1
|
Mani R, Benrashid S, Templeton MD, Druhan LJ, Seegers SL, Chakraborty S, Teague SE, Jaros SC, Yang HT, Foureau DM, Steuerwald NM, Pal D, Ghosh N, Copelan EA, Durden DL, Avalos BR, Park SI. TP53 upregulation via aurora kinase inhibition overcomes primary failure to venetoclax in BCL2-rearranged lymphomas. iScience 2025; 28:112584. [PMID: 40491476 PMCID: PMC12146551 DOI: 10.1016/j.isci.2025.112584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 12/04/2024] [Accepted: 03/29/2025] [Indexed: 06/11/2025] Open
Abstract
Bcl2 inhibition has excellent antitumor activity against hematologic malignancies. However, the clinical results in lymphomas harboring BCL2 gene rearrangements have been disappointing, and the mechanism of this intrinsic resistance remains unknown. Herein, we report that Bcl2 inhibition rapidly repressed p53 with poor response in BCL2-rearranged lymphoma cells. However, concurrent inhibition of aurora kinase (Aurk) overcame this primary resistance to Bcl2 inhibition by restoring the p53/p21 proapoptotic axis via a post-transcriptional increase in p53. Two independent BCL2-rearranged lymphoma murine models showed complete tumor regression in all animals treated with combined Bcl2/Aurk inhibition, whereas mice treated with single-agents demonstrated rapid progression. Transcriptome analysis confirmed that BCL2-rearranged lymphomas rapidly downregulated the p53 target CDKN1A (p21) in response to Bcl2 inhibition in vivo. However, concurrent inhibition of Aurk restored the TP53/CDKN1A pathway, sensitizing the tumors to Bcl2 inhibitor-mediated apoptosis. These data lay the groundwork for evaluation of this combination in the clinical setting.
Collapse
Affiliation(s)
- Rajeswaran Mani
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Samon Benrashid
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Margaret D. Templeton
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Lawrence J. Druhan
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Sara L. Seegers
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Supriya Chakraborty
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Sarah E. Teague
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Scott C. Jaros
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Hsih-Te Yang
- Center for Cancer Biostatistics, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - David M. Foureau
- Immune Monitoring Laboratory, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Nury M. Steuerwald
- Molecular Biology and Genomics Laboratory, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Dhananjaya Pal
- Molecular Targeted Therapeutics, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
| | - Nilanjan Ghosh
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Edward A. Copelan
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Donald L. Durden
- Molecular Targeted Therapeutics, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Belinda R. Avalos
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Steven I. Park
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Charlotte, NC 28204, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
2
|
Yang L, Zhai H, Tian T, Liu B, Ni X, Xia H. Combinatorial biosynthesis of novel gentamicin derivatives with nonsense mutation readthrough activity and low cytotoxicity. Front Pharmacol 2025; 16:1575840. [PMID: 40342992 PMCID: PMC12059486 DOI: 10.3389/fphar.2025.1575840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Background Aminoglycosides (AGs) are one of the initial classes of antibiotics that have been used clinically and possess broad spectrum of activity. Nevertheless, their clinical utilization is restricted by safety issues associated with nephrotoxicity and ototoxicity. Methods Glycosyltransferase (GT) KanM2 was introduced into M. echinospora to produce the gentamicin derivatives, in which a kanosamine sugar ring was introduced to replace the garosamine. The premature termination codon (PTC) readthrough activity of genkamicins (GKs) was compared using dual luciferase reporter assay. The toxicity of GK was assessed in vitro in HEK-293 and NCI-H1299 cells and determined based on cell viability calculated after 48 h of treatment with different concentrations of the compounds. The NCI-H1299 cells harbouring the R213X nonsense mutation were treated with different concentrations of the derivatives to compare their expression of p53 proteins. The expression of p53 and its downstream targets p21 and BAX was detected using Western blotting and qRT-PCR in NCI-H1299 cells containing the R213X nonsense mutation treated with different concentrations of GK-Ae and G418. Finally, immunofluorescence and flow cytometry were used to determine the subcellular localization of full-length p53 protein induced by GK-Ae treatment and its effect on apoptosis in cancer cells. Results Eight gentamicin derivatives were obtained in this study. GK-Ae displayed similar PTC readthrough activity and reduced toxicity compared to natural aminoglycoside G418. Moreover, GK-Ae increased the levels of both p53 and its downstream targets p21 and BAX, and promoted apoptosis of cancer cells. Conclusion These results demonstrate the potential of combinatorial biosynthesis to increase the diversity of structures of AGs and provide directions for the development of new AGs with low toxicity and high PTC readthrough activity.
Collapse
Affiliation(s)
| | | | | | | | - Xianpu Ni
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Huanzhang Xia
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
3
|
Chen H, Shan J, Qi W, Chen L, Zeng X. p53-dependent chromatin relaxation is required for DNA double-strand break repair. Acta Biochim Biophys Sin (Shanghai) 2025; 57:701-711. [PMID: 40007254 DOI: 10.3724/abbs.2025008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
The tumor suppressor p53, an indispensable nuclear transcription factor, plays a central role in orchestrating cellular responses when DNA damage occurs. In this study, we demonstrate that in the initial phases of DNA double-strand break (DSB) repair, p53 is rapidly recruited to sites of damage and the surrounding chromatin, where it enhances DSB repair efficiency. This enhancement occurs through the modulation of chromatin dynamics and the promotion of a more relaxed chromatin configuration, a process influenced by p53 in response to DSB-inducing factors such as etoposide, ultraviolet radiation, and nucleases. These results underscore the pivotal function of p53 as a rapid responder to DSBs, delineating a significant departure from its traditionally recognized role as a downstream transcriptional regulator in DNA damage repair processes. This study emphasizes that the direct engagement of p53 in DNA repair through chromatin structure regulation extends beyond its established involvement in UV irradiation-induced nucleotide excision repair (NER), demonstrating analogous mechanistic attributes in the context of DSB repair. This newly illuminated perspective enhances our understanding of the multifaceted roles of p53 in genome stability and integrity.
Collapse
Affiliation(s)
- Hongyu Chen
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jin Shan
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Wenjing Qi
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
- Department of Bioscience, Changchun Normal University, Changchun 130032, China
| | - Lili Chen
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Xianlu Zeng
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| |
Collapse
|
4
|
Funk JS, Klimovich M, Drangenstein D, Pielhoop O, Hunold P, Borowek A, Noeparast M, Pavlakis E, Neumann M, Balourdas DI, Kochhan K, Merle N, Bullwinkel I, Wanzel M, Elmshäuser S, Teply-Szymanski J, Nist A, Procida T, Bartkuhn M, Humpert K, Mernberger M, Savai R, Soussi T, Joerger AC, Stiewe T. Deep CRISPR mutagenesis characterizes the functional diversity of TP53 mutations. Nat Genet 2025; 57:140-153. [PMID: 39774325 PMCID: PMC11735402 DOI: 10.1038/s41588-024-02039-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
The mutational landscape of TP53, a tumor suppressor mutated in about half of all cancers, includes over 2,000 known missense mutations. To fully leverage TP53 mutation status for personalized medicine, a thorough understanding of the functional diversity of these mutations is essential. We conducted a deep mutational scan using saturation genome editing with CRISPR-mediated homology-directed repair to engineer 9,225 TP53 variants in cancer cells. This high-resolution approach, covering 94.5% of all cancer-associated TP53 missense mutations, precisely mapped the impact of individual mutations on tumor cell fitness, surpassing previous deep mutational scan studies in distinguishing benign from pathogenic variants. Our results revealed even subtle loss-of-function phenotypes and identified promising mutants for pharmacological reactivation. Moreover, we uncovered the roles of splicing alterations and nonsense-mediated messenger RNA decay in mutation-driven TP53 dysfunction. These findings underscore the power of saturation genome editing in advancing clinical TP53 variant interpretation for genetic counseling and personalized cancer therapy.
Collapse
Affiliation(s)
- Julianne S Funk
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Maria Klimovich
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Ole Pielhoop
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Pascal Hunold
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Anna Borowek
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Maxim Noeparast
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Michelle Neumann
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Dimitrios-Ilias Balourdas
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Katharina Kochhan
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Imke Bullwinkel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Julia Teply-Szymanski
- Institute of Pathology, Philipps-University, Marburg University Hospital, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps-University, Marburg, Germany
| | - Tara Procida
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Marek Bartkuhn
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Justus-Liebig-University, Giessen, Germany
| | - Katharina Humpert
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
- Bioinformatics Core Facility, Philipps-University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Lung Microenvironmental Niche in Cancerogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thierry Soussi
- Centre de Recherche Saint-Antoine UMRS_938, INSERM, Sorbonne Université, Paris, France
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Clinical Genetics, Uppsala University Hospital, Uppsala, Sweden
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.
- Genomics Core Facility, Philipps-University, Marburg, Germany.
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.
- Bioinformatics Core Facility, Philipps-University, Marburg, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
5
|
Tu Z, Tang L, Shen H, Hu M, Chen F, Wang X, Abo-Raya MH, Wang Y. Molecular insights into the physiological impact of low-frequency noise on sea slug Onchidium reevesii: Activation of p53 signaling and oxidative stress response. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 373:123481. [PMID: 39626391 DOI: 10.1016/j.jenvman.2024.123481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/12/2024] [Accepted: 11/24/2024] [Indexed: 01/15/2025]
Abstract
To investigate the regulatory role of tumor protein p53 of sea slug (Onchidium reevesii) under oxidative stress conditions, we examined the response mechanisms of O. reevesii to low-frequency noise pollution (1000 Hz) using molecular and cellular biology techniques. We successfully cloned the O. reevesii p53 gene (Orp53) from O. reevesii, obtaining a 3356 bp sequence containing a 2727 bp open reading frame (ORF). Phylogenetic analysis revealed that O. reevesii shares a close evolutionary relationship with other molluscs, including Bulinus truncatus and Elysia marginata. Expression analysis showed that Orp53 is expressed across various tissues, with the highest expression levels in the hepatopancreas. Using RNA interference (RNAi) to silence the Orp53 gene, we found that the mRNA expressions of Orp53 and its downstream apoptosis-related genes, including cytochrome C (Cyt_C), Caspase 9, and Caspase 3, were significantly suppressed until the third day of interference (P < 0.05). Moreover, sip53 treatment resulted in significant reductions in the mRNA expression and protein levels of all studied genes (P < 0.05) compared to the noise-exposed group. In addition, the low-frequency noise exposure decreased central nervous system (CNS) viability while increasing oxidative stress markers, including reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione S-transferase (GST). On the other hand, silencing Orp53 expression via siRNA resulted in significant reductions in CNS cell viability (P < 0.05). Our study establishes a molecular basis for evaluating the consequences of marine noise pollution, confirming that low-frequency noise activates the p53 signaling pathway, oxidative stress, and that p53 can regulate oxidative stress and apoptosis-related genes.
Collapse
Affiliation(s)
- Zhihan Tu
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai, 201306, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Liusiqiao Tang
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai, 201306, China
| | - Heding Shen
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai, 201306, China
| | - Menghong Hu
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai, 201306, China
| | - Feng Chen
- Fisheries Research Institute of Guizhou Academy of Agricultural Sciences, Guiyang, 550025, China
| | - Xiaotong Wang
- Yantai Engineering Laboratory of Development and Utilization of Characteristic Marine Organisms, Ludong University, Yantai, 264025, China
| | - Mohamed H Abo-Raya
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai, 201306, China; Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Youji Wang
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai, 201306, China.
| |
Collapse
|
6
|
Datta D, Navalkar A, Sakunthala A, Paul A, Patel K, Masurkar S, Gadhe L, Manna S, Bhattacharyya A, Sengupta S, Poudyal M, Devi J, Sawner AS, Kadu P, Shaw R, Pandey S, Mukherjee S, Gahlot N, Sengupta K, Maji SK. Nucleo-cytoplasmic environment modulates spatiotemporal p53 phase separation. SCIENCE ADVANCES 2024; 10:eads0427. [PMID: 39661689 PMCID: PMC11633762 DOI: 10.1126/sciadv.ads0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Liquid-liquid phase separation of various transcription factors into biomolecular condensates plays an essential role in gene regulation. Here, using cellular models and in vitro studies, we show the spatiotemporal formation and material properties of p53 condensates that might dictate its function. In particular, p53 forms liquid-like condensates in the nucleus of cells, which can bind to DNA and perform transcriptional activity. However, cancer-associated mutations promote misfolding and partially rigidify the p53 condensates with impaired DNA binding ability. Irrespective of wild-type and mutant forms, the partitioning of p53 into cytoplasm leads to the condensate formation, which subsequently undergoes rapid solidification. In vitro studies show that abundant nuclear components such as RNA and nonspecific DNA promote multicomponent phase separation of the p53 core domain and maintain their liquid-like property, whereas specific DNA promotes its dissolution into tetrameric functional p53. This work provides mechanistic insights into how the life cycle and DNA binding properties of p53 might be regulated by phase separation.
