51
|
Paz MM, Ferretti GDS, Martins-Dinis MMC, Ferreira BIS, Faier-Pereira A, Barnoud T, Moreira OC, Silva JL, Cordeiro Y, Rangel LP. PRIMA-1 inhibits Y220C p53 amyloid aggregation and synergizes with cisplatin in hepatocellular carcinoma. Front Mol Biosci 2023; 10:1165132. [PMID: 37101558 PMCID: PMC10123287 DOI: 10.3389/fmolb.2023.1165132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/22/2023] [Indexed: 04/28/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Although many therapeutic options are available, several factors, including the presence of p53 mutations, impact tumor development and therapeutic resistance. TP53 is the second most frequently mutated gene in HCC, comprising more than 30% of cases. Mutations in p53 result in the formation of amyloid aggregates that promote tumor progression. The use of PRIMA-1, a small molecule capable of restoring p53, is a therapeutic strategy to pharmacologically target the amyloid state mutant p53. In this study, we characterize an HCC mutant p53 model for the study of p53 amyloid aggregation in HCC cell lines, from in silico analysis of p53 mutants to a 3D-cell culture model and demonstrate the unprecedented inhibition of Y220C mutant p53 aggregation by PRIMA-1. In addition, our data show beneficial effects of PRIMA-1 in several "gain of function" properties of mutant-p53 cancer cells, including migration, adhesion, proliferation, and drug resistance. We also demonstrate that the combination of PRIMA-1 and cisplatin is a promising approach for HCC therapy. Taken together, our data support the premise that targeting the amyloid-state of mutant p53 may be an attractive therapeutic approach for HCC, and highlight PRIMA-1 as a new candidate for combination therapy with cisplatin.
Collapse
Affiliation(s)
- Mariana M. Paz
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giulia D. S. Ferretti
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Mafalda M. C. Martins-Dinis
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Beatriz I. S. Ferreira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Amanda Faier-Pereira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Thibaut Barnoud
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Otacilio C. Moreira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jerson L. Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana P. Rangel
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Luciana P. Rangel,
| |
Collapse
|
52
|
Integrin receptor trafficking in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:271-302. [PMID: 36813362 DOI: 10.1016/bs.pmbts.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrins are a family of 24 different heterodimers that are indispensable for multicellular life. Cell polarity, adhesion and migration are controlled by integrins delivered to the cell surface which in turn is regulated by the exo- and endocytic trafficking of integrins. The deep integration between trafficking and cell signaling determines the spatial and temporal output from any biochemical cue. Integrin trafficking plays a key role in development and many pathological conditions, especially cancer. Several novel regulators of integrin traffic have been discovered in recent times, including a novel class of integrin carrying vesicles, the intracellular nanovesicles (INVs). The tight regulation of trafficking pathways by cell signaling, where kinases phosphorylate key small GTPases in the trafficking pathway enable coordination of cell response to the extracellular milieu. Integrin heterodimer expression and trafficking differ in different tissues and contexts. In this Chapter, we discuss recent studies on integrin trafficking and its contribution to normal physiological and pathophysiological states.
Collapse
|
53
|
Sengupta S, Ghufran SM, Khan A, Biswas S, Roychoudhury S. Transition of amyloid/mutant p53 from tumor suppressor to an oncogene and therapeutic approaches to ameliorate metastasis and cancer stemness. Cancer Cell Int 2022; 22:416. [PMID: 36567312 PMCID: PMC9791775 DOI: 10.1186/s12935-022-02831-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/11/2022] [Indexed: 12/27/2022] Open
Abstract
The tumor suppressor p53 when undergoes amyloid formation confers several gain-of-function (GOF) activities that affect molecular pathways crucial for tumorigenesis and progression like some of the p53 mutants. Even after successful cancer treatment, metastasis and recurrence can result in poor survival rates. The major cause of recurrence is mainly the remnant cancer cells with stem cell-like properties, which are resistant to any chemotherapy treatment. Several studies have demonstrated the role of p53 mutants in exacerbating cancer stemness properties and epithelial-mesenchymal transition in these remnant cancer cells. Analyzing the amyloid/mutant p53-mediated signaling pathways that trigger metastasis, relapse or chemoresistance may be helpful for the development of novel or improved individualized treatment plans. In this review, we discuss the changes in the metabolic pathways such as mevalonate pathway and different signaling pathways such as TGF-β, PI3K/AKT/mTOR, NF-κB and Wnt due to p53 amyloid formation, or mutation. In addition to this, we have discussed the role of the regulatory microRNAs and lncRNAs linked with the mutant or amyloid p53 in human malignancies. Such changes promote tumor spread, potential recurrence, and stemness. Importantly, this review discusses the cancer therapies that target either mutant or amyloid p53, restore wild-type functions, and exploit the synthetic lethal interactions with mutant p53.
Collapse
Affiliation(s)
- Shinjinee Sengupta
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Shaikh Maryam Ghufran
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Aqsa Khan
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Subhrajit Biswas
- grid.444644.20000 0004 1805 0217Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, Uttar Pradesh, 201313 India
| | - Susanta Roychoudhury
- grid.489176.50000 0004 1803 6730Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, 700063 India ,grid.417635.20000 0001 2216 5074Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
54
|
Rodriguez Calleja L, Lavaud M, Tesfaye R, Brounais-Le-Royer B, Baud’huin M, Georges S, Lamoureux F, Verrecchia F, Ory B. The p53 Family Members p63 and p73 Roles in the Metastatic Dissemination: Interactions with microRNAs and TGFβ Pathway. Cancers (Basel) 2022; 14:cancers14235948. [PMID: 36497429 PMCID: PMC9741383 DOI: 10.3390/cancers14235948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
TP53 (TP53), p73 (TP73), and p63 (TP63) are members of the p53 transcription factor family, which has many activities spanning from embryonic development through to tumor suppression. The utilization of two promoters and alternative mRNA splicing has been shown to yield numerous isoforms in p53, p63, and p73. TAp73 is thought to mediate apoptosis as a result of nuclear accumulation following chemotherapy-induced DNA damage, according to a number of studies. Overexpression of the nuclear ΔNp63 and ΔNp73 isoforms, on the other hand, suppresses TAp73's pro-apoptotic activity in human malignancies, potentially leading to metastatic spread or inhibition. Another well-known pathway that has been associated to metastatic spread is the TGF pathway. TGFs are a family of structurally related polypeptide growth factors that regulate a variety of cellular functions including cell proliferation, lineage determination, differentiation, motility, adhesion, and cell death, making them significant players in development, homeostasis, and wound repair. Various studies have already identified several interactions between the p53 protein family and the TGFb pathway in the context of tumor growth and metastatic spread, beginning to shed light on this enigmatic intricacy.
Collapse
|
55
|
Gao N, Zheng Q, Wang Y, Li X, Li Z, Xiao H. Wun2-mediated integrin recycling promotes apoptotic cell clearance in Drosophila melanogaster. Cell Death Differ 2022; 29:2545-2561. [PMID: 35840760 PMCID: PMC9751302 DOI: 10.1038/s41418-022-01039-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/25/2022] [Accepted: 07/04/2022] [Indexed: 01/31/2023] Open
Abstract
Apoptotic cell (AC) clearance is a complex process in which phagocytes recognize, engulf, and digest ACs during organismal development and tissue homeostasis. Impaired efferocytosis results in developmental defects and autoimmune diseases. In the current study, we performed RNA-sequencing to systematically identify regulators involved in the phagocytosis of ACs by Drosophila melanogaster macrophage-like S2 cells, followed by targeted RNA interference screening. Wunen2 (Wun2), a homolog of mammalian lipid phosphate phosphatase (LPP), was deemed as required for efferocytosis both in vitro and in vivo. However, efferocytosis was independent of Wun2 phosphatase activity. Proteomic analysis further revealed that Rab11 and its effector Rip11 are interaction partners of Wun2. Therefore, Wun2 collaborates with Rip11 and Rab11 to mediate efficient recycling of the phagocytic receptor βν integrin subunit to the plasma membrane. The loss of Wun2 results in the routing of βv integrin subunit (Itgbn) into lysosomes, leading to its degradation. The deficiency of βv integrin subunit on the cell surface leads to aberrant and disorganized actin cytoskeleton, thereby influencing the formation of macrophage pseudopodia toward ACs and thus failure to engulf them. The findings of this study provide insights that clarify how phagocytes coordinate AC signals and adopt a precise mechanism for the maintenance of engulfment receptors at their cell membrane surface to regulate efferocytosis.
Collapse
Affiliation(s)
- Ning Gao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
- Medical College of Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Qian Zheng
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yanzhe Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Xiaowen Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Zhi Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Hui Xiao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| |
Collapse
|
56
|
Extrinsic cell death pathway plasticity: a driver of clonal evolution in cancer? Cell Death Dis 2022; 8:465. [PMID: 36435845 PMCID: PMC9701215 DOI: 10.1038/s41420-022-01251-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/28/2022]
Abstract
Human cancers are known to adhere to basic evolutionary principles. During their journey from early transformation to metastatic disease, cancer cell populations have proven to be remarkably adaptive to different forms of intra- and extracellular selective pressure, including nutrient scarcity, oxidative stress, and anti-cancer immunity. Adaption may be achieved via the expansion of clones bearing driver mutations that optimize cellular fitness in response to the specific selective scenario, e.g., mutations facilitating evasion of cell death, immune evasion or increased proliferation despite growth suppression, all of which constitute well-established hallmarks of cancer. While great progress concerning the prevention, diagnosis and treatment of clinically apparent disease has been made over the last 50 years, the mechanisms underlying cellular adaption under selective pressure via the immune system during early carcinogenesis and its influence on cancer cell fate or disease severity remain to be clarified. For instance, evasion of cell death is generally accepted as a hallmark of cancer, yet recent decades have revealed that the extrinsic cell death machinery triggered by immune effector cells is composed of an astonishingly complex network of interacting—and sometimes compensating—modes of cell death, whose role in selective processes during early carcinogenesis remains obscure. Based upon recent advances in cell death research, here we propose a concept of cell death pathway plasticity in time shaping cancer evolution prior to treatment in an effort to offer new perspectives on how cancer cell fate may be determined by cell death pathway plasticity during early carcinogenesis.
Collapse
|
57
|
Mutant p53 in cancer: from molecular mechanism to therapeutic modulation. Cell Death Dis 2022; 13:974. [PMID: 36400749 PMCID: PMC9674619 DOI: 10.1038/s41419-022-05408-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
TP53, a crucial tumor suppressor gene, is the most commonly mutated gene in human cancers. Aside from losing its tumor suppressor function, mutant p53 (mutp53) often acquires inherent, novel oncogenic functions, which is termed "gain-of-function". Emerging evidence suggests that mutp53 is highly associated with advanced malignancies and poor prognosis, which makes it a target for development of novel cancer therapies. Herein, we provide a summary of our knowledge of the mutp53 types and mutp53 spectrum in cancers. The mechanisms of mutp53 accumulation and gain-of-function are also summarized. Furthermore, we discuss the gain-of-function of mutp53 in cancers: genetic instability, ferroptosis, microenvironment, and stemness. Importantly, the role of mutp53 in the clinic is also discussed, particularly with regard to chemotherapy and radiotherapy. Last, emphasis is given to emerging strategies on how to target mutp53 for tumor therapy. Thus, this review will contribute to better understanding of the significance of mutp53 as a target for therapeutic strategies.
