1
|
Scimeca M, Rovella V, Palumbo V, Scioli MP, Bonfiglio R, Tor Centre, Melino G, Piacentini M, Frati L, Agostini M, Candi E, Mauriello A. Programmed Cell Death Pathways in Cholangiocarcinoma: Opportunities for Targeted Therapy. Cancers (Basel) 2023; 15:3638. [PMID: 37509299 PMCID: PMC10377326 DOI: 10.3390/cancers15143638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cholangiocarcinoma is a highly aggressive cancer arising from the bile ducts. The limited effectiveness of conventional therapies has prompted the search for new approaches to target this disease. Recent evidence suggests that distinct programmed cell death mechanisms, namely, apoptosis, ferroptosis, pyroptosis and necroptosis, play a critical role in the development and progression of cholangiocarcinoma. This review aims to summarize the current knowledge on the role of programmed cell death in cholangiocarcinoma and its potential implications for the development of novel therapies. Several studies have shown that the dysregulation of apoptotic signaling pathways contributes to cholangiocarcinoma tumorigenesis and resistance to treatment. Similarly, ferroptosis, pyroptosis and necroptosis, which are pro-inflammatory forms of cell death, have been implicated in promoting immune cell recruitment and activation, thus enhancing the antitumor immune response. Moreover, recent studies have suggested that targeting cell death pathways could sensitize cholangiocarcinoma cells to chemotherapy and immunotherapy. In conclusion, programmed cell death represents a relevant molecular mechanism of pathogenesis in cholangiocarcinoma, and further research is needed to fully elucidate the underlying details and possibly identify therapeutic strategies.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valentina Rovella
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Palumbo
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Paola Scioli
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Rita Bonfiglio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Luigi Frati
- Institute Pasteur Italy-Cenci Bolognetti Foundation, Via Regina Elena 291, 00161 Rome, Italy
- IRCCS Neuromed S.p.A., Via Atinense 18, 86077 Pozzilli, Italy
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
2
|
Logotheti S, Pavlopoulou A, Marquardt S, Takan I, Georgakilas AG, Stiewe T. p73 isoforms meet evolution of metastasis. Cancer Metastasis Rev 2022; 41:853-869. [PMID: 35948758 DOI: 10.1007/s10555-022-10057-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/30/2022] [Indexed: 01/25/2023]
Abstract
Cancer largely adheres to Darwinian selection. Evolutionary forces are prominent during metastasis, the final and incurable disease stage, where cells acquire combinations of advantageous phenotypic features and interact with a dynamically changing microenvironment, in order to overcome the metastatic bottlenecks, while therapy exerts additional selective pressures. As a strategy to increase their fitness, tumors often co-opt developmental and tissue-homeostasis programs. Herein, 25 years after its discovery, we review TP73, a sibling of the cardinal tumor-suppressor TP53, through the lens of cancer evolution. The TP73 gene regulates a wide range of processes in embryonic development, tissue homeostasis and cancer via an overwhelming number of functionally divergent isoforms. We suggest that TP73 neither merely mimics TP53 via its p53-like tumor-suppressive functions, nor has black-or-white-type effects, as inferred by the antagonism between several of its isoforms in processes like apoptosis and DNA damage response. Rather, under dynamic conditions of selective pressure, the various p73 isoforms which are often co-expressed within the same cancer cells may work towards a common goal by simultaneously activating isoform-specific transcriptional and non-transcriptional programs. Combinatorial co-option of these programs offers selective advantages that overall increase the likelihood for successfully surpassing the barriers of the metastatic cascade. The p73 functional pleiotropy-based capabilities might be present in subclonal populations and expressed dynamically under changing microenvironmental conditions, thereby supporting clonal expansion and propelling evolution of metastasis. Deciphering the critical p73 isoform patterns along the spatiotemporal axes of tumor evolution could identify strategies to target TP73 for prevention and therapy of cancer metastasis.
Collapse
Affiliation(s)
- Stella Logotheti
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), 15780, Zografou, Greece.
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center (IBG), 35340, Balcova, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340, Balcova, Izmir, Turkey
| | - Stephan Marquardt
- Institute of Translational Medicine for Health Care Systems, Medical School Berlin, Hochschule Für Gesundheit Und Medizin, 14197, Berlin, Germany
| | - Işıl Takan
- Izmir Biomedicine and Genome Center (IBG), 35340, Balcova, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340, Balcova, Izmir, Turkey
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), 15780, Zografou, Greece
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany.,Institute of Lung Health, Giessen, Germany.,German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| |
Collapse
|
3
|
Angelopoulos I, Gakis G, Birmpas K, Kyrousi C, Habeos EE, Kaplani K, Lygerou Z, Habeos I, Taraviras S. Metabolic regulation of the neural stem cell fate: Unraveling new connections, establishing new concepts. Front Neurosci 2022; 16:1009125. [PMID: 36340763 PMCID: PMC9634649 DOI: 10.3389/fnins.2022.1009125] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
The neural stem cell niche is a key regulator participating in the maintenance, regeneration, and repair of the brain. Within the niche neural stem cells (NSC) generate new neurons throughout life, which is important for tissue homeostasis and brain function. NSCs are regulated by intrinsic and extrinsic factors with cellular metabolism being lately recognized as one of the most important ones, with evidence suggesting that it may serve as a common signal integrator to ensure mammalian brain homeostasis. The aim of this review is to summarize recent insights into how metabolism affects NSC fate decisions in adult neural stem cell niches, with occasional referencing of embryonic neural stem cells when it is deemed necessary. Specifically, we will highlight the implication of mitochondria as crucial regulators of NSC fate decisions and the relationship between metabolism and ependymal cells. The link between primary cilia dysfunction in the region of hypothalamus and metabolic diseases will be examined as well. Lastly, the involvement of metabolic pathways in ependymal cell ciliogenesis and physiology regulation will be discussed.
Collapse
Affiliation(s)
| | - Georgios Gakis
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Kyriakos Birmpas
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Christina Kyrousi
- First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Evagelia Eva Habeos
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Konstantina Kaplani
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
| | - Ioannis Habeos
- Division of Endocrinology, Department of Internal Medicine, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
- *Correspondence: Stavros Taraviras,
| |
Collapse
|
4
|
Pereira IC, Mascarenhas IF, Capetini VC, Ferreira PMP, Rogero MM, Torres-Leal FL. Cellular reprogramming, chemoresistance, and dietary interventions in breast cancer. Crit Rev Oncol Hematol 2022; 179:103796. [PMID: 36049616 DOI: 10.1016/j.critrevonc.2022.103796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/16/2022] [Accepted: 08/21/2022] [Indexed: 10/31/2022] Open
Abstract
Breast cancer (BC) diagnosis has been associated with significant risk factors, including family history, late menopause, obesity, poor eating habits, and alcoholism. Despite the advances in the last decades regarding cancer treatment, some obstacles still hinder the effectiveness of therapy. For example, chemotherapy resistance is common in locally advanced or metastatic cancer, reducing treatment options and contributing to mortality. In this review, we provide an overview of BC metabolic changes, including the impact of restrictive diets associated with chemoresistance, the therapeutic potential of the diet on tumor progression, pathways related to metabolic health in oncology, and perspectives on the future in the area of oncological nutrition.
Collapse
Affiliation(s)
- Irislene Costa Pereira
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil; Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Isabele Frazão Mascarenhas
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | - Paulo Michel Pinheiro Ferreira
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, Sao Paulo, Brazil
| | - Francisco Leonardo Torres-Leal
- Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil; Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil.
| |
Collapse
|
5
|
Lena AM, Foffi E, Agostini M, Mancini M, Annicchiarico-Petruzzelli M, Aberdam D, Velletri T, Shi Y, Melino G, Wang Y, Candi E. TAp63 regulates bone remodeling by modulating the expression of TNFRSF11B/Osteoprotegerin. Cell Cycle 2021; 20:2428-2441. [PMID: 34763601 DOI: 10.1080/15384101.2021.1985772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ABBREVIATIONS MSC, mesenchymal stem cells; OPG, osteoprotegerin; RUNX2, Run-trelated transcription factor 2.
