1
|
Butera A, Amelio I. Deciphering the significance of p53 mutant proteins. Trends Cell Biol 2025; 35:258-268. [PMID: 38960851 DOI: 10.1016/j.tcb.2024.06.003] [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: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
Mutations in the p53 gene compromise its role as guardian of genomic integrity, yielding predominantly missense p53 mutant proteins. The gain-of-function hypothesis has long suggested that these mutant proteins acquire new oncogenic properties; however, recent studies challenge this notion, indicating that targeting these mutants may not impact the fitness of cancer cells. Mounting evidence indicates that tumorigenesis involves a cooperative interplay between driver mutations and cellular state, influenced by developmental stage, external insults, and tissue damage. Consistently, the behavior and properties of p53 mutants are altered by the context. This article aims to provide a balanced summary of the evolving evidence regarding the contribution of p53 mutants in the biology of cancer while contemplating alternative frameworks to decipher the complexity of p53 mutants within their physiological contexts.
Collapse
Affiliation(s)
- Alessio Butera
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
2
|
Andrysik Z, Espinosa JM. Harnessing p53 for targeted cancer therapy: new advances and future directions. Transcription 2025; 16:3-46. [PMID: 40031988 DOI: 10.1080/21541264.2025.2452711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 03/05/2025] Open
Abstract
The transcription factor p53 is the most frequently impaired tumor suppressor in human cancers. In response to various stress stimuli, p53 activates transcription of genes that mediate its tumor-suppressive functions. Distinctive characteristics of p53 outlined here enable a well-defined program of genes involved in cell cycle arrest, apoptosis, senescence, differentiation, metabolism, autophagy, DNA repair, anti-viral response, and anti-metastatic functions, as well as facilitating autoregulation within the p53 network. This versatile, anti-cancer network governed chiefly by a single protein represents an immense opportunity for targeted cancer treatment, since about half of human tumors retain unmutated p53. During the last two decades, numerous compounds have been developed to block the interaction of p53 with the main negative regulator MDM2. However, small molecule inhibitors of MDM2 only induce a therapeutically desirable apoptotic response in a limited number of cancer types. Moreover, clinical trials of the MDM2 inhibitors as monotherapies have not met expectations and have revealed hematological toxicity as a characteristic adverse effect across this drug class. Currently, combination treatments are the leading strategy for enhancing efficacy and reducing adverse effects of MDM2 inhibitors. This review summarizes efforts to identify and test therapeutics that work synergistically with MDM2 inhibitors. Two main types of drugs have emerged among compounds used in the following combination treatments: first, modulators of the p53-regulated transcriptome (including chromatin modifiers), translatome, and proteome, and second, drugs targeting the downstream pathways such as apoptosis, cell cycle arrest, DNA repair, metabolic stress response, immune response, ferroptosis, and growth factor signaling. Here, we review the current literature in this field, while also highlighting overarching principles that could guide target selection in future combination treatments.
Collapse
Affiliation(s)
- Zdenek Andrysik
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M Espinosa
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
3
|
Kim M, Yoon J, Choi JY, Park G, Lee JE, Lee GB, Choi BW, Kim P, Kim H, Oh CM, Bae MA, Kim SS, Lee EY, Lee HJ, Kim Y, Kim HW, Lee H, Jeon YH, Ahn JH. Synthesis and Biological Evaluation of Peripheral HTR2A Antagonists for Colorectal Cancer. J Med Chem 2025; 68:1716-1730. [PMID: 39760275 DOI: 10.1021/acs.jmedchem.4c02458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Colorectal cancer is a prevalent and prominent contributor to global cancer-related fatalities with challenges in drug resistance and metastasis. Recent research highlights the potential relationship between serotonin and cancer. 5-Hydroxytryptamine receptor 2A (HTR2A) mRNA expression in colorectal cancer cells was found to be notably elevated compared to that in normal colon cells. We therefore attempted to synthesize and evaluate HTR2A antagonists to find peripherally acting anticancer agents. Among them, 15f showed good in vitro activity (IC50: 42.79 nM). 15f revealed good liver microsomal stability, without significant CYP inhibition and limited blood-brain barrier penetration. 15f also exerted selective cytotoxic effects against various colorectal cancer cells but not normal cells. 15f induced sub-G1 cell cycle arrest and apoptosis in colorectal cancer cells via the activation of p53/p21/caspase 3 signaling. In vivo treatment with 15f led to a marked delay in tumor growth in a colorectal cancer model in a dose-dependent manner.
