
- Gabriela Koifman
- Department of Molecular Cell Biology, the Weizmann Institute of Science, Rehovot, Israel
| | - Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, the Weizmann Institute of Science, Rehovot, Israel.,Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, the Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
212
|
SOX2 and p53 Expression Control Converges in PI3K/AKT Signaling with Versatile Implications for Stemness and Cancer. Int J Mol Sci 2020; 21:ijms21144902. [PMID: 32664542 PMCID: PMC7402325 DOI: 10.3390/ijms21144902] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Stemness and reprogramming involve transcriptional master regulators that suppress cell differentiation while promoting self-renewal. A distinguished example thereof is SOX2, a high mobility group (HMG)-box transcription factor (TF), whose subcellular localization and turnover regulation in embryonic, induced-pluripotent, and cancer stem cells (ESCs, iPSCs, and CSCs, respectively) is mediated by the PI3K/AKT/SOX2 axis, a stem cell-specific branch of the PI3K/AKT signaling pathway. Further effector functions associated with PI3K/AKT induction include cell cycle progression, cellular (mass) growth, and the suppression of apoptosis. Apoptosis, however, is a central element of DNA damage response (DDR), where it provides a default mechanism for cell clearance when DNA integrity cannot be maintained. A key player in DDR is tumor suppressor p53, which accumulates upon DNA-damage and is counter-balanced by PI3K/AKT enforced turnover. Accordingly, stemness sustaining SOX2 expression and p53-dependent DDR mechanisms show molecular–functional overlap in PI3K/AKT signaling. This constellation proves challenging for stem cells whose genomic integrity is a functional imperative for normative ontogenesis. Unresolved mutations in stem and early progenitor cells may in fact provoke transformation and cancer development. Such mechanisms are also particularly relevant for iPSCs, where genetic changes imposed through somatic cell reprogramming may promote DNA damage. The current review aims to summarize the latest advances in the understanding of PI3K/AKT/SOX2-driven stemness and its intertwined relations to p53-signaling in DDR under conditions of pluripotency, reprogramming, and transformation.
Collapse
|
213
|
Yokoi A, Matsumoto T, Oguri Y, Hasegawa Y, Tochimoto M, Nakagawa M, Saegusa M. Upregulation of fibronectin following loss of p53 function is a poor prognostic factor in ovarian carcinoma with a unique immunophenotype. Cell Commun Signal 2020; 18:103. [PMID: 32635925 PMCID: PMC7341596 DOI: 10.1186/s12964-020-00580-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/13/2020] [Indexed: 01/01/2023] Open
Abstract
Background We previously demonstrated that ovarian high grade serous carcinomas (OHGSeCa) and ovarian clear cell carcinomas (OCCCa) with an HNF-1β+/p53+/ARID1A+ immunophenotype were associated with the worst unfavorable prognosis. To clarify the molecular mechanisms underlying this finding, we focused on alterations in the p53 signaling pathway in these tumors. Methods Changes in cell phenotype and function following knockdown of wild-type p53 (p53-KD) were assessed using OCCCa cells expressing endogenous HNF-1β and ARID1A. The prognostic significance of molecules that were deregulated following p53-KD was also examined using 129 OCCCa/OHGSeCa cases. Results p53-KD cells had increased expression of Snail, phospho-Akt (pAkt), and pGSK3β, and decreased E-cadherin expression, leading to epithelial-mesenchymal transition (EMT)/cancer stem cell (CSC) features. The cells also exhibited acceleration of cell motility and inhibition of cell proliferation and apoptosis. Next generation sequencing revealed that fibronectin (FN) expression was significantly increased in the p53 KD-cells, in line with our observation that wild-type p53 (but not mutant p53) repressed FN1 promoter activity. In addition, treatment of OCCCa cells with FN significantly increased cell migration capacity and decreased cell proliferation rate, independent of induction of EMT features. In clinical samples, FN/p53 scores were significantly higher in OCCCa/OHGSeCa with the HNF-1β+/p53+/ARID1A+ immunophenotype when compared to others. Moreover, high FN/high p53 expression was associated with the worst overall survival and progression-free survival in OCCCa/OHGSeCa patients. Conclusion These findings suggest that upregulation of FN following loss of p53 function may impact the biological behavior of OCCCa/OHGSeCa, particularly in tumors with an HNF-1β+/p53+/ARID1A+ immunophenotype, through alterations in cell mobility and cell proliferation. The accompanying induction of EMT/CSC properties and inhibition of apoptosis due to p53 abnormalities also contribute to the establishment and maintenance of tumor phenotypic characteristics. Video Abstract
Collapse
Affiliation(s)
- Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yasuko Oguri
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshinori Hasegawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Laboratory of Clinical Omics Research, 2-6-7 Kazusakamatari, Kisaratsu, Chiba, 292-0818, Japan
| | - Masataka Tochimoto
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
214
|
Man N, Jun Y. Two Mutations in TP53 Promotes Migration, Invasion, and Proliferation of Human Ovarian Cancer A2780 Cells. RUSS J GENET+ 2020. [DOI: 10.1134/s1022795420070091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
215
|
Hsu YC, Luo CW, Huang WL, Wu CC, Chou CL, Chen CI, Chang SJ, Chai CY, Wang HC, Chen TY, Li CF, Pan MR. BMI1-KLF4 axis deficiency improves responses to neoadjuvant concurrent chemoradiotherapy in patients with rectal cancer. Radiother Oncol 2020; 149:249-258. [PMID: 32592893 DOI: 10.1016/j.radonc.2020.06.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Neoadjuvant concurrent chemoradiotherapy (CCRT) is a gold standard treatment for patients with stage II/III rectal cancer. B-cell-specific Moloney murine leukemia virus insertion site 1 (BMI1) is a member of the polycomb group of proteins that are involved in regulating gene expression. High levels of BMI1 have been demonstrated to contribute to the malignant phenotypes of several cancers; however, its relevance in rectal cancer treated with CCRT is largely unknown. METHODS AND MATERIALS We used two patient cohorts to address the clinical relevance of BMI1 in human cancers. In addition, HT-29 and HCT-116 cells were chosen as our in vitro models to verify the role of BMI1 in cell response to ionizing radiation. Stemness-related proteins were analyzed by western blotting and cell survival was determined using clonogenic assays. RESULTS BMI1 overexpression was found to significantly correlate with advanced pre-treatment nodal status (N1-N2; p < 0.001), post-treatment tumor stage (T1-T2; p = 0.015), inferior tumor regression grade (p = 0.001), and also an independent prognosis factor in 172 rectal cancer patients receiving CCRT. Serial cell-based functional examination indicated that BMI1 deficiency sensitized cells to radiation treatment by modulating the gene expression of Kruppel-like factor 4 (KLF4) and enhanced radiosensitivity in microsatellite stable (MSS) colorectal cancers. Overexpression of KLF4 partially overcame BMI1-deficiency-mediated γ-H2AX expression after ionizing radiation exposure. Consistent with in vitro data, an analysis of an additional 30 rectal cancer tissue specimens revealed a positive correlation between BMI1 and KLF4 (p = 0.02). CONCLUSION Higher levels of BMI1 are associated with poor therapeutic response and adverse outcomes in rectal cancer patients receiving CCRT.
Collapse
Affiliation(s)
- Yin-Chou Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chi-Wen Luo
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Lun Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Radiation Oncology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chun-Chieh Wu
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Lin Chou
- Division of Colon & Rectal Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chih-I Chen
- Division of Colon & Rectal Surgery, Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Shu-Jyuan Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Ching Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Yi Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
216
|
Sobhani N, D’Angelo A, Wang X, Young KH, Generali D, Li Y. Mutant p53 as an Antigen in Cancer Immunotherapy. Int J Mol Sci 2020; 21:ijms21114087. [PMID: 32521648 PMCID: PMC7312027 DOI: 10.3390/ijms21114087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022] Open
Abstract
The p53 tumor suppressor plays a pivotal role in cancer and infectious disease. Many oncology treatments are now calling on immunotherapy approaches, and scores of studies have investigated the role of p53 antibodies in cancer diagnosis and therapy. This review summarizes the current knowledge from the preliminary evidence that suggests a potential role of p53 as an antigen in the adaptive immune response and as a key monitor of the innate immune system, thereby speculating on the idea that mutant p53 antigens serve as a druggable targets in immunotherapy. Except in a few cases, the vast majority of published work on p53 antibodies in cancer patients use wild-type p53 as the antigen to detect these antibodies and it is unclear whether they can recognize p53 mutants carried by cancer patients at all. We envision that an antibody targeting a specific mutant p53 will be effective therapeutically against a cancer carrying the exact same mutant p53. To corroborate such a possibility, a recent study showed that a T cell receptor-like (TCLR) antibody, initially made for a wild-type antigen, was capable of discriminating between mutant p53 and wild-type p53, specifically killing more cancer cells expressing mutant p53 than wild-type p53 in vitro and inhibiting the tumour growth of mice injected with mutant p53 cancer cells than mice with wild-type p53 cancer cells. Thus, novel antibodies targeting mutant p53, but not the wild-type isoform, should be pursued in preclinical and clinical studies.