Collapse
Affiliation(s)
- Debalina Datta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajoy Paul
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Shalaka Masurkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Laxmikant Gadhe
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Shouvik Manna
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arpita Bhattacharyya
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Shinjinee Sengupta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Manisha Poudyal
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Jyoti Devi
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ranjit Shaw
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Satyaprakash Pandey
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Kundan Sengupta
- Chromosome Biology Lab, Indian Institute of Science Education and Research, Pune, India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
7
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
8
|
Shao Y, Shah PT, Su Q, Li S, Huang F, Wang J, Wang P, Wu C. Recombinant adenoviruses expressing HPV16/18 E7 upregulate the HDAC6 and DNMT3B genes in C33A cells. Front Cell Infect Microbiol 2024; 14:1459572. [PMID: 39411320 PMCID: PMC11473514 DOI: 10.3389/fcimb.2024.1459572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVE High-risk human papillomavirus (HPV) is a carcinogenic virus associated with nearly all cases of cervical cancer, as well as an increasing number of anal and oral cancers. The two carcinogenic proteins of HPV, E6 and E7, can immortalize keratinocytes and are essential for HPV-related cellular transformation. Currently, the global regulatory effects of these oncogenic proteins on the host proteome are not fully understood, and further exploration of the functions and carcinogenic mechanisms of E6 and E7 proteins is needed. METHODS We used a previously established platform in our laboratory for constructing recombinant adenoviral plasmids expressing the HPV16 E7 gene to further construct recombinant virus particles expressing HPV16/18 E6, E7, and both E6 and E7 genes. These recombinant viruses were used to infect C33A cells to achieve sustained expression of the HPV16/18 E6/E7 genes. Subsequently, total RNA was extracted and RNA-Seq technology was employed for transcriptome sequencing to identify differentially expressed genes associated with HPV infection in cervical cancer. RESULTS RNA-Seq analysis revealed that overexpression of the HPV16/18 E6/E7 genes upregulated GP6, CD36, HDAC6, ESPL1, and DNMT3B among the differentially expressed genes (DEGs) associated with cervical cancer. Spearman correlation analysis revealed a statistically significant correlation between the HDAC6 and DNMT3B genes and key pathways, including DNA replication, tumor proliferation signature, G2M checkpoint, p53 pathways, and PI3K/AKT/mTOR signaling pathways. Further, qRT-PCR and Western blot analyses indicated that both HPV16/18 E7 can upregulate the expression of HDAC6 and DNMT3B, genes associated with HPV infection-related cervical cancer. CONCLUSION The successful expression of HPV16/18 E6/E7 in cells indicates that the recombinant viruses retain the replication and infection capabilities of Ad4. Furthermore, the recombinant viruses expressing HPV16/18 E7 can upregulate the HDAC6 and DNMT3B genes involved in cervical cancer pathways, thereby influencing the cell cycle. Additionally, HDAC6 and DNMT3B are emerging as important therapeutic targets for cancer. This study lays the foundation for further exploration of the oncogenic mechanisms of HPV E6/E7 and may provide new directions for the treatment of HPV-related cancers.
Collapse
Affiliation(s)
- Yunting Shao
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| | - Pir Tariq Shah
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| | - Qisheng Su
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jun Wang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Dalian Medical University Mailing, Dalian, China
| | - Peng Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Chengjun Wu
- Faculty of Medicine, School of Basic Medical Sciences, Dalian University of Technology, Dalian, China
| |
Collapse
|
9
|
Djordjevic S, Itzykson R, Hague F, Lebon D, Legrand J, Ouled‐Haddou H, Jedraszak G, Harbonnier J, Collet L, Paubelle E, Marolleau J, Garçon L, Boyer T. STIM2 is involved in the regulation of apoptosis and the cell cycle in normal and malignant monocytic cells. Mol Oncol 2024; 18:1571-1592. [PMID: 38234211 PMCID: PMC11161727 DOI: 10.1002/1878-0261.13584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Calcium is a ubiquitous messenger that regulates a wide range of cellular functions, but its involvement in the pathophysiology of acute myeloid leukemia (AML) is not widely investigated. Here, we identified, from an analysis of The Cancer Genome Atlas and genotype-tissue expression databases, stromal interaction molecule 2 (STIM2) as being highly expressed in AML with monocytic differentiation and negatively correlated with overall survival. This was confirmed on a validation cohort of 407 AML patients. We then investigated the role of STIM2 in cell proliferation, differentiation, and survival in two leukemic cell lines with monocytic potential and in normal hematopoietic stem cells. STIM2 expression increased at the RNA and protein levels upon monocyte differentiation. Phenotypically, STIM2 knockdown drastically inhibited cell proliferation and induced genomic stress with DNA double-strand breaks, as shown by increased levels of phosphorylate histone H2AXγ (p-H2AXγ), followed by activation of the cellular tumor antigen p53 pathway, decreased expression of cell cycle regulators such as cyclin-dependent kinase 1 (CDK1)-cyclin B1 and M-phase inducer phosphatase 3 (CDC25c), and a decreased apoptosis threshold with a low antiapoptotic/proapoptotic protein ratio. Our study reports STIM2 as a new actor regulating genomic stability and p53 response in terms of cell cycle and apoptosis of human normal and malignant monocytic cells.
Collapse
Affiliation(s)
| | - Raphaël Itzykson
- Département Hématologie et ImmunologieHôpital Saint‐Louis, Assistance Publique‐Hôpitaux de ParisFrance
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRSUniversité Paris CitéFrance
| | - Frédéric Hague
- Laboratoire de Physiologie Cellulaire et Moléculaire UR4667Université Picardie Jules VerneAmiensFrance
| | - Delphine Lebon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Julien Legrand
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | - Guillaume Jedraszak
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Laboratoire de Génétique ConstitutionnelleCHU Amiens‐PicardieFrance
| | | | - Louison Collet
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Etienne Paubelle
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Jean‐Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| | - Thomas Boyer
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| |
Collapse
|
10
|
Ding B, Ye Z, Yin H, Hong XY, Feng SW, Xu JY, Shen Y. Comprehensive single-cell analysis reveals heterogeneity of fibroblast subpopulations in ovarian cancer tissue microenvironment. Heliyon 2024; 10:e27873. [PMID: 38533040 PMCID: PMC10963331 DOI: 10.1016/j.heliyon.2024.e27873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024] Open
Abstract
Background Ovarian cancer, as a highly malignant tumor, features the critical involvement of tumor-associated fibroblasts in the ovarian cancer tissue microenvironment. However, due to the apparent heterogeneity within fibroblast subpopulations, the specific functions of these subpopulations in the ovarian cancer tissue microenvironment remain insufficiently elucidated. Methods In this study, we integrated single-cell sequencing data from 32 ovarian cancer samples derived from four distinct cohorts and 3226 bulk RNA-seq data from GEO and TCGA-OV cohorts. Utilizing computational frameworks such as Seurat, Monocle 2, Cellchat, and others, we analyzed the characteristics of the ovarian cancer tissue microenvironment, focusing particularly on fibroblast subpopulations and their differentiation trajectories. Employing the CIBERSORTX computational framework, we assessed various cellular components within the ovarian cancer tissue microenvironment and evaluated their associations with ovarian cancer prognosis. Additionally, we conducted Mendelian randomization analysis based on cis-eQTL to investigate causal relationships between gene expression and ovarian cancer. Results Through integrative analysis, we identified 13 major cell types present in ovarian cancer tissues, including CD8+ T cells, malignant cells, and fibroblasts. Analysis of the tumor microenvironment (TME) cell proportions revealed a significant increase in the proportion of CD8+ T cells and CD4+ T cells in tumor tissues compared to normal tissues, while fibroblasts predominated in normal tissues. Further subgroup analysis of fibroblasts identified seven subgroups, with the MMP11+Fib subgroup showing the highest activity in the TGFβ signaling pathway. Single-cell analysis suggested that oxidative phosphorylation could be a key pathway driving fibroblast differentiation, and the ATRNL1+KCN + Fib subgroup exhibited chromosomal copy number variations. Prognostic analysis using a large sample size indicated that high infiltration of MMP11+ fibroblasts was associated with poor prognosis in ovarian cancer. SMR analysis identified 132 fibroblast differentiation-related genes, which were linked to pathways such as platinum drug resistance. Conclusions In the context of ovarian cancer, fibroblasts expressing MMP11 emerge as the primary drivers of the TGF-beta signaling pathway. Their presence correlates with an increased risk of adverse ovarian prognoses. Additionally, the genetic regulation governing the differentiation of fibroblasts associated with ovarian cancer correlates with the emergence of drug resistance.
Collapse
Affiliation(s)
- Bo Ding
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Han Yin
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xin-Yi Hong
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Song-wei Feng
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jing-Yun Xu
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Safieh J, Chazan A, Saleem H, Vyas P, Danin-Poleg Y, Ron D, Haran TE. A molecular mechanism for the "digital" response of p53 to stress. Proc Natl Acad Sci U S A 2023; 120:e2305713120. [PMID: 38015851 PMCID: PMC10710088 DOI: 10.1073/pnas.2305713120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023] Open
Abstract
The tumor suppressor protein p53 accumulates in response to cellular stress and consequently orchestrates the expression of multiple genes in a p53-level and time-dependent manner to overcome stress consequences, for which a molecular mechanism is currently unknown. Previously, we reported that DNA torsional flexibility distinguishes among p53 response elements (REs) and that transactivation at basal p53 levels is correlated with p53 REs flexibility. Here, we calculated the flexibility of ~200 p53 REs. By connecting functional outcomes of p53-target genes' activation to the calculated flexibility of their REs, we show that genes known to belong to pathways that are activated rapidly upon stress contain REs that are significantly more flexible relative to REs of genes known to be involved in pathways that are activated later in the response to stress. The global structural properties of several p53 REs belonging to different pathways were experimentally validated. Additionally, reporter-gene expression driven by flexible p53 REs occurred at lower p53 levels and with faster rates than expression from rigid REs. Furthermore, analysis of published endogenous mRNA levels of p53-target genes as a function of REs' flexibility showed that early versus late genes differ significantly in their flexibility properties of their REs and that highly flexible p53 REs enable high-activation level exclusively to early-response genes. Overall, we demonstrate that DNA flexibility of p53 REs contributes significantly to functional selectivity in the p53 system by facilitating the initial steps of p53-dependent target-genes expression, thereby contributing to survival versus death decisions in the p53 system.
Collapse
Affiliation(s)
- Jessy Safieh
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Ariel Chazan
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Hanna Saleem
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Pratik Vyas
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Yael Danin-Poleg
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Dina Ron
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Tali E. Haran
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| |
Collapse
|
12
|
Khan GH, Veltkamp F, Scheper M, Hoebe RA, Claessen N, Butter L, Bouts AHM, Florquin S, Guikema JEJ. Levamisole suppresses activation and proliferation of human T cells by the induction of a p53-dependent DNA damage response. Eur J Immunol 2023; 53:e2350562. [PMID: 37597325 DOI: 10.1002/eji.202350562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 08/21/2023]
Abstract
Levamisole (LMS) is a small molecule used in the treatment of idiopathic nephrotic syndrome (INS). The pathogenesis of INS remains unknown, but evidence points toward an immunological basis of the disease. Recently, LMS has been shown to increase the relapse-free survival in INS patients. While LMS has been hypothesized to exert an immunomodulatory effect, its mechanism of action remains unknown. Here, we show that LMS decreased activation and proliferation of human T cells. T-cell activation-associated cytokines such as IL-2, TNF-α, and IFN-γ were reduced upon LMS treatment, whereas IL-4 and IL-13 were increased. Gene expression profiling confirmed that the suppressive effects of LMS as genes involved in cell cycle progression were downregulated. Furthermore, genes associated with p53 activation were upregulated by LMS. In agreement, LMS treatment resulted in p53 phosphorylation and increased expression of the p53 target gene FAS. Accordingly, LMS sensitized activated T cells for Fas-mediated apoptosis. LMS treatment resulted in a mid-S phase cell cycle arrest accompanied by γH2AX-foci formation and phosphorylation of CHK1. Our findings indicate that LMS acts as an immunosuppressive drug that directly affects the activation and proliferation of human T cells by induction of DNA damage and the activation of a p53-dependent DNA damage response.