Collapse
Affiliation(s)
- Minhee Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Jihyeon Yoon
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jun Young Choi
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), 80 Cheombok-ro Dong-gu, Daegu 41061, Republic of Korea
| | - Geumi Park
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), 80 Cheombok-ro Dong-gu, Daegu 41061, Republic of Korea
| | - Jae-Eon Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), 80 Cheombok-ro Dong-gu, Daegu 41061, Republic of Korea
| | - Gwi Bin Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Byeong Wook Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Pyeongkeun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Seong Soon Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Eun Young Lee
- JD Bioscience Inc., TJS Knowledge Industrial Center Suite 801, 208 Beon-gil Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Hyeok Jae Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yunmin Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyun Woo Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Center for Quantum Technology, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hohjai Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yong Hyun Jeon
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), 80 Cheombok-ro Dong-gu, Daegu 41061, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- JD Bioscience Inc., TJS Knowledge Industrial Center Suite 801, 208 Beon-gil Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| |
Collapse
|
4
|
Bruno PS, Arshad A, Gogu MR, Waterman N, Flack R, Dunn K, Darie CC, Neagu AN. Post-Translational Modifications of Proteins Orchestrate All Hallmarks of Cancer. Life (Basel) 2025; 15:126. [PMID: 39860065 PMCID: PMC11766951 DOI: 10.3390/life15010126] [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: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Post-translational modifications (PTMs) of proteins dynamically build the buffering and adapting interface between oncogenic mutations and environmental stressors, on the one hand, and cancer cell structure, functioning, and behavior. Aberrant PTMs can be considered as enabling characteristics of cancer as long as they orchestrate all malignant modifications and variability in the proteome of cancer cells, cancer-associated cells, and tumor microenvironment (TME). On the other hand, PTMs of proteins can enhance anticancer mechanisms in the tumoral ecosystem or sustain the beneficial effects of oncologic therapies through degradation or inactivation of carcinogenic proteins or/and activation of tumor-suppressor proteins. In this review, we summarized and analyzed a wide spectrum of PTMs of proteins involved in all regulatory mechanisms that drive tumorigenesis, genetic instability, epigenetic reprogramming, all events of the metastatic cascade, cytoskeleton and extracellular matrix (ECM) remodeling, angiogenesis, immune response, tumor-associated microbiome, and metabolism rewiring as the most important hallmarks of cancer. All cancer hallmarks develop due to PTMs of proteins, which modulate gene transcription, intracellular and extracellular signaling, protein size, activity, stability and localization, trafficking, secretion, intracellular protein degradation or half-life, and protein-protein interactions (PPIs). PTMs associated with cancer can be exploited to better understand the underlying molecular mechanisms of this heterogeneous and chameleonic disease, find new biomarkers of cancer progression and prognosis, personalize oncotherapies, and discover new targets for drug development.
Collapse
Affiliation(s)
- Pathea Shawnae Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Aneeta Arshad
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Maria-Raluca Gogu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania;
| | - Natalie Waterman
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Rylie Flack
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Kimberly Dunn
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (P.S.B.); (A.A.); (N.W.); (R.F.); (K.D.)
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
| |
Collapse
|
5
|
Caporali S, Butera A, Ruzza A, Zampieri C, Bantula' M, Scharsich S, Ückert AK, Celardo I, Kouzel IU, Leanza L, Gruber A, Montero J, D'Alessandro A, Brunner T, Leist M, Amelio I. Selective metabolic regulations by p53 mutant variants in pancreatic cancer. J Exp Clin Cancer Res 2024; 43:310. [PMID: 39587609 PMCID: PMC11590503 DOI: 10.1186/s13046-024-03232-3] [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: 09/19/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Approximately half of all human cancers harbour mutations in the p53 gene, leading to the generation of neomorphic p53 mutant proteins. These mutants can exert gain-of-function (GOF) effects, potentially promoting tumour progression. However, the clinical significance of p53 GOF mutations, as well as the selectivity of individual variants, remains controversial and unclear. METHODS To elucidate the metabolic regulations and molecular underpinnings associated with the specific p53R270H and p53R172H mutant variants (the mouse equivalents of human p53R273H and p53R175H, respectively), we employed a comprehensive approach. This included integrating global metabolomic analysis with epigenomic and transcriptomic profiling in mouse pancreatic cancer cells. Additionally, we assessed metabolic parameters such as oxygen consumption rate and conducted analyses of proliferation and cell-cell competition to validate the biological impact of metabolic changes on pancreatic ductal adenocarcinoma (PDAC) phenotype. Our findings were further corroborated through analysis of clinical datasets from human cancer cohorts. RESULTS Our investigation revealed that the p53R270H variant, but not p53R172H, sustains mitochondrial function and energy production while also influencing cellular antioxidant capacity. Conversely, p53R172H, while not affecting mitochondrial metabolism, attenuates the activation of pro-tumorigenic metabolic pathways such as the urea cycle. Thus, the two variants selectively control different metabolic pathways in pancreatic cancer cells. Mechanistically, p53R270H induces alterations in the expression of genes associated with oxidative stress and reduction in mitochondrial respiration. In contrast, p53R172H specifically impacts the expression levels of enzymes involved in the urea metabolism. However, our analysis of cell proliferation and cell competition suggested that the expression of either p53R270H or p53R172H does not influence confer any selective advantage to this cellular model in vitro. Furthermore, assessment of mitochondrial priming indicated that the p53R270H-driven mitochondrial effect does not alter cytochrome c release or the apoptotic propensity of pancreatic cancer cells. CONCLUSIONS Our study elucidates the mutant-specific impact of p53R270H and p53R172H on metabolism of PDAC cancer cells, highlighting the need to shift from viewing p53 mutant variants as a homogeneous group of entities to a systematic assessment of each specific p53 mutant protein. Moreover, our finding underscores the importance of further exploring the significance of p53 mutant proteins using models that more accurately reflect tumor ecology.