Collapse
|
58
|
Bizzarri AR. Conformational Heterogeneity and Frustration of the Tumor Suppressor p53 as Tuned by Punctual Mutations. Int J Mol Sci 2022; 23:12636. [PMID: 36293489 PMCID: PMC9604312 DOI: 10.3390/ijms232012636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 12/02/2022] Open
Abstract
The conformational heterogeneity of the p53 tumor suppressor, the wild-type (p53wt) and mutated forms, was investigated by a computational approach, including the modeling and all atoms of the molecular dynamics (MD) simulations. Four different punctual mutations (p53R175H, p53R248Q, p53R273H, and p53R282W) which are known to affect the DNA binding and belong to the most frequent hot-spot mutations in human cancers, were taken into consideration. The MD trajectories of the wild-type and mutated p53 forms were analyzed by essential dynamics to extract the relevant collective motions and by the frustration method to evaluate the degeneracy of the energy landscape. We found that p53 is characterized by wide collective motions and its energy landscape exhibits a rather high frustration level, especially in the regions involved in the binding to physiological ligands. Punctual mutations give rise to a modulation of both the collective motions and the frustration of p53, with different effects depending on the mutation. The regions of p53wt and of the mutated forms characterized by a high frustration level are also largely involved in the collective motions. Such a correlation is discussed also in connection with the intrinsic disordered character of p53 and with its central functional role.
Collapse
Affiliation(s)
- Anna Rita Bizzarri
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| |
Collapse
|
59
|
GOF Mutant p53 in Cancers: A Therapeutic Challenge. Cancers (Basel) 2022; 14:cancers14205091. [PMID: 36291874 PMCID: PMC9600758 DOI: 10.3390/cancers14205091] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary In normal cells, p53 is a protein which regulates the cell cycle progression to ensure normal cell division, growth, and development. However, in cancer, changes in the p53 DNA sequence, called genetic mutation, results in the protein either losing its normal function or exhibiting advanced pro-tumorigenic functions that lead to cancer. Importantly, cancers with mutations in the p53 protein often represent ones which are more aggressive and more resistant to chemotherapy. As a result, many studies have and continue to investigate multiple ways to target mutant p53-bearing cancer using targeted therapy, gene therapy, immunotherapy, and combination therapies. Knowledge of these strategies is important in improving the overall therapeutic response of cancers with mutant p53. This review highlights new strategies and discusses the progression of such therapies. Abstract TP53 is mutated in the majority of human cancers. Mutations can lead to loss of p53 expression or expression of mutant versions of the p53 protein. These mutant p53 proteins have oncogenic potential. They can inhibit any remaining WTp53 in a dominant negative manner, or they can acquire new functions that promote tumour growth, invasion, metastasis and chemoresistance. In this review we explore some of the mechanisms that make mutant p53 cells resistant to chemotherapy. As mutant p53 tumours are resistant to many traditional chemotherapies, many have sought to explore new ways of targeting mutant p53 tumours and reinstate chemosensitivity. These approaches include targeting of mutant p53 stability, mutant p53 binding partners and downstream pathways, p53 vaccines, restoration of WTp53 function, and WTp53 gene delivery. The current advances and challenges of these strategies are discussed.
Collapse
|
60
|
Drebrin promotes lung adenocarcinoma cell migration through inducing integrin β1 endocytosis. Biochem Biophys Res Commun 2022; 630:175-182. [PMID: 36155064 DOI: 10.1016/j.bbrc.2022.09.050] [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: 07/19/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/22/2022]
Abstract
Lung adenocarcinoma (LUAD) is the most common type of lung cancers, which remains the leading cause of cancer-related death worldwide. Drebrin can promote cell migration and invasion with poor prognosis, but its roes in LUAD tumor progression remains unknown. We showed that the expression of Drebrin was upregulated in clinical LUAD samples. A Kaplan-Meier survival analysis showed that a high expression of Drebrin predicated poor prognosis in LUAD. In vitro, Drebrin promoted anchorage-independent growth and migration of LUAD cells. Drebrin interacted with dynamin through CT domain, and served as an adaptor to promote LUAD cell migration through inducing integrin β1 endocytosis. Thus, this study demonstrated the critical role of Drebrin in LUAD and associated mechanism.
Collapse
|
61
|
Miao TW, Chen FY, Du LY, Xiao W, Fu JJ. Signature based on RNA-binding protein-related genes for predicting prognosis and guiding therapy in non-small cell lung cancer. Front Genet 2022; 13:930826. [PMID: 36118863 PMCID: PMC9479344 DOI: 10.3389/fgene.2022.930826] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Studies have reported that RNA-binding proteins (RBPs) are dysregulated in multiple cancers and are correlated with the progression and prognosis of disease. However, the functions of RBPs in non-small cell lung cancer (NSCLC) remain unclear. The present study aimed to explore the function of RBPs in NSCLC and their prognostic and therapeutic value.Methods: The mRNA expression profiles, DNA methylation data, gene mutation data, copy number variation data, and corresponding clinical information on NSCLC were downloaded from The Cancer Genome Atlas, Gene Expression Omnibus, and the University of California Santa Cruz Xena databases. The differentially expressed RBPs were identified between tumor and control tissues, and the expression and prognostic value of these RBPs were systemically investigated by bioinformatics analysis. A quantitative polymerase chain reaction (qPCR) was performed to validate the dysregulated genes in the prognostic signature.Results: A prognostic RBP-related signature was successfully constructed based on eight RBPs represented as a risk score using least absolute shrinkage and selection operator (LASSO) regression analysis. The high-risk group had a worse overall survival (OS) probability than the low-risk group (p < 0.001) with 1-, 3-, and 5-year area under the receiver operator characteristic curve values of 0.671, 0.638, and 0.637, respectively. The risk score was associated with the stage of disease (p < 0.05) and was an independent prognostic factor for NSCLC when adjusted for age and UICC stage (p < 0.001, hazard ratio (HR): 1.888). The constructed nomogram showed a good predictive value. The P53, focal adhesion, and NOD-like receptor signaling pathways were the primary pathways in the high-risk group (adjusted p value <0.05). The high-risk group was correlated with increased immune infiltration (p < 0.05), upregulated relative expression levels of programmed cell death 1 (PD1) (p = 0.015), cytotoxic T-lymphocyte-associated protein 4 (CTLA4) (p = 0.042), higher gene mutation frequency, higher tumor mutational burden (p = 0.034), and better chemotherapy response (p < 0.001). The signature was successfully validated using the GSE26939, GSE31210, GSE30219, and GSE157009 datasets. Dysregulation of these genes in patients with NSCLC was confirmed using the qPCR in an independent cohort (p < 0.05).Conclusion: An RBP-related signature was successfully constructed to predict prognosis in NSCLC, functioning as a reference for individualized therapy, including immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Ti-Wei Miao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Fang-Ying Chen
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Long-Yi Du
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Xiao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Juan-Juan Fu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Juan-Juan Fu,
| |
Collapse
|
62
|
Parihar K, Nukpezah J, Iwamoto DV, Janmey PA, Radhakrishnan R. Data driven and biophysical insights into the regulation of trafficking vesicles by extracellular matrix stiffness. iScience 2022; 25:104721. [PMID: 35865140 PMCID: PMC9293776 DOI: 10.1016/j.isci.2022.104721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Biomechanical signals from remodeled extracellular matrix (ECM) promote tumor progression. Here, we show that cell-matrix and cell-cell communication may be inherently linked and tuned through mechanisms of mechanosensitive biogenesis of trafficking vesicles. Pan-cancer analysis of cancer cells' mechanical properties (focusing primarily on cell stiffness) on substrates of varied stiffness and composition elucidated a heterogeneous cellular response to mechanical stimuli. Through machine learning, we identified a fingerprint of cytoskeleton-related proteins that accurately characterize cell stiffness in different ECM conditions. Expression of their respective genes correlates with patient prognosis across different tumor types. The levels of selected cytoskeleton proteins indicated that cortical tension mirrors the increase (or decrease) in cell stiffness with a change in ECM stiffness. A mechanistic biophysical model shows that the tendency for curvature generation by curvature-inducing proteins has an ultrasensitive dependence on cortical tension. This study thus highlights the effect of ECM stiffness, mediated by cortical tension, in modulating vesicle biogenesis.
Collapse
Affiliation(s)
- Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Nukpezah
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel V. Iwamoto
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A. Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
63
|
Vadakekolathu J, Boocock DJ, Pandey K, Guinn BA, Legrand A, Miles AK, Coveney C, Ayala R, Purcell AW, McArdle SE. Multi-Omic Analysis of Two Common P53 Mutations: Proteins Regulated by Mutated P53 as Potential Targets for Immunotherapy. Cancers (Basel) 2022; 14:cancers14163975. [PMID: 36010968 PMCID: PMC9406384 DOI: 10.3390/cancers14163975] [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: 06/30/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary TP53 is the most frequently mutated gene in many cancers, but it has failed to be a very effective target for treatment to date. To overcome this, we have examined what else changes in cells when the TP53 gene is mutated. We modified cells that had no TP53 expression to have one of the two most common mutations, either R175H or R273H. We examined how the presence of these TP53 mutations caused cellular changes including microscopic, gene expression and peptide presentation to the immune system. This has allowed us to identify new (secondary) targets that could be used to facilitate the treatment of tumors that harbor p53 mutations. Abstract The p53 protein is mutated in more than 50% of human cancers. Mutated p53 proteins not only lose their normal function but often acquire novel oncogenic functions, a phenomenon termed mutant p53 gain-of-function. Mutant p53 has been shown to affect the transcription of a range of genes, as well as protein–protein interactions with transcription factors and other effectors; however, no one has intensively investigated and identified these proteins, or their MHC presented epitopes, from the viewpoint of their ability to act as targets for immunotherapeutic interventions. We investigated the molecular changes that occurred after the TP53 null osteosarcoma cells, SaOS-2, were transfected with one of two conformational p53-mutants, either R175H or R273H. We then examined the phenotypic and functional changes using macroscopic observations, proliferation, gene expression and proteomics alongside immunopeptidome profiling of peptide antigen presentation in the context of major histocompatibility complex (MHC) class I molecules. We identified several candidate proteins in both TP53 mutant cell lines with differential expression when compared to the TP53 null vector control, SaOS-V. Quantitative SWATH proteomics combined with immune-peptidome analysis of the class-I eluted peptides identified several epitopes presented on pMHC and in silico analysis shortlisted which antigens were expressed in a range of cancerous but not adjacent healthy tissues. Out of all the candidates, KLC1 and TOP2A showed high levels of expression in every tumor type examined. From these proteins, three A2 and four pan HLA-A epitopes were identified in both R175H and R273H from TOP2A. We have now provided a short list of future immunotherapy targets for the treatment of cancers harboring mutated TP53.