Collapse
Affiliation(s)
- Gerarda H Khan
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Floor Veltkamp
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mirte Scheper
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron A Hoebe
- Department of Medical Biology, Amsterdam UMC and Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Nike Claessen
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Loes Butter
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Antonia H M Bouts
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen E J Guikema
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Mauro S, Bolognesi MM, Villa N, Capitoli G, Furia L, Mascadri F, Zucchini N, Totis M, Faretta M, Galimberti S, Bovo G, Cattoretti G. A DNA damage response-like phenotype defines a third of colon cancers at onset. FASEB J 2023; 37:e23020. [PMID: 37342943 DOI: 10.1096/fj.202300132r] [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: 01/25/2023] [Revised: 04/12/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
Colon adenocarcinoma (COAD) has a limited range of diversified, personalized therapeutic opportunities, besides DNA hypermutating cases; thus, both new targets or broadening existing strategies for personalized intervention are of interest. Routinely processed material from 246 untreated COADs with clinical follow-up was probed for evidence of DNA damage response (DDR), that is, the gathering of DDR-associated molecules at discrete nuclear spots, by multiplex immunofluorescence and immunohistochemical staining for DDR complex proteins (γH2AX, pCHK2, and pNBS1). We also tested the cases for type I interferon response, T-lymphocyte infiltration (TILs), and mutation mismatch repair defects (MMRd), known to be associated with defects of DNA repair. FISH analysis for chromosome 20q copy number variations was obtained. A total of 33.7% of COAD display a coordinated DDR on quiescent, non-senescent, non-apoptotic glands, irrespective of TP53 status, chromosome 20q abnormalities, and type I IFN response. Clinicopathological parameters did not differentiate DDR+ cases from the other cases. TILs were equally present in DDR and non-DDR cases. DDR+ MMRd cases were preferentially retaining wild-type MLH1. The outcome after 5FU-based chemotherapy was not different in the two groups. DDR+ COAD represents a subgroup not aligned with known diagnostic, prognostic, or therapeutic categories, with potential new targeted treatment opportunities, exploiting the DNA damage repair pathways.
Collapse
Affiliation(s)
- Stefania Mauro
- Pathology, Vimercate Hospital, ASST-Brianza, Vimercate, Italy
| | - Maddalena M Bolognesi
- Pathology, Department of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Nicoletta Villa
- Genetics, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| | - Giulia Capitoli
- Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, School of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Laura Furia
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesco Mascadri
- Pathology, Department of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Nicola Zucchini
- Pathology, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| | - Mauro Totis
- GI Surgery, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Stefania Galimberti
- Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, School of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
| | - Giorgio Bovo
- Pathology, Vimercate Hospital, ASST-Brianza, Vimercate, Italy
| | - Giorgio Cattoretti
- Pathology, Department of Medicine and Surgery, Universitá di Milano-Bicocca, Monza, Italy
- Pathology, Fondazione IRCCS San Gerardo dei Tintori Monza, Monza, Italy
| |
Collapse
|
14
|
Stillger MN, Chen CY, Lai ZW, Li M, Schäfer A, Pagenstecher A, Nimsky C, Bartsch JW, Schilling O. Changes in calpain-2 expression during glioblastoma progression predisposes tumor cells to temozolomide resistance by minimizing DNA damage and p53-dependent apoptosis. Cancer Cell Int 2023; 23:49. [PMID: 36932402 PMCID: PMC10022304 DOI: 10.1186/s12935-023-02889-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/04/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is characterized by an unfavorable prognosis for patients affected. During standard-of-care chemotherapy using temozolomide (TMZ), tumors acquire resistance thereby causing tumor recurrence. Thus, deciphering essential molecular pathways causing TMZ resistance are of high therapeutic relevance. METHODS Mass spectrometry based proteomics were used to study the GBM proteome. Immunohistochemistry staining of human GBM tissue for either calpain-1 or -2 was performed to locate expression of proteases. In vitro cell based assays were used to measure cell viability and survival of primary patient-derived GBM cells and established GBM cell lines after TMZ ± calpain inhibitor administration. shRNA expression knockdowns of either calpain-1 or calpain-2 were generated to study TMZ sensitivity of the specific subunits. The Comet assay and ɣH2AX signal measurements were performed in order to assess the DNA damage amount and recognition. Finally, quantitative real-time PCR of target proteins was applied to differentiate between transcriptional and post-translational regulation. RESULTS Calcium-dependent calpain proteases, in particular calpain-2, are more abundant in glioblastoma compared to normal brain and increased in patient-matched initial and recurrent glioblastomas. On the cellular level, pharmacological calpain inhibition increased the sensitivities of primary glioblastoma cells towards TMZ. A genetic knockdown of calpain-2 in U251 cells led to increased caspase-3 cleavage and sensitivity to neocarzinostatin, which rapidly induces DNA strand breakage. We hypothesize that calpain-2 causes desensitization of tumor cells against TMZ by preventing strong DNA damage and subsequent apoptosis via post-translational TP53 inhibition. Indeed, proteomic comparison of U251 control vs. U251 calpain-2 knockdown cells highlights perturbed levels of numerous proteins involved in DNA damage response and downstream pathways affecting TP53 and NF-κB signaling. TP53 showed increased protein abundance, but no transcriptional regulation. CONCLUSION TMZ-induced cell death in the presence of calpain-2 expression appears to favor DNA repair and promote cell survival. We conclude from our experiments that calpain-2 expression represents a proteomic mode that is associated with higher resistance via "priming" GBM cells to TMZ chemotherapy. Thus, calpain-2 could serve as a prognostic factor for GBM outcome.
Collapse
Affiliation(s)
- Maren Nicole Stillger
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Chia-Yi Chen
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Zon Weng Lai
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Mujia Li
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Agnes Schäfer
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany
| | - Axel Pagenstecher
- Institute of Neuropathology, Philipps-University, Marburg, Germany.,Center for Mind, Brain and Behavior, CMBB, Marburg University, Hans-Meerwein-Strasse 6, 35032, Marburg, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, CMBB, Marburg University, Hans-Meerwein-Strasse 6, 35032, Marburg, Germany
| | - Jörg Walter Bartsch
- Department of Neurosurgery, Philipps-University Marburg, Marburg, Germany. .,Center for Mind, Brain and Behavior, CMBB, Marburg University, Hans-Meerwein-Strasse 6, 35032, Marburg, Germany. .,Philipps-University Marburg, Laboratory, Department of Neurosurgery, University Hospital Marburg, Baldingerstr., 35033, Marburg, Germany.
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
15
|
Yesil S, Ozdemir C, Arslan M, Gundogdu AC, Kavutcu M, Atan A. Protective effect of cerium oxide on testicular function and oxidative stress after torsion/detorsion in adult male rats. Exp Ther Med 2023; 25:1. [PMID: 36561629 PMCID: PMC9748645 DOI: 10.3892/etm.2022.11700] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Testicular torsion (T)/detorsion (D) can cause testicular injury due to the rotation of the spermatic cord and its vessels, therefore it represents an urological emergency that is surgically treated. Oxidative damage occurs in the testis and distant organs because of the overproduction of free radicals and overexpression of proinflammatory cytokines by reperfusion after surgery. Cerium oxide (CeO2) nanoparticles, a material also known as nanoceria, have regenerative antioxidant properties on oxidative stress. The present study aimed to investigate the effects of nanoceria on testis tissues in testicular T/D in rats. A total of 24 rats were equally and randomly divided into four groups: Control, CeO2, T/D and CeO2-T/D groups. Left inguinoscrotal incision was performed in the control group. In the CeO2 group, 0.5 mg/kg CeO2 was given intraperitoneally 30 min before inguinoscrotal incision. In the T/D group, unilateral testicular T/D was performed through an inguinoscrotal incision and rotating the left testis 720˚ clockwise, which was then left ischemic for 120 min, followed by 120 min of reperfusion. In the CeO2-T/D group, 0.5 mg/kg CeO2 was given intraperitoneally 30 min before testicular T/D. At the end of the experiment, testis tissues were removed for histopathological and biochemical examinations. The samples were histologically examined, Glutathione-s transferase (GST), catalase (CAT), paraoxonase (PON) activities and malondialdehyde (MDA) levels were measured via biochemical analysis methods, while the expression levels of p53, Bax and Bcl-2 were detected using immunohistochemistry. The present results revealed statistically significant inter-group differences in PON, CAT and GST activities and MDA levels. GST, CAT and PON activities were significantly higher, whereas MDA levels in the CeO2-T/D group were significantly lower compared with those in the T/D group. The T/D group had increased Bax and decreased Bcl-2 expression levels in their seminiferous tubules compared with the control and CeO2 groups. CeO2 treatment led to downregulation of Bax and upregulation of Bcl-2. The expression of p53 was high in the T/D group compared with that in the control and CeO2 groups, and was upregulated in all germinal cells. However, compared with that in the T/D group, p53 expression was significantly decreased in the CeO2-T/D group. The testicular injury score significantly increased in the CeO2-T/D group compared with the control and CeO2 groups. Rats in the CeO2-T/D group demonstrated significantly milder tissue lesions compared with those in T/D group. The present findings indicated that nanoceria may protect testis in rats against the harmful effects of T/D. Further studies are required to evaluate how CeO2 reduces oxidative stress and cell death in testis tissue that underwent T/D-related injury.
Collapse
Affiliation(s)
- Suleyman Yesil
- Department of Urology, Medical Faculty, Gazi University, 06510 Ankara, Turkey
| | - Cagri Ozdemir
- Department of Anesthesiology and Reanimation, Medical Faculty, Gazi University, 06510 Ankara, Turkey
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Medical Faculty, Gazi University, 06510 Ankara, Turkey
- Life Sciences Application and Research Center, Gazi University, 06510 Ankara, Turkey
| | - Ayse Cakir Gundogdu
- Department of Histology and Embryology, Medical Faculty, Kutahya Health Sciences University, 43050 Kutahya, Turkey
| | - Mustafa Kavutcu
- Department of Medical Biochemistry, Medical Faculty, Gazi University, 06510 Ankara, Turkey
| | - Ali Atan
- Department of Urology, Medical Faculty, Gazi University, 06510 Ankara, Turkey
| |
Collapse
|
16
|
Prives C. How Terri Grodzicker transformed Genes & Development. Genes Dev 2023; 37:4-5. [PMID: 37061965 DOI: 10.1101/gad.350474.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
GUEST EDITOR.
Collapse
Affiliation(s)
- Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
17
|
Legartová S, Fagherazzi P, Goswami P, Brazda V, Lochmanová G, Koutná I, Bártová E. Irradiation potentiates p53 phosphorylation and p53 binding to the promoter and coding region of the TP53 gene. Biochimie 2023; 204:154-168. [PMID: 36167255 DOI: 10.1016/j.biochi.2022.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/08/2022] [Accepted: 09/21/2022] [Indexed: 01/12/2023]
Abstract
An essential factor of the DNA damage response is 53BP1, a multimeric protein that inhibits the resection-dependent double-strand break (DBS) repair. The p53 protein is a tumor suppressor known as a guardian of the genome. Although the interaction between 53BP1 and its p53 partner is well-known in regulating gene expression, a question remains whether genome injury can affect the interaction between 53BP1 and p53 proteins or p53 binding to DNA. Here, using mass spectrometry, we determine post-translational modifications and interaction properties of 53BP1 and p53 proteins in non-irradiated and γ-irradiated cells. In addition, we used Atomic Force Microscopy (AFM) and Fluorescent Lifetime Imaging Microscopy combined with Fluorescence Resonance Energy Transfer (FLIM-FRET) for studies of p53 binding to DNA. Also, we used local laser microirradiation as a tool of advanced confocal microscopy, showing selected protein accumulation at locally induced DNA lesions. We observed that 53BP1 and p53 proteins accumulate at microirradiated chromatin but with distinct kinetics. The density of 53BP1 (53BP1pS1778) phosphorylated form was lower in DNA lesions than in the non-specified form. By mass spectrometry, we found 22 phosphorylations, 4 acetylation sites, and methylation of arginine 1355 within the DNA-binding domain of the 53BP1 protein (aa1219-1711). The p53 protein was phosphorylated on 8 amino acids and acetylated on the N-terminal domain. Post-translational modifications (PTMs) of 53BP1 were not changed in cells exposed to γ-radiation, while γ-rays increased the level of S6ph and S15ph in p53. Interaction analysis showed that 53BP1 and p53 proteins have 54 identical interaction protein partners, and AFM revealed that p53 binds to both non-specific and TP53-specific sequences (AGACATGCCTA GGCATGTCT). Irradiation by γ-rays enhanced the density of the p53 protein at the AGACATGCCTAGGCATGTCT region, and the binding of p53 S15ph to the TP53 promoter was potentiated in irradiated cells. These findings show that γ-irradiation, in general, strengthens the binding of phosphorylated p53 protein to the encoding gene.