Collapse
Affiliation(s)
- Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Correspondence: (N.S.); (Y.L.)
| | - Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK;
| | - Xu Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ken H. Young
- Department of Pathology, Duke University School of Medicine, Durham, NC 27708, USA;
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada Di Fiume 447, 34149 Trieste, Italy;
| | - Yong Li
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Correspondence: (N.S.); (Y.L.)
| |
Collapse
|
217
|
Kim O, Park EY, Klinkebiel DL, Pack SD, Shin YH, Abdullaev Z, Emerson RE, Coffey DM, Kwon SY, Creighton CJ, Kwon S, Chang EC, Chiang T, Yatsenko AN, Chien J, Cheon DJ, Yang-Hartwich Y, Nakshatri H, Nephew KP, Behringer RR, Fernández FM, Cho CH, Vanderhyden B, Drapkin R, Bast RC, Miller KD, Karpf AR, Kim J. In vivo modeling of metastatic human high-grade serous ovarian cancer in mice. PLoS Genet 2020; 16:e1008808. [PMID: 32497036 PMCID: PMC7297383 DOI: 10.1371/journal.pgen.1008808] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/16/2020] [Accepted: 04/28/2020] [Indexed: 01/03/2023] Open
Abstract
Metastasis is responsible for 90% of human cancer mortality, yet it remains a challenge to model human cancer metastasis in vivo. Here we describe mouse models of high-grade serous ovarian cancer, also known as high-grade serous carcinoma (HGSC), the most common and deadliest human ovarian cancer type. Mice genetically engineered to harbor Dicer1 and Pten inactivation and mutant p53 robustly replicate the peritoneal metastases of human HGSC with complete penetrance. Arising from the fallopian tube, tumors spread to the ovary and metastasize throughout the pelvic and peritoneal cavities, invariably inducing hemorrhagic ascites. Widespread and abundant peritoneal metastases ultimately cause mouse deaths (100%). Besides the phenotypic and histopathological similarities, mouse HGSCs also display marked chromosomal instability, impaired DNA repair, and chemosensitivity. Faithfully recapitulating the clinical metastases as well as molecular and genomic features of human HGSC, this murine model will be valuable for elucidating the mechanisms underlying the development and progression of metastatic ovarian cancer and also for evaluating potential therapies. Rarely does an experimental model fully replicate the clinical metastases of a human malignancy. Faithfully representing the clinical metastases of human high-grade serous ovarian cancer with complete penetrance, coupled with histopathological, molecular, and genomic similarities, these mouse models, particularly one harboring mutant p53, will be vital to elucidating the underlying pathogenesis of human ovarian cancer. In-depth understanding of the development and progression of ovarian cancer is crucial to medical advances in the early detection, effective treatment, and prevention of ovarian cancer. Also, these robust mouse models, as well as cell lines established from the mouse primary and metastatic tumors, will serve as useful preclinical tools to evaluate therapeutic target genes and new therapies in ovarian cancer.
Collapse
Affiliation(s)
- Olga Kim
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Eun Young Park
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - David L. Klinkebiel
- Department of Biochemistry and Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Svetlana D. Pack
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yong-Hyun Shin
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert E. Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Donna M. Coffey
- Department of Pathology and Genomic Medicine, Houston Methodist and Weill Cornell Medical College, Houston, Texas, United States of America
| | - Sun Young Kwon
- Department of Pathology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Chad J. Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sanghoon Kwon
- Research and Development Center, Bioway Inc, Seoul, Republic of Korea
| | - Edmund C. Chang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Theodore Chiang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander N. Yatsenko
- Department of Obstetrics, Gynecology & Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jeremy Chien
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Dong-Joo Cheon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States of America
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Kenneth P. Nephew
- Medical Sciences Program, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Bloomington, Indiana, United States of America
| | - Richard R. Behringer
- Departments of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Facundo M. Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Chi-Heum Cho
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Barbara Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robert C. Bast
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Kathy D. Miller
- Department of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine Indianapolis, Indiana, United States of America
| | - Adam R. Karpf
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jaeyeon Kim
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
218
|
Identification of a Seven-lncRNA Immune Risk Signature and Construction of a Predictive Nomogram for Lung Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7929132. [PMID: 32596372 PMCID: PMC7273488 DOI: 10.1155/2020/7929132] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022]
Abstract
Background The incidence of lung cancer is the highest of all cancers, and it has the highest death rate. Lung adenocarcinoma (LUAD) is a major type of lung cancer. This study is aimed at identifying the prognostic value of immune-related long noncoding RNAs (lncRNAs) in LUAD. Materials and Methods Gene expression profiles and the corresponding clinicopathological features of LUAD patients were obtained from The Cancer Genome Atlas (TCGA). The least absolute shrinkage and selection operator (LASSO) Cox regression algorithm was performed on the prognostic immune-related lncRNAs to calculate the risk scores, and a risk signature was constructed. Survival analysis was performed to assess the prognostic value of the risk signature. A nomogram was also constructed based on the clinicopathological features and risk signature. Results A total of 437 LUAD patients with gene expression data and clinicopathological features were obtained in this study, which was considered the combination set. They were randomly and equally divided into a training set and a validation set. Seven immune-related lncRNAs (AC092794.1, AL034397.3, AC069023.1, AP000695.1, AC091057.1, HLA-DQB1-AS1, and HSPC324) were identified and used to construct a risk signature. The patients were divided into the low- and high-risk groups based on the median risk score of -0.04074. Survival analysis suggested that patients in the low-risk group had a longer overall survival (OS) than those in the high-risk group (p = 1.478e − 02). A nomogram was built that could predict the 1-, 3-, and 5-year survival rates of LUAD patients (C-index of the nomogram was 0.755, and the AUCs for the 1-, 3-, and 5-year survivals were 0.826, 0.719, and 0.724, respectively). The validation and combination sets confirmed these results. Conclusion Our study identified seven novel immune-related lncRNAs and generated a risk signature, as well as a nomogram, that could predict the prognosis of LUAD patients.
Collapse
|
219
|
Nakayama M, Hong CP, Oshima H, Sakai E, Kim SJ, Oshima M. Loss of wild-type p53 promotes mutant p53-driven metastasis through acquisition of survival and tumor-initiating properties. Nat Commun 2020; 11:2333. [PMID: 32393735 PMCID: PMC7214469 DOI: 10.1038/s41467-020-16245-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 04/23/2020] [Indexed: 02/07/2023] Open
Abstract
Missense-type mutant p53 plays a tumor-promoting role through gain-of-function (GOF) mechanism. In addition, the loss of wild-type TP53 through loss of heterozygosity (LOH) is widely found in cancer cells. However, malignant progression induced by cooperation of TP53 GOF mutation and LOH remains poorly understood. Here, we show that mouse intestinal tumors carrying Trp53 GOF mutation with LOH (AKTPM/LOH) are enriched in metastatic lesions when heterozygous Trp53 mutant cells (AKTP+/M) are transplanted. We show that Trp53 LOH is required for dormant cell survival and clonal expansion of cancer cells. Moreover, AKTPM/LOH cells show an increased in vivo tumor-initiating ability compared with AKTPNull and AKTP+/M cells. RNAseq analyses reveal that inflammatory and growth factor/MAPK pathways are specifically activated in AKTPM/LOH cells, while the stem cell signature is upregulated in both AKTPM/LOH and AKTPNull cells. These results indicate that TP53/Trp53 LOH promotes TP53/Trp53 GOF mutation-driven metastasis through the activation of distinct pathway combination.
Collapse
Affiliation(s)
- Mizuho Nakayama
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan.,WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Chang Pyo Hong
- Theragen Etex Bio Institute, Suwon, 16229, Republic of Korea
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan.,WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Eri Sakai
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Seong-Jin Kim
- Theragen Etex Bio Institute, Suwon, 16229, Republic of Korea.,Precision Medicine Research Center, Advanced Institute of Convergence Technology and Department of Transdisciplinary Studies, Seoul National University, Suwon, 16229, Republic of Korea
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan. .,WPI Nano Life Science Institute, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
220
|
Gini B, Thomas N, Blakely CM. Impact of concurrent genomic alterations in epidermal growth factor receptor ( EGFR)-mutated lung cancer. J Thorac Dis 2020; 12:2883-2895. [PMID: 32642201 PMCID: PMC7330397 DOI: 10.21037/jtd.2020.03.78] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Comprehensive characterization of the genomic landscape of epidermal growth factor receptor (EGFR)-mutated lung cancers have identified patterns of secondary mutations beyond the primary oncogenic EGFR mutation. These include concurrent pathogenic alterations affecting p53 (60–65%), RTKs (5–10%), PIK3CA/KRAS (3–23%), Wnt (5–10%), and cell cycle (7–25%) pathways as well as transcription factors such as MYC and NKX2-1 (10–15%). The majority of these co-occurring alterations were detected or enriched in samples collected from patients at resistance to tyrosine kinase inhibitor (TKI) treatment, indicating a potential functional role in driving resistance to therapy. Of note, these co-occurring tumor genomic alterations are not necessarily mutually exclusive, and evidence suggests that multiple clonal and sub-clonal cancer cell populations can co-exist and contribute to EGFR TKI resistance. Computational tools aimed to classify, track and predict the evolution of cancer clonal populations during therapy are being investigated in pre-clinical models to guide the selection of combination therapy switching strategies that may delay the development of treatment resistance. Here we review the most frequently identified tumor genomic alterations that co-occur with mutated EGFR and the evidence that these alterations effect responsiveness to EGFR TKI treatment.
Collapse
Affiliation(s)
- Beatrice Gini
- Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Nicholas Thomas
- Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| |
Collapse
|
221
|
Drokow EK, Chen Y, Waqas Ahmed HA, Oppong TB, Akpabla GS, Pei Y, Kumah MA, Neku EA, Sun K. The relationship between leukemia and TP53 gene codon Arg72Pro polymorphism: analysis in a multi-ethnic population. Future Oncol 2020; 16:923-937. [PMID: 32301350 DOI: 10.2217/fon-2019-0792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Many studies have analyzed the relationship between Arg72Pro polymorphism of TP53 and leukemia; nevertheless, the findings continue to be indeterminate. We, therefore, performed an updated meta-analysis in multi-ethnic groups using specialized software for genome-wide association studies meta-analysis. Materials & methods: PubMed, EMBASE and Google Scholar were searched up to October 2018. An odds ratio (OR) with the corresponding 95% CI was used to evaluate the strength in the association. Results: This meta-analysis included 16 studies with 2337 cases and 9494 controls. In the overall population, significant relationship between Arg72Pro polymorphism of TP53 and leukemia susceptibility was found in two genetic models (recessive model: OR = 1.276, 95% CI = 1.102-1.476; p = 0.01; overdominant model: OR = 0.891, 95% CI = 0.802-0.988; p = 0.03). In stratified studies with ethnicity, a significant association was found in five ethnic groups, including Chinese, Americans, Africans, Japanese and Indians. Conclusion: We demonstrated that an association exist between leukemia risk and TP53 gene codon Arg72Pro polymorphism in the recessive and overdominant genetic models. Also, our findings show that the TP53 Arg72Pro polymorphism may influence leukemia development in different populations.