Collapse
Affiliation(s)
- Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Erica Foffi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy
| | | | | | | | - Tania Velletri
- Cogentech Società Benefit Srl, Parco Scientifico E Tecnologico Della Sicilia, Catania, Italy
| | - Yufang Shi
- Cas Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Ying Wang
- Cas Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy.,IDI-IRCCS, Via dei Monti di Creta, Rome, IT
| |
Collapse
|
6
|
Rozenberg JM, Zvereva S, Dalina A, Blatov I, Zubarev I, Luppov D, Bessmertnyi A, Romanishin A, Alsoulaiman L, Kumeiko V, Kagansky A, Melino G, Ganini C, Barlev NA. The p53 family member p73 in the regulation of cell stress response. Biol Direct 2021; 16:23. [PMID: 34749806 PMCID: PMC8577020 DOI: 10.1186/s13062-021-00307-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
During oncogenesis, cells become unrestrictedly proliferative thereby altering the tissue homeostasis and resulting in subsequent hyperplasia. This process is paralleled by resumption of cell cycle, aberrant DNA repair and blunting the apoptotic program in response to DNA damage. In most human cancers these processes are associated with malfunctioning of tumor suppressor p53. Intriguingly, in some cases two other members of the p53 family of proteins, transcription factors p63 and p73, can compensate for loss of p53. Although both p63 and p73 can bind the same DNA sequences as p53 and their transcriptionally active isoforms are able to regulate the expression of p53-dependent genes, the strongest overlap with p53 functions was detected for p73. Surprisingly, unlike p53, the p73 is rarely lost or mutated in cancers. On the contrary, its inactive isoforms are often overexpressed in cancer. In this review, we discuss several lines of evidence that cancer cells develop various mechanisms to repress p73-mediated cell death. Moreover, p73 isoforms may promote cancer growth by enhancing an anti-oxidative response, the Warburg effect and by repressing senescence. Thus, we speculate that the role of p73 in tumorigenesis can be ambivalent and hence, requires new therapeutic strategies that would specifically repress the oncogenic functions of p73, while keeping its tumor suppressive properties intact.
Collapse
Affiliation(s)
- Julian M Rozenberg
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| | - Svetlana Zvereva
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksandra Dalina
- The Engelhardt Institute of Molecular Biology, Russian Academy of Science, Moscow, Russia
| | - Igor Blatov
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Ilya Zubarev
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daniil Luppov
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | | | - Alexander Romanishin
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia.,School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Lamak Alsoulaiman
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Alexander Kagansky
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Gerry Melino
- Department of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Ganini
- Department of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nikolai A Barlev
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia. .,Institute of Cytology, Russian Academy of Science, Saint-Petersburg, Russia.
| |
Collapse
|
7
|
Sun Q, Melino G, Amelio I, Jiang J, Wang Y, Shi Y. Recent advances in cancer immunotherapy. Discov Oncol 2021; 12:27. [PMID: 35201440 PMCID: PMC8777500 DOI: 10.1007/s12672-021-00422-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/05/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy represents a major advance in the cure of cancer following the dramatic advancements in the development and refinement of chemotherapies and radiotherapies. In the recent decades, together with the development of early diagnostic techniques, immunotherapy has significantly contributed to improving the survival of cancer patients. The immune-checkpoint blockade agents have been proven effective in a significant fraction of standard therapy refractory patients. Importantly, recent advances are providing alternative immunotherapeutic tools that could help overcome their limitations. In this mini review, we provide an overview on the main steps of the discovery of classic immune-checkpoint blockade agents and summarise the most recent development of novel immunotherapeutic strategies, such as tumour antigens, bispecific antibodies and TCR-engineered T cells.
Collapse
Affiliation(s)
- Qiang Sun
- Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, China
- Research Unit of Cell Death Mechanism, Chinese Academy of Medical Science, Beijing, China
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
- DZNE German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Jingting Jiang
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Yufang Shi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| |
Collapse
|
8
|
Panatta E, Zampieri C, Melino G, Amelio I. Understanding p53 tumour suppressor network. Biol Direct 2021; 16:14. [PMID: 34362419 PMCID: PMC8348811 DOI: 10.1186/s13062-021-00298-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
The mutation of TP53 gene affects half of all human cancers, resulting in impairment of the regulation of several cellular functions, including cell cycle progression and cell death in response to genotoxic stress. In the recent years additional p53-mediated tumour suppression mechanisms have been described, questioning the contribution of its canonical pathway for tumour suppression. These include regulation of alternative cell death modalities (i.e. ferroptosis), cell metabolism and the emerging role in RNA stability. Here we briefly summarize our knowledge on p53 “canonical DNA damage response” and discuss the most relevant recent findings describing potential mechanistic explanation of p53-mediated tumour suppression.
Collapse
Affiliation(s)
- Emanuele Panatta
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Carlotta Zampieri
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy. .,School of Life Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
9
|
p53/p73 Protein Network in Colorectal Cancer and Other Human Malignancies. Cancers (Basel) 2021; 13:cancers13122885. [PMID: 34207603 PMCID: PMC8227208 DOI: 10.3390/cancers13122885] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The p53 family of proteins comprises p53, p63, and p73, which share high structural and functional similarity. The two distinct promoters of each locus, the alternative splicing, and the alternative translation initiation sites enable the generation of numerous isoforms with different protein-interacting domains and distinct activities. The co-expressed p53/p73 isoforms have significant but distinct roles in carcinogenesis. Their activity is frequently impaired in human tumors including colorectal carcinoma due to dysregulated expression and a dominant-negative effect accomplished by some isoforms and p53 mutants. The interactions between isoforms are particularly important to understand the onset of tumor formation, progression, and therapeutic response. The understanding of the p53/p73 network can contribute to the development of new targeted therapies. Abstract The p53 tumor suppressor protein is crucial for cell growth control and the maintenance of genomic stability. Later discovered, p63 and p73 share structural and functional similarity with p53. To understand the p53 pathways more profoundly, all family members should be considered. Each family member possesses two promoters and alternative translation initiation sites, and they undergo alternative splicing, generating multiple isoforms. The resulting isoforms have important roles in carcinogenesis, while their expression is dysregulated in several human tumors including colorectal carcinoma, which makes them potential targets in cancer treatment. Their activities arise, at least in part, from the ability to form tetramers that bind to specific DNA sequences and activate the transcription of target genes. In this review, we summarize the current understanding of the biological activities and regulation of the p53/p73 isoforms, highlighting their role in colorectal tumorigenesis. The analysis of the expression patterns of the p53/p73 isoforms in human cancers provides an important step in the improvement of cancer therapy. Furthermore, the interactions among the p53 family members which could modulate normal functions of the canonical p53 in tumor tissue are described. Lastly, we emphasize the importance of clinical studies to assess the significance of combining the deregulation of different members of the p53 family to define the outcome of the disease.
Collapse
|
10
|
Celardo I, Melino G, Amelio I. Commensal microbes and p53 in cancer progression. Biol Direct 2020; 15:25. [PMID: 33213502 PMCID: PMC7678320 DOI: 10.1186/s13062-020-00281-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Aetiogenesis of cancer has not been fully determined. Recent advances have clearly defined a role for microenvironmental factors in cancer progression and initiation; in this context, microbiome has recently emerged with a number of reported correlative and causative links implicating alterations of commensal microbes in tumorigenesis. Bacteria appear to have the potential to directly alter physiological pathways of host cells and in specific circumstances, such as the mutation of the tumour suppressive factor p53, they can also directly switch the function of a gene from oncosuppressive to oncogenic. In this minireview, we report a number of examples on how commensal microbes alter the host cell biology, affecting the oncogenic process. We then discuss more in detail how interaction with the gut microbiome can affect the function of p53 mutant in the intestinal tumorigenesis.