Collapse
Affiliation(s)
- Soňa Legartová
- Department of Cell Biology and Epigenetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Paolo Fagherazzi
- Department of Cell Biology and Epigenetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Pratik Goswami
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Vaclav Brazda
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| | - Gabriela Lochmanová
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Irena Koutná
- The International Clinical Research Center of St. Anne's University Hospital in Brno (FNUSA-ICRC), Pekařská 53, 656 91, Brno, Czech Republic
| | - Eva Bártová
- Department of Cell Biology and Epigenetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic.
| |
Collapse
|
18
|
Patil MR, Bihari A. A comprehensive study of p53 protein. J Cell Biochem 2022; 123:1891-1937. [PMID: 36183376 DOI: 10.1002/jcb.30331] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 01/10/2023]
Abstract
The protein p53 has been extensively investigated since it was found 43 years ago and has become a "guardian of the genome" that regulates the division of cells by preventing the growth of cells and dividing them, that is, inhibits the development of tumors. Initial proof of protein existence by researchers in the mid-1970s was found by altering and regulating the SV40 big T antigen termed the A protein. Researchers demonstrated how viruses play a role in cancer by employing viruses' ability to create T-antigens complex with viral tumors, which was discovered in 1979 following a viral analysis and cancer analog research. Researchers later in the year 1989 explained that in Murine Friend, a virus-caused erythroleukemia, commonly found that p53 was inactivated to suggest that p53 could be a "tumor suppressor gene." The TP53 gene, encoding p53, is one of human cancer's most frequently altered genes. The protein-regulated biological functions of all p53s include cell cycles, apoptosis, senescence, metabolism of the DNA, angiogenesis, cell differentiation, and immunological response. We tried to unfold the history of the p53 protein, which was discovered long back in 1979, that is, 43 years of research on p53, and how p53's function has been developed through time in this article.
Collapse
Affiliation(s)
- Manisha R Patil
- Department of Computer-Applications, School of Information Technology and Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Anand Bihari
- Department of Computational Intelligence, School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
19
|
Mohibi S, Chen X, Zhang J. ZFP14 Regulates Cancer Cell Growth and Migration by Modulating p53 Protein Stability as Part of the MDM2 E3 Ubiquitin Ligase Complex. Cancers (Basel) 2022; 14:cancers14215226. [PMID: 36358645 PMCID: PMC9655198 DOI: 10.3390/cancers14215226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/16/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multi-zinc finger proteins that contain a KRAB domain are part of the biggest family of transcription factors in mammals. However, the physiological or pathological functions for the majority of them are unknown. Here, we showed that ZFP14 (also known as ZNF531) is a p53 target gene that can be induced upon genotoxic stress in a p53-dependent manner. To determine the function of ZFP14 in mouse and human cancer cell lines, we generated multiple cell lines where ZFP14 was knocked out. We showed that ZFP14-KO inhibits cancer cell growth and migration. We also showed that, as a target of p53, ZFP14, in turn, represses p53 expression and that the knockdown of p53 restores the potential of ZFP14-KO cells to proliferate and migrate. Mechanistically, we found that ZFP14 modulates p53 protein stability by increasing its ubiquitination via associating with and possibly enhancing MDM2/p53 complex integrity through its zinc finger domains. Our findings suggest that the reciprocal regulation of p53 and ZFP14 represents a novel p53-ZFP14 regulatory loop and that ZFP14 plays a role in p53-dependent tumor suppression.
Collapse
|
20
|
Gaulin C, Kelemen K, Arana Yi C. Molecular Pathways in Clonal Hematopoiesis: From the Acquisition of Somatic Mutations to Transformation into Hematologic Neoplasm. Life (Basel) 2022; 12:1135. [PMID: 36013314 PMCID: PMC9410004 DOI: 10.3390/life12081135] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cell aging, through the acquisition of somatic mutations, gives rise to clonal hematopoiesis (CH). While a high prevalence of CH has been described in otherwise healthy older adults, CH confers an increased risk of both hematologic and non-hematologic diseases. Classification of CH into clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS) further describes this neoplastic myeloid precursor state and stratifies individuals at risk of developing clinically significant complications. The sequential acquisition of driver mutations, such as DNMT3A, TET2, and ASXL1, provide a selective advantage and lead to clonal expansion. Inflammation, microbiome signatures, and external selective pressures also contribute to clonal evolution. Despite significant progress in recent years, the precise molecular mechanisms driving CH transformation to hematologic neoplasms are not well defined. Further understanding of these complex mechanisms may improve risk stratification and introduce therapeutic interventions in CH. Here we discuss the genetic drivers underpinning CH, mechanisms for clonal evolution, and transformation to hematologic neoplasm.
Collapse
Affiliation(s)
- Charles Gaulin
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Katalin Kelemen
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Cecilia Arana Yi
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| |
Collapse
|
21
|
McCubrey JA, Meher AK, Akula SM, Abrams SL, Steelman LS, LaHair MM, Franklin RA, Martelli AM, Ratti S, Cocco L, Barbaro F, Duda P, Gizak A. Wild type and gain of function mutant TP53 can regulate the sensitivity of pancreatic cancer cells to chemotherapeutic drugs, EGFR/Ras/Raf/MEK, and PI3K/mTORC1/GSK-3 pathway inhibitors, nutraceuticals and alter metabolic properties. Aging (Albany NY) 2022; 14:3365-3386. [PMID: 35477123 PMCID: PMC9085237 DOI: 10.18632/aging.204038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022]
Abstract
TP53 is a master regulator of many signaling and apoptotic pathways involved in: aging, cell cycle progression, gene regulation, growth, apoptosis, cellular senescence, DNA repair, drug resistance, malignant transformation, metastasis, and metabolism. Most pancreatic cancers are classified as pancreatic ductal adenocarcinomas (PDAC). The tumor suppressor gene TP53 is mutated frequently (50-75%) in PDAC. Different types of TP53 mutations have been observed including gain of function (GOF) point mutations and various deletions of the TP53 gene resulting in lack of the protein expression. Most PDACs have point mutations at the KRAS gene which result in constitutive activation of KRas and multiple downstream signaling pathways. It has been difficult to develop specific KRas inhibitors and/or methods that result in recovery of functional TP53 activity. To further elucidate the roles of TP53 in drug-resistance of pancreatic cancer cells, we introduced wild-type (WT) TP53 or a control vector into two different PDAC cell lines. Introduction of WT-TP53 increased the sensitivity of the cells to multiple chemotherapeutic drugs, signal transduction inhibitors, drugs and nutraceuticals and influenced key metabolic properties of the cells. Therefore, TP53 is a key molecule which is critical in drug sensitivity and metabolism of PDAC.
Collapse
Affiliation(s)
- James A. McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Shaw M. Akula
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Stephen L. Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Linda S. Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Michelle M. LaHair
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Richard A. Franklin
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Fulvio Barbaro
- Department of Medicine and Surgery, Re.Mo.Bio.S. Laboratory, Anatomy Section, University of Parma, Parma, Italy
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
22
|
Rapid recruitment of p53 to DNA damage sites directs DNA repair choice and integrity. Proc Natl Acad Sci U S A 2022; 119:e2113233119. [PMID: 35235448 PMCID: PMC8915893 DOI: 10.1073/pnas.2113233119] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Our work focuses on the critical longstanding question of the nontranscriptional role of p53 in tumor suppression. We demonstrate here that poly(ADP-ribose) polymerase (PARP)–dependent modification of p53 enables rapid recruitment of p53 to damage sites, where it in turn directs early repair pathway selection. Specifically, p53-mediated recruitment of 53BP1 at early time points promotes nonhomologous end joining over the more error-prone microhomology end-joining. Similarly, p53 directs nucleotide excision repair by mediating DDB1 recruitment. This property of p53 also correlates with tumor suppression in vivo. Our study provides mechanistic insight into how certain transcriptionally deficient p53 mutants may retain tumor-suppressive functions through regulating the DNA damage response. p53 is primarily known as a downstream transcriptional effector in the DNA damage-response cascade. We report that endogenous p53 rapidly accumulates at DNA damage sites within 2 s of UVA microirradiation. The kinetics of p53 recruitment mimics those of known DNA damage-response proteins, such as Ku70 and poly(ADP-ribose) polymerase (PARP), and precedes recruitment of Nbs1, 53BP1, and DDB1. Mutations in the DNA-binding and C-terminal domains significantly suppress this rapid recruitment. The C-terminal domain of p53 contains key residues for PARP interaction that are required for rapid recruitment of p53 to DNA damage sites, as is PARP-dependent modification. The presence of p53 at damage sites influences the recruitment kinetics of 53BP1 and DDB1 and directs the choice of nonhomologous end joining repair (NHEJ) and nucleotide excision repair. Mutations that suppressed rapid recruitment of p53 promoted error-prone alternative end-joining (alt-NHEJ) and inhibited nucleotide excision repair. Our finding that p53 is a critical early responder to DNA damage stands in contrast with its extensively studied role as a downstream transcriptional regulator in DNA damage repair. We highlight an unrecognized role of p53 in directing DNA repair dynamics and integrity and suggest a parallel mode of p53 tumor suppression apart from its function as a transcription factor.
Collapse
|
23
|
Klimovich B, Meyer L, Merle N, Neumann M, König AM, Ananikidis N, Keber CU, Elmshäuser S, Timofeev O, Stiewe T. Partial p53 reactivation is sufficient to induce cancer regression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:80. [PMID: 35232479 PMCID: PMC8889716 DOI: 10.1186/s13046-022-02269-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/20/2022] [Indexed: 01/21/2023]
Abstract
Background Impaired p53 function is one of the central molecular features of a tumor cell and even a partial reduction in p53 activity can increase the cancer risk in mice and men. From a therapeutic perspective it is noteworthy that tumor cells often become addicted to the absence of p53 providing a rationale for developing p53 reactivating compounds to treat cancer patients. Unfortunately, many of the compounds that are currently undergoing preclinical and clinical testing fail to fully reactivate mutant p53 proteins, raising the crucial question: how much p53 activity is needed to elicit a therapeutic effect? Methods We have genetically modelled partial p53 reactivation using knock-in mice with inducible expression of the p53 variant E177R. This variant has a reduced ability to bind and transactivate target genes and consequently causes moderate cancer susceptibility. We have generated different syngeneically transplanted and autochthonous mouse models of p53-deficient acute myeloid leukemia and B or T cell lymphoma. After cancer manifestation we have activated E177R expression and analyzed the in vivo therapy response by bioluminescence or magnetic resonance imaging. The molecular response was further characterized in vitro by assays for gene expression, proliferation, senescence, differentiation, apoptosis and clonogenic growth. Results We report the conceptually intriguing observation that the p53 variant E177R, which promotes de novo leukemia and lymphoma formation, inhibits proliferation and viability, induces immune cell infiltration and triggers cancer regression in vivo when introduced into p53-deficient leukemia and lymphomas. p53-deficient cancer cells proved to be so addicted to the absence of p53 that even the low-level activity of E177R is detrimental to cancer growth. Conclusions The observation that a partial loss-of-function p53 variant promotes tumorigenesis in one setting and induces regression in another, underlines the highly context-specific effects of individual p53 mutants. It further highlights the exquisite sensitivity of cancer cells to even small changes in p53 activity and reveals that changes in activity level are more important than the absolute level. As such, the study encourages ongoing research efforts into mutant p53 reactivating drugs by providing genetic proof-of-principle evidence that incomplete p53 reactivation may suffice to elicit a therapeutic response. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02269-6.
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Laura Meyer
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Michelle Neumann
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Alexander M König
- Clinic of Diagnostic and Interventional Radiology, Core Facility 7T-small animal MRI, Philipps-University, Marburg, Germany
| | - Nikolaos Ananikidis
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Corinna U Keber
- Institute for Pathology, University Hospital Marburg, Philipps-University, Marburg, Germany
| | - Sabrina Elmshäuser
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany. .,German Center for Lung Research (DZL), Philipps-University, Marburg, Germany.