Collapse
Affiliation(s)
- Emmanuel Kwateng Drokow
- Department of Radiation Oncology, Zhengzhou University People's Hospital & Henan Provincial People's Hospital Henan, 450003 Zhengzhou, PR China
| | - Yuqing Chen
- Department of Haematology, Zhengzhou University People's Hospital & Henan Provincial People's Hospital Henan, 450003 Zhengzhou, PR China
| | - Hafiz Abdul Waqas Ahmed
- Department of Haematology, Zhengzhou University People's Hospital & Henan Provincial People's Hospital Henan, 450003 Zhengzhou, PR China
| | - Timothy Bonney Oppong
- Department of Epidemiology & Biostatistics, College of Public Health, Zhengzhou University, 450001 Zhengzhou, Henan, PR China
| | - Gloria Selorm Akpabla
- Department of Internal Medicine, Tianjin Medical University, 300070 Tianjin, PR China
| | - Yanru Pei
- Department of Haematology, Zhengzhou University People's Hospital & Henan Provincial People's Hospital Henan, 450003 Zhengzhou, PR China
| | - Maame Awoyoe Kumah
- Department of Internal Medicine, University of Ghana Medical School, KB 77 Korle Bu, Accra, Ghana
| | - Enyonam Adjoa Neku
- Department of Pharmacy, Zhengzhou University, 450001 Zhengzhou, Henan, PR China
| | - Kai Sun
- Department of Haematology, Zhengzhou University People's Hospital & Henan Provincial People's Hospital Henan, 450003 Zhengzhou, PR China
| |
Collapse
|
222
|
Lu C, Dong XR, Zhao J, Zhang XC, Chen HJ, Zhou Q, Tu HY, Ai XH, Chen XF, An GL, Bai J, Shan JL, Wang YN, Yang SY, Liu X, Zhuang W, Wu HT, Zhu B, Xia XF, Chen RR, Gu DJ, Xu HM, Wu YL, Yang JJ. Association of genetic and immuno-characteristics with clinical outcomes in patients with RET-rearranged non-small cell lung cancer: a retrospective multicenter study. J Hematol Oncol 2020; 13:37. [PMID: 32295619 PMCID: PMC7160902 DOI: 10.1186/s13045-020-00866-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/27/2020] [Indexed: 01/05/2023] Open
Abstract
Background Rearranged during transfection (RET) has been proven to be a tumorigenic target in non-small cell lung cancers (NSCLCs). In RET-rearranged NSCLCs, molecular features and their impact on prognosis were not well illustrated, and the activity of mainstay therapeutics has not currently been well compared. Methods Patients diagnosed with NSCLCs with RET rearrangements were analyzed for concomitant mutations, tumor mutation burden (TMB), PD-L1 expression, T cell receptor repertoire and clinical outcomes with chemotherapy, immune checkpoint inhibitors (ICIs), and multikinase inhibitors (MKIs). Results Among 129 patients with RET-rearranged NSCLC who were analyzed, 41.1% (53/129) had co-occurring genetic alterations by next-generation sequencing, and concomitant TP53 mutation appeared most frequently (20/53, 37.7%). Patients with concurrent TP53 mutation (n = 15) had shorter overall survival than those without (n = 30; median, 18.4 months [95% CI, 8.6–39.1] vs 24.8 months [95% CI, 11.7–52.8]; P < 0.05). Patients with lower peripheral blood TCR diversity (n = 5) had superior overall survival compared with those with higher diversity (n = 6; median, 18.4 months [95% CI, 16.9–19.9] vs 4.8 months [95% CI, 4.5–5.3]; P = 0.035). An association with overall survival was not observed for PD-L1 expression nor for tumor mutation burden level. Median progression-free survival was not significantly different across chemotherapy, ICIs, and MKIs (median, 3.5 vs 2.5 vs 3.8 months). For patients treated with ICIs, the disease control rate was 60% (6/10) and the objective response rate was 20% (2/10). Conclusions RET-rearranged lung cancers can be heterogeneous in terms of concomitant genetic alterations. Patients with concurrent TP53 mutation or high peripheral blood TCR repertoire diversity have relatively inferior overall survival in this series. Outcomes with traditional systemic therapies in general are suboptimal.
Collapse
Affiliation(s)
- Chang Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiao-Rong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hua-Jun Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xing-Hao Ai
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Feng Chen
- Oncology Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gai-Li An
- Department of Clinical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jun Bai
- Department of Clinical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jin-Lu Shan
- Daping Hospital, Army medical center of PLA, Chongqing, China
| | - Yi-Na Wang
- Department of Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Shuan-Ying Yang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiang Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Wu Zhuang
- Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Hui-Ta Wu
- Department of Oncology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | | | | | - De-Jian Gu
- Geneplus-Beijing Institute, Beijing, China
| | - Hua-Min Xu
- Geneplus-Beijing Institute, Beijing, China
| | - Yi-Long Wu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China. .,Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Jin-Ji Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China. .,Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
223
|
Canale M, Petracci E, Delmonte A, Bronte G, Chiadini E, Ludovini V, Dubini A, Papi M, Baglivo S, De Luigi N, Verlicchi A, Chiari R, Landi L, Metro G, Burgio MA, Crinò L, Ulivi P. Concomitant TP53 Mutation Confers Worse Prognosis in EGFR-Mutated Non-Small Cell Lung Cancer Patients Treated with TKIs. J Clin Med 2020; 9:E1047. [PMID: 32272775 PMCID: PMC7230306 DOI: 10.3390/jcm9041047] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the primary cause of cancer-related deaths worldwide. Epidermal Growth Factor Receptor (EGFR)-mutated patients usually benefit from TKIs treatment, but a significant portion show unresponsiveness due to primary resistance mechanisms. We investigated the role of TP53 mutations in predicting survival and response to EGFR-TKIs in EGFR-mutated NSCLC patients, to confirm, on an independent case series, our previous results. METHODS An independent retrospective cohort study was conducted, on a case series of 136 EGFR-mutated NSCLC patients receiving first or second generation TKIs as a first line therapy, and a smaller fraction of patients who acquired the T790M resistance mutation and were treated with third generation TKIs in the second or further line of treatment. TP53 mutations were evaluated in relation to disease control rate (DCR), objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) of the patients. RESULTS Forty-two patients (30.9%) showed a TP53 mutation. Considered together, TP53 mutations had no significant impact on time-to-event endpoints. Considering the different TP53 mutations separately, exon 8 mutations confirmed their negative effect on PFS (HR 3.16, 95% 1.59-6.28, p = 0.001). In patients who developed the T790M resistance mutation, treated with third generation TKIs, the TP53 exon 8 mutations predicted worse PFS (even though not statistically significant), and OS (HR 4.86, 95% CI: 1.25-18.90, p = 0.023). CONCLUSIONS TP53 exon 8 mutations confirmed their negative prognostic impact in patients treated with first and second generation TKIs and demonstrated a role in affecting clinical outcome in patients treated with third generation TKIs.
Collapse
Affiliation(s)
- Matteo Canale
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (M.C.); (E.C.)
| | - Elisabetta Petracci
- Biostatistics and Clinical Trials Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Angelo Delmonte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.D.); (G.B.); (A.V.); (M.A.B.); (L.C.)
| | - Giuseppe Bronte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.D.); (G.B.); (A.V.); (M.A.B.); (L.C.)
| | - Elisa Chiadini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (M.C.); (E.C.)
| | - Vienna Ludovini
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy; (V.L.); (S.B.); (G.M.)
| | - Alessandra Dubini
- Department of Pathology, Morgagni-Pierantoni Hospital, 47121 Forlì, Italy;
| | - Maximilian Papi
- Department of Medical Oncology, Per gli Infermi Hospital, Rimini 47923, Italy;
| | - Sara Baglivo
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy; (V.L.); (S.B.); (G.M.)
| | - Nicoletta De Luigi
- UOS Oncology, Istituto per la Sicurezza Sociale, State Hospital, Cailungo 47893, San Marino, Italy;
| | - Alberto Verlicchi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.D.); (G.B.); (A.V.); (M.A.B.); (L.C.)
| | - Rita Chiari
- Department of Medical Oncology, Ospedali Riuniti Padova Sud “M. Teresa di Calcutta”, ULSS6 Euganea, 35131 Padova, Italy;
| | - Lorenza Landi
- Department of Medical Oncology, S. Maria delle Croci Hospital, 48121 Ravenna, Italy;
| | - Giulio Metro
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy; (V.L.); (S.B.); (G.M.)
| | - Marco Angelo Burgio
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.D.); (G.B.); (A.V.); (M.A.B.); (L.C.)
| | - Lucio Crinò
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (A.D.); (G.B.); (A.V.); (M.A.B.); (L.C.)
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (M.C.); (E.C.)
| |
Collapse
|
224
|
Boettcher S, Miller PG, Sharma R, McConkey M, Leventhal M, Krivtsov AV, Giacomelli AO, Wong W, Kim J, Chao S, Kurppa KJ, Yang X, Milenkowic K, Piccioni F, Root DE, Rücker FG, Flamand Y, Neuberg D, Lindsley RC, Jänne PA, Hahn WC, Jacks T, Döhner H, Armstrong SA, Ebert BL. A dominant-negative effect drives selection of TP53 missense mutations in myeloid malignancies. Science 2020; 365:599-604. [PMID: 31395785 DOI: 10.1126/science.aax3649] [Citation(s) in RCA: 268] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
TP53, which encodes the tumor suppressor p53, is the most frequently mutated gene in human cancer. The selective pressures shaping its mutational spectrum, dominated by missense mutations, are enigmatic, and neomorphic gain-of-function (GOF) activities have been implicated. We used CRISPR-Cas9 to generate isogenic human leukemia cell lines of the most common TP53 missense mutations. Functional, DNA-binding, and transcriptional analyses revealed loss of function but no GOF effects. Comprehensive mutational scanning of p53 single-amino acid variants demonstrated that missense variants in the DNA-binding domain exert a dominant-negative effect (DNE). In mice, the DNE of p53 missense variants confers a selective advantage to hematopoietic cells on DNA damage. Analysis of clinical outcomes in patients with acute myeloid leukemia showed no evidence of GOF for TP53 missense mutations. Thus, a DNE is the primary unit of selection for TP53 missense mutations in myeloid malignancies.