Collapse
Affiliation(s)
- Ivana Celardo
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy.
- School of Life Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
11
|
Amelio I, Bertolo R, Bove P, Candi E, Chiocchi M, Cipriani C, Di Daniele N, Ganini C, Juhl H, Mauriello A, Marani C, Marshall J, Montanaro M, Palmieri G, Piacentini M, Sica G, Tesauro M, Rovella V, Tisone G, Shi Y, Wang Y, Melino G. Cancer predictive studies. Biol Direct 2020; 15:18. [PMID: 33054808 PMCID: PMC7557058 DOI: 10.1186/s13062-020-00274-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 12/21/2022] Open
Abstract
The identification of individual or clusters of predictive genetic alterations might help in defining the outcome of cancer treatment, allowing for the stratification of patients into distinct cohorts for selective therapeutic protocols. Neuroblastoma (NB) is the most common extracranial childhood tumour, clinically defined in five distinct stages (1–4 & 4S), where stages 3–4 define chemotherapy-resistant, highly aggressive disease phases. NB is a model for geneticists and molecular biologists to classify genetic abnormalities and identify causative disease genes. Despite highly intensive basic research, improvements on clinical outcome have been predominantly observed for less aggressive cancers, that is stages 1,2 and 4S. Therefore, stages 3–4 NB are still complicated at the therapeutic level and require more intense fundamental research. Using neuroblastoma as a model system, here we herein outline how cancer prediction studies can help at steering preclinical and clinical research toward the identification and exploitation of specific genetic landscape. This might result in maximising the therapeutic success and minimizing harmful effects in cancer patients.
Collapse
Affiliation(s)
- Ivano Amelio
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.
| | - Riccardo Bertolo
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.,San Carlo di Nancy Hospital, Rome, Italy
| | - Pierluigi Bove
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.,San Carlo di Nancy Hospital, Rome, Italy
| | - Eleonora Candi
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Marcello Chiocchi
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Chiara Cipriani
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.,San Carlo di Nancy Hospital, Rome, Italy
| | - Nicola Di Daniele
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Carlo Ganini
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | | | - Alessandro Mauriello
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Carla Marani
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.,San Carlo di Nancy Hospital, Rome, Italy
| | - John Marshall
- Medstar Georgetown University Hospital, Georgetown University, Washington DC, USA
| | - Manuela Montanaro
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Giampiero Palmieri
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Mauro Piacentini
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Giuseppe Sica
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Manfredi Tesauro
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Valentina Rovella
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Giuseppe Tisone
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Yufang Shi
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Gerry Melino
- Torvergata Oncoscience Research Centre of Excellence, TOR, Department of Experimental Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
12
|
The Janus-like role of proline metabolism in cancer. Cell Death Discov 2020; 6:104. [PMID: 33083024 PMCID: PMC7560826 DOI: 10.1038/s41420-020-00341-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/18/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
The metabolism of the non-essential amino acid L-proline is emerging as a key pathway in the metabolic rewiring that sustains cancer cells proliferation, survival and metastatic spread. Pyrroline-5-carboxylate reductase (PYCR) and proline dehydrogenase (PRODH) enzymes, which catalyze the last step in proline biosynthesis and the first step of its catabolism, respectively, have been extensively associated with the progression of several malignancies, and have been exposed as potential targets for anticancer drug development. As investigations into the links between proline metabolism and cancer accumulate, the complexity, and sometimes contradictory nature of this interaction emerge. It is clear that the role of proline metabolism enzymes in cancer depends on tumor type, with different cancers and cancer-related phenotypes displaying different dependencies on these enzymes. Unexpectedly, the outcome of rewiring proline metabolism also differs between conditions of nutrient and oxygen limitation. Here, we provide a comprehensive review of proline metabolism in cancer; we collate the experimental evidence that links proline metabolism with the different aspects of cancer progression and critically discuss the potential mechanisms involved.
Collapse
|
13
|
Niklison-Chirou MV, Agostini M, Amelio I, Melino G. Regulation of Adult Neurogenesis in Mammalian Brain. Int J Mol Sci 2020; 21:ijms21144869. [PMID: 32660154 PMCID: PMC7402357 DOI: 10.3390/ijms21144869] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Adult neurogenesis is a multistage process by which neurons are generated and integrated into existing neuronal circuits. In the adult brain, neurogenesis is mainly localized in two specialized niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) adjacent to the lateral ventricles. Neurogenesis plays a fundamental role in postnatal brain, where it is required for neuronal plasticity. Moreover, perturbation of adult neurogenesis contributes to several human diseases, including cognitive impairment and neurodegenerative diseases. The interplay between extrinsic and intrinsic factors is fundamental in regulating neurogenesis. Over the past decades, several studies on intrinsic pathways, including transcription factors, have highlighted their fundamental role in regulating every stage of neurogenesis. However, it is likely that transcriptional regulation is part of a more sophisticated regulatory network, which includes epigenetic modifications, non-coding RNAs and metabolic pathways. Here, we review recent findings that advance our knowledge in epigenetic, transcriptional and metabolic regulation of adult neurogenesis in the SGZ of the hippocampus, with a special attention to the p53-family of transcription factors.
Collapse
Affiliation(s)
- Maria Victoria Niklison-Chirou
- Centre for Therapeutic Innovation (CTI-Bath), Department of Pharmacy & Pharmacology, University of Bath, Bath BA2 7AY, UK;
- Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.A.); (I.A.)
| | - Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.A.); (I.A.)
- School of Life Sciences, University of Nottingham, Nottingham NG7 2HU, UK
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.A.); (I.A.)
- Correspondence:
| |
Collapse
|
14
|
Fritsche-Guenther R, Gloaguen Y, Kirchner M, Mertins P, Tunn PU, Kirwan JA. Progression-Dependent Altered Metabolism in Osteosarcoma Resulting in Different Nutrient Source Dependencies. Cancers (Basel) 2020; 12:cancers12061371. [PMID: 32471029 PMCID: PMC7352851 DOI: 10.3390/cancers12061371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor and OS metastases are mostly found in the lung. The limited understanding of the biology of metastatic processes in OS limits the ability for effective treatment. Alterations to the metabolome and its transformation during metastasis aids the understanding of the mechanism and provides information on treatment and prognosis. The current study intended to identify metabolic alterations during OS progression by using a targeted gas chromatography mass spectrometry approach. Using a female OS cell line model, malignant and metastatic cells increased their energy metabolism compared to benign OS cells. The metastatic cell line showed a faster metabolic flux compared to the malignant cell line, leading to reduced metabolite pools. However, inhibiting both glycolysis and glutaminolysis resulted in a reduced proliferation. In contrast, malignant but non-metastatic OS cells showed a resistance to glycolytic inhibition but a strong dependency on glutamine as an energy source. Our in vivo metabolic approach hinted at a potential sex-dependent metabolic alteration in OS patients with lung metastases (LM), although this will require validation with larger sample sizes. In line with the in vitro results, we found that female LM patients showed a decreased central carbon metabolism compared to metastases from male patients.
Collapse
Affiliation(s)
- Raphaela Fritsche-Guenther
- Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), 13125 Berlin, Germany; (Y.G.); (J.A.K.)
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany; (M.K.); (P.M.)
- Correspondence:
| | - Yoann Gloaguen
- Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), 13125 Berlin, Germany; (Y.G.); (J.A.K.)
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany; (M.K.); (P.M.)
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany; (M.K.); (P.M.)
- Proteomics Platform Berlin Institute of Health (BIH) and Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany; (M.K.); (P.M.)
- Proteomics Platform Berlin Institute of Health (BIH) and Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
| | - Per-Ulf Tunn
- Department of Orthopedic Oncology, Helios Clinic Berlin-Buch, 13125 Berlin, Germany;
| | - Jennifer A. Kirwan
- Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), 13125 Berlin, Germany; (Y.G.); (J.A.K.)