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University, Marburg, Germany. .,German Center for Lung Research (DZL), Philipps-University, Marburg, Germany. .,Genomics Core Facility, Philipps-University, Marburg, Germany.
| |
Collapse
|
24
|
Biglycan Interacts with Type I Insulin-like Receptor (IGF-IR) Signaling Pathway to Regulate Osteosarcoma Cell Growth and Response to Chemotherapy. Cancers (Basel) 2022; 14:cancers14051196. [PMID: 35267503 PMCID: PMC8909324 DOI: 10.3390/cancers14051196] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Osteosarcoma (OS) is an aggressive, primary bone cancer. OS cells produce altered osteoid whose components participate in signaling correlated to the development of this cancer. Biglycan (BGN), a proteoglycan, is correlated to aggressive OS type and resistance to chemotherapy. A constitutive signaling of insulin-like growth factor receptor I (IGF-IR) signaling in sarcoma progression was established. We showed that biglycan binds IGF-IR resulting in prolonged IGF-IR activation, nuclear translocation, and growth response of the poorly-differentiated MG63 cells correlated to increased aggressiveness markers expression and enhanced chemoresistance. This mechanism is not valid in moderately and well-differentiated, biglycan non-expressing U-2OS and Saos-2 OS cells. Abstract Osteosarcoma (OS) is a mesenchymally derived, aggressive bone cancer. OS cells produce an aberrant nonmineralized or partly mineralized extracellular matrix (ECM) whose components participate in signaling pathways connected to specific pathogenic phenotypes of this bone cancer. The expression of biglycan (BGN), a secreted small leucine-rich proteoglycan (SLRP), is correlated to aggressive OS phenotype and resistance to chemotherapy. A constitutive signaling of IGF-IR signaling input in sarcoma progression has been established. Here, we show that biglycan activates the IGF-IR signaling pathway to promote MG63 biglycan-secreting OS cell growth by forming a complex with the receptor. Computational models of IGF-IR and biglycan docking suggest that biglycan binds IGF-IR dimer via its concave surface. Our binding free energy calculations indicate the formation of a stable complex. Biglycan binding results in prolonged IGF-IR activation leading to protracted IGF-IR-dependent cell growth response of the poorly-differentiated MG63 cells. Moreover, biglycan facilitates the internalization (p ≤ 0.01, p ≤ 0.001) and sumoylation-enhanced nuclear translocation of IGF-IR (p ≤ 0.05) and its DNA binding in MG63 cells (p ≤ 0.001). The tyrosine kinase activity of the receptor mediates this mechanism. Furthermore, biglycan downregulates the expression of the tumor-suppressor gene, PTEN (p ≤ 0.01), and increases the expression of endothelial–mesenchymal transition (EMT) and aggressiveness markers vimentin (p ≤ 0.01) and fibronectin (p ≤ 0.01) in MG63 cells. Interestingly, this mechanism is not valid in moderately and well-differentiated, biglycan non-expressing U-2OS and Saos-2 OS cells. Furthermore, biglycan exhibits protective effects against the chemotherapeutic drug, doxorubicin, in MG63 OS cells (p ≤ 0.01). In conclusion, these data indicate a potential direct and adjunct therapeutical role of biglycan in osteosarcoma.
Collapse
|
25
|
Hepatitis B Viral Protein HBx and the Molecular Mechanisms Modulating the Hallmarks of Hepatocellular Carcinoma: A Comprehensive Review. Cells 2022; 11:cells11040741. [PMID: 35203390 PMCID: PMC8870387 DOI: 10.3390/cells11040741] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
With 296 million cases estimated worldwide, chronic hepatitis B virus (HBV) infection is the most common risk factor for hepatocellular carcinoma (HCC). HBV-encoded oncogene X protein (HBx), a key multifunctional regulatory protein, drives viral replication and interferes with several cellular signalling pathways that drive virus-associated hepatocarcinogenesis. This review article provides a comprehensive overview of the role of HBx in modulating the various hallmarks of HCC by supporting tumour initiation, progression, invasion and metastasis. Understanding HBx-mediated dimensions of complexity in driving liver malignancies could provide the key to unlocking novel and repurposed combinatorial therapies to combat HCC.
Collapse
|
26
|
Klimovich B, Merle N, Neumann M, Elmshäuser S, Nist A, Mernberger M, Kazdal D, Stenzinger A, Timofeev O, Stiewe T. p53 partial loss-of-function mutations sensitize to chemotherapy. Oncogene 2022; 41:1011-1023. [PMID: 34907344 PMCID: PMC8837531 DOI: 10.1038/s41388-021-02141-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/23/2022]
Abstract
The tumor suppressive transcription factor p53 is frequently inactivated in cancer cells by missense mutations that cluster in the DNA binding domain. 30% hit mutational hotspot residues, resulting in a complete loss of transcriptional activity and mutant p53-driven chemotherapy resistance. Of the remaining 70% of non-hotspot mutants, many are partial loss-of-function (partial-LOF) mutants with residual transcriptional activity. The therapeutic consequences of a partial-LOF have remained largely elusive. Using a p53 mutation engineered to reduce DNA binding, we demonstrate that partial-LOF is sufficient to enhance oncogene-driven tumorigenesis in mouse models of lung and pancreatic ductal adenocarcinoma and acute myeloid leukemia. Interestingly, mouse and human tumors with partial-LOF mutations showed mutant p53 protein accumulation similar as known for hotspot mutants. Different from the chemotherapy resistance caused by p53-loss, the partial-LOF mutant sensitized to an apoptotic chemotherapy response and led to a survival benefit. Mechanistically, the pro-apoptotic transcriptional activity of mouse and human partial-LOF mutants was rescued at high mutant protein levels, suggesting that accumulation of partial-LOF mutants enables the observed apoptotic chemotherapy response. p53 non-hotspot mutants with partial-LOF, therefore, represent tumorigenic p53 mutations that need to be distinguished from other mutations because of their beneficial impact on survival in a therapy context.
Collapse
MESH Headings
- Animals
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Humans
- Mice
- Loss of Function Mutation
- Drug Resistance, Neoplasm/genetics
- Apoptosis/genetics
- Apoptosis/drug effects
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Michelle Neumann
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Sabrina Elmshäuser
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps-University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany.
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany.
- Genomics Core Facility, Philipps-University, Marburg, Germany.
| |
Collapse
|
27
|
Wang Y, Wang H, Hu L, Chen L. Leptin Gene Protects Against Cold Stress in Antarctic Toothfish. Front Physiol 2021; 12:740806. [PMID: 34975517 PMCID: PMC8715755 DOI: 10.3389/fphys.2021.740806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Leptin is a cytokine-like peptide, predominantly biosynthesized in adipose tissue, which plays an important role in regulating food intake, energy balance and reproduction in mammals. However, how it may have been modified to enable life in the chronic cold is unclear. Here, we identified a leptin-a gene (lepa) in the cold-adapted and neutrally buoyant Antarctic toothfish Dissostichus mawsoni that encodes a polypeptide carrying four α-helices and two cysteine residues forming in-chain disulfide bonds, structures shared by most vertebrate leptins. Quantitative RT-PCR confirmed that mRNA levels of the leptin-a gene of D. mawsoni (DM-lepa) were highest in muscle, followed by kidney and liver; detection levels were low in the gill, brain, intestine, and ovary tissues. Compared with leptin-a genes of fishes living in warmer waters, DM-lepa underwent rapid evolution and was subjected to positive selection. Over-expression of DM-lepa in the zebrafish cell line ZFL resulted in signal accumulation in the cytoplasm and significantly increased cell proliferation both at the normal culture temperature and under cold treatment. DM-lepa over-expression also reduced apoptosis under low-temperature stress and activated the STAT3 signaling pathway, in turn upregulating the anti-apoptotic proteins bcl2l1, bcl2a, myca and mdm2 while downregulating the pro-apoptotic baxa, p53 and caspase-3. These results demonstrate that DM-lepa, through STAT3 signaling, plays a protective role in cold stress by preventing apoptotic damage. Our study reveals a new role of lepa in polar fish.
Collapse
Affiliation(s)
- Ying Wang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
| | - Huamin Wang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
| | - Linghong Hu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
| | - Liangbiao Chen
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai, China
- *Correspondence: Liangbiao Chen,
| |
Collapse
|
28
|
Clay DE, Fox DT. DNA Damage Responses during the Cell Cycle: Insights from Model Organisms and Beyond. Genes (Basel) 2021; 12:1882. [PMID: 34946831 PMCID: PMC8701014 DOI: 10.3390/genes12121882] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
Genome damage is a threat to all organisms. To respond to such damage, DNA damage responses (DDRs) lead to cell cycle arrest, DNA repair, and cell death. Many DDR components are highly conserved, whereas others have adapted to specific organismal needs. Immense progress in this field has been driven by model genetic organism research. This review has two main purposes. First, we provide a survey of model organism-based efforts to study DDRs. Second, we highlight how model organism study has contributed to understanding how specific DDRs are influenced by cell cycle stage. We also look forward, with a discussion of how future study can be expanded beyond typical model genetic organisms to further illuminate how the genome is protected.
Collapse
Affiliation(s)
- Delisa E. Clay
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Donald T. Fox
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA;
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
29
|
Vats A, Trejo-Cerro O, Thomas M, Banks L. Human papillomavirus E6 and E7: What remains? Tumour Virus Res 2021; 11:200213. [PMID: 33716206 PMCID: PMC7972986 DOI: 10.1016/j.tvr.2021.200213] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Decades of research on the human papillomavirus oncogenes, E6 and E7, have given us huge amounts of data on their expression, functions and structures. We know much about the very many cellular proteins and pathways that they influence in one way or another. However, much of this information is quite discrete, referring to one activity examined under one condition. It is now time to join the dots to try to understand a larger picture: how, where and when do all these interactions occur... and why? Examining these questions will also show how many of the yet obscure cellular processes work together for cellular and tissue homeostasis in health and disease.
Collapse
Affiliation(s)
- Arushi Vats
- Tumour Virology Group, ICGEB, AREA Science Park, Trieste, 34149, Italy
| | - Oscar Trejo-Cerro
- Tumour Virology Group, ICGEB, AREA Science Park, Trieste, 34149, Italy
| | - Miranda Thomas
- Tumour Virology Group, ICGEB, AREA Science Park, Trieste, 34149, Italy.
| | - Lawrence Banks
- Tumour Virology Group, ICGEB, AREA Science Park, Trieste, 34149, Italy
| |
Collapse
|
30
|
The influence of hypoxia on the cardiac transcriptomes of two estuarine species - C. variegatus and F. grandis. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100837. [PMID: 33892309 DOI: 10.1016/j.cbd.2021.100837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/21/2021] [Accepted: 04/07/2021] [Indexed: 01/22/2023]
Abstract
Increased nutrient loading has led to eutrophication of coastal shelf waters which has resulted in increased prevalence of persistent hypoxic zones - areas in which the dissolved oxygen content of the water drops below 2 mg/L. The northern Gulf of Mexico, fed primarily by the Mississippi River watershed, undergoes annual establishment of one of the largest hypoxic zones in the world. Exposure to hypoxia can induce physiological impacts in fish cardiac systems that include bradycardia, changes in stroke volume, and altered cardiovascular vessel development. While these impacts have been addressed at the functional level, there is little information regarding the molecular basis for these changes. This study used transcriptomic analysis techniques to interrogate the effects of hypoxia exposure on the developing cardiovascular system in newly hatched larvae of two estuarine species that occupy the same ecological niche - the sheepshead minnow (Cyprinodon variegatus) and the Gulf killifish (Fundulus grandis). Results suggest that while differential gene expression is largely distinct between the two species, downstream impacts on pathways and functional responses such as reduced cardiac hypertrophy, modulation of blood pressure, and increased incidence of apoptosis appear to be conserved. Further, differences in the magnitude of these conserved responses may suggest that the length of embryonic development could impart a level of resiliency to hypoxic perturbation in early life stage fish.
Collapse
|
31
|
Watanabe K, Shibuya S, Ozawa Y, Toda T, Shimizu T. Pathological Relationship between Intracellular Superoxide Metabolism and p53 Signaling in Mice. Int J Mol Sci 2021; 22:3548. [PMID: 33805584 PMCID: PMC8037821 DOI: 10.3390/ijms22073548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/16/2022] Open
Abstract
Intracellular superoxide dismutases (SODs) maintain tissue homeostasis via superoxide metabolism. We previously reported that intracellular reactive oxygen species (ROS), including superoxide accumulation caused by cytoplasmic SOD (SOD1) or mitochondrial SOD (SOD2) insufficiency, induced p53 activation in cells. SOD1 loss also induced several age-related pathological changes associated with increased oxidative molecules in mice. To evaluate the contribution of p53 activation for SOD1 knockout (KO) (Sod1-/-) mice, we generated SOD1 and p53 KO (double-knockout (DKO)) mice. DKO fibroblasts showed increased cell viability with decreased apoptosis compared with Sod1-/- fibroblasts. In vivo experiments revealed that p53 insufficiency was not a great contributor to aging-like tissue changes but accelerated tumorigenesis in Sod1-/- mice. Furthermore, p53 loss failed to improve dilated cardiomyopathy or the survival in heart-specific SOD2 conditional KO mice. These data indicated that p53 regulated ROS-mediated apoptotic cell death and tumorigenesis but not ROS-mediated tissue degeneration in SOD-deficient models.