Collapse
Affiliation(s)
- Steffen Boettcher
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter G Miller
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rohan Sharma
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marie McConkey
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew Leventhal
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrei V Krivtsov
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrew O Giacomelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Waihay Wong
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jesi Kim
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sherry Chao
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Department of Biomedical Informatics, Harvard University, Boston, MA 02115, USA
| | - Kari J Kurppa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xiaoping Yang
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Kirsten Milenkowic
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Federica Piccioni
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - David E Root
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Frank G Rücker
- Department of Internal Medicine III, University of Ulm, 89081 Ulm, Germany
| | - Yael Flamand
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - R Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, 89081 Ulm, Germany
| | - Scott A Armstrong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA. .,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.,Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
225
|
Long noncoding RNA AGPG regulates PFKFB3-mediated tumor glycolytic reprogramming. Nat Commun 2020; 11:1507. [PMID: 32198345 PMCID: PMC7083971 DOI: 10.1038/s41467-020-15112-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/20/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor cells often reprogram their metabolism for rapid proliferation. The roles of long noncoding RNAs (lncRNAs) in metabolism remodeling and the underlying mechanisms remain elusive. Through screening, we found that the lncRNA Actin Gamma 1 Pseudogene (AGPG) is required for increased glycolysis activity and cell proliferation in esophageal squamous cell carcinoma (ESCC). Mechanistically, AGPG binds to and stabilizes 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3). By preventing APC/C-mediated ubiquitination, AGPG protects PFKFB3 from proteasomal degradation, leading to the accumulation of PFKFB3 in cancer cells, which subsequently activates glycolytic flux and promotes cell cycle progression. AGPG is also a transcriptional target of p53; loss or mutation of TP53 triggers the marked upregulation of AGPG. Notably, inhibiting AGPG dramatically impaired tumor growth in patient-derived xenograft (PDX) models. Clinically, AGPG is highly expressed in many cancers, and high AGPG expression levels are correlated with poor prognosis, suggesting that AGPG is a potential biomarker and cancer therapeutic target. PFKFB3 enhances glycolysis to promote cancer cell proliferation. Here, the authors identify a long noncoding RNA in esophageal squamous cell carcinoma, AGPG, which interacts with PFKFB3 and promotes its stability, leading to increased glycolysis and proliferation.
Collapse
|
226
|
Di Agostino S. The Impact of Mutant p53 in the Non-Coding RNA World. Biomolecules 2020; 10:biom10030472. [PMID: 32204575 PMCID: PMC7175150 DOI: 10.3390/biom10030472] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), micro RNAs (miRNAs), and extracellular RNAs (exRNAs) are new groups of RNAs with regulation activities that have low or no protein-coding ability. Emerging evidence suggests that deregulated expression of these non-coding RNAs is associated with the induction and progression of diverse tumors throughout epigenetic, transcriptional, and post-transcriptional modifications. A consistent number of non-coding RNAs (ncRNAs) has been shown to be regulated by p53, the most important tumor suppressor of the cells frequently mutated in human cancer. It has been shown that some mutant p53 proteins are associated with the loss of tumor suppressor activity and the acquisition of new oncogenic functions named gain-of-function activities. In this review, we highlight recent lines of evidence suggesting that mutant p53 is involved in the expression of specific ncRNAs to gain oncogenic functions through the creation of a complex network of pathways that influence each other.
Collapse
|
227
|
Barta JA, Pauley K, Kossenkov AV, McMahon SB. The lung-enriched p53 mutants V157F and R158L/P regulate a gain of function transcriptome in lung cancer. Carcinogenesis 2020; 41:67-77. [PMID: 31067569 PMCID: PMC7316406 DOI: 10.1093/carcin/bgz087] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 04/24/2019] [Accepted: 05/07/2019] [Indexed: 01/03/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the USA, and alterations in the tumor suppressor gene TP53 are the most frequent somatic mutation among all histologic subtypes of lung cancer. Mutations in TP53 frequently result in a protein that exhibits not only loss of tumor suppressor capability but also oncogenic gain-of-function (GOF). The canonical p53 hotspot mutants R175H and R273H, for example, confer upon tumors a metastatic phenotype in murine models of mutant p53. To the best of our knowledge, GOF phenotypes of the less often studied V157, R158 and A159 mutants-which occur with higher frequency in lung cancer compared with other solid tumors-have not been defined. In this study, we aimed to define whether the lung mutants are simply equivalent to full loss of the p53 locus, or whether they additionally acquire the ability to drive new downstream effector pathways. Using a publicly available human lung cancer dataset, we characterized patients with V157, R158 and A159 p53 mutations. In addition, we show here that cell lines with mutant p53-V157F, p53-R158L and p53-R158P exhibit a loss of expression of canonical wild-type p53 target genes. Furthermore, these lung-enriched p53 mutants regulate genes not previously linked to p53 function including PLAU. Paradoxically, mutant p53 represses genes associated with increased cell viability, migration and invasion. These findings collectively represent the first demonstration that lung-enriched p53 mutations at V157 and R158 regulate a novel transcriptome in human lung cancer cells and may confer de novo function.
Collapse
Affiliation(s)
- Julie A Barta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kristen Pauley
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- Bioinformatics Core Facility, The Wistar Institute, Philadelphia, PA, USA
| | - Steven B McMahon
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
228
|
Dutta S, Pregartner G, Rücker FG, Heitzer E, Zebisch A, Bullinger L, Berghold A, Döhner K, Sill H. Functional Classification of TP53 Mutations in Acute Myeloid Leukemia. Cancers (Basel) 2020; 12:E637. [PMID: 32164171 PMCID: PMC7139772 DOI: 10.3390/cancers12030637] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 12/17/2022] Open
Abstract
Mutations of the TP53 gene occur in a subset of patients with acute myeloid leukemia (AML) and confer an exceedingly adverse prognosis. However, whether different types of TP53 mutations exert a uniformly poor outcome has not been investigated yet. Here, we addressed this issue by analyzing data of 1537 patients intensively treated within protocols of the German-Austrian AML study group. We classified TP53 mutations depending on their impact on protein structure and according to the evolutionary action (EAp53) score and the relative fitness score (RFS). In 98/1537 (6.4%) patients, 108 TP53 mutations were detected. While the discrimination depending on the protein structure and the EAp53 score did not show a survival difference, patients with low-risk and high-risk AML-specific RFS showed a different overall survival (OS; median, 12.9 versus 5.5 months, p = 0.017) and event-free survival (EFS; median, 7.3 versus 5.2 months, p = 0.054). In multivariable analyses adjusting for age, gender, white blood cell count, cytogenetic risk, type of AML, and TP53 variant allele frequency, these differences were statistically significant for both OS (HR, 2.14; 95% CI, 1.15-4.0; p = 0.017) and EFS (HR, 1.97; 95% CI, 1.06-3.69; p = 0.033). We conclude that the AML-specific RFS is of prognostic value in patients with TP53-mutated AML and a useful tool for therapeutic decision-making.
Collapse
Affiliation(s)
- Sayantanee Dutta
- Division of Hematology, Medical University of Graz, A-8036 Graz, Austria
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, A-8036 Graz, Austria
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, D-89081 Ulm, Germany
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, A-8010 Graz, Austria
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, A-8036 Graz, Austria
- Otto-Loewi-Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, A-8010 Graz, Austria
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, D-10117 Berlin, Germany
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, A-8036 Graz, Austria
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, D-89081 Ulm, Germany
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, A-8036 Graz, Austria
| |
Collapse
|
229
|
Huang M, Jin J, Zhang F, Wu Y, Xu C, Ying L, Su D. Non-disruptive mutation in TP53 DNA-binding domain is a beneficial factor of esophageal squamous cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:316. [PMID: 32355760 PMCID: PMC7186752 DOI: 10.21037/atm.2020.02.142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background TP53 is frequently altered in esophageal squamous cell carcinoma (ESCC). However, the landscape of TP53 mutation and its effects on patients remain controversial. Methods Somatic mutations of TP53 in 161 patients with resectable ESCC were identified by next-generation sequencing (NGS) and verified by immunohistochemistry (IHC). Patients were stratified into seven TP53 mutations, and depending on the extent of the effect on the encoded protein, it was divided into "disruptive" and "non-disruptive" types. The association of TP53 mutation with clinicopathological properties and disease outcome was investigated. Results TP53 mutations were discovered in 85.7% patients, of which 68.9% carried mutations in the DNA-binding domain (DBD). A total of 47.8% and 37.9% patients had disruptive and non-disruptive TP53 mutations, respectively. Most patients carried only one TP53 mutation, but 15.5% had double mutations. TP53 mutations were dominant in exons 5 to 8. Missense mutation was the most frequent (97/163, 59.5%), and the top five frequently occurring variations included R273X, Y220X, H193, H179X, and R175H. Multivariable analysis revealed non-disruptive mutation in TP53 DBD as the independent prognostic predictor for progression-free survival (PFS) and overall survival (OS). The expression of p53 positively correlated with non-disruptive mutation in DBD. Patients with high p53 protein expression showed better outcomes. Conclusions Non-disruptive mutation in TP53 DBD serves as an independent beneficial prognostic factor of prolonged survival in resectable ESCC.
Collapse
Affiliation(s)
- Minran Huang
- Department of Oncology, The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China.,Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jiaoyue Jin
- Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Fanrong Zhang
- Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Breast Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yingxue Wu
- Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Chenyang Xu
- Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Lisha Ying
- Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Cancer Hospital of University of Chinese Academy of Sciences, 310022, China
| | - Dan Su
- Institute of Cancer and Basic Medical (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
230
|
Analysis of the radio-pathological factors of triple negative breast cancer and determination of risk profiles. RADIOLOGIA 2020; 62:365-375. [PMID: 32093905 DOI: 10.1016/j.rx.2020.01.001] [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: 04/18/2019] [Revised: 12/14/2019] [Accepted: 01/07/2020] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Triple-negative tumors are the most aggressive type of breast cancer. We aimed to analyze the main radiologic and histopathologic factors of these tumors to create a risk profile. MATERIALS AND METHODS We analyzed data from 140 patients diagnosed with triple-negative breast cancer between January 2007 and December 2016, with follow-up through April 2018. We analyzed the following variables in the breast MRI done for staging: size, necrosis, associated findings, adenopathies, and perfusion and diffusion parameters. We analyzed the following variables in histopathologic studies of biopsy specimens: histological type, Scarf-Bloom, Ki67, and p53 in the infiltrating component as well as in the in situ component. We analyzed the following variables in histopathologic studies of positive lymph nodes and surgical specimens: size, lymphovascular/perineural invasion, and microglandular adenosis. We analyzed the relation between the radiologic and histopathologic factors and recurrence and disease-free survival. RESULTS MRI tumor size>25mm, non-nodular enhancement, breast edema, areola-nipple complex retraction, and lymph-node involvement were associated with recurrence and lower disease-free survival. Invasive lobular carcinoma, postsurgical size>20mm, and p53<15% were also associated with recurrence and lower disease-free survival. Histologically positive lymph nodes were associated with a greater percentage of recurrence and lymphovascular invasion and with lower disease-free survival. The multivariate analysis found that the variables MRI size>25mm, non-nodular enhancement, adenopathies on MRI, and p53 expression <15% were independent predictors of lower disease-free survival. CONCLUSIONS In triple-negative breast tumors, factors associated with lower disease-free survival are non-nodular enhancement, size>25mm, and adenopathies on MRI, and p53 expression <15% on histopathologic study.
