1
|
Tseng YH, Tran TTM, Tsai Chang J, Huang YT, Nguyen AT, Chang IYF, Chen YT, Hsieh HW, Juang YL, Chang PMH, Huang TY, Chang YC, Chen YM, Liu H, Huang CYF. Utilizing TP53 hotspot mutations as effective predictors of gemcitabine treatment outcome in non-small-cell lung cancer. Cell Death Discov 2025; 11:26. [PMID: 39870629 PMCID: PMC11772833 DOI: 10.1038/s41420-025-02300-7] [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: 03/27/2024] [Revised: 12/11/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
TP53 mutations are recognized to correlate with a worse prognosis in individuals with non-small cell lung cancer (NSCLC). There exists an immediate necessity to pinpoint selective treatment for patients carrying TP53 mutations. Potential drugs were identified by comparing drug sensitivity differences, represented by the half-maximal inhibitory concentration (IC50), between TP53 mutant and wild-type NSCLC cell lines using database analysis. In addition, clinical data from NSCLC patients were collected to evaluate both their TP53 status and their response to gemcitabine, thereby facilitating further validation. Subsequently, NSCLC cell lines with different TP53 status (A549 and H1299) were subjected to gemcitabine treatment to investigate the association between TP53 mutations and gemcitabine response. According to the dataset, NSCLC cell lines carrying TP53 mutations displayed heightened sensitivity to gemcitabine. From a clinical standpoint, patients exhibiting TP53 hotspot mutations demonstrated prolonged overall survival upon gemcitabine treatment. In vitro, overexpressing various hotspot TP53 mutations significantly sensitized H1299 cells to gemcitabine. Moreover, the knockdown of TP53 in A549 cells notably augmented sensitivity to gemcitabine treatment, as evidenced by cell viability and reproductive cell death assays. Conversely, the overexpression of wild-type TP53 in H1299 cells led to an increased resistance against gemcitabine. Gemcitabine is a treatment option for patients with non-small cell lung cancer (NSCLC) who carry TP53 hotspot mutations. This potential effectiveness might arise from its ability to disrupt DNA damage repair processes, leading to G2/M phase cell cycle arrest or an augmentation of mitotic abnormalities, eventually cause cell death. As a result, when planning treatment strategies for NSCLC patients possessing TP53 hotspot mutations, gemcitabine should be considered to incorporate into the indication.
Collapse
Affiliation(s)
- Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Trieu Thi My Tran
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - Yu-Tang Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Biomedical Industry Ph.D. Program, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Anh Thuc Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Tung Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Wen Hsieh
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yue-Li Juang
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Peter Mu-Hsin Chang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Yi Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hsuan Liu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Chong Hin Loon Memorial Cancer and Biotherapy Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
2
|
Nakayama M, Saito H, Murakami K, Oshima H, Oshima M. Missense Mutant p53 Transactivates Wnt/β-Catenin Signaling in Neighboring p53-Destabilized Cells through the COX-2/PGE2 Pathway. CANCER RESEARCH COMMUNICATIONS 2025; 5:13-23. [PMID: 39641656 PMCID: PMC11695814 DOI: 10.1158/2767-9764.crc-24-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
SIGNIFICANCE There is intratumor heterogeneity in the stabilization of missense mutant p53, and it has been thought that only cells with nuclear accumulation of mutant p53 have oncogenic function. However, using mouse intestinal tumor-derived organoids, we show that mutant p53-stabilized cells transactivate Wnt/β-catenin signaling in neighboring p53-destabilized cells through activating the COX-2/PGE2 pathway. These results suggest that both p53-stabilized cells and p53-destabilized cells contribute to malignant progression through interaction within the intratumor microenvironment.
Collapse
Affiliation(s)
- Mizuho Nakayama
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Hiroshi Saito
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Japan
| | - Kazuhiro Murakami
- Division of Epithelial Stem Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
3
|
Morrissey RL, Thompson AM, Lozano G. Is loss of p53 a driver of ductal carcinoma in situ progression? Br J Cancer 2022; 127:1744-1754. [PMID: 35764786 DOI: 10.1038/s41416-022-01885-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/17/2022] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive carcinoma. Multiple studies have shown that DCIS lesions typically possess a driver mutation associated with cancer development. Mutation in the TP53 tumour suppressor gene is present in 15-30% of pure DCIS lesions and in ~30% of invasive breast cancers. Mutations in TP53 are significantly associated with high-grade DCIS, the most likely form of DCIS to progress to invasive carcinoma. In this review, we summarise published evidence on the prevalence of mutant TP53 in DCIS (including all DCIS subtypes), discuss the availability of mouse models for the study of DCIS and highlight the need for functional studies of the role of TP53 in the development of DCIS and progression from DCIS to invasive disease.
Collapse
Affiliation(s)
- Rhiannon L Morrissey
- Genetics and Epigenetics Program at The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alastair M Thompson
- Division of Surgical Oncology, Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Guillermina Lozano
- Genetics and Epigenetics Program at The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA. .,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
P63 and P73 Activation in Cancers with p53 Mutation. Biomedicines 2022; 10:biomedicines10071490. [PMID: 35884795 PMCID: PMC9313412 DOI: 10.3390/biomedicines10071490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 12/27/2022] Open
Abstract
The members of the p53 family comprise p53, p63, and p73, and full-length isoforms of the p53 family have a tumor suppressor function. However, p53, but not p63 or p73, has a high mutation rate in cancers causing it to lose its tumor suppressor function. The top and second-most prevalent p53 mutations are missense and nonsense mutations, respectively. In this review, we discuss possible drug therapies for nonsense mutation and a missense mutation in p53. p63 and p73 activators may be able to replace mutant p53 and act as anti-cancer drugs. Herein, these p63 and p73 activators are summarized and how to improve these activator responses, particularly focusing on p53 gain-of-function mutants, is discussed.
Collapse
|
5
|
Chiang YT, Chien YC, Lin YH, Wu HH, Lee DF, Yu YL. The Function of the Mutant p53-R175H in Cancer. Cancers (Basel) 2021; 13:4088. [PMID: 34439241 PMCID: PMC8391618 DOI: 10.3390/cancers13164088] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
Wild-type p53 is known as "the guardian of the genome" because of its function of inducing DNA repair, cell-cycle arrest, and apoptosis, preventing the accumulation of gene mutations. TP53 is highly mutated in cancer cells and most TP53 hotspot mutations are missense mutations. Mutant p53 proteins, encoded by these hotspot mutations, lose canonical wild-type p53 functions and gain functions that promote cancer development, including promoting cancer cell proliferation, migration, invasion, initiation, metabolic reprogramming, angiogenesis, and conferring drug resistance to cancer cells. Among these hotspot mutations, p53-R175H has the highest occurrence. Although losing the transactivating function of the wild-type p53 and prone to aggregation, p53-R175H gains oncogenic functions by interacting with many proteins. In this review, we summarize the gain of functions of p53-R175H in different cancer types, the interacting proteins of p53-R175H, and the downstream signaling pathways affected by p53-R175H to depict a comprehensive role of p53-R175H in cancer development. We also summarize treatments that target p53-R175H, including reactivating p53-R175H with small molecules that can bind to p53-R175H and alter it into a wild-type-like structure, promoting the degradation of p53-R175H by targeting heat-shock proteins that maintain the stability of p53-R175H, and developing immunotherapies that target the p53-R175H-HLA complex presented by tumor cells.