Collapse
Affiliation(s)
- Kenji Watanabe
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan; (K.W.); (S.S.)
| | - Shuichi Shibuya
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan; (K.W.); (S.S.)
| | - Yusuke Ozawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-8677, Chiba, Japan; (Y.O.); (T.T.)
| | - Toshihiko Toda
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-8677, Chiba, Japan; (Y.O.); (T.T.)
| | - Takahiko Shimizu
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan; (K.W.); (S.S.)
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-8677, Chiba, Japan; (Y.O.); (T.T.)
| |
Collapse
|
32
|
Freewoman JM, Snape R, Cui F. Temporal gene regulation by p53 is associated with the rotational setting of its binding sites in nucleosomes. Cell Cycle 2021; 20:792-807. [PMID: 33764853 PMCID: PMC8098069 DOI: 10.1080/15384101.2021.1904554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The tumor suppressor protein p53 is a DNA-binding transcription factor (TF) that, once activated, coordinates the expression of thousands of target genes. Increased p53 binding to gene promoters occurs shortly after p53 activation. Intriguingly, gene transcription exhibits differential kinetics with some genes being induced early (early genes) and others being induced late (late genes). To understand pre-binding factors contributing to the temporal gene regulation by p53, we performed time-course RNA sequencing experiments in human colon cancer cell line HCT116 treated with fluorouracil to identify early and late genes. Published p53 ChIP fragments co-localized with the early or late genes were used to uncover p53 binding sites (BS). We demonstrate that the BS associated with early genes are clustered around gene starts with decreased nucleosome occupancy. DNA analysis shows that these BS are likely exposed on nucleosomal surface if wrapped into nucleosomes, thereby facilitating stable interactions with and fast induction by p53. By contrast, p53 BS associated with late genes are distributed uniformly across the genes with increased nucleosome occupancy. Predicted rotational settings of these BS show limited accessibility. We therefore propose a hypothetical model in which the BS are fully, partially or not accessible to p53 in the nucleosomal context. The partial accessibility of the BS allows subunits of a p53 tetramer to bind, but the resulting p53-DNA complex may not be stable enough to recruit cofactors, which leads to delayed induction. Our work highlights the importance of DNA conformations of p53 BS in gene expression dynamics.
Collapse
Affiliation(s)
- Julia M Freewoman
- Thomas H. Gosnell School of Life Sciences, College of Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Rajiv Snape
- Thomas H. Gosnell School of Life Sciences, College of Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Feng Cui
- Thomas H. Gosnell School of Life Sciences, College of Science, Rochester Institute of Technology, Rochester, NY, USA
| |
Collapse
|
33
|
Centromeric chromatin integrity is compromised by loss of Cdk5rap2, a transcriptional activator of CENP-A. Biomed Pharmacother 2021; 138:111463. [PMID: 33725591 DOI: 10.1016/j.biopha.2021.111463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 11/21/2022] Open
Abstract
Centromeres are chromosomal loci where kinetochores assemble to ensure faithful chromosome segregation during mitosis. CENP-A defines the loci by serving as an epigenetic marker that recruits other centromere components for a functional structure. However, the mechanism that controls CENP-A regulation of centromeric chromatin integrity remains to be explored. Separate studies have shown that loss of CENP-A or the Cdk5 regulatory subunit associated protein 2 (Cdk5rap2), a key player in mitotic progression, triggers the occurrence of lagging chromosomes. This prompted us to investigate a potential link between CENP-A and Cdk5rap2 in the maintenance of centromeric chromatin integrity. Here, we demonstrate that loss of Cdk5rap2 causes reduced CENP-A expression while exogenous Cdk5rap2 expression in cells depleted of endogenous Cdk5rap2 restores CENP-A expression. Indeed, we show that Cdk5rap2 is a nuclear protein that acts as a positive transcriptional regulator of CENP-A. Cdk5rap2 interacts with the CENP-A promoter and upregulates CENP-A transcription. Accordingly, loss of Cdk5rap2 causes reduced level of centromeric CENP-A. Exogenous CENP-A expression partially inhibits the occurrence of lagging chromosomes in Cdk5rap2 knockdown cells, indicating that lagging chromosomes induced by loss of Cdk5rap2 is due, in part, to loss of CENP-A. Aside from manifesting lagging chromosomes, cells depleted of Cdk5rap2, and thus CENP-A, show increased micronuclei and chromatin bridge formation. Altogether, our findings indicate that Cdk5rap2 serves to maintain centromeric chromatin integrity partly through CENP-A.
Collapse
|
34
|
Abrams SL, Akula SM, Martelli AM, Cocco L, Ratti S, Libra M, Candido S, Montalto G, Cervello M, Gizak A, Rakus D, Steelman LS, McCubrey JA. Sensitivity of pancreatic cancer cells to chemotherapeutic drugs, signal transduction inhibitors and nutraceuticals can be regulated by WT-TP53. Adv Biol Regul 2021; 79:100780. [PMID: 33451973 DOI: 10.1016/j.jbior.2020.100780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic malignancy. Approximately 85% of pancreatic cancers are classified as PDACs. The survival of PDAC patients is very poor and only 5-10% of patients survive 5 years after diagnosis. Mutations at the KRAS and TP53 gene are frequently observed in PDAC patients. The PANC-28 cell line lacks wild-type (WT) TP53. In the following study, we have investigated the effects of restoration of WT TP53 activity on the sensitivity of PANC-28 pancreatic cancer cells to various drugs which are used to treat PDAC patients as well as other cancer patients. In addition, we have examined the effects of signal transduction inhibitors which target critical pathways frequently deregulated in cancer. The effects of the anti-diabetes drug metformin and the anti-malarial drug chloroquine were also examined as these drugs may be repurposed to treat other diseases. Finally, the effects of certain nutraceuticals which are used to treat various ailments were also examined. Introduction of WT-TP53 activity in PANC-28 PDAC cells, can increase their sensitivity to various drugs. Attempts are being made clinically to increase TP53 activity in various cancer types which will often inhibit cell growth by multiple mechanisms.
Collapse
Affiliation(s)
- Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Massimo Libra
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Saverio Candido
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Montalto
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834.
| |
Collapse
|
35
|
Xie H, Ma K, Zhang K, Zhou J, Li L, Yang W, Gong Y, Cai L, Gong K. Cell-cycle arrest and senescence in TP53-wild type renal carcinoma by enhancer RNA-P53-bound enhancer regions 2 (p53BER2) in a p53-dependent pathway. Cell Death Dis 2021; 12:1. [PMID: 33414393 PMCID: PMC7791070 DOI: 10.1038/s41419-020-03229-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/24/2022]
Abstract
TP53 is a classic tumor suppressor, but its role in kidney cancer remains unclear. In our study, we tried to explain the role of p53 in kidney cancer through the p53-related enhancer RNA pathway. Functional experiments were used to explore whether P53-bound enhancer regions 2 (p53BER2) has a role in the cell cycle and senescence response of TP53-wild type (WT) renal cancer cells in vitro or vivo. RNA-sequencing was used to identify the potential target of p53BER2. The results showed that the expression level of P53BER2 was downregulated in renal cancer tissues and cell lines, further dual-luciferase experiments and APR-256-reactivated experiments showed p53BER2 expresses in a p53-dependent way. Moreover, knockdown p53BER2 could reverse nutlin-3-induced cytotoxic effect in TP53-WT cell lines. Further exploration showed the downregulation of p53BER2 could reverse nutlin-3-induced G1-arrest and senescence in TP53-WT cell lines. What is more, the knockdown of p53BER2 showed resistance to nutlin-3 treatment in vivo. Additionally, we found BRCA2 could be regulated by p53BER2 in vitro and vivo; further experiment showed p53BER2 could induce cell-cycle arrest and DNA repair by mediating BRCA2. In summary, the p53-associated enhancer RNA-p53BER2 mediates the cell cycle and senescence of p53 in TP53-WT renal cancer cells.
Collapse
Affiliation(s)
- Haibiao Xie
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Urology, Peking University, Beijing, People's Republic of China.,National Urological Cancer Center, Beijing, People's Republic of China.,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China
| | - Kaifang Ma
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Urology, Peking University, Beijing, People's Republic of China.,National Urological Cancer Center, Beijing, People's Republic of China.,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China
| | - Kenan Zhang
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Urology, Peking University, Beijing, People's Republic of China.,National Urological Cancer Center, Beijing, People's Republic of China.,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China
| | - Jingcheng Zhou
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Urology, Peking University, Beijing, People's Republic of China.,National Urological Cancer Center, Beijing, People's Republic of China.,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China
| | - Lei Li
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Urology, Peking University, Beijing, People's Republic of China.,National Urological Cancer Center, Beijing, People's Republic of China.,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China
| | - Wuping Yang
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Urology, Peking University, Beijing, People's Republic of China.,National Urological Cancer Center, Beijing, People's Republic of China.,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China. .,Institute of Urology, Peking University, Beijing, People's Republic of China. .,National Urological Cancer Center, Beijing, People's Republic of China.
| | - Lin Cai
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China. .,Institute of Urology, Peking University, Beijing, People's Republic of China. .,National Urological Cancer Center, Beijing, People's Republic of China.
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing, People's Republic of China. .,Institute of Urology, Peking University, Beijing, People's Republic of China. .,National Urological Cancer Center, Beijing, People's Republic of China. .,Hereditary Kidney Cancer Research Center, Peking University First Hospital, Beijing, People's Republic of China.
| |
Collapse
|
36
|
Chen X, Peng F, Ji Y, Xiang H, Wang X, Liu T, Wang H, Han Y, Wang C, Zhang Y, Kong X, Lang JY. Brca2 deficiency drives gastrointestinal tumor formation and is selectively inhibited by mitomycin C. Cell Death Dis 2020; 11:812. [PMID: 32980867 PMCID: PMC7519908 DOI: 10.1038/s41419-020-03013-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022]
Abstract
BRCA2 is crucial for repairing DNA double-strand breaks with high fidelity, and loss of BRCA2 increases the risks of developing breast and ovarian cancers. Herein, we show that BRCA2 is inactively mutated in 10% of gastric and 7% of colorectal adenocarcinomas, and that this inactivation is significantly correlated with microsatellite instability. Villin-driven Brca2 depletion promotes mouse gastrointestinal tumor formation when genome instability is increased. Whole-genome screening data showed that these BRCA2 monoallelic and biallelic mutant tumors were selectively inhibited by mitomycin C. Mechanistically, mitomycin C provoked double-strand breaks in cancer cells that often recruit wild-type BRCA2 for repair; the failure to repair double-strand breaks caused cell-cycle arrest at the S phase and p53-mediated cell apoptosis of BRCA2 monoallelic and biallelic mutant tumor cells. Our study unveils the role of BRCA2 loss in the development of gastrointestinal tumors and provides a potential therapeutic strategy to eliminate BRCA2 monoallelic and biallelic mutant tumors through mitomycin C.