Collapse
Affiliation(s)
- Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK or
| | - David J. Boocock
- Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham NG1 4FQ, UK
| | - Kirti Pandey
- Infection and Immunology Program, Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Barbara-ann Guinn
- Department of Biomedical Sciences, University of Hull, Hull HU6 7RX, UK
| | - Antoine Legrand
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK or
| | - Amanda K. Miles
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK or
- Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham NG1 4FQ, UK
| | - Clare Coveney
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK or
| | - Rochelle Ayala
- Infection and Immunology Program, Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Anthony W. Purcell
- Infection and Immunology Program, Biomedicine Discovery Institute, Melbourne, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Stephanie E. McArdle
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK or
- Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham NG1 4FQ, UK
- Correspondence:
| |
Collapse
|
64
|
Proteasomal and autophagy-mediated degradation of mutp53 proteins through mitochondria-targeting aggregation-induced-emission materials. Acta Biomater 2022; 150:402-412. [PMID: 35931280 DOI: 10.1016/j.actbio.2022.07.057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022]
Abstract
Close to half of human cancers harbor point mutations in the tumor-suppressor p53 gene, giving rise to the cellular accumulation of mutant p53 (mutp53) proteins with novel neomorphic gain-of-function (GOF) properties. The destruction of mutp53 proteins through either autophagic or proteasomal degradation is a viable strategy for the targeted therapy of p53-mutated cancers. Several nanomaterials, including zinc-iron and ZIF-8 nanoparticles (NPs), have been reported to induce the proteasomal degradation of mutp53 proteins. However, how autophagy, the other major cellular degradative pathway, influences NP-induced mutp53 degradation has not been investigated. This article shows that AIE-Mit-TPP, a mitochondria-targeting material with aggregation-induced emission (AIE) characteristics, elicits ubiquitination-dependent proteasomal degradation of a broad range of mutp53 proteins. Meanwhile, AIE-Mit-TPP also induces massive mitochondrial damage and autophagy. The inhibition of autophagy further increases AIE-Mit-TPP-elicited mutp53 degradation, revealing the negative impact of autophagy on AIE-Mit-TPP-induced mutp53 degradation. As expected, the degradation of mutp53 proteins by AIE-Mit-TPP abrogated mutp53-manifested GOF, leading to reductions in cell proliferation and migration and increases in cell cycle arrest and cell death. Consequently, AIE-Mit-TPP inhibited the growth of mutp53 tumors. This paper unravels the interesting interplay between the proteasomal and autophagic degradative pathways and pinpoints the modulation of autophagy as a potential strategy for optimizing NP-induced mutp53 degradation and p53-targeted cancer therapy. STATEMENT OF SIGNIFICANCE: We have designed three different types of AIE materials: non-targeting (AIE-Br), mitochondria-targeting (AIE-Mit-TPP), lysosome-targeting (AIE-Lyso). Our results proved that mitochondria-targeting AIE material induced degradation of mutp53 proteins via the proteasome degradation pathway and abrogated mutp53-conferred GOF phenotypes. Furthermore, we performed in vitro studies on the effect of the tested materials in mutp53-expressing cancer cells and demonstrated our findings via in vivo investigations in a mouse subcutaneous p53R175H TOV112D ovarian cancer model. Our results confirmed the link between the proteasome pathway and autophagy and thus proposed a strategy of combining AIE-Mit-TPP with autophagy inhibitors for the targeted treatment of mutp53-associated tumors. Finally, we found that AIE-Mit-TPP could induce degradation of a wide-spectrum mutp53 proteins, which makes mitochondria-targeting AIE materials an effective therapeutic strategy for p53-mutated cancers.
Collapse
|
65
|
Li Y, Liu B, Yin X, Jiang Z, Fang C, Chen N, Zhang B, Dai L, Yin Y. Targeted demethylation of the SLC5A7 promotor inhibits colorectal cancer progression. Clin Epigenetics 2022; 14:92. [PMID: 35858918 PMCID: PMC9301853 DOI: 10.1186/s13148-022-01308-5] [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: 11/15/2021] [Accepted: 07/04/2022] [Indexed: 02/08/2023] Open
Abstract
Background SLC5A7 (solute carrier family 5 member 7), also known as choline transporter 1 (CHT1), is downregulated in colorectal cancer (CRC) and functions as a tumor suppressor. However, the mechanisms underlying the inactivation of SLC5A7 in CRC remain to be elucidated. Results In the present study, two broad-spectrum demethylation agents (azacitidine and decitabine) employed to treat CRC cells significantly upregulated SLC5A7 expression. Further results based on the CRC cohort and TCGA database indicated that SLC5A7 promoter methylation inversely correlated with SLC5A7 expression, and the status of SLC5A7 promotor methylation showed a promising prognostic value for patients with CRC. Next, the dCas9-multiGCN4/scFv-TET1CD-based precision demethylation system was constructed, which could significantly and specifically promote SLC5A7 expression in CRC cells through sgRNA targeting the SLC5A7 promoter. Both in vitro and in vivo experiments demonstrated that targeted demethylation of SLC5A7 by dCas9-multiGCN4/scFv-TET1CD-sgSLC5A7 inhibited tumor growth by stabilizing p53 and regulating downstream targets. Conclusions Collectively, DNA promoter methylation caused inactivation of SLC5A7 in CRC, and targeted demethylation of SLC5A7 might be a therapeutic target for CRC and other cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01308-5.
Collapse
Affiliation(s)
- Yang Li
- Department of Gastrointestinal Surgery, Guang'an People's Hospital, Guang'an, 638500, Sichuan, People's Republic of China.,Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Baike Liu
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhiyuan Jiang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.,School of Pharmacy, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
66
|
Mouse Models for Application in Colorectal Cancer: Understanding the Pathogenesis and Relevance to the Human Condition. Biomedicines 2022; 10:biomedicines10071710. [PMID: 35885015 PMCID: PMC9313309 DOI: 10.3390/biomedicines10071710] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancer (CRC) is a malignant disease that is the second most common cancer worldwide. CRC arises from the complex interactions among a variety of genetic and environmental factors. To understand the mechanism of colon tumorigenesis, preclinical studies have developed various mouse models including carcinogen-induced and transgenic mice to recapitulate CRC in humans. Using these mouse models, scientific breakthroughs have been made on the understanding of the pathogenesis of this complex disease. Moreover, the availability of transgenic knock-in or knock-out mice further increases the potential of CRC mouse models. In this review, the overall features of carcinogen-induced (focusing on azoxymethane and azoxymethane/dextran sulfate sodium) and transgenic (focusing on ApcMin/+) mouse models, as well as their mechanisms to induce colon tumorigenesis, are explored. We also discuss limitations of these mouse models and their applications in the evaluation and study of drugs and treatment regimens against CRC. Through these mouse models, a better understanding of colon tumorigenesis can be achieved, thereby facilitating the discovery of novel therapeutic strategies against CRC.
Collapse
|
67
|
Seo SU, Woo SM, Lee SG, Kim MY, Lee HS, Choi YH, Kim SH, Chang YC, Min KJ, Kwon TK. BAP1 phosphorylation-mediated Sp1 stabilization plays a critical role in cathepsin K inhibition-induced C-terminal p53-dependent Bax upregulation. Redox Biol 2022; 53:102336. [PMID: 35584569 PMCID: PMC9117696 DOI: 10.1016/j.redox.2022.102336] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 11/25/2022] Open
Abstract
Cathepsin K inhibitor (odanacatib; ODN) and cathepsin K knockdown (siRNA) enhance oxaliplatin-induced apoptosis through p53-dependent Bax upregulation. However, its underlying mechanisms remain unclear. In this study, we elucidated the mechanism behind enhancement of oxaliplatin-induced apoptosis by ODN. We also investigated the molecular mechanisms of ODN-induced Bax upregulation. Here, we demonstrated that ODN-induced Bax upregulation required p53, but it was independent of p53 transcriptional activity. Various mutants of the DNA-binding domain of p53 induced Bax upregulation in ODN-treated cells. p53 functional domain analysis showed that the C-terminal domain of p53 participates in the physical interaction and stabilization of Sp1, a major transcription factor of Bax. We screened a specific siRNA encoding 50 deubiquitinases and identified that BAP1 stabilizes Sp1. The knockdown or catalytic mutant form of BAP1 abolished the ODN-induced upregulation of Sp1 and Bax expression. Mechanistically, ODN induced BAP1 phosphorylation and enhanced Sp1-BAP1 interaction, resulting in Sp1 ubiquitination and degradation. Interestingly, ODN-induced BAP1 phosphorylation and DNA damage were modulated by the production of mitochondrial reactive oxygen species (ROS). Mitochondrial ROS scavengers prevented DNA damage, BAP1-mediated Sp1 stabilization, and Bax upregulation by ODN. BAP1 downregulation by siRNA inhibited apoptosis induced by the combined treatment of ODN and oxaliplatin/etoposide. Therefore, Sp1 is a crucial transcription factor for ODN-induced Bax upregulation, and Sp1 stabilization is regulated by BAP1. Odanacatib (ODN) enhances oxaliplatin-induced apoptosis by upregulating Bax. ODN-mediated Bax upregulation is independent of p53 transcriptional activity. C-terminal domain of p53 induces Sp1 stabilization linked to BAP1 phosphorylation. ODN-mediated mitochondrial ROS generation causes BAP1 phosphorylation and DNA damage.
Collapse
Affiliation(s)
- Seung Un Seo
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, South Korea
| | - Seon Min Woo
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, South Korea
| | - Seul Gi Lee
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, South Korea
| | - Min Yeong Kim
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan, 47227, South Korea
| | - Hyun Shik Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, 41566, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan, 47227, South Korea
| | - Sang Hyun Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, 42472, South Korea
| | - Kyoung-Jin Min
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, South Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, South Korea.
| |
Collapse
|
68
|
Chen M, Choi S, Wen T, Chen C, Thapa N, Lee JH, Cryns VL, Anderson RA. A p53-phosphoinositide signalosome regulates nuclear AKT activation. Nat Cell Biol 2022; 24:1099-1113. [PMID: 35798843 PMCID: PMC9833102 DOI: 10.1038/s41556-022-00949-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 05/26/2022] [Indexed: 01/13/2023]
Abstract
The tumour suppressor p53 and PI3K-AKT pathways have fundamental roles in the regulation of cell growth and apoptosis, and are frequently mutated in cancer. Here, we show that genotoxic stress induces nuclear AKT activation through a p53-dependent mechanism that is distinct from the canonical membrane-localized PI3K-AKT pathway. Following genotoxic stress, a nuclear PI3K binds p53 in the non-membranous nucleoplasm to generate a complex of p53 and phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3), which recruits AKT, PDK1 and mTORC2 to activate AKT and phosphorylate FOXO proteins, thereby inhibiting DNA damage-induced apoptosis. Wild-type p53 activates nuclear AKT in an on/off fashion following stress, whereas mutant p53 dose-dependently stimulates high basal AKT activity. The p53-PtdIns(3,4,5)P3 complex is dephosphorylated to p53-phosphatidylinositol 4,5-bisphosphate by PTEN to inhibit AKT activation. The nuclear p53-phosphoinositide signalosome is distinct from the canonical membrane-localized pathway and insensitive to PI3K inhibitors currently in the clinic, which underscores its therapeutic relevance.
Collapse
Affiliation(s)
- Mo Chen
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Suyong Choi
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Tianmu Wen
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Changliang Chen
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Narendra Thapa
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Jeong Hyo Lee
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Vincent L Cryns
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA.
| | - Richard A Anderson
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
69
|
The Therapeutic Potential of the Restoration of the p53 Protein Family Members in the EGFR-Mutated Lung Cancer. Int J Mol Sci 2022; 23:ijms23137213. [PMID: 35806218 PMCID: PMC9267050 DOI: 10.3390/ijms23137213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the recent development of precision medicine and targeted therapies, lung cancer remains the top cause of cancer-related mortality worldwide. The patients diagnosed with metastatic disease have a five-year survival rate lower than 6%. In metastatic disease, EGFR is the most common driver of mutation, with the most common co-driver hitting TP53. EGFR-positive patients are offered the frontline treatment with tyrosine kinase inhibitors, yet the development of resistance and the lack of alternative therapies make this group of patients only fit for clinical trial participation. Since mutant p53 is the most common co-driver in the metastatic setting, therapies reactivating the p53 pathway might serve as a promising alternative therapeutic approach in patients who have developed a resistance to tyrosine kinase inhibitors. This review focuses on the molecular background of EGFR-mutated lung cancer and discusses novel therapeutic options converging on the reactivation of p53 tumor suppressor pathways.