Collapse
Affiliation(s)
- Yen-Ting Chiang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
| | - Yi-Chung Chien
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
- Program for Translational Medicine, China Medical University, Taichung 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
- Drug Development Center, Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Yu-Heng Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
| | - Hui-Hsuan Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yung-Luen Yu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
- Program for Translational Medicine, China Medical University, Taichung 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
- Drug Development Center, Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
6
|
Alvarado-Ortiz E, de la Cruz-López KG, Becerril-Rico J, Sarabia-Sánchez MA, Ortiz-Sánchez E, García-Carrancá A. Mutant p53 Gain-of-Function: Role in Cancer Development, Progression, and Therapeutic Approaches. Front Cell Dev Biol 2021; 8:607670. [PMID: 33644030 PMCID: PMC7905058 DOI: 10.3389/fcell.2020.607670] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/23/2020] [Indexed: 02/05/2023] Open
Abstract
Frequent p53 mutations (mutp53) not only abolish tumor suppressor capacities but confer various gain-of-function (GOF) activities that impacts molecules and pathways now regarded as central for tumor development and progression. Although the complete impact of GOF is still far from being fully understood, the effects on proliferation, migration, metabolic reprogramming, and immune evasion, among others, certainly constitute major driving forces for human tumors harboring them. In this review we discuss major molecular mechanisms driven by mutp53 GOF. We present novel mechanistic insights on their effects over key functional molecules and processes involved in cancer. We analyze new mechanistic insights impacting processes such as immune system evasion, metabolic reprogramming, and stemness. In particular, the increased lipogenic activity through the mevalonate pathway (MVA) and the alteration of metabolic homeostasis due to interactions between mutp53 and AMP-activated protein kinase (AMPK) and Sterol regulatory element-binding protein 1 (SREBP1) that impact anabolic pathways and favor metabolic reprograming. We address, in detail, the impact of mutp53 over metabolic reprogramming and the Warburg effect observed in cancer cells as a consequence, not only of loss-of-function of p53, but rather as an effect of GOF that is crucial for the imbalance between glycolysis and oxidative phosphorylation. Additionally, transcriptional activation of new targets, resulting from interaction of mutp53 with NF-kB, HIF-1α, or SREBP1, are presented and discussed. Finally, we discuss perspectives for targeting molecules and pathways involved in chemo-resistance of tumor cells resulting from mutp53 GOF. We discuss and stress the fact that the status of p53 currently constitutes one of the most relevant criteria to understand the role of autophagy as a survival mechanism in cancer, and propose new therapeutic approaches that could promote the reduction of GOF effects exercised by mutp53 in cancer.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Karen Griselda de la Cruz-López
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
- Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jared Becerril-Rico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sánchez
- Programa de Posgrado en Ciencias Bioquímicas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Alejandro García-Carrancá
- Laboratorio de Virus and Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| |
Collapse
|
7
|
Shi Y, Norberg E, Vakifahmetoglu-Norberg H. Mutant p53 as a Regulator and Target of Autophagy. Front Oncol 2021; 10:607149. [PMID: 33614491 PMCID: PMC7886977 DOI: 10.3389/fonc.2020.607149] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/15/2020] [Indexed: 12/30/2022] Open
Abstract
One of the most notoriously altered genes in human cancer is the tumor-suppressor TP53, which is mutated with high frequency in more cancers than any other tumor suppressor gene. Beyond the loss of wild-type p53 functions, mutations in the TP53 gene often lead to the expression of full-length proteins with new malignant properties. Among the defined oncogenic functions of mutant p53 is its effect on cell metabolism and autophagy. Due to the importance of autophagy as a stress adaptive response, it is frequently dysfunctional in human cancers. However, the role of p53 is enigmatic in autophagy regulation. While the complex action of the wild-type p53 on autophagy has extensively been described in literature, in this review, we focus on the conceivable role of distinct mutant p53 proteins in regulating different autophagic pathways and further discuss the available evidence suggesting a possible autophagy stimulatory role of mutant p53. Moreover, we describe the involvement of different autophagic pathways in targeting and degrading mutant p53 proteins, exploring the potential strategies of targeting mutant p53 in cancer by autophagy.
Collapse
Affiliation(s)
- Yong Shi
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Norberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
8
|
Caporossi D, Parisi A, Fantini C, Grazioli E, Cerulli C, Dimauro I. AlphaB-crystallin and breast cancer: role and possible therapeutic strategies. Cell Stress Chaperones 2021; 26:19-28. [PMID: 33111264 PMCID: PMC7736448 DOI: 10.1007/s12192-020-01175-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/10/2020] [Accepted: 10/20/2020] [Indexed: 01/18/2023] Open
Abstract
AlphaB-crystallin (HSPB5) is one of the most prominent and well-studied members of the small heat shock protein (sHsp) family. To date, it is known that this protein modulates significant cellular processes and therefore, it is not surprising that its deregulation is involved in various human pathologies, including cancer diseases. Despite the pathogenic significance of HSPB5 in cancer and its regulatory mechanism related to aggressiveness is poorly understood, several reports describe the association of breast carcinoma progression with HSPB5, whose expression is also considered an independent predictor of breast cancer metastasis to the brain. Indeed, numerous authors indicate HSPB5 as a new valuable biomarker for clinicopathological parameters and poor prognosis in breast cancer. Considering the cytoprotective, anti-apoptotic, pro-angiogenic, and pro-metastatic properties of the sHsps, it is not surprising that they are considered as promising targets for anticancer treatment, even though, at present, a deeper understanding of their mode of action is needed to allow the development of precise therapeutic interventions. Data on the direct inhibition of different sHsps demonstrate promising results in cancer pathologies; however, specific strategies against HSPB5 have not been considered. This review highlights the most relevant findings on HSPB5 and its role in breast cancer, as well as the possible strategies in using HSPB5 inhibition for therapeutic purposes.
Collapse
Affiliation(s)
- Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Attilio Parisi
- Unit of Sport Medicine, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Cristina Fantini
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Elisa Grazioli
- Unit of Sport Medicine, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Claudia Cerulli
- Unit of Sport Medicine, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy.
| |
Collapse
|
9
|
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: 177] [Impact Index Per Article: 35.4] [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
|
10
|
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: 4.0] [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
|
11
|
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: 6.4] [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
|
12
|
Thayer KM, Carcamo C. Homologs of the Tumor Suppressor Protein p53: A Bioinformatics Study for Drug Design. MOJ PROTEOMICS & BIOINFORMATICS 2020; 9:5-14. [PMID: 34532721 PMCID: PMC8442938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sequence and structure of proteins related to the tumor suppressor protein p53 were studied from the perspective of gaining insight for the development of therapeutic drugs. Our study addresses two major issues encumber bringing novel drugs to market: side effects and artifacts from animal models. In the first phase of our study, we performed a genome-wide search to identify potentially similar proteins to p53 that may be susceptible to off target effects. In the second phase, we chose a selection of common model organisms that could potentially be available to undergraduate researchers in the university setting to assess which ones utilize p53 most similar to humans on the basis of sequence homology and structural similarity from predicted structures. Our results confirm the proteins in the humans significantly similar to p53 are known paralogs within the p53 family. In considering model organisms, murine p53 bore great similarity to human p53 in terms of both sequence and structure, but others performed similarly well. We discuss the findings against the background of other structural benchmarks and point out potential benefits and drawbacks of various alternatives for use in future drug design pilot studies.
Collapse
Affiliation(s)
- Kelly M Thayer
- Vassar College, 124 Raymond Avenue, Poughkeepsie, NY 12604, USA
| | - Claudia Carcamo
- Vassar College, 124 Raymond Avenue, Poughkeepsie, NY 12604, USA
| |
Collapse
|
13
|
Behring M, Vazquez AI, Cui X, Irvin MR, Ojesina AI, Agarwal S, Manne U, Shrestha S. Gain of function in somatic TP53 mutations is associated with immune-rich breast tumors and changes in tumor-associated macrophages. Mol Genet Genomic Med 2019; 7:e1001. [PMID: 31637877 PMCID: PMC6900370 DOI: 10.1002/mgg3.1001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Somatic mutations in TP53 are present in 20%-30% of all breast tumors. While there are numerous population-based analyses of TP53, yet none have examined the relationship between somatic mutations in TP53 and tumor invasive immune cells. METHODS Clinical and genetic data from 601 women drawn from The Cancer Genome Atlas (TCGA) were used to test the association between somatic TP53 mutation and immune-rich or immune-poor tumor status; determined using the CIBERSORT-based gene expression signature of 22 immune cell types. Our validation dataset, the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC), used a pathologist-determined measure of lymphocyte infiltration. RESULTS Within TP53-mutated samples, a mutation at codon p.R175H was shown to be present at higher frequency in immune-rich tumors. In validation analysis, any somatic mutation in TP53 was associated with immune-rich status, and the mutation at p.R175H had a significant association with tumor-invasive lymphocytes. TCGA-only analysis of invasive immune cell type identified an increase in M0 macrophages associated with p.R175H. CONCLUSIONS These findings suggest that TP53 somatic mutations, particularly at codon p.R175H, are enriched in tumors with infiltrating immune cells. Our results confirm recent research showing inflammation-related gain of function in specific TP53 mutations.