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany; (M.K.); (P.M.)
| |
Collapse
|
15
|
Amelio I, Melino G. Context is everything: extrinsic signalling and gain-of-function p53 mutants. Cell Death Discov 2020; 6:16. [PMID: 32218993 PMCID: PMC7090043 DOI: 10.1038/s41420-020-0251-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023] Open
Abstract
The TP53 genomic locus is a target of mutational events in at least half of cancers. Despite several decades of study, a full consensus on the relevance of the acquisition of p53 gain-of-function missense mutants has not been reached. Depending on cancer type, type of mutations and other unidentified factors, the relevance for tumour development and progression of the oncogenic signalling directed by p53 mutants might significantly vary, leading to inconsistent observations that have fuelled a long and fierce debate in the field. Here, we discuss how interaction with the microenvironment and stressors might dictate the gain-of-function effects exerted by individual mutants. We report evidence from the most recent literature in support of the context dependency of p53 mutant biology. This perspective article aims to raise a discussion in the field on the relevance that context might have on p53 gain-of-function mutants, assessing whether this should generally be considered a cell non-autonomous process.
Collapse
Affiliation(s)
- Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome ’’Tor Vergata”, 00133 Rome, Italy
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome ’’Tor Vergata”, 00133 Rome, Italy
- Toxicology Unit, University of Cambridge, Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP UK
| |
Collapse
|
16
|
Pham TD, Fan C, Pfeifer D, Zhang H, Sun XF. Image-Based Network Analysis of DNp73 Expression by Immunohistochemistry in Rectal Cancer Patients. Front Physiol 2020; 10:1551. [PMID: 31969833 PMCID: PMC6960186 DOI: 10.3389/fphys.2019.01551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 12/09/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Rectal cancer is a disease characterized with tumor heterogeneity. The combination of surgery, radiotherapy, and chemotherapy can reduce the risk of local recurrence. However, there is a significant difference in the response to radiotherapy among rectal cancer patients even they have the same tumor stage. Despite rapid advances in knowledge of cellular functions affecting radiosensitivity, there is still a lack of predictive factors for local recurrence and normal tissue damage. The tumor protein DNp73 is thought as a biomarker in colorectal cancer, but its clinical significance is still not sufficiently investigated, mainly due to the limitation of human-based pathology analysis. In this study, we investigated the predictive value of DNp73 in patients with rectal adenocarcinoma using image-based network analysis. Methods: The fuzzy weighted recurrence network of time series was extended to handle multi-channel image data, and applied to the analysis of immunohistochemistry images of DNp73 expression obtained from a cohort of 25 rectal cancer patients who underwent radiotherapy before surgery. Two mathematical weighted network properties, which are the clustering coefficient and characteristic path length, were computed for the image-based networks of the primary tumor (obtained after operation) and biopsy (obtained before operation) of each cancer patient. Results: The ratios of two weighted recurrence network properties of the primary tumors to biopsies reveal the correlation of DNp73 expression and long survival time, and discover the non-effective radiotherapy to a cohort of rectal cancer patients who had short survival time. Conclusion: Our work contributes to the elucidation of the predictive value of DNp73 expression in rectal cancer patients who were given preoperative radiotherapy. Mathematical properties of fuzzy weighted recurrence networks of immunohistochemistry images are not only able to show the predictive factor of DNp73 expression in the patients, but also reveal the identification of non-effective application of radiotherapy to those who had poor overall survival outcome.
Collapse
Affiliation(s)
- Tuan D Pham
- Department of Biomedical Engineering, Linkoping University, Linkoping, Sweden.,The Center for Artificial Intelligence, Prince Mohammad Bin Fahd University, Al Khobar, Saudi Arabia
| | - Chuanwen Fan
- Department of Oncology, Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden.,Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Daniella Pfeifer
- Department of Oncology, Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| | - Hong Zhang
- Department of Medical Sciences, Orebro University, Orebro, Sweden
| | - Xiao-Feng Sun
- Department of Oncology, Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| |
Collapse
|
17
|
Lopriore P, Capitanio N, Panatta E, Di Daniele N, Gambacurta A, Melino G, Amelio I. TAp73 regulates ATP7A: possible implications for ageing-related diseases. Aging (Albany NY) 2019; 10:3745-3760. [PMID: 30530920 PMCID: PMC6326685 DOI: 10.18632/aging.101669] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
The p53 family member p73 controls a wide range of cellular function. Deletion of p73 in mice results in increased tumorigenesis, infertility, neurological defects and altered immune system. Despite the extensive effort directed to define the molecular underlying mechanism of p73 function a clear definition of its transcriptional signature and the extent of overlap with the other p53 family members is still missing. Here we describe a novel TAp73 target, ATP7A a member of a large family of P-type ATPases implicated in human neurogenerative conditions and cancer chemoresistance. Modulation of TAp73 expression influences basal expression level of ATP7A in different cellular models and chromatin immunoprecipitation confirmed a physical direct binding of TAp73 on ATP7A genomic regions. Bioinformatic analysis of expression profile datasets of human lung cancer patients suggests a possible implication of TAp73/ATP7A axis in human cancer. These data provide a novel TAp73-dependent target which might have implications in ageing-related diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Piervito Lopriore
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 7HB, United Kingdom.,Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Emanuele Panatta
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 7HB, United Kingdom
| | - Nicola Di Daniele
- Department of Systems Medicine, Nephrology and Hypertension Unit, Tor Vergata University Hospital, Rome, Italy
| | - Alessandra Gambacurta
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Gerry Melino
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 7HB, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Ivano Amelio
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 7HB, United Kingdom
| |
Collapse
|
18
|
Nemajerova A, Moll UM. Tissue-specific roles of p73 in development and homeostasis. J Cell Sci 2019; 132:132/19/jcs233338. [PMID: 31582429 DOI: 10.1242/jcs.233338] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
p73 (TP73) belongs to the p53 family of transcription factors. Its gene locus encodes two opposing types of isoforms, the transcriptionally active TAp73 class and the dominant-negative DNp73 class, which both play critical roles in development and homeostasis in an astonishingly diverse array of biological systems within specific tissues. While p73 has functions in cancer, this Review focuses on the non-oncogenic activities of p73. In the central and peripheral nervous system, both isoforms cooperate in complex ways to regulate neural stem cell survival, self-renewal and terminal differentiation. In airways, oviduct and to a lesser extent in brain ependyma, TAp73 is the master transcriptional regulator of multiciliogenesis, enabling fluid and germ cell transport across tissue surfaces. In male and female reproduction, TAp73 regulates gene networks that control cell-cell adhesion programs within germinal epithelium to enable germ cell maturation. Finally, p73 participates in the control of angiogenesis in development and cancer. While many open questions remain, we discuss here key findings that provide insight into the complex functions of this gene at the organismal, cellular and molecular level.
Collapse
Affiliation(s)
- Alice Nemajerova
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8691, USA
| | - Ute M Moll
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8691, USA
| |
Collapse
|
19
|
Palombo R, Caporali S, Falconi M, Iacovelli F, Morozzo Della Rocca B, Lo Surdo A, Campione E, Candi E, Melino G, Bernardini S, Terrinoni A. Luteolin-7- O-β-d-Glucoside Inhibits Cellular Energy Production Interacting with HEK2 in Keratinocytes. Int J Mol Sci 2019; 20:ijms20112689. [PMID: 31159225 PMCID: PMC6600217 DOI: 10.3390/ijms20112689] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 01/10/2023] Open
Abstract
Flavonoids have been demonstrated to affect the activity of many mammalian enzyme systems. Their functional phenolic groups are able to mediate antioxidant effects by scavenging free radicals. Molecules of this class have been found able to modulate the activity of kinases, phospholipase A2, cyclooxygenases, lipoxygenase, glutathione S-transferase, and many others. Recently, it has been demonstrated that luteolin, in the form of Luteolin-7-O-β-d-glucoside (LUT-7G) is able to induce the keratinocyte differentiation process in vitro. This flavonoid is able to counteract the proliferative effects of IL-22/IL6 pathway by the inhibition of STAT3 activity also in vivo in a psoriatic mouse model. Observations on energy metabolism changes of differentiating cells led us to perform a complete metabolomics analysis using human primary keratinocytes treated with LUT-7G. Our results show that LUT-7G, is not only able to impair the nuclear translocation of STAT3, but it also blocks the energy metabolism pathway, depressing the glycolytic and Krebs pathway by the inhibition of hexokinase 2 activity. These data confirm that LUT-7G can be proposed as a potential candidate for the treatment of inflammatory and proliferative diseases, but its role as a hexokinase 2 (HEK2) inhibitor opens new perspectives in nutritional science, and especially in cancer therapy, in which the inhibition of the Warburg effect could be relevant.