Collapse
|
231
|
Pavlakis E, Stiewe T. p53's Extended Reach: The Mutant p53 Secretome. Biomolecules 2020; 10:biom10020307. [PMID: 32075247 PMCID: PMC7072272 DOI: 10.3390/biom10020307] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 02/08/2023] Open
Abstract
p53 suppresses tumorigenesis by activating a plethora of effector pathways. While most of these operate primarily inside of cells to limit proliferation and survival of incipient cancer cells, many extend to the extracellular space. In particular, p53 controls expression and secretion of numerous extracellular factors that are either soluble or contained within extracellular vesicles such as exosomes. As part of the cellular secretome, they execute key roles in cell-cell communication and extracellular matrix remodeling. Mutations in the p53-encoding TP53 gene are the most frequent genetic alterations in cancer cells, and therefore, have profound impact on the composition of the tumor cell secretome. In this review, we discuss how the loss or dominant-negative inhibition of wild-type p53 in concert with a gain of neomorphic properties observed for many mutant p53 proteins, shapes a tumor cell secretome that creates a supportive microenvironment at the primary tumor site and primes niches in distant organs for future metastatic colonization.
Collapse
|
232
|
Ramesh PS, Devegowda D, Singh A, Thimmulappa RK. NRF2, p53, and p16: Predictive biomarkers to stratify human papillomavirus associated head and neck cancer patients for de-escalation of cancer therapy. Crit Rev Oncol Hematol 2020; 148:102885. [PMID: 32062315 DOI: 10.1016/j.critrevonc.2020.102885] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/09/2020] [Accepted: 01/25/2020] [Indexed: 01/18/2023] Open
Abstract
Patients with HPV associated (HPV+ve) head and neck squamous cell carcinoma (HNSCC), particularly oropharyngeal cancer, show better treatment response, higher survival rates, and lower risks of recurrence as compared to HPV-ve HNSCC patients. Despite increased sensitivity to treatment modality, HPV+ve HNSCC patients are subjected to the same intensive anti-cancer therapy as HPV-ve HNSCC patients and thus subjecting them to unwarranted long-term toxicity. To identify predictive biomarkers for risk-stratification, we have analyzed the mutational spectrum, and the evidence suggests that gain-of-function mutations in the NRF2 pathway are highly prevalent in HPV-ve HNSCC. At the same time, it is rare in HPV+ve HNSCC tumors. We have reviewed the importance of gain-of-NRF2 function and loss of p53 in the prognosis of HNSCC patients and discussed a predictive scoring system using a combination of HPV status (p16), NRF2 pathway and p53 to stratify HPV+ve HNSCC into good versus poor responders, which could immensely help in guiding future de-escalation treatment approaches in patients with HPV+ve HNSCC.
Collapse
Affiliation(s)
- Pushkal S Ramesh
- Centre of Excellence in Molecular Biology & Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, India
| | - Devananda Devegowda
- Centre of Excellence in Molecular Biology & Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, India
| | - Anju Singh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, United States
| | - Rajesh K Thimmulappa
- Centre of Excellence in Molecular Biology & Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, India.
| |
Collapse
|
233
|
Zeng F, Heng J, Guo X, Wang Y, Wu W, Tang L, Chen M, Wang S, Deng H, Wang J. The novel TP53 3'-end methylation pattern associated with its expression would be a potential biomarker for breast cancer detection. Breast Cancer Res Treat 2020; 180:237-245. [PMID: 31983017 DOI: 10.1007/s10549-020-05536-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Deficiency or silence of TP53 is an early event in breast tumorigenesis. Aberrant methylation and mutation in regulatory regions were considered as crucial regulators of gene expression. METHODS Using multiplex-PCR and next-generation sequencing technology, we analyzed TP53 mutation spectrum in its promoter region. Using PCR target sequence enrichment and next-generation bisulfite sequencing technology, we analyzed the methylation profile of the promoter and 3'-end regions of TP53 gene in paired breast tumor and normal tissues from 120 breast cancer patients. The expression of TP53 and the flanking gene ATP1B2 was explored with qPCR method in the same cohort. RESULTS No promoter mutation of TP53 gene was found in the cohort of the 120 breast cancer patients. The 3'-end of TP53 gene was hyper-methylated (average 78.71%) compared with the promoter region (average less than 1%) in breast tumor tissues. TP53 was significantly lower expressed (P = 1.68E-15) and hyper-methylated in 3'-end (P = 1.82E-18) in tumor. Negative cis correlation was found between the TP53 expression and its 3'-end methylation (P = 9.02E-8, R = 0.337). TP53 expression was significantly associated with PR status (P = 0.0128), Ki67 level (P = 0.0091), and breast cancer subtypes (P = 0.0109). TP53 3'-end methylation and expression showed a good performance in discriminating breast cancer and normal tissues with an AUC of 0.930. CONCLUSIONS The 3'-end methylation of TP53 might be a crucial regulator for its expression in breast cancer, suggesting that TP53 3'-end hyper-methylation associated with its lower expression could be a potential biomarker for breast cancer diagnosis.
Collapse
Affiliation(s)
- Feng Zeng
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jianfu Heng
- Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
- School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Xinwu Guo
- Sansure Biotech Inc., Changsha, 410205, Hunan, China
| | - Yue Wang
- School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Wenhan Wu
- School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Lili Tang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Min Chen
- Sansure Biotech Inc., Changsha, 410205, Hunan, China
| | - Shouman Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongyu Deng
- Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, China
| | - Jun Wang
- School of Life Sciences, Central South University, Changsha, 410013, Hunan, China.
- Sansure Biotech Inc., Changsha, 410205, Hunan, China.
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
234
|
Tuna M, Ju Z, Yoshihara K, Amos CI, Tanyi JL, Mills GB. Clinical relevance of TP53 hotspot mutations in high-grade serous ovarian cancers. Br J Cancer 2020; 122:405-412. [PMID: 31780779 PMCID: PMC7000721 DOI: 10.1038/s41416-019-0654-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/28/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mutation of TP53 is the most frequent genetic alteration in high-grade serous ovarian cancer (HGSOC). The impact of hotspot mutations of TP53 and protein levels on patient outcomes in HGSOC has not been fully elucidated. METHODS The study population (n = 791) comprised of HGSOC samples with TP53 mutation from TCGA and other publicly available data. Univariate and multivariate cox proportional hazards regression analyses were used to select variables that were correlated with patient survival. RESULTS We assessed the effects of TP53 mutations based on type and individual hotspot mutations on patient outcomes in HGSOC. Only hotspot mutations were associated with outcomes. Three hotspot mutations: G266, Y163C, and R282, in aggregate were associated with a worsened overall and recurrence-free survival compared with other hotspot mutations (p < 0.0001 and p = 0.001), other non-hotspot missense mutations (p < 0.0001 and p = 0.008), truncated mutations (p < 0.0001 and p = 0.001), and all other mutations (p < 0.0001 and p = 0.001). Specific hotspot mutations were associated with different protein expression patterns consistent with different functions. CONCLUSIONS This study provides evidence that individual TP53 hotspot mutations have different impact on HGSOC patient outcomes and potentially TP53 function. Thus the status of particular TP53 aberrations could influence response to therapy and selection of therapeutic agents.
Collapse
Affiliation(s)
- Musaffe Tuna
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Precision Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University, Niigata, Japan
| | | | - Janos L Tanyi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gordon B Mills
- Precision Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
235
|
TP53/miR-34a-associated signaling targets SERPINE1 expression in human pancreatic cancer. Aging (Albany NY) 2020; 12:2777-2797. [PMID: 31986125 PMCID: PMC7041729 DOI: 10.18632/aging.102776] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/12/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease of aging. The TP53 gene product regulates cell growth, aging, and cancer. To determine the important targets of TP53 in PDAC, we examined the expression of 440 proteins on a reverse phase protein array (RPPA) in PDAC-derived MIA-PaCa-2 cells which either had WT-TP53 or lacked WT-TP53. MIA-PaCa-2 cells have a TP53 mutation as well as mutant KRAS and represent a good in vitro model to study PDAC. RPPA analysis demonstrated expression of tumor promoting proteins in cells that lacked WT-TP53; and this feature could be reversed significantly when the cells were transfected with vector encoding WT-TP53 or treated with berberine or a modified berberine (BBR). Expression of miR-34a-associated signaling was elevated in cells expressing WT-TP53 compared to cells expressing mTP53. Results from in vivo studies using human PDAC specimens confirmed the in vitro results as the expression of miR-34a and associated signaling was significantly decreased in PDAC specimens compared to non-cancerous tissues. This study determined SERPINE1 as a miR-34a target with relevance to the biology of PDAC. Thus, we have identified a key target (SERPINE1) of the TP53/miR-34a axis that may serve as a potential biomarker for early detection of pancreatic cancer.
Collapse
|
236
|
Prognosis, Biology, and Targeting of TP53 Dysregulation in Multiple Myeloma. Cells 2020; 9:cells9020287. [PMID: 31991614 PMCID: PMC7072230 DOI: 10.3390/cells9020287] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/10/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological cancer and is characterized by genetic features including translocations, chromosomal copy number aberrations, and mutations in key oncogene and tumor suppressor genes. Dysregulation of the tumor suppressor TP53 is important in the pathogenesis of many cancers, including MM. In newly-diagnosed MM patients, TP53 dysregulation occurs in three subsets: monoallelic deletion as part of deletion of chromosome 17p (del17p) (~8%), monoallelic mutations (~6%), and biallelic inactivation (~4%). Del17p is an established high-risk feature in MM and is included in current disease staging criteria. Biallelic inactivation and mutation have also been reported in MM patients but are not yet included in disease staging criteria for high-risk disease. Emerging clinical and genomics data suggest that the biology of high-risk disease is complex, and so far, traditional drug development efforts to target dysregulated TP53 have not been successful. Here we review the TP53 dysregulation literature in cancer and in MM, including the three segments of TP53 dysregulation observed in MM patients. We propose a reverse translational approach to identify novel targets and disease drivers from TP53 dysregulated patients to address the unmet medical need in this setting.