Collapse
Affiliation(s)
- Michael Behring
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Pathology and Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ana I Vazquez
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Xiangqin Cui
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Akinyemi I Ojesina
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA.,HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sumit Agarwal
- Department of Pathology and Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Upender Manne
- Department of Pathology and Surgery, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadeep Shrestha
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
14
|
FKBP12 is a predictive biomarker for efficacy of anthracycline-based chemotherapy in breast cancer. Cancer Chemother Pharmacol 2019; 84:861-872. [PMID: 31428819 PMCID: PMC6769097 DOI: 10.1007/s00280-019-03923-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/06/2019] [Indexed: 12/29/2022]
Abstract
Background FK506-binding protein 12 (FKBP12) is abundant, ubiquitously expressed cytoplasmic protein with multiple functions in cell signaling transduction. Recently, we reported a novel function for FKBP12 in oncoprotein mouse double minute 2 (MDM2) self-ubiquitination and degradation, which greatly enhanced the sensitivity of cancer cells to chemotherapy. However, the clinical relevance remains unclear. Methods An immunohistochemical analysis of FKBP12 expression was performed in a cohort of 524 patients with invasive breast cancer. The correlations of FKBP12 expression with patient survival and chemoresponse were statistically analyzed. MDA-MB-468 cells were transfected with FKBP12 siRNA or Myc-tagged FKBP12, and then, the tumor cells were treated with doxorubicin followed by western blot, cell viability, and apoptosis assay. Results The expression of FKBP12 was decreased in breast cancer tissues, and there was a significant correlation between FKBP12 loss and MDM2 overexpression. Furthermore, FKBP12 loss was specifically correlated with poor prognosis and increased resistance to anthracycline-based chemotherapy. Kaplan–Meier survival analysis showed that overall survival (OS) and disease-free survival (DFS) were both significantly lower in the low FKBP12 expression group than those in the high FKBP12 expression group. In patients treated with anthracycline-based preoperative chemotherapy, low FKBP12 expression patients had a significant lower rate of pathologic complete response (pCR). Importantly, these results seemed to be driven mainly by MDM2. These observations were especially prominent in the MDM2-positive subgroup. Univariate and multivariate analyses revealed that FKBP12 loss was an independent factor for predicting prognosis and pCR. In in vitro assay, FKBP12 silence led to significant upregulation of MDM2. Accordingly, MDA-MB-468 cells with FKBP12 silence were less responsive to doxorubicin-induced cytotoxic and apoptotic effect. In contrast, in FKBP12-transfected MDA-MB-468 cells, MDM2 was more greatly inhibited by doxorubicin, resulting in greater cytotoxic and apoptotic effect. Conclusions We propose that FKBP12 loss, which can be enhanced by MDM2 overexpression, predicts poor prognosis and chemoresistance. Increasing the expression of FKBP12 may be a valuable strategy to add to anthracycline-based chemotherapy, especially in MDM2-overexpressed patients.
Collapse
|
15
|
Gandhi N, Das GM. Metabolic Reprogramming in Breast Cancer and Its Therapeutic Implications. Cells 2019; 8:E89. [PMID: 30691108 PMCID: PMC6406734 DOI: 10.3390/cells8020089] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 12/22/2022] Open
Abstract
Current standard-of-care (SOC) therapy for breast cancer includes targeted therapies such as endocrine therapy for estrogen receptor-alpha (ERα) positive; anti-HER2 monoclonal antibodies for human epidermal growth factor receptor-2 (HER2)-enriched; and general chemotherapy for triple negative breast cancer (TNBC) subtypes. These therapies frequently fail due to acquired or inherent resistance. Altered metabolism has been recognized as one of the major mechanisms underlying therapeutic resistance. There are several cues that dictate metabolic reprogramming that also account for the tumors' metabolic plasticity. For metabolic therapy to be efficacious there is a need to understand the metabolic underpinnings of the different subtypes of breast cancer as well as the role the SOC treatments play in targeting the metabolic phenotype. Understanding the mechanism will allow us to identify potential therapeutic vulnerabilities. There are some very interesting questions being tackled by researchers today as they pertain to altered metabolism in breast cancer. What are the metabolic differences between the different subtypes of breast cancer? Do cancer cells have a metabolic pathway preference based on the site and stage of metastasis? How do the cell-intrinsic and -extrinsic cues dictate the metabolic phenotype? How do the nucleus and mitochondria coordinately regulate metabolism? How does sensitivity or resistance to SOC affect metabolic reprogramming and vice-versa? This review addresses these issues along with the latest updates in the field of breast cancer metabolism.
Collapse
Affiliation(s)
- Nishant Gandhi
- Department of Pharmacology and Therapeutics, Center for Genetics & Pharmacology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Gokul M Das
- Department of Pharmacology and Therapeutics, Center for Genetics & Pharmacology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
16
|
Alaee M, Nool K, Pasdar M. Plakoglobin restores tumor suppressor activity of p53 R175H mutant by sequestering the oncogenic potential of β-catenin. Cancer Sci 2018; 109:1876-1888. [PMID: 29660231 PMCID: PMC5989865 DOI: 10.1111/cas.13612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 12/16/2022] Open
Abstract
Tumor suppressor/transcription factor p53 is mutated in over 50% of all cancers. Some mutant p53 proteins have not only lost tumor suppressor activities but they also gain oncogenic functions (GOF). One of the most frequently expressed GOF p53 mutants is Arg175His (p53R175H ) with well-documented roles in cancer development and progression. Plakoglobin is a cell adhesion and signaling protein and a paralog of β-catenin. Unlike β-catenin that has oncogenic function through its role in the Wnt pathway, plakoglobin generally acts as a tumor/metastasis suppressor. We have shown that plakoglobin interacted with wild type and a number of p53 mutants in various carcinoma cell lines. Plakoglobin and mutant p53 interacted with the promoter and regulated the expression of several p53 target genes. Furthermore, plakoglobin interactions with p53 mutants restored their tumor suppressor/metastasis activities in vitro. GOF p53 mutants induce accumulation and oncogenic activation of β-catenin. Previously, we showed that one mechanism by which plakoglobin may suppress tumorigenesis is by sequestering β-catenin's oncogenic activity. Here, we examined the effects of p53R175H expression on β-catenin accumulation and transcriptional activation and their modifications by plakoglobin coexpression. We showed that p53R175H expression in plakoglobin null cells increased total and nuclear levels of β-catenin and its transcriptional activity. Coexpression of plakoglobin in these cells promoted β-catenin's proteasomal degradation, and decreased its nuclear levels and transactivation. Wnt/β-catenin targets, c-MYC and S100A4 were upregulated in p53R175H cells and were downregulated when plakoglobin was coexpressed. Plakoglobin-p53R175H cells also showed significant reduction in their migration and invasion in vitro.