Collapse
Affiliation(s)
- Ramona Palombo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
- Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy.
| | - Sabrina Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Mattia Falconi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Blasco Morozzo Della Rocca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Alessandro Lo Surdo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Elena Campione
- Department of Systems Medicine, Dermatologic Unit, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
- IDI-IRCCS, Biochemistry Laboratory, via dei Monti di Creta, 104, 00167 Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Alessandro Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| |
Collapse
|
20
|
Fedorova NE, Chernoryzh YY, Vinogradskaya GR, Emelianova SS, Zavalyshina LE, Yurlov KI, Zakirova NF, Verbenko VN, Kochetkov SN, Kushch AA, Ivanov AV. Inhibitor of polyamine catabolism MDL72.527 restores the sensitivity to doxorubicin of monocytic leukemia Thp-1 cells infected with human cytomegalovirus. Biochimie 2018; 158:82-89. [PMID: 30578923 DOI: 10.1016/j.biochi.2018.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
Leukemic cells from different patients exhibit different sensitivity to anticancer drugs including doxorubicin (DOX). Resistance to chemotherapy decreases efficacy of the treatment and promotes cancer recurrence and metastases. One of the approaches to overcome drug resistance includes E2F1-mediated regulation of the р73 protein that belongs to the р53 family. Its ΔNp73 isoform exhibits pro-oncogenic effects, and TAp73 - anti-oncogenic effects. Human cytomegalovirus (HCMV), often found in tumors, suppresses pro-apoptotic pathways and E2F1/p73 in particular. The activity of E2F1 and p73 transcription factors is linked to metabolism of biogenic polyamines. Therefore, it could be suggested that compounds that target polyamine-metabolizing enzymes can sensitize HCMV-infected hematological malignancies to doxorubicin. Here we report that HCMV infection of ТНР-1 monocytic leukemic cells considerably elevates E2F1 levels and shifts the balance between the р73 isoforms towards ΔNp73 leading to survival of DOX-treated leukemic cells. In contrast, MDL72.527, an inhibitor of polyamine catabolism, decreases ΔNp73/ТАр73 ratio and thus restores sensitivity of the cells to DOX. Our findings indicate the combination of doxorubicin and MDL72.527 may present a novel strategy for therapy of leukemia in patients with and without HCMV infection.
Collapse
Affiliation(s)
- Natalia E Fedorova
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Yana Yu Chernoryzh
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Galina R Vinogradskaya
- Konstantinov Petersburg Nuclear Physics Institute, National Research Center "Kurchatov Institute", Gatchina, Leningrad Region, Russia
| | - Svetlana S Emelianova
- Konstantinov Petersburg Nuclear Physics Institute, National Research Center "Kurchatov Institute", Gatchina, Leningrad Region, Russia
| | - Larisa E Zavalyshina
- Educational Institution of Further Professional Education «Russian Medical Academy of Continuous Professional Education» of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Kirill I Yurlov
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Natalia F Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Valery N Verbenko
- Konstantinov Petersburg Nuclear Physics Institute, National Research Center "Kurchatov Institute", Gatchina, Leningrad Region, Russia
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alla A Kushch
- Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
21
|
Rotblat B, Agostini M, Niklison-Chirou MV, Amelio I, Willis AE, Melino G. Sustained protein synthesis and reduced eEF2K levels in TAp73 -\- mice brain: a possible compensatory mechanism. Cell Cycle 2018; 17:2637-2643. [PMID: 30507330 DOI: 10.1080/15384101.2018.1553341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The transcription factor p73 is a member of the p53 family, of which the transactivation domain containing isoform (TAp73) plays key roles in brain development and neuronal stem cells. TAp73 also facilitates homoeostasis and prevents oxidative damage in vivo by inducing the expression of its target genes. Recently, we found that in addition to its role in regulation of transcription, TAp73 also affects mRNA translation. In cultured cells, acute TAp73 depletion activates eEF2K, which phosphorylates eEF2 reducing mRNA translation elongation. As a consequence, there is a reduction in global proteins synthesis rates and reprogramming of the translatome, leading to a selective decrease in the translation of rRNA processing factors. Given the dramatic effects of Tap73 depletion in vitro it was important to determine whether similar effects were observed in vivo. Here, we report the surprising finding that in brains of TAp73 KO mice there is a reduced level of eEF2K, which allows protein synthesis rates to be maintained suggesting a compensation model. These data provide new insights to the role of TAp73 in translation regulation and the eEF2K pathway in the brain.
Collapse
Affiliation(s)
- Barak Rotblat
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,b Department of Life Sciences , Ben Gurion University in the Negev , Beer Sheva , Israel
| | - Massimiliano Agostini
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,c Department of Experimental Medicine and Surgery, IDI-IRCCS , University of Rome Tor Vergata , Rome , Italy
| | - Maria Victoria Niklison-Chirou
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,d Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry , Queen Mary University of London , London , UK
| | - Ivano Amelio
- a MRC Toxicology Unit , University of Cambridge , Rome , UK
| | - Anne E Willis
- a MRC Toxicology Unit , University of Cambridge , Rome , UK
| | - Gerry Melino
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,c Department of Experimental Medicine and Surgery, IDI-IRCCS , University of Rome Tor Vergata , Rome , Italy
| |
Collapse
|
22
|
Carbone M, Amelio I, Affar EB, Brugarolas J, Cannon-Albright LA, Cantley LC, Cavenee WK, Chen Z, Croce CM, Andrea AD, Gandara D, Giorgi C, Jia W, Lan Q, Mak TW, Manley JL, Mikoshiba K, Onuchic JN, Pass HI, Pinton P, Prives C, Rothman N, Sebti SM, Turkson J, Wu X, Yang H, Yu H, Melino G. Consensus report of the 8 and 9th Weinman Symposia on Gene x Environment Interaction in carcinogenesis: novel opportunities for precision medicine. Cell Death Differ 2018; 25:1885-1904. [PMID: 30323273 PMCID: PMC6219489 DOI: 10.1038/s41418-018-0213-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022] Open
Abstract
The relative contribution of intrinsic genetic factors and extrinsic environmental ones to cancer aetiology and natural history is a lengthy and debated issue. Gene-environment interactions (G x E) arise when the combined presence of both a germline genetic variant and a known environmental factor modulates the risk of disease more than either one alone. A panel of experts discussed our current understanding of cancer aetiology, known examples of G × E interactions in cancer, and the expanded concept of G × E interactions to include somatic cancer mutations and iatrogenic environmental factors such as anti-cancer treatment. Specific genetic polymorphisms and genetic mutations increase susceptibility to certain carcinogens and may be targeted in the near future for prevention and treatment of cancer patients with novel molecularly based therapies. There was general consensus that a better understanding of the complexity and numerosity of G × E interactions, supported by adequate technological, epidemiological, modelling and statistical resources, will further promote our understanding of cancer and lead to novel preventive and therapeutic approaches.