Collapse
Affiliation(s)
- Xiaomin Chen
- 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, 200031, Shanghai, China
| | - Fangfei Peng
- 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, 200031, Shanghai, China
| | - Yan Ji
- Bioinformatics Core, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Honggang Xiang
- Department of General Surgery, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Science, 201299, Shanghai, China
| | - Xiang 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, 200031, Shanghai, China
| | - Tingting Liu
- 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, 200031, Shanghai, China
| | - Heng 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, 200031, Shanghai, China
| | - Yumin Han
- 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, 200031, Shanghai, China
| | - Changxu 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, 200031, Shanghai, China
| | - Yongfeng Zhang
- 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, 200031, Shanghai, China
| | - Xiangyin Kong
- 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, 200031, Shanghai, China
| | - Jing-Yu Lang
- 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, 200031, Shanghai, China.
| |
Collapse
|
37
|
Zhao H, Li R, Wang X, Lu X, Hu M, Zhang J, Zhao X, Song X, Liu Y. The role of apatinib combined with paclitaxel (aluminum binding type) in platinum-resistant ovarian cancer. J Ovarian Res 2020; 13:113. [PMID: 32958014 PMCID: PMC7507263 DOI: 10.1186/s13048-020-00719-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To assess the anti-tumor activity and side effects of different dosages of paclitaxel (albumin binding type) (hereinafter referred to as nab-P) combined with Apatinib (hereinafter referred to as AP) in platinum-resistant ovarian cancer cell line and xenograft models. METHODS SKOV-3/DDP cell line was selected as the research object in cytology experiment. Firstly, we divided it into three groups for experiments to explore the individual effects of nab-P and AP. a): Control group, blank control, no drug intervention; b): nab-P group, nab-P 40 μmol/l; c): AP group, AP 50 μmol/l (Drug doses were IC-50 values that detected by MTT assay). Apoptosis related protein (Bax, bcl-2), vascular related protein(p-VEGFR-2), invasion related protein (MMP-2) expression were detected by Western blot and Cellular immunofluorescence, the invasion ability of tumor cells were detected by Transwell and Cell scratch test. Based on these dates, secondly, establishing different doses of nab-P combined with Ap to explore the curative effect of combination therapy. a): Control group, blank control, no drug intervention; b): Group-1, nab-P 5 μmol/l + AP 10 μmol/l, c): Group-2, nab-P 4.5 μmol/l + AP 10 μmol/l, d): Group-3, nab-P 4 μmol/l + AP 10 μmol/l, e): nab-P group, nab-P 5 μmol/l, f): AP group, AP 10 μmol/l (MTT assay). The combination index was analyzed by Compusyn software, Western blot, Immunofluescence, Transwell and Cell scratch test also were also chose to observe of inhibition effect. Thirdly, we used xenograft models to verify the results of cytological experiments. Tumor-forming BALB/c female nude mice were randomly divided into 4 groups, a): Control group, no drug intervention, only saline injection, b): nab-P 20 mg/kg + AP 150 mg/kg, c): nab-P 18 mg/kg + AP 150 mg/kg, d): nab-P 16 mg/kg + AP 150 mg/kg (The doses were guided by the pharmaceutical manufacturers). The tumor growth curve was analyzed during the experiment. And the apoptosis related protein (Bax, bcl-2), angiogenesis related protein (CD31, p-VEGFR-2) and invasion related protein (MMP-2) were observed by Western blot, Immunofluescence and Immunohistochemistry to analysis the ant-tumor effects. The quality of life in nude mice were observed to analysed the drug-induced side effects. RESULT In the separate medication section, (1) The IC-50 value of nab-P was 45.53 ± 4.06 μmol/l, while the AP was 50.66 ± 4.96 umol/L (48 h). (2) The expressions of bcl-2 (nab-P group, AP group), p-VEGFR-2 (AP group), MMP-2(nab-P group, AP group) were higher than Control group, while Bax (nab-P group, AP group) lower (P < 0.01). (3) The cell invasive ability was decreased after the nab-P and AP intervation (P < 0.01). In the combination medication section, (1) Compusyn showed the Combination index (Cl) were all below 1 (Cl < 1), that means nab-P and AP are synergism. (2) The combination IC-50 value was nab-P 5.28 μmol/l + AP 10.56 μmol/l (48 h). (3) In the detection of related protein expression, the combination of drugs can improve the anti-tumor effect, otherwise, after combined with AP, when nab-P were reduced dose in proper quantity, there were no obvious different in drug effect. (4) After reducing the doses of nab-P, the average food intake of nude mice increased from 4.50 g ± 0.17 to 5.55 g ± 0.13, and the one-hour activity increased from 6.11 min ±0.16 to 6.34 min ±0.13. CONCLUSION nab-P, a chemotherapeutic agent, can play an anti-tumor role in platinum-resistant ovarian cancer, but it can cause adverse effects that increase with dose. When combined with AP, the two drugs have synergistic effect, which can improve the anti-tumor effects of single drug. In addition, when combined with AP, the doses of nab-P can be appropriately reduced under the standard of recommended to reduce the toxicity of chemotherapy drugs, without affecting the anti-tumor effect.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, No. 56 Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, China.
| | - Rong Li
- Department of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, No. 56 Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, China
| | - Xiaoyan Wang
- Department of Gynecology, Shanxi Cancer Hospital, Taiyuan, China
| | - Xin Lu
- Department of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, No. 56 Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, China
| | - Min Hu
- Department of Biochemistry and Molecular Biology, Basic Medical College, Shanxi Medical University, No. 56 Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, China
| | - Jinbin Zhang
- Department of Gynecology, Shanxi Cancer Hospital, Taiyuan, China
| | - Xia Zhao
- Shanxi province center for disease control and prevention, Taiyuan, China
| | - Xiaoqin Song
- The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Yangyang Liu
- The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
38
|
Suarez OJ, Vega CJ, Sanchez EN, González-Santiago AE, Rodríguez-Jorge O, Alanis AY, Chen G, Hernandez-Vargas EA. Pinning Control for the p53-Mdm2 Network Dynamics Regulated by p14ARF. Front Physiol 2020; 11:976. [PMID: 32982771 PMCID: PMC7485292 DOI: 10.3389/fphys.2020.00976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 07/17/2020] [Indexed: 01/26/2023] Open
Abstract
p53 regulates the cellular response to genotoxic damage and prevents carcinogenic events. Theoretical and experimental studies state that the p53-Mdm2 network constitutes the core module of regulatory interactions activated by cellular stress induced by a variety of signaling pathways. In this paper, a strategy to control the p53-Mdm2 network regulated by p14ARF is developed, based on the pinning control technique, which consists into applying local feedback controllers to a small number of nodes (pinned ones) in the network. Pinned nodes are selected on the basis of their importance level in a topological hierarchy, their degree of connectivity within the network, and the biological role they perform. In this paper, two cases are considered. For the first case, the oscillatory pattern under gamma-radiation is recovered; afterward, as the second case, increased expression of p53 level is taken into account. For both cases, the control law is applied to p14ARF (pinned node based on a virtual leader methodology), and overexpressed Mdm2-mediated p53 degradation condition is considered as carcinogenic initial behavior. The approach in this paper uses a computational algorithm, which opens an alternative path to understand the cellular responses to stress, doing it possible to model and control the gene regulatory network dynamics in two different biological contexts. As the main result of the proposed control technique, the two mentioned desired behaviors are obtained.
Collapse
Affiliation(s)
- Oscar J. Suarez
- Electrical Engineering Department, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Guadalajara, Mexico
| | - Carlos J. Vega
- Electrical Engineering Department, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Guadalajara, Mexico
| | - Edgar N. Sanchez
- Electrical Engineering Department, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Guadalajara, Mexico
| | - Ana E. González-Santiago
- Biomedical Sciences Department, Centro de Investigación Multidisciplinario en Salud, Universidad de Guadalajara, Tonalá, Mexico
| | - Otoniel Rodríguez-Jorge
- Biochemistry and Molecular Biology Department, Instituto de Investigaciones Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Alma Y. Alanis
- Computer Sciences Department, Universidad de Guadalajara, Guadalajara, Mexico
| | - Guanrong Chen
- Electrical Engineering Department, City University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
39
|
Humpton T, Vousden KH. Taking up the reins of power: metabolic functions of p53. J Mol Cell Biol 2020; 11:610-614. [PMID: 31282931 PMCID: PMC6736434 DOI: 10.1093/jmcb/mjz065] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/23/2022] Open
|
40
|
SOX2 and p53 Expression Control Converges in PI3K/AKT Signaling with Versatile Implications for Stemness and Cancer. Int J Mol Sci 2020; 21:ijms21144902. [PMID: 32664542 PMCID: PMC7402325 DOI: 10.3390/ijms21144902] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Stemness and reprogramming involve transcriptional master regulators that suppress cell differentiation while promoting self-renewal. A distinguished example thereof is SOX2, a high mobility group (HMG)-box transcription factor (TF), whose subcellular localization and turnover regulation in embryonic, induced-pluripotent, and cancer stem cells (ESCs, iPSCs, and CSCs, respectively) is mediated by the PI3K/AKT/SOX2 axis, a stem cell-specific branch of the PI3K/AKT signaling pathway. Further effector functions associated with PI3K/AKT induction include cell cycle progression, cellular (mass) growth, and the suppression of apoptosis. Apoptosis, however, is a central element of DNA damage response (DDR), where it provides a default mechanism for cell clearance when DNA integrity cannot be maintained. A key player in DDR is tumor suppressor p53, which accumulates upon DNA-damage and is counter-balanced by PI3K/AKT enforced turnover. Accordingly, stemness sustaining SOX2 expression and p53-dependent DDR mechanisms show molecular–functional overlap in PI3K/AKT signaling. This constellation proves challenging for stem cells whose genomic integrity is a functional imperative for normative ontogenesis. Unresolved mutations in stem and early progenitor cells may in fact provoke transformation and cancer development. Such mechanisms are also particularly relevant for iPSCs, where genetic changes imposed through somatic cell reprogramming may promote DNA damage. The current review aims to summarize the latest advances in the understanding of PI3K/AKT/SOX2-driven stemness and its intertwined relations to p53-signaling in DDR under conditions of pluripotency, reprogramming, and transformation.
Collapse
|
41
|
Wilson Y, Morris ID, Kimber SJ, Brison DR. The role of Trp53 in the mouse embryonic response to DNA damage. Mol Hum Reprod 2020; 25:397-407. [PMID: 31227838 DOI: 10.1093/molehr/gaz029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/23/2019] [Accepted: 05/31/2019] [Indexed: 12/23/2022] Open
Abstract
Apoptosis occurs primarily in the blastocyst inner cell mass, cells of which go on to form the foetus. Apoptosis is likely to play a role in ensuring the genetic integrity of the foetus, yet little is known about its regulation. In this study, the role of the mouse gene, transformation-related protein 53 (Trp53) in the response of embryos to in vitro culture and environmentally induced DNA damage was investigated using embryos from a Trp53 knockout mouse model. In vivo-derived blastocysts were compared to control embryos X-irradiated at the two-cell stage and cultured to Day 5. An analysis of DNA by comet assay demonstrated that 1.5 Gy X-irradiation directly induced damage in cultured two-cell mouse embryos; this was correlated with retarded development to blastocyst stage and increased apoptosis at the blastocyst stage but not prior to this. Trp53 null embryos developed to blastocysts at a higher frequency and with higher cell numbers than wild-type embryos. Trp53 also mediates apoptosis in conditions of low levels of DNA damage, in vivo or in vitro in the absence of irradiation. However, following DNA damage induced by X-irradiation, apoptosis is induced by Trp53 independent as well as dependent mechanisms. These data suggest that Trp53 and apoptosis play important roles in normal mouse embryonic development both in vitro and in vivo and in response to DNA damage. Therefore, clinical ART practices that alter apoptosis in human embryos and/or select embryos for transfer, which potentially lack a functional Trp53 gene, need to be carefully considered.
Collapse
Affiliation(s)
- Yvonne Wilson
- Department of Reproductive Medicine, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9WL, UK
| | - Ian D Morris
- Hull York Medical School, University of York, Heslington, York YO10 5DD, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Daniel R Brison
- Department of Reproductive Medicine, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9WL, UK.,Maternal and Fetal Health Research, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
42
|
PABPN1, a Target of p63, Modulates Keratinocyte Differentiation through Regulation of p63α mRNA Translation. J Invest Dermatol 2020; 140:2166-2177.e6. [PMID: 32243883 DOI: 10.1016/j.jid.2020.03.942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 01/25/2023]
Abstract
p63 is expressed from two promoters and produces two N-terminal isoforms, TAp63 and ΔNp63. Alternative splicing creates three C-terminal isoforms p63α, p63β, and p63δ, whereas alternative polyadenylation (APA) in coding sequence creates two more C-terminal isoforms p63γ and p63ε. Although several transcription factors have been identified to differentially regulate the N-terminal p63 isoforms, it is unclear how the C-terminal p63 isoforms are regulated. Thus, we determined whether PABPN1, a key regulator of APA, may differentially regulate the C-terminal p63 isoforms. We found that PABPN1 deficiency increases p63γ mRNA through APA in coding sequence. We also found that PABPN1 is necessary for p63α translation by modulating the binding of translation initiation factors eIF4E and eIF4G to p63α mRNA. Moreover, we found that the p53 family, especially p63α, regulates PABPN1 transcription, suggesting that the mutual regulation between p63 and PABPN1 forms a feedback loop. Furthermore, we found that PABPN1 deficiency inhibits keratinocyte cell growth, which can be rescued by ectopic ΔNp63α. Finally, we found that PABPN1 controls the terminal differentiation of HaCaT keratinocytes by modulating ΔNp63α expression. Taken together, our findings suggest that PABPN1 is a key regulator of the C-terminal p63 isoforms through APA in coding sequence and mRNA translation and that the p63-PABPN1 loop modulates p63 activity and the APA landscape.