Collapse
Affiliation(s)
- Mahsa Alaee
- Department of OncologyUniversity of AlbertaEdmontonCanada
| | - Kristina Nool
- Department of OncologyUniversity of AlbertaEdmontonCanada
| | - Manijeh Pasdar
- Department of OncologyUniversity of AlbertaEdmontonCanada
| |
Collapse
|
17
|
Kuykendall A, Duployez N, Boissel N, Lancet JE, Welch JS. Acute Myeloid Leukemia: The Good, the Bad, and the Ugly. Am Soc Clin Oncol Educ Book 2018; 38:555-573. [PMID: 30231330 DOI: 10.1200/edbk_199519] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Acute myeloid leukemia (AML) was initially subdivided according to morphology (the French-American-British system), which proved helpful in pathologic categorization. Subsequently, clinical and genomic factors were found to correlate with response to chemotherapy and with overall survival. These included a history of antecedent hematologic disease, a history of chemotherapy or radiation therapy, the presence of various recurrent cytogenetic abnormalities, and, more recently, the presence of specific point mutations. This article reviews the biology and responses of one AML subgroup with consistent response and good outcomes following chemotherapy (core-binding factor leukemia), and two subgroups with persistently bad, and even ugly, outcomes (secondary AML and TP53-mutated AML).
Collapse
MESH Headings
- Alleles
- Biomarkers, Tumor
- Chromosome Aberrations
- Combined Modality Therapy
- Core Binding Factors/genetics
- Core Binding Factors/metabolism
- Gene Frequency
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/mortality
- Mutation
- Neoplasm, Residual/diagnosis
- Neoplasms, Second Primary/diagnosis
- Neoplasms, Second Primary/epidemiology
- Neoplasms, Second Primary/etiology
- Neoplasms, Second Primary/therapy
- Signal Transduction
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Andrew Kuykendall
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Nicolas Duployez
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Nicolas Boissel
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Jeffrey E Lancet
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - John S Welch
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Zhang Y, Hu Y, Fang JY, Xu J. Gain-of-function miRNA signature by mutant p53 associates with poor cancer outcome. Oncotarget 2017; 7:11056-66. [PMID: 26840456 PMCID: PMC4905457 DOI: 10.18632/oncotarget.7090] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/15/2016] [Indexed: 01/05/2023] Open
Abstract
Missense mutation of p53 not only impairs its tumor suppression function, but also causes oncogenic gain of function (GOF). The molecular underpinning of mutant p53 (mutp53) GOF is not fully understood, especially for the potential roles of non-coding genes. Here we identify the microRNA expression profile (microRNAome) of mutp53 on Arg282 by controlled microarray experiments, and clarify the prognostic significance of mutp53-regulated miRNAs in cancers. A predominant repression effect on miRNA expression was found for mutant p53, with 183 significantly downregulated and only 12 upregulated miRNAs. Mutp53 and wild-type (wtp53) commonly upregulate let-7i, and other two miRNAs were upregulated by wtp53 but repressed by mutp53 (miR-610 and miR-3065–3p). Based the mutp53-regulated miRNA signature, a non-negative matrix factorization (NMF) model classified gastric cancer (GC) cases into subgroups with significantly different Disease-free survival (Kaplan-Meier test, P = 0.013). In contrast, the NMF model based on all miRNAs did not associate with cancer outcome. The mutp53 miRNA signature associated with the outcomes of breast cancer (P = 0.024) and hepatocellular cancer (P = 0.012). The miRPath analysis revealed that mutp53-suppressed miRNAs associate with Hippo, TGF-β and stem cell signaling pathways. Taken together, our results highlight a miRNA-mediated GOF mechanism of mutant p53 on Arg282, and suggest the prognostic potential of mutp53-associated miRNA signature.
Collapse
Affiliation(s)
- Yao Zhang
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Ye Hu
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China.,Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Jie Xu
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| |
Collapse
|
19
|
Zhao D, Tahaney WM, Mazumdar A, Savage MI, Brown PH. Molecularly targeted therapies for p53-mutant cancers. Cell Mol Life Sci 2017; 74:4171-4187. [PMID: 28643165 PMCID: PMC5664959 DOI: 10.1007/s00018-017-2575-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/30/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
Abstract
The tumor suppressor p53 is lost or mutated in approximately half of human cancers. Mutant p53 not only loses its anti-tumor transcriptional activity, but also often acquires oncogenic functions to promote tumor proliferation, invasion, and drug resistance. Traditional strategies have been taken to directly target p53 mutants through identifying small molecular compounds to deplete mutant p53, or to restore its tumor suppressive function. Accumulating evidence suggest that cancer cells with mutated p53 often exhibit specific functional dependencies on secondary genes or pathways to survive, providing alternative targets to indirectly treat p53-mutant cancers. Targeting these genes or pathways, critical for survival in the presence of p53 mutations, holds great promise for cancer treatment. In addition, mutant p53 often exhibits novel gain-of-functions to promote tumor growth and metastasis. Here, we review and discuss strategies targeting mutant p53, with focus on targeting the mutant p53 protein directly, and on the progress of identifying genes and pathways required in p53-mutant cells.
Collapse
Affiliation(s)
- Dekuang Zhao
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - William M Tahaney
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Michelle I Savage
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
20
|
Androgen induces G3BP2 and SUMO-mediated p53 nuclear export in prostate cancer. Oncogene 2017; 36:6272-6281. [PMID: 28692047 DOI: 10.1038/onc.2017.225] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/27/2017] [Accepted: 05/27/2017] [Indexed: 12/16/2022]
Abstract
The androgen receptor (AR) has a central role in prostate cancer progression, particularly treatment-resistance disease including castration-resistant prostate cancer. Loss of the p53 tumor suppressor, a nuclear transcription factor, is also known to contribute to prostate malignancy. Here we report that p53 is translocated to the cytoplasm by androgen-mediated induction of G3BP2, a newly described direct target gene of AR. G3BP2 induces both cell cycle progression and blocks apoptosis. Translocation of p53 is regulated by androgen-dependent sumoylation mediated by the G3BP2-interacting SUMO-E3 ligase, RanBP2. G3BP2 knockdown results in reduced tumor growth and increased nuclear p53 accumulation in mouse xenograft models of prostate cancer with or without long-term androgen deprivation. Moreover, strong cytoplasmic p53 localization is correlated clinically with elevated G3BP2 expression and predicts poor prognosis and disease progression to the hormone-refractory state. Our findings reveal a new AR-mediated mechanism of p53 inhibition that promotes treatment-resistant prostate cancer.
Collapse
|
21
|
Xiong J, Li J, Yang Q, Wang J, Su T, Zhou S. Gossypol has anti-cancer effects by dual-targeting MDM2 and VEGF in human breast cancer. Breast Cancer Res 2017; 19:27. [PMID: 28274247 PMCID: PMC5343402 DOI: 10.1186/s13058-017-0818-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/14/2017] [Indexed: 01/14/2023] Open
Abstract
Background Mouse double minute 2 (MDM2) and vascular endothelial growth factor (VEGF) are important molecules involved in tumor progression. We researched potential inhibitors that simultaneously target MDM2 and VEGF. In our recent study involving the performance of high-throughput screening with a fluorescence polarization assay, gossypol was identified as one of the top hits that inhibit protein-RNA binding activity. Because MDM2 is an RNA-binding protein and its targets include VEGF mRNA, we investigated whether gossypol has an inhibitory effect on MDM2-VEGF. Methods UV cross-linking and RNA binding assay, isothermal titration calorimetry assay, and ubiquitination assay were performed to determine mechanisms by which gossypol functions as a dual inhibitor of MDM2 and VEGF. The effect of gossypol on MDM2 and VEGF expression, cancer cell apoptosis, tumor growth and VEGF-mediated angiogenesis were studied in vitro and in vivo in different human breast cancer models with a different p53 status. Results We observed that gossypol inhibited expression of both MDM2 and VEGF in human breast cancer cells with either wild-type or mutant p53. A nechanistic study further demonstrated that, through disrupting the interaction between MDM2 protein and VEGF mRNA, gossypol induced MDM2 self-ubiquitination and decreased VEGF translation simultaneously, which resulted in both apoptosis and anti-angiogenesis effects. In vitro, regardless of p53 status, gossypol induced cancer cell apoptosis. In nude mouse xenograft in vivo models, gossypol suppressed tumor growth and VEGF-mediated angiogenesis. Conclusion Gossypol has anti-cancer effects by dual-targeting MDM2 and VEGF in human breast cancer. Our study reveals a novel mechanism by which gossypol functions as an anticancer agent. We believe that MDM2-VEGF targeting represents a novel strategy for improving cancer outcome.