Collapse
Affiliation(s)
| | | | - El Bachir Affar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, Quebec, H1T 2M4, Canada
| | - James Brugarolas
- Department of Internal Medicine, Hematology-Oncology Division, Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, 413 E. 69(th) Street, New York, NY, 10021, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zhijian Chen
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alan D' Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David Gandara
- Thoracic Oncology, UC Davis, Sacramento, CA, 96817, USA
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Wei Jia
- Hawaii Cancer Center, Honolulu, HI, USA
| | - Qing Lan
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Tak Wah Mak
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Jose N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
| | - Harvey I Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York, 10027, USA
| | - Nathaniel Rothman
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Said M Sebti
- Drug Discovery Department, Moffitt Cancer Center, and Department of Oncologic Sciences, University of South Florida, Tampa, FL, 33612, USA
| | | | - Xifeng Wu
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Gerry Melino
- MRC Toxicology Unit, Leicester, UK.
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
23
|
Xie N, Vikhreva P, Annicchiarico-Petruzzelli M, Amelio I, Barlev N, Knight RA, Melino G. Integrin-β4 is a novel transcriptional target of TAp73. Cell Cycle 2018; 17:589-594. [PMID: 29233040 DOI: 10.1080/15384101.2017.1403684] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As a member of p53 family, p73 has attracted intense investigations due to its structural and functional similarities to p53. Among more than ten p73 variants, the transactivation (TA) domain-containing isoform TAp73 is the one that imitates the p53's behavior most. TAp73 induces apoptosis and cell cycle arrest, which endows it the capacity of tumour suppression. Also, it can exert diverse biological influences on cells through activating a complex and context dependent transcriptional programme. The transcriptional activities further broaden its roles in more intricate biological processes. In this article, we report that p73 is a positive regulator of a cell adhesion related gene named integrin β4 (ITGB4). This finding may have implications for the dissection of the biological mechanisms underlining p73 functions.
Collapse
Affiliation(s)
- Ningxia Xie
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom.,b Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Rome 00133 , Italy
| | - Polina Vikhreva
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom
| | | | - Ivano Amelio
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom
| | - Nicolai Barlev
- d Institute of Cytology Russian Academy of Sciences , Saint-Petersburg , 194064 , Russia
| | - Richard A Knight
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom
| | - Gerry Melino
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom.,b Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Rome 00133 , Italy.,d Institute of Cytology Russian Academy of Sciences , Saint-Petersburg , 194064 , Russia
| |
Collapse
|
24
|
Petrova V, Annicchiarico-Petruzzelli M, Melino G, Amelio I. The hypoxic tumour microenvironment. Oncogenesis 2018; 7:10. [PMID: 29362402 PMCID: PMC5833859 DOI: 10.1038/s41389-017-0011-9] [Citation(s) in RCA: 675] [Impact Index Per Article: 112.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/04/2017] [Indexed: 12/13/2022] Open
Abstract
Cancer progression often benefits from the selective conditions present in the tumour microenvironment, such as the presence of cancer-associated fibroblasts (CAFs), deregulated ECM deposition, expanded vascularisation and repression of the immune response. Generation of a hypoxic environment and activation of its main effector, hypoxia-inducible factor-1 (HIF-1), are common features of advanced cancers. In addition to the impact on tumour cell biology, the influence that hypoxia exerts on the surrounding cells represents a critical step in the tumorigenic process. Hypoxia indeed enables a number of events in the tumour microenvironment that lead to the expansion of aggressive clones from heterogeneous tumour cells and promote a lethal phenotype. In this article, we review the most relevant findings describing the influence of hypoxia and the contribution of HIF activation on the major components of the tumour microenvironment, and we summarise their role in cancer development and progression.
Collapse
Affiliation(s)
- Varvara Petrova
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK
| | | | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK.
| |
Collapse
|
25
|
Agostini M, Annicchiarico-Petruzzelli M, Melino G, Rufini A. Metabolic pathways regulated by TAp73 in response to oxidative stress. Oncotarget 2017; 7:29881-900. [PMID: 27119504 PMCID: PMC5058650 DOI: 10.18632/oncotarget.8935] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/16/2016] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species are involved in both physiological and pathological processes including neurodegeneration and cancer. Therefore, cells have developed scavenging mechanisms to maintain redox homeostasis under control. Tumor suppressor genes play a critical role in the regulation of antioxidant genes. Here, we investigated whether the tumor suppressor gene TAp73 is involved in the regulation of metabolic adaptations triggered in response to oxidative stress. H2O2 treatment resulted in numerous biochemical changes in both control and TAp73 knockout (TAp73−/−) mouse embryonic fibroblasts, however the extent of these changes was more pronounced in TAp73−/− cells when compared to control cells. In particular, loss of TAp73 led to alterations in glucose, nucleotide and amino acid metabolism. In addition, H2O2 treatment resulted in increased pentose phosphate pathway (PPP) activity in null mouse embryonic fibroblasts. Overall, our results suggest that in the absence of TAp73, H2O2 treatment results in an enhanced oxidative environment, and at the same time in an increased pro-anabolic phenotype. In conclusion, the metabolic profile observed reinforces the role of TAp73 as tumor suppressor and indicates that TAp73 exerts this function, at least partially, by regulation of cellular metabolism.
Collapse
Affiliation(s)
- Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | | | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandro Rufini
- Department of Cancer Studies, CRUK Leicester Cancer Centre, University of Leicester, Leicester, UK
| |
Collapse
|
26
|
p73 promotes glioblastoma cell invasion by directly activating POSTN (periostin) expression. Oncotarget 2017; 7:11785-802. [PMID: 26930720 PMCID: PMC4914248 DOI: 10.18632/oncotarget.7600] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma Multiforme is one of the most highly metastatic cancers and constitutes 70% of all gliomas. Despite aggressive treatments these tumours have an exceptionally bad prognosis, mainly due to therapy resistance and tumour recurrence. Here we show that the transcription factor p73 confers an invasive phenotype by directly activating expression of POSTN (periostin, HGNC:16953) in glioblastoma cells. Knock down of endogenous p73 reduces invasiveness and chemo-resistance, and promotes differentiation in vitro. Using chromatin immunoprecipitation and reporter assays we demonstrate that POSTN, an integrin binding protein that has recently been shown to play a major role in metastasis, is a transcriptional target of TAp73. We further show that POSTN overexpression is sufficient to rescue the invasive phenotype of glioblastoma cells after p73 knock down. Additionally, bioinformatics analysis revealed that an intact p73/POSTN axis, where POSTN and p73 expression is correlated, predicts bad prognosis in several cancer types. Taken together, our results support a novel role of TAp73 in controlling glioblastoma cell invasion by regulating the expression of the matricellular protein POSTN.
Collapse
|
27
|
Nemajerova A, Amelio I, Gebel J, Dötsch V, Melino G, Moll UM. Non-oncogenic roles of TAp73: from multiciliogenesis to metabolism. Cell Death Differ 2017; 25:144-153. [PMID: 29077094 PMCID: PMC5729534 DOI: 10.1038/cdd.2017.178] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 01/24/2023] Open
Abstract
The p53 family of transcription factors (p53, p63 and p73) covers a wide range of functions critical for development, homeostasis and health of mammals across their lifespan. Beside the well-established tumor suppressor role, recent evidence has highlighted novel non-oncogenic functions exerted by p73. In particular, p73 is required for multiciliated cell (MCC) differentiation; MCCs have critical roles in brain and airways to move fluids across epithelial surfaces and to transport germ cells in the reproductive tract. This novel function of p73 provides a unifying cellular mechanism for the disparate inflammatory and immunological phenotypes of p73-deficient mice. Indeed, mice with Trp73 deficiency suffer from hydrocephalus, sterility and chronic respiratory tract infections due to profound defects in ciliogenesis and complete loss of mucociliary clearance since MCCs are essential for cleaning airways from inhaled pollutants, pathogens and allergens. Cross-species genomic analyses and functional rescue experiments identify TAp73 as the master transcriptional integrator of ciliogenesis, upstream of previously known central nodes. In addition, TAp73 shows a significant ability to regulate cellular metabolism and energy production through direct transcriptional regulation of several metabolic enzymes, such as glutaminase-2 and glucose-6 phosphate dehydrogenase. This recently uncovered role of TAp73 in the regulation of cellular metabolism strongly affects oxidative balance, thus potentially influencing all the biological aspects associated with p73 function, including development, homeostasis and cancer. Although through different mechanisms, p63 isoforms also contribute to regulation of cellular metabolism, thus indicating a common route used by all family members to control cell fate. At the structural level, the complexity of p73's function is further enhanced by its ability to form heterotetramers with some p63 isoforms, thus indicating the existence of an intrafamily crosstalk that determines the global outcome of p53 family function. In this review, we have tried to summarize all the recent evidence that have emerged on the novel non-oncogenic roles of p73, in an attempt to provide a unified view of the complex function of this gene within its family.