Collapse
|
43
|
Goligorsky MS. Chronic Kidney Disease: A Vicarious Relation to Premature Cell Senescence. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1164-1171. [PMID: 32194054 DOI: 10.1016/j.ajpath.2020.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/25/2020] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease (CKD), commonly fostering nonrenal complications, themselves more life threatening than renal pathology, remains enigmatic. Despite more than a century of intense research, therapeutic options to halt or reverse renal disease are rather limited. Recently, similarity between manifestations of progressive CKD and aging kidney has attracted investigative attention that revealed senescent cells and secreting proinflammatory and profibrotic mediators in all renal compartments, even at young age, in patients with kidney maladies. The overlapping features of these categories have been noticed previously and are briefly summarized herein. I propose two hypothetical scenarios for interactive association of kidney diseases and cell senescence, both culminating in progressive deterioration of renal function. Persistence of senescent cells is considered as a critical contributor to this association; and the mechanisms explaining persistence, such as activation of cell cycle regulators, anti-apoptotic stimuli, metabolic aberrations, and their interactions, are discussed. The mutual encroachment of underlying kidney disease and cell senescence bring about the conclusion that both entities merge along the natural history of the disease. This putative interpretation of vicarious relation between cell senescence and CKD may expand the arsenal of pharmacotherapy to include the judicious use of senotherapeutics in the management of renal disease.
Collapse
Affiliation(s)
- Michael S Goligorsky
- Renal Research Institute, and the Departments of Medicine, Pharmacology, and Physiology, New York Medical College at Touro University, Valhalla, New York.
| |
Collapse
|
44
|
Yue Z, Lam HC, Chen K, Siridechakorn I, Liu Y, Pudhom K, Lei X. Biomimetic Synthesis of Rhytidenone A and Mode of Action of Cytotoxic Rhytidenone F. Angew Chem Int Ed Engl 2020; 59:4115-4120. [PMID: 31868281 DOI: 10.1002/anie.201914257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/07/2019] [Indexed: 12/20/2022]
Abstract
The rhytidenone family comprises spirobisnaphthalene natural products isolated from the mangrove endophytic fungus Rhytidhysteron rufulum AS21B. The biomimetic synthesis of rhytidenone A was achieved by a Michael reaction/aldol/lactonization cascade in a single step from the proposed biosynthetic precursor rhytidenone F. Moreover, the mode of action of the highly cytotoxic rhytidenone F was investigated. The pulldown assay coupled with mass spectrometry analysis revealed the target protein PA28γ is covalently attached to rhytidenone F at the Cys92 residue. The interactions of rhytidenone F with PA28γ lead to the accumulation of p53, which is an essential tumor suppressor in humans. Consequently, the Fas-dependent signaling pathway is activated to initiate cellular apoptosis. These studies have identified the first small-molecule inhibitor targeting PA28γ, suggesting rhytidenone F may serve as a promising natural product lead for future anticancer drug development.
Collapse
Affiliation(s)
- Zongwei Yue
- School of Life Sciences, Peking University, Beijing, 100871, P. R. China.,Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Hiu C Lam
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Kaiqi Chen
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Ittipon Siridechakorn
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Yaxi Liu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Khanitha Pudhom
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| |
Collapse
|
45
|
Qiu B, Zhou T, Zhang J. Stochastic fluctuations in apoptotic threshold of tumour cells can enhance apoptosis and combat fractional killing. ROYAL SOCIETY OPEN SCIENCE 2020; 7:190462. [PMID: 32257298 PMCID: PMC7062090 DOI: 10.1098/rsos.190462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 01/20/2020] [Indexed: 06/11/2023]
Abstract
Fractional killing, which is a significant impediment to successful chemotherapy, is observed even in a population of genetically identical cancer cells exposed to apoptosis-inducing agents. This phenomenon arises not from genetic mutation but from cell-to-cell variation in the activation timing and level of the proteins that regulates apoptosis. To understand the mechanism behind the phenomenon, we formulate complex fractional killing processes as a first-passage time (FPT) problem with a stochastically fluctuating boundary. Analytical calculations are performed for the FPT distribution in a toy model of stochastic p53 gene expression, where the cancer cell is killed only when the p53 expression level crosses an active apoptotic threshold. Counterintuitively, we find that threshold fluctuations can effectively enhance cellular killing by significantly decreasing the mean time that the p53 protein reaches the threshold level for the first time. Moreover, faster fluctuations lead to the killing of more cells. These qualitative results imply that fluctuations in threshold are a non-negligible stochastic source, and can be taken as a strategy for combating fractional killing of cancer cells.
Collapse
Affiliation(s)
- Baohua Qiu
- School of Mathematics, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
- Key Laboratory of Computational Mathematics, Guangzhou, Guangdong Province, People's Republic of China
| | - Tianshou Zhou
- School of Mathematics, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
- Key Laboratory of Computational Mathematics, Guangzhou, Guangdong Province, People's Republic of China
| | - Jiajun Zhang
- School of Mathematics, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
- Key Laboratory of Computational Mathematics, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
46
|
Yue Z, Lam HC, Chen K, Siridechakorn I, Liu Y, Pudhom K, Lei X. Biomimetic Synthesis of Rhytidenone A and Mode of Action of Cytotoxic Rhytidenone F. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Zongwei Yue
- School of Life Sciences Peking University Beijing 100871 P. R. China
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Natural and Biomimetic Drugs Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Department of Chemical Biology College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences Peking University Beijing 100871 P. R. China
| | - Hiu C. Lam
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Natural and Biomimetic Drugs Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Department of Chemical Biology College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences Peking University Beijing 100871 P. R. China
| | - Kaiqi Chen
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Natural and Biomimetic Drugs Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Department of Chemical Biology College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences Peking University Beijing 100871 P. R. China
| | - Ittipon Siridechakorn
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Natural and Biomimetic Drugs Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Department of Chemical Biology College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences Peking University Beijing 100871 P. R. China
| | - Yaxi Liu
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Natural and Biomimetic Drugs Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Department of Chemical Biology College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences Peking University Beijing 100871 P. R. China
| | - Khanitha Pudhom
- Department of Chemistry Faculty of Science Chulalongkorn University Bangkok 10330 Thailand
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences State Key Laboratory of Natural and Biomimetic Drugs Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education Department of Chemical Biology College of Chemistry and Molecular Engineering Synthetic and Functional Biomolecules Center, and Peking-Tsinghua Center for Life Sciences Peking University Beijing 100871 P. R. China
| |
Collapse
|
47
|
Kiang KMY, Zhang P, Li N, Zhu Z, Jin L, Leung GKK. Loss of cytoskeleton protein ADD3 promotes tumor growth and angiogenesis in glioblastoma multiforme. Cancer Lett 2020; 474:118-126. [PMID: 31958485 DOI: 10.1016/j.canlet.2020.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
Adducin 3 (ADD3) is a crucial assembly factor in the actin cytoskeleton and has been found to be aberrantly expressed in various cancers, including glioblastoma multiforme (GBM). It has previously been studied in array-based studies with controversial findings as to its functional role in glioma. In microarray analyses of 452 glioma specimens, we found significant downregulation of ADD3 in GBM, but not in less malignant gliomas, compared to normal brain tissue, which suggests that its downregulation might underlie critical events during malignant progression. We also found that ADD3 was functionally dependent on cell-matrix interaction. In our in vivo study, the proliferative and angiogenic capacity of ADD3-depleted GBM cells was promoted, possibly through PCNA, while p53 and p21 expression was suppressed, and pro-angiogenic signals were induced through VEGF-VEGFR-2-mediated activation in endothelial cells. With correlative in vitro, in vivo, and clinical data, we provide compelling evidence on the putative tumor-suppressive role of ADD3 in modulating GBM growth and angiogenesis. As a preclinical study, our research offers a better understanding of the pathogenesis of glioma malignant progression for the benefit of future investigations.
Collapse
Affiliation(s)
- Karrie Mei-Yee Kiang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Pingde Zhang
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ning Li
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Zhiyuan Zhu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Lei Jin
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Gilberto Ka-Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong.
| |
Collapse
|
48
|
Miura H, Kondo Y, Matsuda M, Aoki K. Cell-to-Cell Heterogeneity in p38-Mediated Cross-Inhibition of JNK Causes Stochastic Cell Death. Cell Rep 2019; 24:2658-2668. [PMID: 30184500 DOI: 10.1016/j.celrep.2018.08.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/17/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
Abstract
The stress-activated protein kinases c-Jun N-terminal kinase (JNK) and p38 are important players in cell-fate decisions in response to environmental stress signals. Crosstalk signaling between JNK and p38 is emerging as an important regulatory mechanism in inflammatory and stress responses. However, it is unknown how this crosstalk affects signaling dynamics, cell-to-cell variation, and cellular responses at the single-cell level. We established a multiplexed live-cell imaging system based on kinase translocation reporters to simultaneously monitor JNK and p38 activities with high specificity and sensitivity at single-cell resolution. Various stresses activated JNK and p38 with various dynamics. In all cases, p38 suppressed JNK activity in a cross-inhibitory manner. We demonstrate that p38 antagonizes JNK through both transcriptional and post-translational mechanisms. This cross-inhibition generates cellular heterogeneity in JNK activity after stress exposure. Our data indicate that this heterogeneity in JNK activity plays a role in fractional killing in response to UV stress.
Collapse
Affiliation(s)
- Haruko Miura
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Yohei Kondo
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
49
|
Klimovich B, Stiewe T, Timofeev O. Inactivation of Mdm2 restores apoptosis proficiency of cooperativity mutant p53 in vivo. Cell Cycle 2019; 19:109-123. [PMID: 31749402 DOI: 10.1080/15384101.2019.1693748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
TP53 mutations are found in 50% of all cancers and mutated TP53 status is considered poor for treatment. However, some TP53 mutations exhibit only partial loss-of-function (LOF), meaning they retain residual transcriptional and non-transcriptional activities that are potentially beneficial for therapy. Earlier we have characterized a knock-in mouse model for the partial LOF mutant Trp53E177R (p53RR). Reduced DNA binding cooperativity of this mutant led to the loss of p53-dependent apoptosis, while p53 functions in cell cycle control, senescence, metabolism, and antioxidant defense remained intact. Concomitantly, tumor suppression was evident but strongly compromised compared to wild-type mice. Here we used the Trp53E177R mouse as a model to investigate whether residual functions of mutant p53 can be engaged to induce cell death, which is considered the most desirable outcome of tumor therapy. We made use of Mdm2 knock-out in developing embryos as a sensitive tool for detecting remaining p53 activities. Genetic ablation of Mdm2 led to embryonic lethality in Trp53E177R/E177R homozygotes at days 9.5-11.5. This effect was not rescued by concomitant p21-knockout, indicating its independence of p21-mediated cell cycle arrest. Instead, immunohistochemical analysis showed widespread apoptosis in tissues of defective embryos accompanied by persistent accumulation of p53RR protein. This led to partial restoration of the mutant's proficiency in transcriptional induction of the pro-apoptotic genes Bbc3 (Puma) and Bax. These data indicate that increased quantity can compensate for qualitative defects of p53 mutants and suggest that Mdm2-targeting (potentially in combination with other drugs) might be effective against cells bearing p53 partial LOF mutants.
Collapse
Affiliation(s)
- Boris Klimovich
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| | - Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps-University, Marburg, Germany
| |
Collapse
|
50
|
Timofeev O, Klimovich B, Schneikert J, Wanzel M, Pavlakis E, Noll J, Mutlu S, Elmshäuser S, Nist A, Mernberger M, Lamp B, Wenig U, Brobeil A, Gattenlöhner S, Köhler K, Stiewe T. Residual apoptotic activity of a tumorigenic p53 mutant improves cancer therapy responses. EMBO J 2019; 38:e102096. [PMID: 31483066 PMCID: PMC6792016 DOI: 10.15252/embj.2019102096] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53−/− mice. Surprisingly, stabilization of p53R178E in Mdm2−/− mice nevertheless triggers extensive apoptosis, indicative of residual wild‐type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E;Mdm2−/− embryos, it proves insufficient for suppression of spontaneous and oncogene‐driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53−/− mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53−/− ones, resulting in enhanced eradication of p53‐mutated tumor cells. Together, this provides genetic proof‐of‐principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Boris Klimovich
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Jean Schneikert
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany
| | | | - Julia Noll
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Samet Mutlu
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Boris Lamp
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Ulrich Wenig
- Institute of Pathology, Justus Liebig University, Giessen, Germany
| | | | | | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University, Giessen, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,Genomics Core Facility, Philipps University, Marburg, Germany
| |
Collapse
|