Collapse
Affiliation(s)
- Jing Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiansha Li
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Pathology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Yang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tiefen Su
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Pathology, School of Basic Medical Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Sheng Zhou
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
22
|
Olivos DJ, Mayo LD. Emerging Non-Canonical Functions and Regulation by p53: p53 and Stemness. Int J Mol Sci 2016; 17:ijms17121982. [PMID: 27898034 PMCID: PMC5187782 DOI: 10.3390/ijms17121982] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/15/2023] Open
Abstract
Since its discovery nearly 40 years ago, p53 has ascended to the forefront of investigated genes and proteins across diverse research disciplines and is recognized most exclusively for its role in cancer as a tumor suppressor. Levine and Oren (2009) reviewed the evolution of p53 detailing the significant discoveries of each decade since its first report in 1979. In this review, we will highlight the emerging non-canonical functions and regulation of p53 in stem cells. We will focus on general themes shared among p53's functions in non-malignant stem cells and cancer stem-like cells (CSCs) and the influence of p53 on the microenvironment and CSC niche. We will also examine p53 gain of function (GOF) roles in stemness. Mutant p53 (mutp53) GOFs that lead to survival, drug resistance and colonization are reviewed in the context of the acquisition of advantageous transformation processes, such as differentiation and dedifferentiation, epithelial-to-mesenchymal transition (EMT) and stem cell senescence and quiescence. Finally, we will conclude with therapeutic strategies that restore wild-type p53 (wtp53) function in cancer and CSCs, including RING finger E3 ligases and CSC maintenance. The mechanisms by which wtp53 and mutp53 influence stemness in non-malignant stem cells and CSCs or tumor-initiating cells (TICs) are poorly understood thus far. Further elucidation of p53's effects on stemness could lead to novel therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lindsey D Mayo
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
23
|
p53 deficiency induces cancer stem cell pool expansion in a mouse model of triple-negative breast tumors. Oncogene 2016; 36:2355-2365. [PMID: 27775073 DOI: 10.1038/onc.2016.396] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/14/2016] [Accepted: 09/16/2016] [Indexed: 12/16/2022]
Abstract
Triple-negative breast cancer is a heterogeneous disease characterized by the expression of basal cell markers, no estrogen or progesterone receptor expression and a lack of HER2 overexpression. Triple-negative tumors often display activated Wnt/β-catenin signaling and most have impaired p53 function. We studied the interplay between p53 loss and Wnt/β-catenin signaling in stem cell function and tumorigenesis, by deleting p53 from the mammary epithelium of K5ΔNβcat mice displaying a constitutive activation of Wnt/β-catenin signaling in basal cells. K5ΔNβcat transgenic mice present amplification of the basal stem cell pool and develop triple-negative mammary carcinomas. The loss of p53 in K5ΔNβcat mice led to an early expansion of mammary stem/progenitor cells and accelerated the formation of triple-negative tumors. In particular, p53-deficient tumors expressed high levels of integrins and extracellular matrix components and were enriched in cancer stem cells. They also overexpressed the tyrosine kinase receptor Met, a feature characteristic of human triple-negative breast tumors. The inhibition of Met kinase activity impaired tumorsphere formation, demonstrating the requirement of Met signaling for cancer stem cell growth in this model. Human basal-like breast cancers with predicted mutated p53 status had higher levels of MET expression than tumors with wild-type p53. These results connect p53 loss and β-catenin activation to stem cell regulation and tumorigenesis in triple-negative cancer and highlight the role of Met signaling in maintaining cancer stem cell properties, revealing new cues for targeted therapies.
Collapse
|
24
|
Abstract
High-throughput sequencing of cancer genomes is increasingly becoming an essential tool of clinical oncology that facilitates target identification and targeted therapy within the context of precision medicine. The cumulative profiles of somatic mutations in cancer yielded by comprehensive molecular studies also constitute a fingerprint of historical exposures to exogenous and endogenous mutagens, providing insight into cancer evolution and etiology. Mutational signatures that were first established by inspection of the TP53 gene somatic landscape have now been confirmed and expanded by comprehensive sequencing studies. Further, the degree of granularity achieved by deep sequencing allows detection of low-abundance mutations with clinical relevance. In tumors, they represent the emergence of small aggressive clones; in normal tissues, they signal a mutagenic exposure related to cancer risk; and, in blood, they may soon become effective surveillance tools for diagnostic purposes and for monitoring of cancer prognosis and recurrence.
Collapse
Affiliation(s)
- Ana I Robles
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Jin Jen
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, and Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
25
|
Xie C, Wu B, Chen B, Shi Q, Guo J, Fan Z, Huang Y. Histone deacetylase inhibitor sodium butyrate suppresses proliferation and promotes apoptosis in osteosarcoma cells by regulation of the MDM2-p53 signaling. Onco Targets Ther 2016; 9:4005-13. [PMID: 27445491 PMCID: PMC4938147 DOI: 10.2147/ott.s105418] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Histone deacetylase inhibitors have been reported to induce tumor cell growth arrest, differentiation, and apoptosis. This study aimed to investigate the effects of one histone deacetylase inhibitor - sodium butyrate (SB) - on osteosarcoma (OS) cell proliferation and apoptosis and also the molecular mechanisms by which SB exerts regulatory effects on OS cells. U2OS and MG63 cells were treated with SB at various concentrations. Then, cell proliferation and apoptosis were determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide and flow cytometry assays, respectively; the expression of Ki67, Bax, Bcl-2, MDM2, and p53 proteins was determined by using Western blot assay. The results showed that SB suppressed proliferation in a concentration-dependent manner and promoted apoptosis of OS cells. In addition, SB enhanced p53 expression and decreased MDM2 expression, indicating that SB can regulate MDM2-p53 feedback loop. p53 inhibited proliferation and promoted apoptosis, whereas MDM2 promoted proliferation and suppressed apoptosis, which indicated that functional effect of SB on OS cell lines at least in part depended on the MDM2-p53 signaling. We also explored the effect of SB on OS cells in vivo and found that SB suppressed the growth of OS cells with no noticeable effect on activity and body weight of mice in vivo. These findings will offer new clues for OS development and progression and offer SB as a potent targeted agent for OS treatment.
Collapse
Affiliation(s)
- Chuhai Xie
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Boyi Wu
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Binwei Chen
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qunwei Shi
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jianhong Guo
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ziwen Fan
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
26
|
Ahronian LG, Driscoll DR, Klimstra DS, Lewis BC. The p53R172H mutant does not enhance hepatocellular carcinoma development and progression. PLoS One 2015; 10:e0123816. [PMID: 25885474 PMCID: PMC4401698 DOI: 10.1371/journal.pone.0123816] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/07/2015] [Indexed: 01/09/2023] Open
Abstract
Hepatocellular carcinoma is a highly deadly malignancy, accounting for approximately 800,000 deaths worldwide every year. Mutation of the p53 tumor suppressor gene is a common genetic change in HCC, present in 30% of cases. p53R175H (corresponding to p53R172H in mice) is a hotspot for mutation that demonstrates “prometastatic” gain-of-function in other cancer models. Since the frequency of p53 mutation increases with tumor grade in HCC, we hypothesized that p53R172H is a gain-of-function mutation in HCC that contributes to a decrease in tumor-free survival and an increase in metastasis. In an HCC mouse model, we found that p53R172H/flox mice do not have decreased survival, increased tumor incidence, or increased metastasis, relative to p53flox/flox littermates. Analysis of cell lines derived from both genotypes indicated that there are no differences in anchorage-independent growth and cell migration. However, shRNA-mediated knockdown of mutant p53 in p53R172H-expressing HCC cell lines resulted in decreased cell migration and anchorage-independent growth. Thus, although p53 mutant-expressing cells and tumors do not have enhanced properties relative to their p53 null counterparts, p53R172H-expressing HCC cells depend on this mutant for their transformation. p53 mutants have been previously shown to bind and inhibit the p53 family proteins p63 and p73. Interestingly, we find that the levels of p63 and p73 target genes are similar in p53 mutant and p53 null HCC cells. These data suggest that pathways regulated by these p53 family members are similarly impacted by p53R172H in mutant expressing cells, and by alternate mechanisms in p53 null cells, resulting in equivalent phenotypes. Consistent with this, we find that p53 null HCC cell lines display lower levels of the TA isoforms of p63 and p73 and higher levels of ΔNp63. Taken together these data point to the importance of p63 and p73 in constraining HCC progression.