Collapse
Affiliation(s)
- Alice Nemajerova
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, PO Box 138, Leicester LE1 9HN, UK
| | - Jakob Gebel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Hodgkin Building, Lancaster Road, PO Box 138, Leicester LE1 9HN, UK.,Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome 00133, Italy
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
28
|
Min L, Choy E, Tu C, Hornicek F, Duan Z. Application of metabolomics in sarcoma: From biomarkers to therapeutic targets. Crit Rev Oncol Hematol 2017; 116:1-10. [PMID: 28693790 PMCID: PMC5527996 DOI: 10.1016/j.critrevonc.2017.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/25/2017] [Accepted: 05/09/2017] [Indexed: 02/05/2023] Open
Affiliation(s)
- Li Min
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA; Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan, 610041, China
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA
| | - Chongqi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan, 610041, China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, MA 02114, USA.
| |
Collapse
|
29
|
Marini A, Lena AM, Panatta E, Ivan C, Han L, Liang H, Annicchiarico-Petruzzelli M, Di Daniele N, Calin GA, Candi E, Melino G. Ultraconserved long non-coding RNA uc.63 in breast cancer. Oncotarget 2017; 8:35669-35680. [PMID: 27447964 PMCID: PMC5482607 DOI: 10.18632/oncotarget.10572] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022] Open
Abstract
Transcribed-ultraconserved regions (T-UCRs) are long non-coding RNAs (lncRNA) encoded by a subset of long ultraconserved stretches in the human genome. Recent studies revealed that the expression of several T-UCRs is altered in cancer and growing evidences underline the importance of T-UCRs in oncogenesis, offering also potential new strategies for diagnosis and prognosis. We found that overexpression of one specific T-UCRs named uc.63 is associated with bad outcome in luminal A subtype of breast cancer patients. uc.63 is localized in the third intron of exportin-1 gene (XPO1) and is transcribed in the same orientation of its host gene. Interestingly, silencing of uc.63 induces apoptosis in vitro. However, silencing of host gene XPO1 does not cause the same effect suggesting that the transcription of uc.63 is independent of XPO1. Our results reveal an important role of uc.63 in promoting breast cancer cells survival and offer the prospect to identify a signature associated with poor prognosis.
Collapse
Affiliation(s)
- Alberto Marini
- Medical Research Council, Toxicology Unit, Hodgkin Building, University of Leicester, Leicester, UK
| | - Anna Maria Lena
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - Emanuele Panatta
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - Cristina Ivan
- Department of Experimental Therapeutics and The Center for RNA interference and non-coding RNA, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Nicola Di Daniele
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - George A. Calin
- Department of Experimental Therapeutics and The Center for RNA interference and non-coding RNA, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Eleonora Candi
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
- IDI-IRCCS, Biochemistry Laboratory, Rome, Italy
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Hodgkin Building, University of Leicester, Leicester, UK
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
30
|
Agostini M, Niklison-Chirou MV, Annicchiarico-Petruzzelli MM, Grelli S, Di Daniele N, Pestlikis I, Knight RA, Melino G, Rufini A. p73 Regulates Primary Cortical Neuron Metabolism: a Global Metabolic Profile. Mol Neurobiol 2017; 55:3237-3250. [PMID: 28478509 DOI: 10.1007/s12035-017-0517-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 04/04/2017] [Indexed: 12/20/2022]
Abstract
The transcription factor p73 has been demonstrated to play a significant role in survival and differentiation of neuronal stem cells. In this report, by employing comprehensive metabolic profile and mitochondrial bioenergetics analysis, we have explored the metabolic alterations in cortical neurons isolated from p73 N-terminal isoform specific knockout animals. We found that loss of the TAp73 or ΔNp73 triggers selective biochemical changes. In particular, p73 isoforms regulate sphingolipid and phospholipid biochemical pathway signaling. Indeed, sphinganine and sphingosine levels were reduced in p73-depleted cortical neurons, and decreased levels of several membrane phospholipids were also observed. Moreover, in line with the complexity associated with p73 functions, loss of the TAp73 seems to increase glycolysis, whereas on the contrary, loss of ΔNp73 isoform reduces glucose metabolism, indicating an isoform-specific differential effect on glycolysis. These changes in glycolytic flux were not reflected by parallel alterations of mitochondrial respiration, as only a slight increase of mitochondrial maximal respiration was observed in p73-depleted cortical neurons. Overall, our findings reinforce the key role of p73 in regulating cellular metabolism and point out that p73 exerts its functions in neuronal biology at least partially through the regulation of metabolic pathways.
Collapse
Affiliation(s)
- Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, LE1 9HN, UK.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Maria Victoria Niklison-Chirou
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, LE1 9HN, UK.,Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | | | - Sandro Grelli
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, Nephrology and Hypertension Unit, "Tor Vergata" University Hospital, Rome, Italy
| | - Ilias Pestlikis
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Richard A Knight
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, LE1 9HN, UK
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, LE1 9HN, UK. .,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - Alessandro Rufini
- Department of Cancer Studies, University of Leicester, Leicester, LE2 7LX, UK.
| |
Collapse
|
31
|
Vikhreva P, Petrova V, Gokbulut T, Pestlikis I, Mancini M, Di Daniele N, Knight RA, Melino G, Amelio I. TAp73 upregulates IL-1β in cancer cells: Potential biomarker in lung and breast cancer? Biochem Biophys Res Commun 2017; 482:498-505. [PMID: 28212736 PMCID: PMC5243147 DOI: 10.1016/j.bbrc.2016.10.085] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/19/2016] [Accepted: 10/23/2016] [Indexed: 02/06/2023]
Abstract
p73 is a transcription factor belonging to the p53 tumour suppressor family. p73−/− mice exhibit a range of phenotypes including neurological, reproductive and inflammatory defects. Although the role of p73 in the control of genomic stability explains part of these phenotypes, a clear mechanism of how p73 participates in the inflammatory response is still elusive. Interleukin-1β (IL-1β) has a crucial role in mediating the inflammatory response. Because of its high potency to induce inflammation, the activation and secretion of IL-1β is tightly regulated by large protein complexes, named inflammasomes. Inflammasomes regulate activation of proinflammatory caspase-1, which in turn proteolytically processes its substrates, including pro-IL-1β. Caspase-1 gene transcription is strongly activated by p53 protein family members including p73. Here, we have addressed whether p73 might be directly involved in IL-1β regulation and therefore in the control of the inflammatory response. Our results show that TAp73β upregulates pro-IL-1β mRNA and processed IL-1β protein. In addition, analysis of breast and lung cancer patient cohorts demonstrated that interaction between p73 and IL-1β predicts a negative survival outcome in these human cancers. The p53 family member p73 controls a wide a range of biological processes required for its tumour suppressor functions. p73 regulates IL-1β expression, thus potentially affecting inflammasomes and inflammatory response. p73/IL-1β axis correlates with poor prognosis in lung and breast cancer.