Collapse
|
237
|
Welponer H, Tsibulak I, Wieser V, Degasper C, Shivalingaiah G, Wenzel S, Sprung S, Marth C, Hackl H, Fiegl H, Zeimet AG. The miR-34 family and its clinical significance in ovarian cancer. J Cancer 2020; 11:1446-1456. [PMID: 32047551 PMCID: PMC6995379 DOI: 10.7150/jca.33831] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor suppressor miR-34 family is transcriptionally induced by p53. Clinical significance of the various miR-34 family members has not been studied in ovarian cancer. In 228 ovarian cancers and in 19 non-neoplastic fallopian tube samples we analysed miR-34 a/b/c expression in relation to clinicopathological characteristics and clinical outcome. We found significantly lower levels of miR-34 a/b/c in ovarian cancers as compared to control-tissues (P=0.002, P<0.001, P<0.001, respectively). Expression of miR-34 b/c revealed an inverse correlation with BRCA1/2 mRNA-expression (BRCA1: miR34 b/c P=0.002 each; BRCA2: miR-34 b/c P<0.001 each), the same was true for miR-34a and BRCA2 mRNA-expression (P<0.001). The miR-34 family expression was found to be significantly lower in type 2 in comparison to type 1 cancers (P<0.001) and in TP53-mutated compared with TP53-wild-type ovarian cancers (P<0.001, P=0.002, P=0.004, respectively). When low grade serous ovarian cancers were compared with high grade serous cancers the respective miR-34 a/b/c expression was 2.6-, 40.8- and 32.3-fold higher. The expression of each of the miR-34 family members was revealed to be of independent prognostic relevance regarding progression free survival (PFS); miR-34a: HR 0.6, P=0.033; miR-34b: HR 0.2, P=0.001 and miR-34c: HR 0.3, P=0.002, respectively). For overall survival (OS) independency of the prognostic value was confined to miR-34b (HR 0.4, P=0.016) and miR-34c (HR 0.6, P=0.049). The independency of the prognostic value of our identified thresholds was confirmed for PFS for miR-34c in a publicly available dataset (NCBI Gene Expression Omnibus GSE73582). Our findings suggest that downregulation of miR-34 family is a crucial part in ovarian cancer development. Low miR-34 levels are linked to a worse overall survival and progression free survival and may indicate a more aggressive disease.
Collapse
Affiliation(s)
- Hannah Welponer
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Irina Tsibulak
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Verena Wieser
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Christine Degasper
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Giridhar Shivalingaiah
- Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Sören Wenzel
- Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Susanne Sprung
- Institute of Pathology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Christian Marth
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Hubert Hackl
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Heidelinde Fiegl
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| | - Alain G Zeimet
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Tyrol, 6020, Austria
| |
Collapse
|
238
|
Bang S, Kaur S, Kurokawa M. Regulation of the p53 Family Proteins by the Ubiquitin Proteasomal Pathway. Int J Mol Sci 2019; 21:E261. [PMID: 31905981 PMCID: PMC6981958 DOI: 10.3390/ijms21010261] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/24/2019] [Indexed: 12/25/2022] Open
Abstract
The tumor suppressor p53 and its homologues, p63 and p73, play a pivotal role in the regulation of the DNA damage response, cellular homeostasis, development, aging, and metabolism. A number of mouse studies have shown that a genetic defect in the p53 family could lead to spontaneous tumor development, embryonic lethality, or severe tissue abnormality, indicating that the activity of the p53 family must be tightly regulated to maintain normal cellular functions. While the p53 family members are regulated at the level of gene expression as well as post-translational modification, they are also controlled at the level of protein stability through the ubiquitin proteasomal pathway. Over the last 20 years, many ubiquitin E3 ligases have been discovered that directly promote protein degradation of p53, p63, and p73 in vitro and in vivo. Here, we provide an overview of such E3 ligases and discuss their roles and functions.
Collapse
Affiliation(s)
| | | | - Manabu Kurokawa
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA; (S.B.); (S.K.)
| |
Collapse
|
239
|
p53-induced ARVCF modulates the splicing landscape and supports the tumor suppressive function of p53. Oncogene 2019; 39:2202-2211. [DOI: 10.1038/s41388-019-1133-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 01/09/2023]
|
240
|
Chen S, Wang Q, Yu H, Capitano ML, Vemula S, Nabinger SC, Gao R, Yao C, Kobayashi M, Geng Z, Fahey A, Henley D, Liu SZ, Barajas S, Cai W, Wolf ER, Ramdas B, Cai Z, Gao H, Luo N, Sun Y, Wong TN, Link DC, Liu Y, Boswell HS, Mayo LD, Huang G, Kapur R, Yoder MC, Broxmeyer HE, Gao Z, Liu Y. Mutant p53 drives clonal hematopoiesis through modulating epigenetic pathway. Nat Commun 2019; 10:5649. [PMID: 31827082 PMCID: PMC6906427 DOI: 10.1038/s41467-019-13542-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 11/11/2019] [Indexed: 01/16/2023] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) increases with age and is associated with increased risks of hematological malignancies. While TP53 mutations have been identified in CHIP, the molecular mechanisms by which mutant p53 promotes hematopoietic stem and progenitor cell (HSPC) expansion are largely unknown. Here we discover that mutant p53 confers a competitive advantage to HSPCs following transplantation and promotes HSPC expansion after radiation-induced stress. Mechanistically, mutant p53 interacts with EZH2 and enhances its association with the chromatin, thereby increasing the levels of H3K27me3 in genes regulating HSPC self-renewal and differentiation. Furthermore, genetic and pharmacological inhibition of EZH2 decreases the repopulating potential of p53 mutant HSPCs. Thus, we uncover an epigenetic mechanism by which mutant p53 drives clonal hematopoiesis. Our work will likely establish epigenetic regulator EZH2 as a novel therapeutic target for preventing CHIP progression and treating hematological malignancies with TP53 mutations.
Collapse
Affiliation(s)
- Sisi Chen
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, 46202, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Qiang Wang
- Department of Biochemistry and Molecular Biology, the Cancer Institute, College of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hao Yu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University, Indianapolis, IN, 46202, USA
| | - Sasidhar Vemula
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Sarah C Nabinger
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Rui Gao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Chonghua Yao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Michihiro Kobayashi
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Zhuangzhuang Geng
- Department of Biochemistry and Molecular Biology, the Cancer Institute, College of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA
| | - Aidan Fahey
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Danielle Henley
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Stephen Z Liu
- Department of Medical Genetics, Indiana University, Indianapolis, IN, 46202, USA
| | - Sergio Barajas
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, 46202, USA
| | - Wenjie Cai
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, 46202, USA
| | - Eric R Wolf
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Baskar Ramdas
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Zhigang Cai
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical Genetics, Indiana University, Indianapolis, IN, 46202, USA
| | - Na Luo
- Department of Ophthalmology, Indiana University, Indianapolis, IN, 46202, USA
| | - Yang Sun
- Department of Ophthalmology, Indiana University, Indianapolis, IN, 46202, USA
| | - Terrence N Wong
- Siteman Cancer Center, Washington University, St. Louis, MO, 63110, USA
| | - Daniel C Link
- Siteman Cancer Center, Washington University, St. Louis, MO, 63110, USA
| | - Yunlong Liu
- Department of Medical Genetics, Indiana University, Indianapolis, IN, 46202, USA
| | - H Scott Boswell
- Department of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Lindsey D Mayo
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Gang Huang
- Division of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Mervin C Yoder
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University, Indianapolis, IN, 46202, USA
| | - Zhonghua Gao
- Department of Biochemistry and Molecular Biology, the Cancer Institute, College of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA.
| | - Yan Liu
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
241
|
Gain-of-Function Mutant p53: All the Roads Lead to Tumorigenesis. Int J Mol Sci 2019; 20:ijms20246197. [PMID: 31817996 PMCID: PMC6940767 DOI: 10.3390/ijms20246197] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/25/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023] Open
Abstract
The p53 protein is mutated in about 50% of human cancers. Aside from losing the tumor-suppressive functions of the wild-type form, mutant p53 proteins often acquire inherent, novel oncogenic functions, a phenomenon termed mutant p53 gain-of-function (GOF). A growing body of evidence suggests that these pro-oncogenic functions of mutant p53 proteins are mediated by affecting the transcription of various genes, as well as by protein-protein interactions with transcription factors and other effectors. In the current review, we discuss the various GOF effects of mutant p53, and how it may serve as a central node in a network of genes and proteins, which, altogether, promote the tumorigenic process. Finally, we discuss mechanisms by which "Mother Nature" tries to abrogate the pro-oncogenic functions of mutant p53. Thus, we suggest that targeting mutant p53, via its reactivation to the wild-type form, may serve as a promising therapeutic strategy for many cancers that harbor mutant p53. Not only will this strategy abrogate mutant p53 GOF, but it will also restore WT p53 tumor-suppressive functions.
Collapse
|
242
|
Sborchia M, Keun HC, Phillips DH, Arlt VM. The Impact of p53 on Aristolochic Acid I-Induced Gene Expression In Vivo. Int J Mol Sci 2019; 20:ijms20246155. [PMID: 31817608 PMCID: PMC6940885 DOI: 10.3390/ijms20246155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to aristolochic acid (AA) is linked to kidney disease and urothelial cancer in humans. The major carcinogenic component of the AA plant extract is aristolochic acid I (AAI). The tumour suppressor p53 is frequently mutated in AA-induced tumours. We previously showed that p53 protects from AAI-induced renal proximal tubular injury, but the underlying mechanism(s) involved remain to be further explored. In the present study, we investigated the impact of p53 on AAI-induced gene expression by treating Trp53(+/+), Trp53(+/-), and Trp53(-/-) mice with 3.5 mg/kg body weight (bw) AAI daily for six days. The Clariom™ S Assay microarray was used to elucidate gene expression profiles in mouse kidneys after AAI treatment. Analyses in Qlucore Omics Explorer showed that gene expression in AAI-exposed kidneys is treatment-dependent. However, gene expression profiles did not segregate in a clear-cut manner according to Trp53 genotype, hence further investigations were performed by pathway analysis with MetaCore™. Several pathways were significantly altered to varying degrees for AAI-exposed kidneys. Apoptotic pathways were modulated in Trp53(+/+) kidneys; whereas oncogenic and pro-survival pathways were significantly altered for Trp53(+/-) and Trp53(-/-) kidneys, respectively. Alterations of biological processes by AAI in mouse kidneys could explain the mechanisms by which p53 protects from or p53 loss drives AAI-induced renal injury in vivo.