Collapse
Affiliation(s)
- Leanne G. Ahronian
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - David R. Driscoll
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - David S. Klimstra
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Brian C. Lewis
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
27
|
Yallowitz AR, Li D, Lobko A, Mott D, Nemajerova A, Marchenko N. Mutant p53 Amplifies Epidermal Growth Factor Receptor Family Signaling to Promote Mammary Tumorigenesis. Mol Cancer Res 2015; 13:743-54. [PMID: 25573952 DOI: 10.1158/1541-7786.mcr-14-0360] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/17/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED The EGFR family (ErbB2/Her2 and EGFR/ErbB1/Her1) often modulates the transcriptional program involved in promoting mammary tumorigenesis. In humans, the majority of ErbB2-positive sporadic breast cancers harbor p53 mutations, which correlate with poor prognosis. Also, the extremely high incidence of ErbB2-positive breast cancer in women with p53 germline mutations (Li-Fraumeni syndrome) suggests a key role of mutant p53 specifically in ErbB2-mediated mammary tumorigenesis. To examine the role of mutant p53 during ErbB2-mediated mammary tumorigenesis, a mutant p53 allele (R172H) was introduced into the (MMTV)-ErbB2/Neu mouse model system. Interestingly, we show in heterozygous p53 mice that mutant p53 R172H is a more potent activator of ErbB2-mediated mammary tumorigenesis than simple loss of p53. The more aggressive disease in mutant p53 animals was reflected by earlier tumor onset, increased mammary tumor multiplicity, and shorter survival. These in vivo and in vitro data provide mechanistic evidence that mutant p53 amplifies ErbB2 and EGFR signaling to promote the expansion of mammary stem cells and induce cell proliferation. IMPLICATIONS This study identifies mutant p53 as an essential player in ErbB2 and EGFR-mediated mammary tumorigenesis and indicates the potential translational importance of targeting mutant p53 in this subset of patients with breast cancer.
Collapse
Affiliation(s)
- Alisha R Yallowitz
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Dun Li
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Anthony Lobko
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Daniel Mott
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Alice Nemajerova
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Natalia Marchenko
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York.
| |
Collapse
|
28
|
Alexandrova EM, Marchenko ND. Mutant p53 - Heat Shock Response Oncogenic Cooperation: A New Mechanism of Cancer Cell Survival. Front Endocrinol (Lausanne) 2015; 6:53. [PMID: 25954247 PMCID: PMC4406088 DOI: 10.3389/fendo.2015.00053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/03/2015] [Indexed: 01/06/2023] Open
Abstract
The main tumor suppressor function of p53 as a "guardian of the genome" is to respond to cellular stress by transcriptional activation of apoptosis, growth arrest, or senescence in damaged cells. Not surprisingly, mutations in the p53 gene are the most frequent genetic alteration in human cancers. Importantly, mutant p53 (mutp53) proteins not only lose their wild-type tumor suppressor activity but also can actively promote tumor development. Two main mechanisms accounting for mutp53 proto-oncogenic activity are inhibition of the wild-type p53 in a dominant-negative fashion and gain of additional oncogenic activities known as gain-of-function (GOF). Here, we discuss a novel mechanism of mutp53 GOF, which relies on its oncogenic cooperation with the heat shock machinery. This coordinated adaptive mechanism renders cancer cells more resistant to proteotoxic stress and provides both, a strong survival advantage to cancer cells and a promising means for therapeutic intervention.
Collapse
Affiliation(s)
| | - Natalia D. Marchenko
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
- *Correspondence: Natalia D. Marchenko, Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
29
|
Targeting TopBP1 at a convergent point of multiple oncogenic pathways for cancer therapy. Nat Commun 2014; 5:5476. [PMID: 25400145 PMCID: PMC4254804 DOI: 10.1038/ncomms6476] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 10/06/2014] [Indexed: 01/12/2023] Open
Abstract
The progression of many solid tumors is driven by de-regulation of multiple common pathways, particularly Rb, PI (3) K/Akt and p53. Prior studies identified TopBP1as a key mediator for the oncogenic gain-of-function activities of mutant p53 (mutp53) in cancer. In Akt-hyperactive cancer, TopBP1 forms oligomers and represses E2F1-dependent apoptosis. Here we perform a molecular docking screening and identify a lead compound, calcein, capable of blocking TopBP1 oligomerization and p53 binding, resulting in re-activation of E2F1-dependent apoptosis and blockade of mutp53 gain-of-function. Calcein AM, the cell permeable derivative of calcein, shows significant anti-tumor activity in a wide-spectrum of cultured cancer cells harboring high TopBP1 levels. These biochemical findings are recapitulated in breast cancer xenograft models. Thus, our study provides proof-of-concept evidence for targeting TopBP1, a convergent point of multiple pathways, as a cancer therapy.
Collapse
|
30
|
Ali A, Shah AS, Ahmad A. Gain-of-function of mutant p53: mutant p53 enhances cancer progression by inhibiting KLF17 expression in invasive breast carcinoma cells. Cancer Lett 2014; 354:87-96. [PMID: 25111898 DOI: 10.1016/j.canlet.2014.07.045] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/04/2014] [Accepted: 07/30/2014] [Indexed: 01/02/2023]
Abstract
Kruppel-like-factor 17 (KLF17) is a negative regulator of metastasis and epithelial-mesenchymal-transition (EMT). However, its expression is downregulated in metastatic breast cancer that contains p53 mutations. Here, we show that mutant-p53 plays a key role to suppress KLF17 and thereby enhances cancer progression, which defines novel gain-of-function (GOF) of mutant-p53. Mutant-p53 interacts with KLF17 and antagonizes KLF17 mediated EMT genes transcription. Depletion of KLF17 promotes cell viability, decreases apoptosis and induces drug resistance in metastatic breast cancer cells. KLF17 suppresses cell migration and invasion by decreasing CD44, PAI-1 and Cyclin-D1 expressions. Taken together, our results show that KLF17 is important for the suppression of metastasis and could be a potential therapeutic target during chemotherapy.