Collapse
|
70
|
|
71
|
Cross-talk between mutant p53 and p62/SQSTM1 augments cancer cell migration by promoting the degradation of cell adhesion proteins. Proc Natl Acad Sci U S A 2022; 119:e2119644119. [PMID: 35439056 PMCID: PMC9173583 DOI: 10.1073/pnas.2119644119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Missense mutations in the TP53 gene, encoding the p53 tumor suppressor, are very frequent in human cancer. Some of those mutations, particularly the more common (“hotspot”) ones, not only abrogate p53’s tumor suppressor activities but also endow the mutant protein with oncogenic gain of function (GOF). We report that p53R273H, the most common p53 mutant in pancreatic cancer, interacts with the SQSTM1/p62 protein to accelerate the degradation of cell adhesion proteins. This enables pancreatic cancer cells to detach from the epithelial sheet and engage in individualized cell migration, probably augmenting metastatic spread. By providing insights into mechanisms that underpin mutant p53 GOF, this study may suggest ways to interfere with the progression of cancers bearing particular p53 mutants. Missense mutations in the p53 tumor suppressor abound in human cancer. Common (“hotspot”) mutations endow mutant p53 (mutp53) proteins with oncogenic gain of function (GOF), including enhanced cell migration and invasiveness, favoring cancer progression. GOF is usually attributed to transcriptional effects of mutp53. To elucidate transcription-independent effects of mutp53, we characterized the protein interactome of the p53R273H mutant in cells derived from pancreatic ductal adenocarcinoma (PDAC), where p53R273H is the most frequent p53 mutant. We now report that p53R273H, but not the p53R175H hotspot mutant, interacts with SQSTM1/p62 and promotes cancer cell migration and invasion in a p62-dependent manner. Mechanistically, the p53R273H-p62 axis drives the proteasomal degradation of several cell junction–associated proteins, including the gap junction protein Connexin 43, facilitating scattered cell migration. Concordantly, down-regulation of Connexin 43 augments PDAC cell migration, while its forced overexpression blunts the promigratory effect of the p53R273H-p62 axis. These findings define a mechanism of mutp53 GOF.
Collapse
|
72
|
Liu S, Yu J, Zhang H, Liu J. TP53 Co-Mutations in Advanced EGFR-Mutated Non-Small Cell Lung Cancer: Prognosis and Therapeutic Strategy for Cancer Therapy. Front Oncol 2022; 12:860563. [PMID: 35444951 PMCID: PMC9013831 DOI: 10.3389/fonc.2022.860563] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/16/2022] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. As the most prevalent molecular mutation subtypes in non-small cell lung cancer (NSCLC), EGFR-TKIs are currently a standard first-line therapy for targeting the mutated EGFR in advanced NSCLC patients. However, 20-30% of this subset of patients shows primary resistance to EGFR-TKIs. Patients with co-mutations of EGFR and several other genes have a poor response to EGFR-TKIs, whereas the prognostic and predictive significance of EGFR/TP53 co-mutation in NSCLC patients remains controversial. Meanwhile, little is known about how to choose an optimal therapeutic strategy for this subset of patients. Presently, no drugs targeting TP53 mutations are available on the market, and some p53 protein activators are in the early stage of clinical trials. A combination of EGFR-TKIs with antiangiogenic agents or chemotherapy or other agents might be a more appropriate strategy to tackle the problem. In this review, we describe the prognostic and predictive value of EGFR/TP53 co-mutation in NSCLC patients, investigate the mechanisms of this co-mutation affecting the response to EGFR-TKIs, and further explore optimal regimens effectively to prolong the survival time of the NSCLC patients harboring this co-mutation.
Collapse
Affiliation(s)
- Surui Liu
- Department of Oncology, Jinan Central Hospital, Jinan, China.,Department of Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jin Yu
- Department of Oncology, Jinan Central Hospital, Jinan, China
| | - Hui Zhang
- Department of Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Liu
- Department of Oncology, Jinan Central Hospital, Jinan, China
| |
Collapse
|
73
|
Ferrero A, Borghese M, Restaino S, Puppo A, Vizzielli G, Biglia N. Predicting Response to Anthracyclines in Ovarian Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:4260. [PMID: 35409939 PMCID: PMC8998349 DOI: 10.3390/ijerph19074260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022]
Abstract
(1) Background: Anthracyclines are intriguing drugs, representing one of the cornerstones of both first and subsequent-lines of chemotherapy in ovarian cancer (OC). Their efficacy and mechanisms of action are related to the hot topics of OC clinical research, such as BRCA status and immunotherapy. Prediction of response to anthracyclines is challenging and no markers can predict certain therapeutic success. The current narrative review provides a summary of the clinical and biological mechanisms involved in the response to anthracyclines. (2) Methods: A MEDLINE search of the literature was performed, focusing on papers published in the last two decades. (3) Results and Conclusions: BRCA mutated tumors seem to show a higher response to anthracyclines compared to sporadic tumors and the severity of hand-foot syndrome and mucositis may be a predictive marker of PLD efficacy. CA125 can be a misleading marker of clinical response during treatment with anthracyclines, the response of which also appears to depend on OC histology. Immunochemistry, in particular HER-2 expression, could be of some help in predicting the response to such drugs, and high levels of mutated p53 appear after exposure to anthracyclines and impair their antitumor effect. Finally, organoids from OC are promising for drug testing and prediction of response to chemotherapy.
Collapse
Affiliation(s)
- Annamaria Ferrero
- Academic Department of Gynaecology and Obstetrics, Mauriziano Hospital, 10128 Torino, Italy;
| | - Martina Borghese
- Department of Gynecology and Obstetrics, Santa Croce and Carle Hospital, 12100 Cuneo, Italy; (M.B.); (A.P.)
| | - Stefano Restaino
- Obstetrics and Gynecology Unit, Department of Obstetrics, Gynecology and Pediatrics, Department of Medical Area DAME, Udine University Hospital, 33100 Udine, Italy;
| | - Andrea Puppo
- Department of Gynecology and Obstetrics, Santa Croce and Carle Hospital, 12100 Cuneo, Italy; (M.B.); (A.P.)
| | - Giuseppe Vizzielli
- Clinic of Obstetrics and Gynaecology, Department of Medical Area (DAME), University of Udine, “Santa Maria della Misericordia” Hospital, Azienda Sanitaria Ospedaliera Friuli Centrale, 33100 Udine, Italy;
| | - Nicoletta Biglia
- Academic Department of Gynaecology and Obstetrics, Mauriziano Hospital, 10128 Torino, Italy;
| |
Collapse
|
74
|
Wang C, Zhang S, Ma B, Fu Y, Luo Y. TP53 mutations upregulate RCP expression via Sp1/3 to drive lung cancer progression. Oncogene 2022; 41:2357-2371. [PMID: 35256783 DOI: 10.1038/s41388-022-02260-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 11/10/2022]
Abstract
Mutant p53 (mtp53) can exert cancer-promoting activities via "gain-of-function", which has become a popular research target. Although lots of researchers focus on the tumor-suppressor role for p53, the regulation of mutant p53 remains unknown. Here, we report a mechanism by which mtp53 regulate the transcription of Rab coupling protein (RCP) to influence lung cancer behavior. First, we show that RCP is specifically expressed at high levels in lung cancer tissues and cells, and RCP knockout suppresses tumor growth and metastasis. Further mass spectrometry and functional analysis identify that Sp1, Sp3 and Stat3 are the transcriptional activators of RCP. Moreover, p53 is involved in modulating RCP expression in an Sp1/3 dependent manner. Mechanistically, in contrast to wild-type p53 suppression of RCP transcription by decreasing Sp1/3 proteins, TP53 mutations have changed on Sp1/3 expression via "loss-of-function". Surprisingly, the DNA contact mutants of p53 further robustly enhance their binding ability with Sp1/3 to drive RCP expression through the "gain-of-function" activity. Collectively, we reveal a mechanism by which p53 regulating the transcription of RCP to influence lung cancer progression, which provides new insights for treating p53 mutant lung cancer.
Collapse
Affiliation(s)
- Caihong Wang
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The National Engineering Research Center for Protein Technology, Tsinghua University, Beijing, 100084, China
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, 100084, China
| | - Shaosen Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The National Engineering Research Center for Protein Technology, Tsinghua University, Beijing, 100084, China
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, 100084, China
- Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Boyuan Ma
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The National Engineering Research Center for Protein Technology, Tsinghua University, Beijing, 100084, China
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, 100084, China
| | - Yan Fu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- The National Engineering Research Center for Protein Technology, Tsinghua University, Beijing, 100084, China
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, 100084, China
| | - Yongzhang Luo
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- The National Engineering Research Center for Protein Technology, Tsinghua University, Beijing, 100084, China.
- Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
75
|
Huang HM, Huang XY, Wu SP, Chen CK, He XH, Zhang YF. Parecoxib inhibits esophageal squamous cell carcinoma progression via the PDK1-AKT pathway. Cell Mol Biol Lett 2022; 27:28. [PMID: 35305553 PMCID: PMC8933956 DOI: 10.1186/s11658-022-00324-w] [Citation(s) in RCA: 3] [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: 09/30/2021] [Accepted: 02/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Parecoxib plays an important role in inhibition of human cancer. However, the effect of parecoxib on esophageal squamous cell carcinoma (ESCC) is still not well known. The purpose of this study was to investigate the effect of parecoxib on ESCC and its underlying mechanism. METHODS RNA-sequence analysis was performed to identify functional alterations and mechanisms. Cell cycle, proliferation, invasion, and migration were assessed using flow cytometry, CCK-8 assay, colony formation, transwell, and wound healing assays. Extracellular matrix (ECM) degradation was detected by substrate gel zymography and 3D cell culture assay. Western blotting was used to detect parecoxib-dependent mechanisms involving cell cycle, proliferation, invasion, and migration. Tumor formation in vivo was detected by mouse assay. RESULTS Functional experiments indicated that parecoxib induced ESCC cell cycle arrest in G2 phase, and inhibited cell proliferation, invasion, and migration in vitro. Western blotting revealed that parecoxib downregulated the phosphorylation levels of AKT and PDK1, as well as the expression of the mutant p53, cyclin B1, and CDK1, while upregulating p21waf1. Parecoxib inhibited matrix metalloproteinase-2 (MMP2) secretion and invadopodia formation, which were related to ECM degradation. Furthermore, we found that parecoxib suppressed ESCC growth in heterotopic tumor models. CONCLUSION Parecoxib inhibits ESCC progression, including cell cycle, proliferation, invasion, and migration, via the PDK1-AKT signaling pathway.
Collapse
Affiliation(s)
- Han-Ming Huang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xiao-Yu Huang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Shao-Ping Wu
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Can-Keng Chen
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xin-Hua He
- Department of Physiology, Shantou University Medical College, Shantou, 515041, People's Republic of China.
| | - Yong-Fa Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China.
| |
Collapse
|
76
|
Islam SU, Ahmed MB, Sonn JK, Jin EJ, Lee YS. PRP4 Induces Epithelial–Mesenchymal Transition and Drug Resistance in Colon Cancer Cells via Activation of p53. Int J Mol Sci 2022; 23:ijms23063092. [PMID: 35328513 PMCID: PMC8955441 DOI: 10.3390/ijms23063092] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/16/2022] Open
Abstract
Pre-mRNA processing factor 4B (PRP4) promotes pre-mRNA splicing and signal transduction. Recent studies have shown that PRP4 modulates the assembly of actin cytoskeleton in cancer cells and induces epithelial–mesenchymal transition (EMT) and drug resistance. PRP4 displays kinase domain-like cyclin-dependent kinases and mitogen-activated protein kinases, making it capable of phosphorylating p53 and other target proteins. In the current study, we report that PRP4 induces drug resistance and EMT via direct binding to the p53 protein, inducing its phosphorylation. Moreover, PRP4 overexpression activates the transcription of miR-210 in a hypoxia-inducible factor 1α (HIF-1α)-dependent manner, which activates p53. The involvement of miR-210 in the activation of p53 was confirmed by utilizing si-miR210. si-miR210 blocked the PRP4-activated cell survival pathways and reversed the PRP4-induced EMT phenotype. Moreover, we used deferoxamine as a hypoxia-mimetic agent, and si-HIF to silence HIF-1α. This procedure demonstrated that PRP4-induced EMT and drug resistance emerged in response to consecutive activation of HIF-1α, miR-210, and p53 by PRP4 overexpression. Collectively, our findings suggest that the PRP4 contributes to EMT and drug resistance induction via direct interactions with p53 and actions that promote upregulation of HIF-1α and miR-210. We conclude that PRP4 is an essential factor promoting cancer development and progression. Specific PRP4 inhibition could benefit patients with colon cancer.