Collapse
Affiliation(s)
- Polina Vikhreva
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Varvara Petrova
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Tarik Gokbulut
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom; Erciyes University, Faculty of Science, Department of Biology, 38039 Kayseri, Turkey
| | - Ilias Pestlikis
- Department of Experimental Medicine and Surgery, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Mara Mancini
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Nicola Di Daniele
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Richard A Knight
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Gerry Melino
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom; Department of Experimental Medicine and Surgery, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Ivano Amelio
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom.
| |
Collapse
|
32
|
He Z, Agostini M, Liu H, Melino G, Simon HU. p73 regulates basal and starvation-induced liver metabolism in vivo. Oncotarget 2016; 6:33178-90. [PMID: 26375672 PMCID: PMC4741757 DOI: 10.18632/oncotarget.5090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/26/2015] [Indexed: 12/30/2022] Open
Abstract
As a member of the p53 gene family, p73 regulates cell cycle arrest, apoptosis, neurogenesis, immunity and inflammation. Recently, p73 has been shown to transcriptionally regulate selective metabolic enzymes, such as cytochrome c oxidase subunit IV isoform 1, glucose 6-phosphate dehydrogenase and glutaminase-2, resulting in significant effects on metabolism, including hepatocellular lipid metabolism, glutathione homeostasis and the pentose phosphate pathway. In order to further investigate the metabolic effect of p73, here, we compared the global metabolic profile of livers from p73 knockout and wild-type mice under both control and starvation conditions. Our results show that the depletion of all p73 isoforms cause altered lysine metabolism and glycolysis, distinct patterns for glutathione synthesis and Krebs cycle, as well as an elevated pentose phosphate pathway and abnormal lipid accumulation. These results indicate that p73 regulates basal and starvation-induced fuel metabolism in the liver, a finding that is likely to be highly relevant for metabolism-associated disorders, such as diabetes and cancer.
Collapse
Affiliation(s)
- Zhaoyue He
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - He Liu
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Yen CY, Huang HW, Shu CW, Hou MF, Yuan SSF, Wang HR, Chang YT, Farooqi AA, Tang JY, Chang HW. DNA methylation, histone acetylation and methylation of epigenetic modifications as a therapeutic approach for cancers. Cancer Lett 2016; 373:185-92. [DOI: 10.1016/j.canlet.2016.01.036] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/23/2015] [Accepted: 01/18/2016] [Indexed: 02/09/2023]
|
34
|
TAp73 loss favors Smad-independent TGF-β signaling that drives EMT in pancreatic ductal adenocarcinoma. Cell Death Differ 2016; 23:1358-70. [PMID: 26943320 DOI: 10.1038/cdd.2016.18] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/29/2022] Open
Abstract
Advances made in pancreatic cancer therapy have been far from sufficient and have allowed only a slight improvement in global survival of patients with pancreatic ductal adenocarcinoma (PDA). Recent progresses in chemotherapy have offered some hope for an otherwise gloomy outlook, however, only a limited number of patients are eligible because of important cytotoxicity. In this context, enhancing our knowledge on PDA initiation and evolution is crucial to highlight certain weaknesses on which to specifically target therapy. We found that loss of transcriptionally active p73 (TAp73), a p53 family member, impacted PDA development. In two relevant and specific engineered pancreatic cancer mouse models, we observed that TAp73 deficiency reduced survival and enhanced epithelial-to-mesenchymal transition (EMT). Through proteomic analysis of conditioned media from TAp73 wild-type (WT) and deficient pancreatic tumor cells, we identified a secreted protein, biglycan (BGN), which is necessary and sufficient to mediate this pro-EMT effect. Interestingly, BGN is modulated by and modulates the transforming growth factor-β (TGF-β) pathway, a key regulator of the EMT process. We further examined this link and revealed that TAp73 impacts the TGF-β pathway by direct regulation of BGN expression and Sma and Mad-related proteins (SMADs) expression/activity. Absence of TAp73 leads to activation of TGF-β signaling through a SMAD-independent pathway, favoring oncogenic TGF-β effects and EMT. Altogether, our data highlight the implication of TAp73 in the aggressiveness of pancreatic carcinogenesis through modulation of the TGF-β signaling. By suggesting TAp73 as a predictive marker for response to TGF-β inhibitors, our study could improve the classification of PDA patients with a view to offering combined therapy involving TGF-β inhibitors.
Collapse
|
35
|
Affiliation(s)
- Gerry Melino
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
36
|
Petrova V, Mancini M, Agostini M, Knight RA, Annicchiarico-Petruzzelli M, Barlev NA, Melino G, Amelio I. TAp73 transcriptionally represses BNIP3 expression. Cell Cycle 2015; 14:2484-93. [PMID: 25950386 PMCID: PMC4612661 DOI: 10.1080/15384101.2015.1044178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 03/26/2015] [Accepted: 04/18/2015] [Indexed: 01/07/2023] Open
Abstract
TAp73 is a tumor suppressor transcriptional factor, belonging to p53 family. Alteration of TAp73 in tumors might lead to reduced DNA damage response, cell cycle arrest and apoptosis. Carcinogen-induced TAp73(-/-) tumors display also increased angiogenesis, associated to hyperactivition of hypoxia inducible factor signaling. Here, we show that TAp73 suppresses BNIP3 expression, directly binding its gene promoter. BNIP3 is a hypoxia responsive protein, involved in a variety of cellular processes, such as autophagy, mitophagy, apoptosis and necrotic-like cell death. Therefore, through different cellular process altered expression of BNIP3 may differently contribute to cancer development and progression. We found a significant upregulation of BNIP3 in human lung cancer datasets, and we identified a direct association between BNIP3 expression and survival rate of lung cancer patients. Our data therefore provide a novel transcriptional target of TAp73, associated to its antagonistic role on HIF signaling in cancer, which might play a role in tumor suppression.
Collapse
Affiliation(s)
- Varvara Petrova
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Molecular Pharmacology Laboratory; Saint-Petersburg Institute of Technology; Saint-Petersburg, Russia
| | - Mara Mancini
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
| | - Massimiliano Agostini
- Department of Experimental Medicine and Surgery; University of Rome “Tor Vergata”; Rome, Italy
| | - Richard A Knight
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
| | | | - Nikolai A Barlev
- Molecular Pharmacology Laboratory; Saint-Petersburg Institute of Technology; Saint-Petersburg, Russia
- Gene Expression Laboratory; Institute of Cytology; Saint-Petersburg, Russia
| | - Gerry Melino
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Molecular Pharmacology Laboratory; Saint-Petersburg Institute of Technology; Saint-Petersburg, Russia
- Department of Experimental Medicine and Surgery; University of Rome “Tor Vergata”; Rome, Italy
- Biochemistry Laboratory IDI-IRCC; Rome, Italy
| | - Ivano Amelio
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
| |
Collapse
|
37
|
Amelio I, Melino G. The p53 family and the hypoxia-inducible factors (HIFs): determinants of cancer progression. Trends Biochem Sci 2015; 40:425-34. [PMID: 26032560 DOI: 10.1016/j.tibs.2015.04.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/17/2015] [Accepted: 04/29/2015] [Indexed: 12/20/2022]
Abstract
HIFs have long been associated with resistance to therapy, metastasis, and poor survival rates in cancer patients. In parallel, although the tumor-suppressor p53 acts as the first barrier against tumor transformation, its inactivation also appears to be crucial for enabling cancer progression at advanced stages. p53 has been proposed to antagonize HIF, and emerging evidence suggests that the p53 siblings p63 and p73 also participate in this interplay. Crosstalk between HIFs and the p53 family acts as a determinant of cancer progression through regulating angiogenesis, the tumor microenvironment, dormancy, metastasis, and recurrence. We discuss the possible mechanisms underlying this regulation and the controversies in this field in an attempt to provide a unified view of current knowledge.
Collapse
Affiliation(s)
- Ivano Amelio
- Medical Research Council Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Gerry Melino
- Medical Research Council Toxicology Unit, Leicester University, Leicester LE1 9HN, UK; Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata (IDI), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', 00133 Rome, Italy.
| |
Collapse
|