Collapse
Affiliation(s)
- Mateja Sborchia
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
| | - Hector C. Keun
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London W12 0NN, UK;
| | - David H. Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
| | - Volker M. Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
- Correspondence:
| |
Collapse
|
243
|
Zhang J, Sun W, Kong X, Zhang Y, Yang HJ, Ren C, Jiang Y, Chen M, Chen X. Mutant p53 antagonizes p63/p73-mediated tumor suppression via Notch1. Proc Natl Acad Sci U S A 2019; 116:24259-24267. [PMID: 31712410 PMCID: PMC6883818 DOI: 10.1073/pnas.1913919116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
p53 is the most frequently mutated gene in human cancers and mutant p53 has a gain of function (GOF) that promotes tumor progression and therapeutic resistance. One of the major GOF activities of mutant p53 is to suppress 2 other p53 family proteins, p63 and p73. However, the molecular basis is not fully understood. Here, we examined whether mutant p53 antagonizes p63/p73-mediated tumor suppression in vivo by using mutant p53-R270H knockin and TAp63/p73-deficient mouse models. We found that knockin mutant p53-R270H shortened the life span of p73+/- mice and subjected TAp63+/- or p73+/- mice to T lymphoblastic lymphomas (TLBLs). To unravel the underlying mechanism, we showed that mutant p53 formed a complex with Notch1 intracellular domain (NICD) and antagonized p63/p73-mediated repression of HES1 and ECM1. As a result, HES1 and ECM1 were overexpressed in TAp63+/- ;p53R270H/- and p73+/- ;p53R270H/- TLBLs, suggesting that normal function of HES1 and ECM1 in T cell activation is hyperactivated, leading to lymphomagenesis. Together, our data reveal a previously unappreciated mechanism by which GOF mutant p53 hijacks the p63/p73-regulated transcriptional program via the Notch1 pathway.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616;
| | - Wenqiang Sun
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Hee Jung Yang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Cong Ren
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Yuqian Jiang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616;
| |
Collapse
|
244
|
Kron A, Alidousty C, Scheffler M, Merkelbach-Bruse S, Seidel D, Riedel R, Ihle MA, Michels S, Nogova L, Fassunke J, Heydt C, Kron F, Ueckeroth F, Serke M, Krüger S, Grohe C, Koschel D, Benedikter J, Kaminsky B, Schaaf B, Braess J, Sebastian M, Kambartel KO, Thomas R, Zander T, Schultheis AM, Büttner R, Wolf J. Impact of TP53 mutation status on systemic treatment outcome in ALK-rearranged non-small-cell lung cancer. Ann Oncol 2019; 29:2068-2075. [PMID: 30165392 PMCID: PMC6225899 DOI: 10.1093/annonc/mdy333] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background We analyzed whether co-occurring mutations influence the outcome of systemic therapy in ALK-rearranged non-small-cell lung cancer (NSCLC). Patients and methods ALK-rearranged stage IIIB/IV NSCLC patients were analyzed with next-generation sequencing and fluorescence in situ hybridization analyses on a centralized diagnostic platform. Median progression-free survival (PFS) and overall survival (OS) were determined in the total cohort and in treatment-related sub-cohorts. Cox regression analyses were carried out to exclude confounders. Results Among 216 patients with ALK-rearranged NSCLC, the frequency of pathogenic TP53 mutations was 23.8%, while other co-occurring mutations were rare events. In ALK/TP53 co-mutated patients, median PFS and OS were significantly lower compared with TP53 wildtype patients [PFS 3.9 months (95% CI: 2.4–5.6) versus 10.3 months (95% CI: 8.6–12.0), P < 0.001; OS 15.0 months (95% CI: 5.0–24.9) versus 50.0 months (95% CI: 22.9–77.1), P = 0.002]. This difference was confirmed in all treatment-related subgroups including chemotherapy only [PFS first-line chemotherapy 2.6 months (95% CI: 1.3–4.1) versus 6.2 months (95% CI: 1.8–10.5), P = 0.021; OS 2.0 months (95% CI: 0.0–4.6) versus 9.0 months (95% CI: 6.1–11.9), P = 0.035], crizotinib plus chemotherapy [PFS crizotinib 5.0 months (95% CI: 2.9–7.2) versus 14.0 months (95% CI: 8.0–20.1), P < 0.001; OS 17.0 months (95% CI: 6.7–27.3) versus not reached, P = 0.049] and crizotinib followed by next-generation ALK-inhibitor [PFS next-generation inhibitor 5.4 months (95% CI: 0.1–10.7) versus 9.9 months (95% CI: 6.4–13.5), P = 0.039; OS 7.0 months versus 50.0 months (95% CI: not reached), P = 0.001). Conclusions In ALK-rearranged NSCLC co-occurring TP53 mutations predict an unfavorable outcome of systemic therapy. Our observations encourage future research to understand the underlying molecular mechanisms and to improve treatment outcome of the ALK/TP53 co-mutated subgroup.
Collapse
Affiliation(s)
- A Kron
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - C Alidousty
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - M Scheffler
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - S Merkelbach-Bruse
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - D Seidel
- Center for Integrated Oncology Köln Bonn, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - R Riedel
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - M A Ihle
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - S Michels
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - L Nogova
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - J Fassunke
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - C Heydt
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - F Kron
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - F Ueckeroth
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - M Serke
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Lungenklinik Hemer des Deutschen Gemeinschafts-Diakonieverbandes GmbH, Hemer, Germany
| | - S Krüger
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Florence Nightingale Hospital, Düsseldorf, Germany
| | - C Grohe
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Evangelische Lungenklinik Berlin (Paul Gerhardt Diakonie), Berlin, Germany
| | - D Koschel
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Fachkrankenhaus Coswig, Coswig, Germany
| | - J Benedikter
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Klinikum Bogenhausen, Munich, Germany
| | - B Kaminsky
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Krankenhaus Bethanien, Solingen, Germany
| | - B Schaaf
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Center, Klinikum Dortmund GmbH, Dortmund, Germany
| | - J Braess
- Network Genomic Medicine, Cologne, Germany; Department of Oncology and Hematology, Krankenhaus Barmherzige Brueder, Regensburg, Germany
| | - M Sebastian
- Network Genomic Medicine, Cologne, Germany; Department of Oncology and Hematology, University Hospital Frankfurt (Johannes-Wolfgang Goethe Institute), Frankfurt am Main, Germany
| | - K-O Kambartel
- Network Genomic Medicine, Cologne, Germany; Department of Pneumology, Bethanien Hospital Moers-Lungenzentrum, Moers, Germany
| | - R Thomas
- Network Genomic Medicine, Cologne, Germany; Cologne Center for Genomics, University Hospital of Cologne, Cologne, Germany
| | - T Zander
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany
| | - A M Schultheis
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - R Büttner
- Network Genomic Medicine, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany; Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - J Wolf
- Network Genomic Medicine, Cologne, Germany; Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln Bonn, Cologne, Germany.
| |
Collapse
|
245
|
Klimovich B, Stiewe T, Timofeev O. Inactivation of Mdm2 restores apoptosis proficiency of cooperativity mutant p53 in vivo. Cell Cycle 2019; 19:109-123. [PMID: 31749402 DOI: 10.1080/15384101.2019.1693748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
TP53 mutations are found in 50% of all cancers and mutated TP53 status is considered poor for treatment. However, some TP53 mutations exhibit only partial loss-of-function (LOF), meaning they retain residual transcriptional and non-transcriptional activities that are potentially beneficial for therapy. Earlier we have characterized a knock-in mouse model for the partial LOF mutant Trp53E177R (p53RR). Reduced DNA binding cooperativity of this mutant led to the loss of p53-dependent apoptosis, while p53 functions in cell cycle control, senescence, metabolism, and antioxidant defense remained intact. Concomitantly, tumor suppression was evident but strongly compromised compared to wild-type mice. Here we used the Trp53E177R mouse as a model to investigate whether residual functions of mutant p53 can be engaged to induce cell death, which is considered the most desirable outcome of tumor therapy. We made use of Mdm2 knock-out in developing embryos as a sensitive tool for detecting remaining p53 activities. Genetic ablation of Mdm2 led to embryonic lethality in Trp53E177R/E177R homozygotes at days 9.5-11.5. This effect was not rescued by concomitant p21-knockout, indicating its independence of p21-mediated cell cycle arrest. Instead, immunohistochemical analysis showed widespread apoptosis in tissues of defective embryos accompanied by persistent accumulation of p53RR protein. This led to partial restoration of the mutant's proficiency in transcriptional induction of the pro-apoptotic genes Bbc3 (Puma) and Bax. These data indicate that increased quantity can compensate for qualitative defects of p53 mutants and suggest that Mdm2-targeting (potentially in combination with other drugs) might be effective against cells bearing p53 partial LOF mutants.