Collapse
Affiliation(s)
- Salman Ul Islam
- Department of Pharmacy, Cecos University, Hayatabad, Peshawar 25000, Pakistan;
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Muhammad Bilal Ahmed
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Jong-Kyung Sonn
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Eun-Jung Jin
- Department of Biological Science, College of Natural Science, Wonkwang University, Iksan 54538, Korea
- Correspondence: (E.-J.J.); (Y.-S.L.); Tel.: +82-63-8500-6197(E.-J.J.); +82-53-950-6353 (Y.-S.L.); Fax: +82-53-943-2762 (E.-J.J.)
| | - Young-Sup Lee
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea;
- Correspondence: (E.-J.J.); (Y.-S.L.); Tel.: +82-63-8500-6197(E.-J.J.); +82-53-950-6353 (Y.-S.L.); Fax: +82-53-943-2762 (E.-J.J.)
| |
Collapse
|
77
|
Zampieri C, Panatta E, Corbo V, Mauriello A, Melino G, Amelio I. p53 mutations define the chromatin landscape to confer drug tolerance in pancreatic cancer. Mol Oncol 2022; 16:1259-1271. [PMID: 34919788 PMCID: PMC8936522 DOI: 10.1002/1878-0261.13161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/11/2021] [Accepted: 12/10/2021] [Indexed: 11/09/2022] Open
Abstract
Somatic inactivation of p53 (TP53) mainly occurs as missense mutations that lead to the acquisition of neomorphic mutant protein forms. p53 mutants have been postulated to exert gain-of-function (GOF) effects, including promotion of metastasis and drug tolerance, which generally contribute to the acquisition of the lethal phenotype. Here, by integrating a p53R270H -dependent transcriptomic analysis with chromatin accessibility (ATAC-seq) profiling, we shed light on the molecular basis of a p53 mutant-dependent drug-tolerant phenotype in pancreatic cancer. p53R270H finely tunes chromatin accessibility in specific genomic loci, orchestrating a transcriptional programme that participates in phenotypic evolution of the cancer. We specifically focused on the p53R270H -dependent regulation of the tyrosine kinase receptor macrophage-stimulating protein receptor (MST1r). MST1r deregulation substantially impinged on drug response in the experimental model, recapitulating the p53R270H -dependent phenotype, and strongly correlated with p53 mutant and aggressive phenotype in pancreatic cancer patients. As cellular plasticity in the final stages of the evolution of pancreatic cancer seems to predominantly originate from epigenetic mechanisms, we propose that mutant p53 participates in the acquisition of a lethal phenotype by fine-tuning the chromatin landscape.
Collapse
Affiliation(s)
- Carlotta Zampieri
- Department of Experimental MedicineTOR, University of Rome Tor VergataRomeItaly
| | - Emanuele Panatta
- Department of Experimental MedicineTOR, University of Rome Tor VergataRomeItaly
| | | | | | - Gerry Melino
- Department of Experimental MedicineTOR, University of Rome Tor VergataRomeItaly
| | - Ivano Amelio
- Department of Experimental MedicineTOR, University of Rome Tor VergataRomeItaly
- School of Life SciencesUniversity of NottinghamUK
| |
Collapse
|
78
|
p53 Signaling on Microenvironment and Its Contribution to Tissue Chemoresistance. MEMBRANES 2022; 12:membranes12020202. [PMID: 35207121 PMCID: PMC8877489 DOI: 10.3390/membranes12020202] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/06/2023]
Abstract
Chemoresistance persists as a significant, unresolved clinical challenge in many cancer types. The tumor microenvironment, in which cancer cells reside and interact with non-cancer cells and tissue structures, has a known role in promoting every aspect of tumor progression, including chemoresistance. However, the molecular determinants of microenvironment-driven chemoresistance are mainly unknown. In this review, we propose that the TP53 tumor suppressor, found mutant in over half of human cancers, is a crucial regulator of cancer cell-microenvironment crosstalk and a prime candidate for the investigation of microenvironment-specific modulators of chemoresistance. Wild-type p53 controls the secretion of factors that inhibit the tumor microenvironment, whereas altered secretion or mutant p53 interfere with p53 function to promote chemoresistance. We highlight resistance mechanisms promoted by mutant p53 and enforced by the microenvironment, such as extracellular matrix remodeling and adaptation to hypoxia. Alterations of wild-type p53 extracellular function may create a cascade of spatial amplification loops in the tumor tissue that can influence cellular behavior far from the initial oncogenic mutation. We discuss the concept of chemoresistance as a multicellular/tissue-level process rather than intrinsically cellular. Targeting p53-dependent crosstalk mechanisms between cancer cells and components of the tumor environment might disrupt the waves of chemoresistance that spread across the tumor tissue, increasing the efficacy of chemotherapeutic agents.
Collapse
|
79
|
Marques MA, de Andrade GC, Silva JL, de Oliveira GAP. Protein of a thousand faces: The tumor-suppressive and oncogenic responses of p53. Front Mol Biosci 2022; 9:944955. [PMID: 36090037 PMCID: PMC9452956 DOI: 10.3389/fmolb.2022.944955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/18/2022] [Indexed: 12/30/2022] Open
Abstract
The p53 protein is a pleiotropic regulator working as a tumor suppressor and as an oncogene. Depending on the cellular insult and the mutational status, p53 may trigger opposing activities such as cell death or survival, senescence and cell cycle arrest or proliferative signals, antioxidant or prooxidant activation, glycolysis, or oxidative phosphorylation, among others. By augmenting or repressing specific target genes or directly interacting with cellular partners, p53 accomplishes a particular set of activities. The mechanism in which p53 is activated depends on increased stability through post-translational modifications (PTMs) and the formation of higher-order structures (HOS). The intricate cell death and metabolic p53 response are reviewed in light of gaining stability via PTM and HOS formation in health and disease.
Collapse
Affiliation(s)
- Mayra A. Marques
- *Correspondence: Mayra A. Marques, ; Guilherme A. P. de Oliveira,
| | | | | | | |
Collapse
|
80
|
OUP accepted manuscript. Carcinogenesis 2022; 43:494-503. [DOI: 10.1093/carcin/bgac015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 01/08/2022] [Accepted: 01/28/2022] [Indexed: 11/12/2022] Open
|
81
|
Marei HE, Althani A, Afifi N, Hasan A, Caceci T, Pozzoli G, Morrione A, Giordano A, Cenciarelli C. p53 signaling in cancer progression and therapy. Cancer Cell Int 2021; 21:703. [PMID: 34952583 PMCID: PMC8709944 DOI: 10.1186/s12935-021-02396-8] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022] Open
Abstract
The p53 protein is a transcription factor known as the "guardian of the genome" because of its critical function in preserving genomic integrity. The TP53 gene is mutated in approximately half of all human malignancies, including those of the breast, colon, lung, liver, prostate, bladder, and skin. When DNA damage occurs, the TP53 gene on human chromosome 17 stops the cell cycle. If p53 protein is mutated, the cell cycle is unrestricted and the damaged DNA is replicated, resulting in uncontrolled cell proliferation and cancer tumours. Tumor-associated p53 mutations are usually associated with phenotypes distinct from those caused by the loss of the tumor-suppressing function exerted by wild-type p53protein. Many of these mutant p53 proteins have oncogenic characteristics, and therefore modulate the ability of cancer cells to proliferate, escape apoptosis, invade and metastasize. Because p53 deficiency is so common in human cancer, this protein is an excellent option for cancer treatment. In this review, we will discuss some of the molecular pathways by which mutant p53 proteins might perform their oncogenic activities, as well as prospective treatment methods based on restoring tumor suppressive p53 functions.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Asmaa Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Thomas Caceci
- Biomedical Sciences, Virginia Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Giacomo Pozzoli
- Pharmacology Unit, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine. Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine. Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | | |
Collapse
|
82
|
von Grabowiecki Y, Phatak V, Aschauer L, Muller PAJ. Rab11-FIP1/RCP Functions as a Major Signalling Hub in the Oncogenic Roles of Mutant p53 in Cancer. Front Oncol 2021; 11:804107. [PMID: 35757381 PMCID: PMC9231559 DOI: 10.3389/fonc.2021.804107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/03/2021] [Indexed: 12/22/2022] Open
Abstract
Rab11-FIP1 is a Rab effector protein that is involved in endosomal recycling and trafficking of various molecules throughout the endocytic compartments of the cell. The consequence of this can be increased secretion or increased membrane expression of those molecules. In general, expression of Rab11-FIP1 coincides with more tumourigenic and metastatic cell behaviour. Rab11-FIP1 can work in concert with oncogenes such as mutant p53, but has also been speculated to be an oncogene in its own right. In this perspective, we will discuss and speculate upon our observations that mutant p53 promotes Rab11-FIP1 function to not only promote invasive behaviour, but also chemoresistance by regulating a multitude of different proteins.
Collapse
Affiliation(s)
- Yannick von Grabowiecki
- Tumour Suppressors Group, Cancer Research United Kingdom (UK) Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| | - Vinaya Phatak
- Medical Research Council (MRC) Toxicology Unit, Cambridge, United Kingdom
- Avacta Life Sciences, Cambridge, United Kingdom
| | - Lydia Aschauer
- Medical Research Council (MRC) Toxicology Unit, Cambridge, United Kingdom
- Orbit Discovery, Oxford, United Kingdom
| | - Patricia A. J. Muller
- Tumour Suppressors Group, Cancer Research United Kingdom (UK) Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
- Department of Biosciences, Faculty of Science, Durham University, Durham, United Kingdom
- *Correspondence: Patricia A. J. Muller,
| |
Collapse
|
83
|
Kaida A, Iwakuma T. Regulation of p53 and Cancer Signaling by Heat Shock Protein 40/J-Domain Protein Family Members. Int J Mol Sci 2021; 22:13527. [PMID: 34948322 PMCID: PMC8706882 DOI: 10.3390/ijms222413527] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/27/2022] Open
Abstract
Heat shock proteins (HSPs) are molecular chaperones that assist diverse cellular activities including protein folding, intracellular transportation, assembly or disassembly of protein complexes, and stabilization or degradation of misfolded or aggregated proteins. HSP40, also known as J-domain proteins (JDPs), is the largest family with over fifty members and contains highly conserved J domains responsible for binding to HSP70 and stimulation of the ATPase activity as a co-chaperone. Tumor suppressor p53 (p53), the most frequently mutated gene in human cancers, is one of the proteins that functionally interact with HSP40/JDPs. The majority of p53 mutations are missense mutations, resulting in acquirement of unexpected oncogenic activities, referred to as gain of function (GOF), in addition to loss of the tumor suppressive function. Moreover, stability and levels of wild-type p53 (wtp53) and mutant p53 (mutp53) are crucial for their tumor suppressive and oncogenic activities, respectively. However, the regulatory mechanisms of wtp53 and mutp53 are not fully understood. Accumulating reports demonstrate regulation of wtp53 and mutp53 levels and/or activities by HSP40/JDPs. Here, we summarize updated knowledge related to the link of HSP40/JDPs with p53 and cancer signaling to improve our understanding of the regulation of tumor suppressive wtp53 and oncogenic mutp53 GOF activities.