Collapse
Affiliation(s)
- Amjad Ali
- Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China.
| | - Abdus Saboor Shah
- Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China
| | - Ayaz Ahmad
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Department of Biotechnology, Abdul Wali Khan University, Mardan, 23200, Pakistan.
| |
Collapse
|
31
|
Kluth M, Harasimowicz S, Burkhardt L, Grupp K, Krohn A, Prien K, Gjoni J, Haß T, Galal R, Graefen M, Haese A, Simon R, Hühne-Simon J, Koop C, Korbel J, Weischenfeld J, Huland H, Sauter G, Quaas A, Wilczak W, Tsourlakis MC, Minner S, Schlomm T. Clinical significance of different types of p53 gene alteration in surgically treated prostate cancer. Int J Cancer 2014; 135:1369-80. [PMID: 24523142 DOI: 10.1002/ijc.28784] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 11/22/2013] [Accepted: 12/19/2013] [Indexed: 01/14/2023]
Abstract
Despite a multitude of p53 immunohistochemistry (IHC) studies, data on the combined effect of nuclear p53 protein accumulation and TP53 genomic inactivation are lacking for prostate cancer. A tissue microarray including 11,152 prostate cancer samples was analyzed by p53 IHC and fluorescence in situ hybridization. Nuclear p53 accumulation was found in 10.1% of patients including 1.4% with high-level and 8.7% with low-level immunostaining. TP53 sequencing revealed that 17 of 22 (77%) cases with high-level p53 immunostaining, but only 3% (1 of 31) low-level p53 cases carried putative dominant-negative mutations. TP53 deletions occurred in 14.8% of cancers. Both deletions and protein accumulation were linked to unfavorable tumor phenotype and prostate specific antigen (PSA) recurrence (p<0.0001 each). The combination of both methods revealed subgroups with remarkable differences in their clinical course. Tumors with either TP53 deletion (14%) or low-level p53 positivity (8.7%) had identical risks of PSA recurrence, which were markedly higher than in cancers without p53 alterations (p<0.0001). Tumors with both p53 deletion and low-level p53 positivity (1.5%) had a worse prognosis than patients with only one of these alterations (p<0.0001). Tumors with strong p53 immunostaining or homozygous inactivation through deletion of one allele and disrupting translocation involving the second allele had the worst outcome, independent from clinical and pathological parameters. These data demonstrate a differential clinical impact of various TP53 alterations in prostate cancer. Strong p53 immunostaining-most likely accompanying dominant negative or oncogenic p53 mutation-has independent prognostic relevance and may thus represent a clinical useful molecular feature of prostate cancer.
Collapse
Affiliation(s)
- Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Shetzer Y, Solomon H, Koifman G, Molchadsky A, Horesh S, Rotter V. The paradigm of mutant p53-expressing cancer stem cells and drug resistance. Carcinogenesis 2014; 35:1196-208. [PMID: 24658181 DOI: 10.1093/carcin/bgu073] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is well accepted that expression of mutant p53 involves the gain of oncogenic-specific activities accentuating the malignant phenotype. Depending on the specific cancer type, mutant p53 can contribute to either the early or the late events of the multiphase process underlying the transformation of a normal cell into a cancerous one. This multifactorial system is evident in ~50% of human cancers. Mutant p53 was shown to interfere with a variety of cellular functions that lead to augmented cell survival, cellular plasticity, aberration of DNA repair machinery and other effects. All these effects culminate in the acquisition of drug resistance often seen in cancer cells. Interestingly, drug resistance has also been suggested to be associated with cancer stem cells (CSCs), which reside within growing tumors. The notion that p53 plays a regulatory role in the life of stem cells, coupled with the observations that p53 mutations may contribute to the evolvement of CSCs makes it challenging to speculate that drug resistance and cancer recurrence are mediated by CSCs expressing mutant p53.
Collapse
Affiliation(s)
- Yoav Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hilla Solomon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gabriela Koifman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alina Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Stav Horesh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
33
|
Muller PAJ, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 2014; 25:304-17. [PMID: 24651012 PMCID: PMC3970583 DOI: 10.1016/j.ccr.2014.01.021] [Citation(s) in RCA: 1149] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/13/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022]
Abstract
Many different types of cancer show a high incidence of TP53 mutations, leading to the expression of mutant p53 proteins. There is growing evidence that these mutant p53s have both lost wild-type p53 tumor suppressor activity and gained functions that help to contribute to malignant progression. Understanding the functions of mutant p53 will help in the development of new therapeutic approaches that may be useful in a broad range of cancer types.
Collapse
Affiliation(s)
- Patricia A J Muller
- Medical Research Council Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK.
| | - Karen H Vousden
- CR-UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
34
|
Garcia PB, Attardi LD. Illuminating p53 function in cancer with genetically engineered mouse models. Semin Cell Dev Biol 2014; 27:74-85. [PMID: 24394915 DOI: 10.1016/j.semcdb.2013.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/06/2013] [Accepted: 12/23/2013] [Indexed: 01/23/2023]
Abstract
The key role of the p53 protein in tumor suppression is highlighted by its frequent mutation in human cancers and by the completely penetrant cancer predisposition of p53 null mice. Beyond providing definitive evidence for the critical function of p53 in tumor suppression, genetically engineered mouse models have offered numerous additional insights into p53 function. p53 knock-in mice expressing tumor-derived p53 mutants have revealed that these mutants display gain-of-function activities that actively promote carcinogenesis. The generation of p53 knock-in mutants with alterations in different domains of p53 has helped further elucidate the cellular and biochemical activities of p53 that are most fundamental for tumor suppression. In addition, modulation of p53 post-translational modification (PTM) status by generating p53 knock-in mouse strains with mutations in p53 PTM sites has revealed a subtlety and complexity to p53 regulation. Analyses of mouse models perturbing upstream regulators of p53 have solidified the notion that the p53 pathway can be compromised by means other than direct p53 mutation. Finally, switchable p53 models that allow p53 reactivation in tumors have helped evaluate the potential of p53 restoration therapy for cancer treatment. Collectively, mouse models have greatly enhanced our understanding of physiological p53 function and will continue to provide new biological and clinical insights in future investigations.
Collapse
Affiliation(s)
- Patty B Garcia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
35
|
Rustighi A, Zannini A, Tiberi L, Sommaggio R, Piazza S, Sorrentino G, Nuzzo S, Tuscano A, Eterno V, Benvenuti F, Santarpia L, Aifantis I, Rosato A, Bicciato S, Zambelli A, Del Sal G. Prolyl-isomerase Pin1 controls normal and cancer stem cells of the breast. EMBO Mol Med 2013; 6:99-119. [PMID: 24357640 PMCID: PMC3936488 DOI: 10.1002/emmm.201302909] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Mammary epithelial stem cells are fundamental to maintain tissue integrity. Cancer stem cells (CSCs) are implicated in both treatment resistance and disease relapse, and the molecular bases of their malignant properties are still poorly understood. Here we show that both normal stem cells and CSCs of the breast are controlled by the prolyl-isomerase Pin1. Mechanistically, following interaction with Pin1, Notch1 and Notch4, key regulators of cell fate, escape from proteasomal degradation by their major ubiquitin-ligase Fbxw7α. Functionally, we show that Fbxw7α acts as an essential negative regulator of breast CSCs' expansion by restraining Notch activity, but the establishment of a Notch/Pin1 active circuitry opposes this effect, thus promoting breast CSCs self-renewal, tumor growth and metastasis in vivo. In human breast cancers, despite Fbxw7α expression, high levels of Pin1 sustain Notch signaling, which correlates with poor prognosis. Suppression of Pin1 holds promise in reverting aggressive phenotypes, through CSC exhaustion as well as recovered drug sensitivity carrying relevant implications for therapy of breast cancers.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- F-Box Proteins/genetics
- F-Box Proteins/metabolism
- F-Box-WD Repeat-Containing Protein 7
- Female
- Humans
- Mammary Glands, Human/cytology
- Mice
- Mice, Knockout
- Mice, SCID
- NIMA-Interacting Peptidylprolyl Isomerase