Collapse
Affiliation(s)
- Sabrina Caporali
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alessio Butera
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alessia Ruzza
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Carlotta Zampieri
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, San Diego, CA, USA
| | - Marina Bantula'
- Department of Biomedical Sciences, Universitat de Barcelona, Casanova 143, Barcelona, 08036, Spain
| | - Sandra Scharsich
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Anna-Katerina Ückert
- Chair for in Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Ivana Celardo
- Chair for in Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Ian U Kouzel
- Applied Bioinformatic Group, University of Konstanz, Constance, Germany
| | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy
| | - Andreas Gruber
- Applied Bioinformatic Group, University of Konstanz, Constance, Germany
| | - Joan Montero
- Department of Biomedical Sciences, Universitat de Barcelona, Casanova 143, Barcelona, 08036, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain
| | | | - Thomas Brunner
- Chair for in Biochemical Pharmacology, University of Konstanz, Constance, Germany
| | - Marcel Leist
- Chair for in Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Ivano Amelio
- Chair for Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
6
|
Abad E, Sandoz J, Romero G, Zadra I, Urgel-Solas J, Borredat P, Kourtis S, Ortet L, Martínez CM, Weghorn D, Sdelci S, Janic A. The TP53-activated E3 ligase RNF144B is a tumour suppressor that prevents genomic instability. J Exp Clin Cancer Res 2024; 43:127. [PMID: 38685100 PMCID: PMC11057071 DOI: 10.1186/s13046-024-03045-4] [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: 12/04/2023] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND TP53, the most frequently mutated gene in human cancers, orchestrates a complex transcriptional program crucial for cancer prevention. While certain TP53-dependent genes have been extensively studied, others, like the recently identified RNF144B, remained poorly understood. This E3 ubiquitin ligase has shown potent tumor suppressor activity in murine Eμ Myc-driven lymphoma, emphasizing its significance in the TP53 network. However, little is known about its targets and its role in cancer development, requiring further exploration. In this work, we investigate RNF144B's impact on tumor suppression beyond the hematopoietic compartment in human cancers. METHODS Employing TP53 wild-type cells, we generated models lacking RNF144B in both non-transformed and cancerous cells of human and mouse origin. By using proteomics, transcriptomics, and functional analysis, we assessed RNF144B's impact in cellular proliferation and transformation. Through in vitro and in vivo experiments, we explored proliferation, DNA repair, cell cycle control, mitotic progression, and treatment resistance. Findings were contrasted with clinical datasets and bioinformatics analysis. RESULTS Our research underscores RNF144B's pivotal role as a tumor suppressor, particularly in lung adenocarcinoma. In both human and mouse oncogene-expressing cells, RNF144B deficiency heightened cellular proliferation and transformation. Proteomic and transcriptomic analysis revealed RNF144B's novel function in mediating protein degradation associated with cell cycle progression, DNA damage response and genomic stability. RNF144B deficiency induced chromosomal instability, mitotic defects, and correlated with elevated aneuploidy and worse prognosis in human tumors. Furthermore, RNF144B-deficient lung adenocarcinoma cells exhibited resistance to cell cycle inhibitors that induce chromosomal instability. CONCLUSIONS Supported by clinical data, our study suggests that RNF144B plays a pivotal role in maintaining genomic stability during tumor suppression.
Collapse
Affiliation(s)
- Etna Abad
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
| | - Jérémy Sandoz
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
| | - Gerard Romero
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Ivan Zadra
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
| | - Julia Urgel-Solas
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
| | - Pablo Borredat
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
| | - Savvas Kourtis
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
| | - Laura Ortet
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain
| | - Carlos M Martínez
- Pathology Platform, Instituto Murciano de Investigación Biosanitaria (IMIB-Pascual Parrilla), Murcia, 30120, Spain
| | - Donate Weghorn
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
| | - Sara Sdelci
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
| | - Ana Janic
- Department of Medicine and Life Sciences, Universidad Pompeu Fabra, Barcelona, 08003, Spain.
| |
Collapse
|