Collapse
Affiliation(s)
- Atsushi Kaida
- Department of Oral Radiation Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan;
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Pediatrics, Children’s Mercy Research Institute, Kansas City, MO 64108, USA
| |
Collapse
|
84
|
García-Garrido E, Cordani M, Somoza Á. Modified Gold Nanoparticles to Overcome the Chemoresistance to Gemcitabine in Mutant p53 Cancer Cells. Pharmaceutics 2021; 13:2067. [PMID: 34959348 PMCID: PMC8703659 DOI: 10.3390/pharmaceutics13122067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 12/29/2022] Open
Abstract
Mutant p53 proteins result from missense mutations in the TP53 gene, the most mutated in human cancer, and have been described to contribute to cancer initiation and progression. Therapeutic strategies for targeting mutant p53 proteins in cancer cells are limited and have proved unsuitable for clinical application due to problems related to drug delivery and toxicity to healthy tissues. Therefore, the discovery of efficient and safe therapeutic strategies that specifically target mutant p53 remains challenging. In this study, we generated gold nanoparticles (AuNPs) chemically modified with low molecular branched polyethylenimine (bPEI) for the efficient delivery of gapmers targeting p53 mutant protein. The AuNPs formulation consists of a combination of polymeric mixed layer of polyethylene glycol (PEG) and PEI, and layer-by-layer assembly of bPEI through a sensitive linker. These nanoparticles can bind oligonucleotides through electrostatic interactions and release them in the presence of a reducing agent as glutathione. The nanostructures generated here provide a non-toxic and powerful system for the delivery of gapmers in cancer cells, which significantly downregulated mutant p53 proteins and altered molecular markers related to cell growth and apoptosis, thus overcoming chemoresistance to gemcitabine.
Collapse
Affiliation(s)
- Eduardo García-Garrido
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
| | - Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
- Unidad Asociada al Centro Nacional de Biotecnología (CSIC), Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
85
|
Zhang X, Katsakhyan L, LiVolsi VA, Roth JJ, Rassekh CH, Bagley SJ, Nasrallah MP. TP53 Mutation and Extraneural Metastasis of Glioblastoma: Insights From an Institutional Experience and Comprehensive Literature Review. Am J Surg Pathol 2021; 45:1516-1526. [PMID: 34366423 DOI: 10.1097/pas.0000000000001762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Extraneural metastases of glioblastoma (GBM), although rare, are becoming an increasingly recognized occurrence. Currently, the biological mechanism underlying this rare occurrence is not understood. To explore the potential genomic drivers of extraneural metastasis in GBM, we present the molecular features of 4 extraneural metastatic GBMs, along with a comprehensive review and analysis of previously reported cases that had available molecular characterization. In addition to our 4 cases, 42 patients from 35 publications are reviewed. To compare the molecular profiles between GBM cases with extraneural metastasis and the general GBM population, genomic data from GBM samples in The Cancer Genome Atlas (TCGA) database were also analyzed. We found that 64.5% (20/31) of the cases with extraneural metastasis that were tested for TP53 changes had at least 1 TP53 pathogenic variant detected in either 1 or both primary and metastatic tumors. In contrast, TP53 mutation was significantly less frequent in the unselected GBM from TCGA (22.6%, 56/248) (P=0.000). In addition, O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation was more common in unselected TCGA GBM cases (48.6%, 170/350) than in cases with extraneural metastasis (31.8%, 7/22), although not statistically significant. Although isocitrate dehydrogenase (IDH) mutation is a rare occurrence in high-grade astrocytomas, IDH-mutant grade 4 astrocytomas are at least as likely to metastasize as IDH wild-type GBMs; 3 metastatic cases definitively harbored an IDH1 (p.R132H) mutation in our analysis. Our findings not only provide potential biomarkers for earlier screening of extraneural metastasis, but could also suggest clues to understanding biological mechanisms underlying GBM metastasis, and for the development of therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | | | | | - Stephen J Bagley
- Hematology Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
86
|
Yin Y, Jiang Z, Fu J, Li Y, Fang C, Yin X, Chen Y, Chen N, Li J, Ji Y, Su X, Qiu M, Huang W, Zhang B, Deng H, Dai L. Choline-induced SLC5A7 impairs colorectal cancer growth by stabilizing p53 protein. Cancer Lett 2021; 525:55-66. [PMID: 34562520 DOI: 10.1016/j.canlet.2021.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023]
Abstract
The members of the solute carrier (SLC) superfamily are vital membrane transporters in human cells. In the present study, we determine the expression and function of SLC5 family members in colorectal cancer (CRC). Expression analysis based on The Cancer Genome Atlas database and potential clinical relation analysis based on the Oncomine database indicate that SLC5A7 is downregulated and is predicted to correlate with the staging, and prognosis response of CRC. Additional results demonstrate that SLC5A7 is downregulated and correlates with good prognosis in patients with CRC. Ectopic expression of SLC5A7 either by overexpression, or uptake of choline efficiently inhibits CRC growth. Examination of the molecular mechanism reveals that SLC5A7 promotes p53 protein expression by directly interacting with and modifying p53 and disrupting the interaction between p53 and MDM2 in wild type p53 CRC cells. Our findings establish the clear correlation between SLC5A7 and tumour growth, providing a novel potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Yuan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Zhiyuan Jiang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Jiamei Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yang Li
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China; Department of Gastrointestinal Surgery, Guang'an People's Hospital, Guang'an, Sichuan, 638500, People's Republic of China
| | - Chao Fang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ye Chen
- Department of Medical Oncology, Cancer Center, The State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Junshu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanhong Ji
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China
| | - Meng Qiu
- Department of Medical Oncology, Cancer Center, The State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
87
|
Piipponen M, Riihilä P, Nissinen L, Kähäri VM. The Role of p53 in Progression of Cutaneous Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13184507. [PMID: 34572732 PMCID: PMC8466956 DOI: 10.3390/cancers13184507] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Skin cancers are the most common types of cancer worldwide, and their incidence is increasing. Melanoma, basal cell carcinoma (BCC), and cutaneous squamous cell carcinoma (cSCC) are the three major types of skin cancer. Melanoma originates from melanocytes, whereas BCC and cSCC originate from epidermal keratinocytes and are therefore called keratinocyte carcinomas. Chronic exposure to ultraviolet radiation (UVR) is a common risk factor for skin cancers, but they differ with respect to oncogenic mutational profiles and alterations in cellular signaling pathways. cSCC is the most common metastatic skin cancer, and it is associated with poor prognosis in the advanced stage. An important early event in cSCC development is mutation of the TP53 gene and inactivation of the tumor suppressor function of the tumor protein 53 gene (TP53) in epidermal keratinocytes, which then leads to accumulation of additional oncogenic mutations. Additional genomic and proteomic alterations are required for the progression of premalignant lesion, actinic keratosis, to invasive and metastatic cSCC. Recently, the role of p53 in the invasion of cSCC has also been elucidated. In this review, the role of p53 in the progression of cSCC and as potential new therapeutic target for cSCC will be discussed.
Collapse
Affiliation(s)
- Minna Piipponen
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland; (M.P.); (P.R.); (L.N.)
- FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Center for Molecular Medicine, Department of Medicine Solna, Dermatology and Venereology Division, Karolinska Institute, 17176 Stockholm, Sweden
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland; (M.P.); (P.R.); (L.N.)
- FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland; (M.P.); (P.R.); (L.N.)
- FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Hämeentie 11 TE6, FI-20520 Turku, Finland; (M.P.); (P.R.); (L.N.)
- FICAN West Cancer Centre Research Laboratory, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Correspondence: ; Tel.: +358-2-3131600
| |
Collapse
|
88
|
Xiong YJ, Zhu Y, Liu YL, Zhao YF, Shen X, Zuo WQ, Lin F, Liang ZQ. P300 Participates in Ionizing Radiation-Mediated Activation of Cathepsin L by Mutant p53. J Pharmacol Exp Ther 2021; 378:276-286. [PMID: 34253647 DOI: 10.1124/jpet.121.000639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Our previous studies have shown that cathepsin L (CTSL) is involved in the ability of tumors to resist ionizing radiation (IR), but the specific mechanisms responsible for this remain unknown. We report here that mutant p53 (mut-p53) is involved in IR-induced transcription of CTSL. We found that irradiation caused activation of CTSL in mut-p53 cell lines, whereas there was almost no activation in p53 wild-type cell lines. Additionally, luciferase reporter gene assay results demonstrated that IR induced the p53 binding region on the CTSL promoter. We further demonstrated that the expression of p300 and early growth response factor-1 (Egr-1) was upregulated in mut-p53 cell lines after IR treatment. Accordingly, the expression of Ac-H3, Ac-H4, AcH3K9 was upregulated after IR treatment in mut-p53 cell lines, whereas histone deacetylase (HDAC) 4 and HDAC6 were reciprocally decreased. Moreover, knockdown of either Egr-1 or p300 abolished the binding of mut-p53 to the promoter of CTSL. Chromatin immunoprecipitation assay results showed that the IR-activated transcription of CTSL was dependent on p300. To further delineate the clinical relevance of interactions between Egr-1/p300, mut-p53, and CTSL, we accessed primary tumor samples to evaluate the relationships between mut-p53, CTSL, and Egr-1/p300 ex vivo. The results support the notion that mut-p53 is correlated with CTSL transcription involving the Egr-1/p300 pathway. Taken together, the results of our study revealed that p300 is an important target in the process of IR-induced transcription of CTSL, which confirms that CTSL participates in mut-p53 gain-of-function. SIGNIFICANCE STATEMENT: Transcriptional activation of cathepsin L by ionizing radiation required the involvement of mutated p53 and Egr-1/p300. Interference with Egr-1 or p300 could inhibit the expression of cathepsin L induced by ionizing radiation. The transcriptional activation of cathepsin L by p300 may be mediated by p53 binding sites on the cathepsin L promoter.
Collapse
Affiliation(s)
- Ya-Jie Xiong
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Ying Zhu
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Ya-Li Liu
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Yi-Fan Zhao
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Xiao Shen
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Wen-Qing Zuo
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Fang Lin
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| | - Zhong-Qin Liang
- Department of Pharmacology, Soochow University, Suzhou, China (Y.X., Y.L., Y.Zha., X.S., Q.Z., F.L., Z.L.), and Department of Pharmacy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China (Y.Zhu)
| |
Collapse
|
89
|
Redman-Rivera LN, Shaver TM, Jin H, Marshall CB, Schafer JM, Sheng Q, Hongo RA, Beckermann KE, Wheeler FC, Lehmann BD, Pietenpol JA. Acquisition of aneuploidy drives mutant p53-associated gain-of-function phenotypes. Nat Commun 2021; 12:5184. [PMID: 34465782 PMCID: PMC8408227 DOI: 10.1038/s41467-021-25359-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
p53 is mutated in over half of human cancers. In addition to losing wild-type (WT) tumor-suppressive function, mutant p53 proteins are proposed to acquire gain-of-function (GOF) activity, leading to novel oncogenic phenotypes. To study mutant p53 GOF mechanisms and phenotypes, we genetically engineered non-transformed and tumor-derived WT p53 cell line models to express endogenous missense mutant p53 (R175H and R273H) or to be deficient for p53 protein (null). Characterization of the models, which initially differed only by TP53 genotype, revealed that aneuploidy frequently occurred in mutant p53-expressing cells. GOF phenotypes occurred clonally in vitro and in vivo, were independent of p53 alteration and correlated with increased aneuploidy. Further, analysis of outcome data revealed that individuals with aneuploid-high tumors displayed unfavorable prognoses, regardless of the TP53 genotype. Our results indicate that genetic variation resulting from aneuploidy accounts for the diversity of previously reported mutant p53 GOF phenotypes.