- Neoplastic Stem Cells/cytology
- Neoplastic Stem Cells/metabolism
- Peptidylprolyl Isomerase/antagonists & inhibitors
- Peptidylprolyl Isomerase/genetics
- Peptidylprolyl Isomerase/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch4
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Stem Cells/cytology
- Stem Cells/metabolism
- Transplantation, Heterologous
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Alessandra Rustighi
- Laboratorio Nazionale CIB (LNCIB), Area Science ParkTrieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di TriesteTrieste, Italy
| | - Alessandro Zannini
- Laboratorio Nazionale CIB (LNCIB), Area Science ParkTrieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di TriesteTrieste, Italy
| | - Luca Tiberi
- Laboratorio Nazionale CIB (LNCIB), Area Science ParkTrieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di TriesteTrieste, Italy
| | - Roberta Sommaggio
- Dipartimento di Scienze Oncologiche e Chirurgiche, Università degli Studi di Padova e Istituto Oncologico Veneto IRCCSPadova, Italy
| | - Silvano Piazza
- Laboratorio Nazionale CIB (LNCIB), Area Science ParkTrieste, Italy
| | - Giovanni Sorrentino
- Laboratorio Nazionale CIB (LNCIB), Area Science ParkTrieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di TriesteTrieste, Italy
| | - Simona Nuzzo
- Center for Genome Research, Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio EmiliaModena, Italy
| | | | - Vincenzo Eterno
- Oncology Department IRCCS Fondazione Salvatore MaugeriPavia, Italy
| | - Federica Benvenuti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science ParkTrieste, Italy
| | - Libero Santarpia
- Translational Research Unit, Istituto Toscano TumoriPrato, Italy
| | - Iannis Aifantis
- Howard Hughes Medical Institute and Department of Pathology, NYU School of MedicineNew York, NY, USA
| | - Antonio Rosato
- Dipartimento di Scienze Oncologiche e Chirurgiche, Università degli Studi di Padova e Istituto Oncologico Veneto IRCCSPadova, Italy
| | - Silvio Bicciato
- Center for Genome Research, Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio EmiliaModena, Italy
| | - Alberto Zambelli
- Oncology Department IRCCS Fondazione Salvatore MaugeriPavia, Italy
| | - Giannino Del Sal
- Laboratorio Nazionale CIB (LNCIB), Area Science ParkTrieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di TriesteTrieste, Italy
- Corresponding author: Tel: +39 040 3756801; Fax +39 040 398990; E-mail:
| |
Collapse
|
36
|
Jin H, Liang L, Liu L, Deng W, Liu J. HDAC inhibitor DWP0016 activates p53 transcription and acetylation to inhibit cell growth in U251 glioblastoma cells. J Cell Biochem 2013; 114:1498-509. [PMID: 23297003 DOI: 10.1002/jcb.24491] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 12/18/2012] [Indexed: 11/11/2022]
Abstract
Here we report a hydroacid named DWP0016, which exhibited HDAC inhibition and induced p53 acetylation in U251 glioblastoma cells. DWP0016 effectively inhibited the cell growth of U251 cells and other four carcinoma cell lines but did not affect the normal cells. Cell cycle distribution analysis showed DWP0016 arrested at G1 phase cell cycle dose-dependently in U251 cells. DWP0016 induced caspase-dependent and independent apoptosis in U251 cells, which was identified by flow cytometry analysis, caspases activity analysis, Western blotting assay, and caspases inhibition. Mechanisms research suggested that DWP0016 activated transcription and acetylation of tumor suppressor p53. DWP0016 regulated p300, CBP, and PCAF to facilitate p53 acetylation at lys382 in U251 cells. In addition, activation of p53 by DWP0016 promoted PUMA to catalyze mitochondrial pathway. Besides, siRNA assay indicated p53 was the key gene to induce growth inhibition, cell cycle arrest, and apoptosis in DWP0016 treated U251 cells. Conclusively, our results show DWP0016 is a potent HDAC inhibitor and the anti-tumor activity is consistent with its intended p53 activation mechanisms. These findings indicate the promising antitumor potential of DWP0016 for further glioblastoma treatment applications.
Collapse
Affiliation(s)
- Hui Jin
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
37
|
Mello SS, Attardi LD. Not all p53 gain-of-function mutants are created equal. Cell Death Differ 2013; 20:855-7. [PMID: 23749181 DOI: 10.1038/cdd.2013.53] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
38
|
Effects of MDM2 inhibitors on vascular endothelial growth factor-mediated tumor angiogenesis in human breast cancer. Angiogenesis 2013; 17:37-50. [PMID: 23907365 DOI: 10.1007/s10456-013-9376-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/29/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND Mouse double minute 2 (MDM2) is overexpressed in many malignant tumors, and MDM2 levels are associated with poor prognosis of several human cancers, including breast cancer. In the present study, we investigated the function of MDM2 in vascular endothelial growth factor (VEGF)-mediated tumor angiogenesis of breast cancer and the potential value of MDM2 as an anti-angiogenic therapy target for cancer therapy by inhibiting MDM2 with antisense oligonucleotides (ASO) or other antagonist nutlin-3. METHODS Anti-MDM2 ASO and nutlin-3 were evaluated for their in vitro and in vivo anti-angiogenesis activities in different human breast cancer models with a different p53 status: MCF-7 cell line containing wild-type p53 and MDA-MB-468 cell line containing mutant p53. MCF-7 and MDA-MB-468 cells were incubated with different concentrations of ASO or nutlin-3 for various periods of time. VEGF gene and protein expression in tumor cells was measured by qPCR and Western blot. The level of VEGF protein secreted in the culture supernatant of treated cells was quantified by enzyme-linked immunosorbent assay (ELISA). Nude mouse xenograft models were further established to determine their effects on tumor growth and angiogenesis. Serum levels of VEGF were measured by ELISA. VEGF expression and microvessel density in tumor tissues were studied by immunohistochemistry. Both angiogenesis and tumor growth were digitally quantified. RESULTS In both MCF-7 and MDA-MB-468 cells, VEGF expression and secretion were reduced, resulting from specific inhibition of MDM2 expression by ASO. In vivo assay, after administration of ASO, VEGF production reduced and anti-angiogenesis activity occurred in nude mice bearing MCF-7 or MDA-MB-468 xenograft. However, in both models treated with nutlin-3, VEGF production was not changed and anti-angiogenesis activity was not observed. CONCLUSION In summary, the ASO construct targeting MDM2 specifically suppresses VEGF expression in vitro and VEGF-mediated tumor angiogenesis in vivo in breast cancer. Furthermore, the suppression of VEGF expression subsequent to inhibition of MDM2 in p53 mutant cells suggests that MDM2 has a regulatory role on VEGF expression through a p53-independent mechanism.
Collapse
|
39
|
Grugan KD, Vega ME, Wong GS, Diehl JA, Bass AJ, Wong KK, Nakagawa H, Rustgi AK. A common p53 mutation (R175H) activates c-Met receptor tyrosine kinase to enhance tumor cell invasion. Cancer Biol Ther 2013; 14:853-9. [PMID: 23792586 DOI: 10.4161/cbt.25406] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive forms of human cancer with poor prognosis due to late diagnosis and metastasis. Common genomic alterations in ESCC include p53 mutation, p120ctn inactivation, and overexpression of oncogenes such as cyclin D1, EGFR, and c-Met. Using esophageal epithelial cells transformed by the overexpression of EGFR and p53(R175H), we find novel evidence of a functional link between p53(R175H) and the c-Met receptor tyrosine kinase to mediate tumor cell invasion. Increased c-Met receptor activation was observed upon p53(R175H) expression and enhanced further upon subsequent EGFR overexpression. We inhibited c-Met phosphorylation, resulting in diminished invasion of the genetically transformed primary esophageal epithelial cells (EPC-hTERT-EGFR-p53(R175H)), suggesting that the mechanism of increased invasiveness upon EGFR and p53(R175H) expression may be the result of increased c-Met activation. These results suggest that the use of therapeutics directed at c-Met in ESCC and other squamous cell cancers.
Collapse
Affiliation(s)
- Katharine D Grugan
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Maria E Vega
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Gabrielle S Wong
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - J Alan Diehl
- The Abramson Family Cancer Research Institute; University of Pennsylvania; Philadelphia, PA USA
| | - Adam J Bass
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston, MA USA
| | - Kwok K Wong
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston, MA USA
| | - Hiroshi Nakagawa
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA
| | - Anil K Rustgi
- Gastroenterology Division; Department of Medicine; University of Pennsylvania; Philadelphia, PA USA; Abramson Cancer Center; University of Pennsylvania; Philadelphia, PA USA; Department of Genetics; University of Pennsylvania; Philadelphia, PA USA
| |
Collapse
|