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| |
Collapse
|
246
|
Sborchia M, De Prez EG, Antoine MH, Bienfait L, Indra R, Valbuena G, Phillips DH, Nortier JL, Stiborová M, Keun HC, Arlt VM. The impact of p53 on aristolochic acid I-induced nephrotoxicity and DNA damage in vivo and in vitro. Arch Toxicol 2019; 93:3345-3366. [PMID: 31602497 PMCID: PMC6823306 DOI: 10.1007/s00204-019-02578-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022]
Abstract
Exposure to aristolochic acid (AA) is associated with human nephropathy and urothelial cancer. The tumour suppressor TP53 is a critical gene in carcinogenesis and frequently mutated in AA-induced urothelial tumours. We investigated the impact of p53 on AAI-induced nephrotoxicity and DNA damage in vivo by treating Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice with 3.5 mg/kg body weight (bw) AAI daily for 2 or 6 days. Renal histopathology showed a gradient of intensity in proximal tubular injury from Trp53(+/+) to Trp53(-/-) mice, especially after 6 days. The observed renal injury was supported by nuclear magnetic resonance (NMR)-based metabonomic measurements, where a consistent Trp53 genotype-dependent trend was observed for urinary metabolites that indicate aminoaciduria (i.e. alanine), lactic aciduria (i.e. lactate) and glycosuria (i.e. glucose). However, Trp53 genotype had no impact on AAI-DNA adduct levels, as measured by 32P-postlabelling, in either target (kidney and bladder) or non-target (liver) tissues, indicating that the underlying mechanisms of p53-related AAI-induced nephrotoxicity cannot be explained by differences in AAI genotoxicity. Performing gas chromatography-mass spectrometry (GC-MS) on kidney tissues showed metabolic pathways affected by AAI treatment, but again Trp53 status did not clearly impact on such metabolic profiles. We also cultured primary mouse embryonic fibroblasts (MEFs) derived from Trp53(+/+), Trp53(+/-) and Trp53(-/-) mice and exposed them to AAI in vitro (50 µM for up to 48 h). We found that Trp53 genotype impacted on the expression of NAD(P)H:quinone oxidoreductase (Nqo1), a key enzyme involved in AAI bioactivation. Nqo1 induction was highest in Trp53(+/+) MEFs and lowest in Trp53(-/-) MEFs; and it correlated with AAI-DNA adduct formation, with lowest adduct levels being observed in AAI-exposed Trp53(-/-) MEFs. Overall, our results clearly demonstrate that p53 status impacts on AAI-induced renal injury, but the underlying mechanism(s) involved remain to be further explored. Despite the impact of p53 on AAI bioactivation and DNA damage in vitro, such effects were not observed in vivo.
Collapse
Affiliation(s)
- Mateja Sborchia
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK
| | - Eric G De Prez
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Marie-Hélène Antoine
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Lucie Bienfait
- Department of Pathology, Erasme University Hospital, 1070, Brussels, Belgium
| | - Radek Indra
- Department of Biochemistry, Faculty of Science, Charles University Prague, 128 40, Prague, Czech Republic
| | - Gabriel Valbuena
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK
| | - Joëlle L Nortier
- Laboratory of Experimental Nephrology, Department of Experimental Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 1070, Brussels, Belgium
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University Prague, 128 40, Prague, Czech Republic
| | - Hector C Keun
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, London, SE1 9NH, UK.
| |
Collapse
|
247
|
A negative feedback regulatory loop between miR-138 and TP53 is mediated by USP10. Oncotarget 2019; 10:6288-6296. [PMID: 31695837 PMCID: PMC6824876 DOI: 10.18632/oncotarget.27275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/10/2019] [Indexed: 02/05/2023] Open
Abstract
TP53 is a critical tumor suppressor. In approximately 50% of human cancers the TP53 gene is either lost or mutated. The expression level of TP53 in the cells is tightly controlled by a fine-tune machinery, mainly through the Mdm2-mediated ubiquitination pathway. On the other side, the ubiquitinated TP53 could be reversed and stabilized by USP7 and USP10, to keep the amount of TP53 in check. MicroRNAs can negatively regulate TP53 expression levels through direct targeting or positively regulate TP53 function through the repression of negative regulators of TP53. Here we report that microRNA-138 controls TP53 expression by directly targeting USP10. Furthermore, TP53 represses microRNA-138 expression, forming a negative feedback regulatory loop. This finding adds another layer of complexity to the TP53 network.
Collapse
|
248
|
Feng L, Li J, Li F, Li H, Bei S, Zhang X, Yang Z. Long noncoding RNA VCAN-AS1 contributes to the progression of gastric cancer via regulating p53 expression. J Cell Physiol 2019; 235:4388-4398. [PMID: 31637706 DOI: 10.1002/jcp.29315] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 09/30/2019] [Indexed: 12/30/2022]
Abstract
Gastric cancer (GC) is one of the most frequent malignancies worldwide. Long noncoding RNAs (lncRNAs) are found to be largely implicated in various cancers, including GC. However, the function of lncRNA VCAN antisense RNA 1 (VCAN-AS1) in GC remains unclear. Herein, we observed a low level of VCAN-AS1 in normal gastric tissues through NCBI and UCSC, and that VCAN-AS1 upregulation in GC tissues was related to poor prognosis by TCGA. Furthermore, VCAN-AS1 was found markedly enhanced in GC tissues and cell lines, while its upregulation was related with clinical outcomes of GC patients. Besides this, silencing VCAN-AS1 represses cell proliferation, migration, and invasion but enhances apoptosis. More important, we discovered that VCAN-AS1 expression was negatively correlated with wild-type p53 levels in GC tissues and that p53 was negatively modulated by VCAN-AS1 in GC cells. Furthermore, p53 suppression reversed the repression of VCAN-AS1 silence on the biological processes of AGS cells. Intriguingly, we identified that both VCAN-AS1 and TP53 can bind with eIF4A3, one of the core proteins in the exon junction complex. Also, we confirmed that VCAN-AS1 negatively regulates TP53 expression by competitively binding with eIF4A3. Our findings disclosed that VCAN-AS1 contributes to GC progression through interacting with eIF4A3 to downregulate TP53 expression, indicating that VCAN-AS1 is a novel therapeutic strategy for GC treatment.
Collapse
Affiliation(s)
- Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Huanqin Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Songhua Bei
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaohong Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhen Yang
- Surgical Department, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
249
|
He MY, Xu SB, Qu ZH, Guo YM, Liu XC, Cong XX, Wang JF, Low BC, Li L, Wu Q, Lin P, Yan SG, Bao Z, Zhou YT, Zheng LL. Hsp90β interacts with MDM2 to suppress p53-dependent senescence during skeletal muscle regeneration. Aging Cell 2019; 18:e13003. [PMID: 31313490 PMCID: PMC6718578 DOI: 10.1111/acel.13003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/02/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Cellular senescence plays both beneficial and detrimental roles in embryonic development and tissue regeneration, while the underlying mechanism remains elusive. Recent studies disclosed the emerging roles of heat-shock proteins in regulating muscle regeneration and homeostasis. Here, we found that Hsp90β, but not Hsp90α isoform, was significantly upregulated during muscle regeneration. RNA-seq analysis disclosed a transcriptional elevation of p21 in Hsp90β-depleted myoblasts, which is due to the upregulation of p53. Moreover, knockdown of Hsp90β in myoblasts resulted in p53-dependent cellular senescence. In contrast to the notion that Hsp90 interacts with and protects mutant p53 in cancer, Hsp90β preferentially bound to wild-type p53 and modulated its degradation via a proteasome-dependent manner. Moreover, Hsp90β interacted with MDM2, the chief E3 ligase of p53, to regulate the stability of p53. In line with these in vitro studies, the expression level of p53-p21 axis was negatively correlated with Hsp90β in aged mice muscle. Consistently, administration of 17-AAG, a Hsp90 inhibitor under clinical trial, impaired muscle regeneration by enhancing injury-induced senescence in vivo. Taken together, our finding revealed a previously unappreciated role of Hsp90β in regulating p53 stability to suppress senescence both in vitro and in vivo.
Collapse
Affiliation(s)
- Min Yi He
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
| | - Shui Bo Xu
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
| | - Zi Hao Qu
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Yue Mei Guo
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
| | - Xiao Ceng Liu
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
| | - Xiao Xia Cong
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
| | - Jian Feng Wang
- Department of Respiratory Medicine, The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Boon Chuan Low
- Mechanobiology Institute, Department of Biological Sciences National University of Singapore Singapore
| | - Li Li
- Institute of Aging Research Hangzhou Normal University Hangzhou China
| | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine Macau University of Science and Technology Macau China
| | - Peng Lin
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Shi Gui Yan
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Zhang Bao
- Department of Respiratory Medicine, The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Yi Ting Zhou
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
- ZJU‐UoE Institute Zhejiang University School of Medicine Hangzhou China
| | - Li Ling Zheng
- Department of Biochemistry and Molecular Biology, Department of Orthopaedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine Zhejiang University School of Medicine Hangzhou China
| |
Collapse
|
250
|
Trp53 null and R270H mutant alleles have comparable effects in regulating invasion, metastasis, and gene expression in mouse colon tumorigenesis. J Transl Med 2019; 99:1454-1469. [PMID: 31148594 PMCID: PMC6759392 DOI: 10.1038/s41374-019-0269-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/22/2019] [Accepted: 04/07/2019] [Indexed: 12/25/2022] Open
Abstract
Somatic APC (adenomatous polyposis coli), TP53, KRAS mutations are present in roughly 80%, 60%, and 40%, respectively, of human colorectal cancers (CRCs). Most TP53 mutant alleles in CRCs encode missense mutant proteins with loss-of-function (LOF) of p53's transcriptional activity and dominant negative (DN) effects on wild-type p53 function. Missense mutant p53 proteins have been reported to exert gain-of-function (GOF) effects in cancer. We compared the phenotypic effects of the common human cancer-associated TP53 R273H missense mutation to p53 null status in a genetically engineered mouse CRC model. Inactivation of one allele of Apc together with activation of a Kras mutant allele in mouse colon epithelium instigated development of serrated and hyperplastic epithelium and adenomas (AK mice). Addition of a Trp53R270H or Trp53null mutant allele to the model (AKP mice) led to markedly shortened survival and increased tumor burden relative to that of AK mice, including adenocarcinomas in AKP mice. Comparable life span and tumor burden were seen in AKP mice carrying Trp53R270H or Trp53null alleles, along with similar frequencies of spontaneous metastasis to lymph nodes, lung, and liver. The fraction of adenocarcinomas with submucosa or deeper invasion was higher in AKP270/fl mice than in AKPfl/fl mice, but the incidence of adenocarcinomas per mouse did not differ significantly between AKPfl/fl and AKP270/fl mice. In line with their comparable biological behaviors, mouse primary tumors and tumor-derived organoids with the Trp53R270H or Trp53null alleles had highly similar gene expression profiles. Human CRCs with TP53 R273 missense mutant or null alleles also had essentially homogeneous gene expression patterns. Our findings indicate the R270H/R273H p53 mutant protein does not manifest definite GOF biological effects in mouse and human CRCs, suggesting possible GOF effects of mutant p53 in cancer phenotypes are likely allele-specific and/or context-dependent.
Collapse
|