Collapse
Affiliation(s)
- Lindsay N. Redman-Rivera
- grid.152326.10000 0001 2264 7217Department of Biochemistry, Vanderbilt University, Nashville, TN USA
| | - Timothy M. Shaver
- grid.152326.10000 0001 2264 7217Department of Biochemistry, Vanderbilt University, Nashville, TN USA ,Inscripta, Inc, Boulder, CO USA
| | - Hailing Jin
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - Clayton B. Marshall
- grid.152326.10000 0001 2264 7217Department of Biochemistry, Vanderbilt University, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| | - Johanna M. Schafer
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA ,grid.261331.40000 0001 2285 7943Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH USA
| | - Quanhu Sheng
- grid.412807.80000 0004 1936 9916Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Rachel A. Hongo
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Kathryn E. Beckermann
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Ferrin C. Wheeler
- grid.412807.80000 0004 1936 9916Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Brian D. Lehmann
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jennifer A. Pietenpol
- grid.152326.10000 0001 2264 7217Department of Biochemistry, Vanderbilt University, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
90
|
Zhang C, Liu J, Xu D, Zhang T, Hu W, Feng Z. Gain-of-function mutant p53 in cancer progression and therapy. J Mol Cell Biol 2021; 12:674-687. [PMID: 32722796 PMCID: PMC7749743 DOI: 10.1093/jmcb/mjaa040] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/28/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
p53 is a key tumor suppressor, and loss of p53 function is frequently a prerequisite for cancer development. The p53 gene is the most frequently mutated gene in human cancers; p53 mutations occur in >50% of all human cancers and in almost every type of human cancers. Most of p53 mutations in cancers are missense mutations, which produce the full-length mutant p53 (mutp53) protein with only one amino acid difference from wild-type p53 protein. In addition to loss of the tumor-suppressive function of wild-type p53, many mutp53 proteins acquire new oncogenic activities independently of wild-type p53 to promote cancer progression, termed gain-of-function (GOF). Mutp53 protein often accumulates to very high levels in cancer cells, which is critical for its GOF. Given the high mutation frequency of the p53 gene and the GOF activities of mutp53 in cancer, therapies targeting mutp53 have attracted great interest. Further understanding the mechanisms underlying mutp53 protein accumulation and GOF will help develop effective therapies treating human cancers containing mutp53. In this review, we summarize the recent advances in the studies on mutp53 regulation and GOF as well as therapies targeting mutp53 in human cancers.
Collapse
Affiliation(s)
- Cen Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Dandan Xu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Tianliang Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| |
Collapse
|
91
|
Lai ZY, Tsai KY, Chang SJ, Chuang YJ. Gain-of-Function Mutant TP53 R248Q Overexpressed in Epithelial Ovarian Carcinoma Alters AKT-Dependent Regulation of Intercellular Trafficking in Responses to EGFR/MDM2 Inhibitor. Int J Mol Sci 2021; 22:ijms22168784. [PMID: 34445495 PMCID: PMC8395913 DOI: 10.3390/ijms22168784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 01/27/2023] Open
Abstract
As the most common gene mutation found in cancers, p53 mutations are detected in up to 96% of high-grade serous ovarian carcinoma (HGSOC). Meanwhile, mutant p53 overexpression is known to drive oncogenic phenotypes in cancer patients and to sustain the activation of EGFR signaling. Previously, we have demonstrated that the combined inhibition of EGFR and MDM2-p53 pathways, by gefitinib and JNJ-26854165, exerts a strong synergistic lethal effect on HGSOC cells. In this study, we investigated whether the gain-of-function p53 mutation (p53R248Q) overexpression could affect EGFR-related signaling and the corresponding drug inhibition outcome in HGSOC. The targeted inhibition responses of gefitinib and JNJ-26854165, in p53R248Q-overexpressing cells, were extensively evaluated. We found that the phosphorylation of AKT increased when p53R248Q was transiently overexpressed. Immunocytochemistry analysis further showed that upon p53R248Q overexpression, several AKT-related regulatory proteins translocated in unique intracellular patterns. Subsequent analysis revealed that, under the combined inhibition of gefitinib and JNJ-26854165, the cytonuclear trafficking of EGFR and MDM2 was disrupted. Next, we analyzed the gefitinib and JNJ-26854165 responses and found differential sensitivity to the single- or combined-drug inhibitions in p53R248Q-overexpressing cells. Our findings suggested that the R248Q mutation of p53 in HGSOC caused significant changes in signaling protein function and trafficking, under EGFR/MDM2-targeted inhibition. Such knowledge could help to advance our understanding of the role of mutant p53 in ovarian carcinoma and to improve the prognosis of patients receiving EGFR/MDM2-targeted therapies.
Collapse
Affiliation(s)
- Zih-Yin Lai
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Z.-Y.L.); (K.-Y.T.)
| | - Kai-Yun Tsai
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Z.-Y.L.); (K.-Y.T.)
| | - Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu 30071, Taiwan
- Correspondence: (S.-J.C.); (Y.-J.C.); Tel.: +886-3-6119595 (S.-J.C.); +886-3-5742764 (Y.-J.C.); Fax: +886-3-6110900 (S.-J.C.); +886-3-5715934 (Y.-J.C.)
| | - Yung-Jen Chuang
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Z.-Y.L.); (K.-Y.T.)
- Correspondence: (S.-J.C.); (Y.-J.C.); Tel.: +886-3-6119595 (S.-J.C.); +886-3-5742764 (Y.-J.C.); Fax: +886-3-6110900 (S.-J.C.); +886-3-5715934 (Y.-J.C.)
| |
Collapse
|
92
|
Xia Y, Li X, Sun W. Applications of Recombinant Adenovirus-p53 Gene Therapy for Cancers in the Clinic in China. Curr Gene Ther 2021; 20:127-141. [PMID: 32951572 DOI: 10.2174/1566523220999200731003206] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 01/30/2023]
Abstract
Suppression of TP53 function is nearly ubiquitous in human cancers, and a significant fraction of cancers have mutations in the TP53 gene itself. Therefore, the wild-type TP53 gene has become an important target gene for transformation research of cancer gene therapy. In 2003, the first anti-tumor gene therapy drug rAd-p53 (recombinant human p53 adenovirus), trade name Gendicine™, was approved by the China Food and Drug Administration (CFDA) for treatment of head and neck squamous cell carcinoma (HNSCC) in combination with radiotherapy. The recombinant human TP53 gene is delivered into cancer cells by an adenovirus vector constructed to express the functional p53 protein. Although the only currently approved used of Gendicine is in combination with radiotherapy for treatment of HNSCC, clinical studies have been carried out for more than 20 other applications of Gendicine in treating cancer, including treatment of advanced lung cancer, advanced liver cancer, malignant gynecological tumors, and soft tissue sarcomas. Currently more than 30,000 patients have been treated with Gendicine. This review provides an overview of the clinical applications of Gendicine in China. We summarize a total of 48 studies with 2,561 patients with solid tumors, including 34 controlled clinical studies and 14 open clinical studies, i.e., clinical studies without a control group. There are 11 studies for head and neck cancer, 10 for liver cancer, 6 for malignant gynecological tumors, 4 for non-small cell lung cancer, 4 for soft tissue sarcoma, 4 for malignant effusion, 2 for gastrointestinal tumors, and 7 for other types of cancer. In all the reported clinical studies, the most common side effect was self-limited fever. Intratumoral injection and intra-arterial infusion were the most common routes of administration. Overall, Gendicine combined with chemotherapy, radiotherapy, or other conventional treatment regimens demonstrated significantly higher response rates compared to standard therapies alone. Some of the published studies also showed that Gendicine combination regimens demonstrated longer progression-free survival times than conventional treatments alone. To date, Gendicine has been clinically used in China for treatment of cancers other than HNSCC for more than ten years, mainly for patients with advanced or unresectable malignant tumors. However, the establishment of standard treatment regimens using TP53 gene therapy is still needed in order to advance its use in clinical practice.
Collapse
Affiliation(s)
- Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Xiuqin Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Wei Sun
- Radiology Department, Shengjing Hospital of China Medical University, Sanhao, China
| |
Collapse
|
93
|
Chiang YT, Chien YC, Lin YH, Wu HH, Lee DF, Yu YL. The Function of the Mutant p53-R175H in Cancer. Cancers (Basel) 2021; 13:4088. [PMID: 34439241 PMCID: PMC8391618 DOI: 10.3390/cancers13164088] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
Wild-type p53 is known as "the guardian of the genome" because of its function of inducing DNA repair, cell-cycle arrest, and apoptosis, preventing the accumulation of gene mutations. TP53 is highly mutated in cancer cells and most TP53 hotspot mutations are missense mutations. Mutant p53 proteins, encoded by these hotspot mutations, lose canonical wild-type p53 functions and gain functions that promote cancer development, including promoting cancer cell proliferation, migration, invasion, initiation, metabolic reprogramming, angiogenesis, and conferring drug resistance to cancer cells. Among these hotspot mutations, p53-R175H has the highest occurrence. Although losing the transactivating function of the wild-type p53 and prone to aggregation, p53-R175H gains oncogenic functions by interacting with many proteins. In this review, we summarize the gain of functions of p53-R175H in different cancer types, the interacting proteins of p53-R175H, and the downstream signaling pathways affected by p53-R175H to depict a comprehensive role of p53-R175H in cancer development. We also summarize treatments that target p53-R175H, including reactivating p53-R175H with small molecules that can bind to p53-R175H and alter it into a wild-type-like structure, promoting the degradation of p53-R175H by targeting heat-shock proteins that maintain the stability of p53-R175H, and developing immunotherapies that target the p53-R175H-HLA complex presented by tumor cells.
Collapse
Affiliation(s)
- Yen-Ting Chiang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
| | - Yi-Chung Chien
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
- Program for Translational Medicine, China Medical University, Taichung 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
- Drug Development Center, Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Yu-Heng Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
| | - Hui-Hsuan Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yung-Luen Yu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-T.C.); (Y.-C.C.); (Y.-H.L.); (H.-H.W.)
- Program for Translational Medicine, China Medical University, Taichung 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
- Drug Development Center, Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
94
|
Mammarella E, Zampieri C, Panatta E, Melino G, Amelio I. NUAK2 and RCan2 participate in the p53 mutant pro-tumorigenic network. Biol Direct 2021; 16:11. [PMID: 34348766 PMCID: PMC8335924 DOI: 10.1186/s13062-021-00296-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/29/2021] [Indexed: 02/04/2023] Open
Abstract
Most inactivating mutations in TP53 gene generates neomorphic forms of p53 proteins that experimental evidence and clinical observations suggest to exert gain-of-function effects. While massive effort has been deployed in the dissection of wild type p53 transcriptional programme, p53 mutant pro-tumorigenic gene network is still largely elusive. To help dissecting the molecular basis of p53 mutant GOF, we performed an analysis of a fully annotated genomic and transcriptomic human pancreatic adenocarcinoma to select candidate players of p53 mutant network on the basis their differential expression between p53 mutant and p53 wild-type cohorts and their prognostic value. We identified NUAK2 and RCan2 whose p53 mutant GOF-dependent regulation was further validated in pancreatic cancer cellular model. Our data demonstrated that p53R270H can physically bind RCan2 gene locus in regulatory regions corresponding to the chromatin permissive areas where known binding partners of p53 mutant, such as p63 and Srebp, bind. Overall, starting from clinically relevant data and progressing into experimental validation, our work suggests NUAK2 and RCan2 as novel candidate players of the p53 mutant pro-tumorigenic network whose prognostic and therapeutic interest might attract future studies.
Collapse
Affiliation(s)
- Eleonora Mammarella
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlotta Zampieri
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Emanuele Panatta
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
- School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
95
|
D-Propranolol Impairs EGFR Trafficking and Destabilizes Mutant p53 Counteracting AKT Signaling and Tumor Malignancy. Cancers (Basel) 2021; 13:cancers13143622. [PMID: 34298835 PMCID: PMC8305715 DOI: 10.3390/cancers13143622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Cancer progression is frequently driven by altered functions of EGFR belonging to the tyrosine-kinase family of growth factor receptors and by the transcription factor p53, which is called the “genome guardian”. We report that D-Propranolol, previously used for other purposes in human patients, has antitumor effects involving a redistribution of cell surface EGFR to intracellular compartments and degradation of gain-of-function mutants of p53 (GOF-mutp53). These effects can be seen in cancer cell lines expressing EGFR and GOF-mutp53 and are reproduced in vivo, reducing tumor growth and prolonging survival of xenografted mice. D-Propranolol is proposed as a prototype drug for a new strategy against highly aggressive EGFR- and mutp53-expressing tumors. Abstract Cancer therapy may be improved by the simultaneous interference of two or more oncogenic pathways contributing to tumor progression and aggressiveness, such as EGFR and p53. Tumor cells expressing gain-of-function (GOF) mutants of p53 (mutp53) are usually resistant to EGFR inhibitors and display invasive migration and AKT-mediated survival associated with enhanced EGFR recycling. D-Propranolol (D-Prop), the non-beta blocker enantiomer of propranolol, was previously shown to induce EGFR internalization through a PKA inhibitory pathway that blocks the recycling of the receptor. Here, we first show that D-Prop decreases the levels of EGFR at the surface of GOF mutp53 cells, relocating the receptor towards recycling endosomes, both in the absence of ligand and during stimulation with high concentrations of EGF or TGF-α. D-Prop also inactivates AKT signaling and reduces the invasive migration and viability of these mutp53 cells. Unexpectedly, mutp53 protein, which is stabilized by interaction with the chaperone HSP90 and mediates cell oncogenic addiction, becomes destabilized after D-Prop treatment. HSP90 phosphorylation by PKA and its interaction with mutp53 are decreased by D-Prop, releasing mutp53 towards proteasomal degradation. Furthermore, a single daily dose of D-Prop reproduces most of these effects in xenografts of aggressive gallbladder cancerous G-415 cells expressing GOF R282W mutp53, resulting in reduced tumor growth and extended mice survival. D-Prop then emerges as an old drug endowed with a novel therapeutic potential against EGFR- and mutp53-driven tumor traits that are common to a large variety of cancers.
Collapse
|
96
|
Nassif EF, Auclin E, Bahleda R, Honoré C, Mir O, Dumont S, Mery B, Hodroj K, Brahmi M, Trédan O, Ray-Coquard I, Blay JY, Massard C, Le Cesne A, Dufresne A. TP53 Mutation as a Prognostic and Predictive Marker in Sarcoma: Pooled Analysis of MOSCATO and ProfiLER Precision Medicine Trials. Cancers (Basel) 2021; 13:3362. [PMID: 34282771 PMCID: PMC8268242 DOI: 10.3390/cancers13133362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: locally resected high-grade sarcomas relapse in 40% of cases. There is no prognostic or predictive genomic marker for response to peri-operative chemotherapy. (2) Methods: MOSCATO and ProfiLER are pan-tumor prospective precision medicine trials for advanced tumors. Molecular analysis in both trials comprised targeted next-generation sequencing and comparative genomic hybridization array. We investigated if molecular alterations identified in these trials in sarcomas were associated with disease-free survival (DFS) and response to anthracyclines. (3) Results: this analysis included 215 sarcomas, amongst which 53 leiomyosarcomas, 27 rhabdomyosarcomas, 20 undifferentiated pleomorphic sarcomas, and 17 liposarcomas. The most frequently altered gene was TP53 (46 mutations and eight deletions). There were 149 surgically resected localized sarcomas. Median DFS in TP53 wild type (WT), deleted, and mutated sarcomas was 16, 10, and 10 months, respectively (p = 0.028; deletions: HR = 1.55; 95% CI = 0.75-3.19; mutations: HR = 1.70; 95%CI = 1.13-2.64). In multivariate analysis, TP53 mutations remained associated with shorter DFS (p = 0.027; HR = 2.30; 95%CI = 1.10-4.82). There were 161 localized and advanced sarcomas evaluable for response to anthracyclines. Objective response rates were 35% and 55% in TP53 WT and mutated sarcomas, respectively (OR = 2.24; 95%CI = 1.01-5.03; p = 0.05). In multivariate analysis, TP53 mutations remained associated with increased response (OR = 3.24; 95%CI = 1.30-8.45; p = 0.01). (4) Conclusions: TP53 mutations are associated with shorter DFS and increased response to anthracyclines. Post-validation, these findings could assist in decision-making for peri-operative treatments.
Collapse
Affiliation(s)
- Elise F. Nassif
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Edouard Auclin
- Oncology Department, Hopital Européen Georges Pompidou, 75015 Paris, France;
| | - Rastilav Bahleda
- DITEP (Département d’Innovation Therapeutique et d’Essais Précoces), Drug Development Department, Gustave Roussy, 94805 Villejuif, France; (R.B.); (C.M.)
| | - Charles Honoré
- Surgical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Olivier Mir
- Ambulatory Cancer Care Department, Gustave Roussy, 94805 Villejuif, France;
| | - Sarah Dumont
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Benoite Mery
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Khalil Hodroj
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Mehdi Brahmi
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Olivier Trédan
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Isabelle Ray-Coquard
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Jean-Yves Blay
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Christophe Massard
- DITEP (Département d’Innovation Therapeutique et d’Essais Précoces), Drug Development Department, Gustave Roussy, 94805 Villejuif, France; (R.B.); (C.M.)
| | - Axel Le Cesne
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Armelle Dufresne
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| |
Collapse
|
97
|
Isermann T, Şener ÖÇ, Stender A, Klemke L, Winkler N, Neesse A, Li J, Wegwitz F, Moll UM, Schulz-Heddergott R. Suppression of HSF1 activity by wildtype p53 creates a driving force for p53 loss-of-heterozygosity. Nat Commun 2021; 12:4019. [PMID: 34188043 PMCID: PMC8242083 DOI: 10.1038/s41467-021-24064-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
The vast majority of human tumors with p53 mutations undergo loss of the remaining wildtype p53 allele (loss-of-heterozygosity, p53LOH). p53LOH has watershed significance in promoting tumor progression. However, driving forces for p53LOH are poorly understood. Here we identify the repressive WTp53-HSF1 axis as one driver of p53LOH. We find that the WTp53 allele in AOM/DSS chemically-induced colorectal tumors (CRC) of p53R248Q/+ mice retains partial activity and represses heat-shock factor 1 (HSF1), the master regulator of the proteotoxic stress response (HSR) that is ubiquitously activated in cancer. HSR is critical for stabilizing oncogenic proteins including mutp53. WTp53-retaining CRC tumors, tumor-derived organoids and human CRC cells all suppress the tumor-promoting HSF1 program. Mechanistically, retained WTp53 activates CDKN1A/p21, causing cell cycle inhibition and suppression of E2F target MLK3. MLK3 links cell cycle with the MAPK stress pathway to activate the HSR response. In p53R248Q/+ tumors WTp53 activation by constitutive stress represses MLK3, thereby weakening the MAPK-HSF1 response necessary for tumor survival. This creates selection pressure for p53LOH which eliminates the repressive WTp53-MAPK-HSF1 axis and unleashes tumor-promoting HSF1 functions, inducing mutp53 stabilization enabling invasion.
Collapse
Affiliation(s)
- Tamara Isermann
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Özge Çiçek Şener
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Adrian Stender
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Luisa Klemke
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadine Winkler
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany
| | - Jinyu Li
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Ute M Moll
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
98
|
Hussain Z, Nigri J, Tomasini R. The Cellular and Biological Impact of Extracellular Vesicles in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13123040. [PMID: 34207163 PMCID: PMC8235245 DOI: 10.3390/cancers13123040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The increased incidence and global failure of ongoing therapies project pancreatic cancer as the second deadliest cancer worldwide. While our knowledge of pancreatic cancer cells’ abilities and specificities has drastically improved based on multi-scaled omics, one must consider that much more remains to be uncovered on the role and impact of stromal cells and the established network of communication with tumor cells. This review article discusses how tumor cells communicate with the various cells composing the stroma and its implication in tumor cells’ abilities, PDA (pancreatic ductal adenocarcinoma) carcinogenesis and therapeutic response. We will focus on extracellular vesicles-mediated crosstalk and how this multifaceted dialogue impacts both cellular compartments and its subsequent impact on PDA biology. Abstract Deciphering the interactions between tumor and stromal cells is a growing field of research to improve pancreatic cancer-associated therapies and patients’ care. Indeed, while accounting for 50 to 90% of the tumor mass, many pieces of evidence reported that beyond their structural role, the non-tumoral cells composing the intra-tumoral microenvironment influence tumor cells’ proliferation, metabolism, cell death and resistance to therapies, among others. Simultaneously, tumor cells can influence non-tumoral neighboring or distant cells in order to shape a tumor-supportive and immunosuppressive environment as well as influencing the formation of metastatic niches. Among intercellular modes of communication, extracellular vesicles can simultaneously transfer the largest variety of signals and were recently reported as key effectors of cell–cell communication in pancreatic cancer, from its development to its evolution as well as its ability to resist available treatments. This review focuses on extracellular vesicles-mediated communication between different cellular components of pancreatic tumors, from the modulation of cellular activities and abilities to their biological and physiological relevance. Taking into consideration the intra-tumoral microenvironment and its extracellular-mediated crosstalk as main drivers of pancreatic cancer development should open up new therapeutic windows.
Collapse
|
99
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
100
|
Bhatta B, Luz I, Krueger C, Teo FX, Lane DP, Sabapathy K, Cooks T. Cancer Cells Shuttle Extracellular Vesicles Containing Oncogenic Mutant p53 Proteins to the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13122985. [PMID: 34203762 PMCID: PMC8232660 DOI: 10.3390/cancers13122985] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary In addition to the classical cell-to-cell communication patterns, extracellular vesicles (EVs) are instrumental in conveying molecular messages across cell types and have the potential to mediate changes at a tissue level. Since it is now appreciated that carcinomas are fundamentally reliant on two-way communication with activated cells in the tumor microenvironment, elucidating the roles of EVs exchange and of the cargo that is transferred is essential to obtain a thorough understanding of tumor progression. This study reveals that mutant p53 proteins—the result of the most frequent mutated gene in human cancer—are packed into EVs and delivered to neighboring cells with the potential to reprogram immune cells and subsequently establish a positive feedback loop that will enhance tumor progression. This non-cell autonomous role of mutant p53 is evidence of an extra layer of communication that is orchestrated by smaller vesicles that transfer oncogenic elements between cellular entities. Building on the foundation of our work on mutant p53, future studies may aim to characterize the potential activation of additional oncogenes, thus opening new paths of research at the interface of extracellular vesicles, cancer, and evolution. Abstract Extracellular vesicles (EVs) shed by cancer cells play a major role in mediating the transfer of molecular information by reprogramming the tumor microenvironment (TME). TP53 (encoding the p53 protein) is the most mutated gene across many cancer types. Mutations in TP53 not only result in the loss of its tumor-suppressive properties but also results in the acquisition of novel gain-of-functions (GOF) that promote the growth of cancer cells. Here, we demonstrate that GOF mutant p53 proteins can be transferred via EVs to neighboring cancer cells and to macrophages, thus modulating them to release tumor supportive cytokines. Our data from pancreatic, lung, and colon carcinoma cell lines demonstrate that the mutant p53 protein can be selectively sorted into EVs. More specifically, mutant p53 proteins in EVs can be taken up by neighboring cells and mutant p53 expression is found in non-tumor cells in both human cancers and in non-human tissues in human xenografts. Our findings shed light on the intricate methods in which specific GOF p53 mutants can promote oncogenic mechanisms by reprogramming and then recruiting non-cancerous elements for tumor progression.
Collapse
Affiliation(s)
- Bibek Bhatta
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.B.); (I.L.)
| | - Ishai Luz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.B.); (I.L.)
| | - Christian Krueger
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore; (C.K.); (F.X.T.); (K.S.)
| | - Fanny Xueting Teo
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore; (C.K.); (F.X.T.); (K.S.)
| | - David P. Lane
- p53 Laboratory (p53Lab), Agency for Science, Technology, and Research (A*STAR), Singapore 138648, Singapore;
| | - Kanaga Sabapathy
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore; (C.K.); (F.X.T.); (K.S.)
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (B.B.); (I.L.)
- Correspondence:
| |
Collapse
|