150 results on '"G. Selivanova"'
Search Results
52. [Clinical aspects and diagnosis of chronic subdural hematomas]
- Author
-
N A, Popov, I A, Zakharov, and I G, Selivanova
- Subjects
Adult ,Diagnosis, Differential ,Male ,Hematoma, Subdural ,Chronic Disease ,Methods ,Humans ,Female ,Intracranial Embolism and Thrombosis ,Middle Aged - Published
- 1969
53. The Antioxidant Properties of Pectin Fractions Isolated from Vegetables Using a Simulated Gastric Fluid
- Author
-
V. Smirnov, Vasily, V. Golovchenko, Victoria, V. Vityazev, Fedor, A. Patova, Olga, Yu. Selivanov, Nikolay, G. Selivanova, Olga, and V. Popov, Sergey
- Abstract
The antioxidant properties of vegetable pectin fractions against intraluminal reactive oxygen species were elucidated in vitro in conjunction with their structural features. The pectin fractions were isolated using a simulated gastric fluid (pH 1.5, pepsin 0.5 g/L, 37°C, 4 h) from fresh white cabbage, carrot, onion, and sweet pepper. The fraction from onion was found to inhibit the production of superoxide radicals by inhibiting the xanthine oxidase. The high molecular weight of onion pectin and a large number of galactose residues in its side chains appeared to participate in interaction with xanthine oxidase. All the isolated pectic polysaccharides were found to be associated with protein (2–9%) and phenolics (0.5–0.7%) as contaminants; these contaminants were shown to be responsible for the antioxidant effect of vegetable pectin fractions against the hydroxyl and 1,1-diphenyl-2-picrylhydrazyl radicals.
- Published
- 2017
- Full Text
- View/download PDF
54. Translating p53-based therapies for cancer into the clinic.
- Author
-
Peuget S, Zhou X, and Selivanova G
- Subjects
- Humans, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53, Mutation, Neoplasms drug therapy, Neoplasms genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Inactivation of the most important tumour suppressor gene TP53 occurs in most, if not all, human cancers. Loss of functional wild-type p53 is achieved via two main mechanisms: mutation of the gene leading to an absence of tumour suppressor activity and, in some cases, gain-of-oncogenic function; or inhibition of the wild-type p53 protein mediated by overexpression of its negative regulators MDM2 and MDMX. Because of its high potency as a tumour suppressor and the dependence of at least some established tumours on its inactivation, p53 appears to be a highly attractive target for the development of new anticancer drugs. However, p53 is a transcription factor and therefore has long been considered undruggable. Nevertheless, several innovative strategies have been pursued for targeting dysfunctional p53 for cancer treatment. In mutant p53-expressing tumours, the predominant strategy is to restore tumour suppressor function with compounds acting either in a generic manner or otherwise selective for one or a few specific p53 mutations. In addition, approaches to deplete mutant p53 or to target vulnerabilities created by mutant p53 expression are currently under development. In wild-type p53 tumours, the major approach is to protect p53 from the actions of MDM2 and MDMX by targeting these negative regulators with inhibitors. Although the results of at least some clinical trials of MDM2 inhibitors and mutant p53-restoring compounds are promising, none of the agents has yet been approved by the FDA. Alternative strategies, based on a better understanding of p53 biology, the mechanisms of action of compounds and treatment regimens as well as the development of new technologies are gaining interest, such as proteolysis-targeting chimeras for MDM2 degradation. Other approaches are taking advantage of the progress made in immune-based therapies for cancer. In this Review, we present these ongoing clinical trials and emerging approaches to re-evaluate the current state of knowledge of p53-based therapies for cancer., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
55. MDM2-PROTAC versus MDM2 Inhibitors: Beyond p53 Reactivation.
- Author
-
Peuget S and Selivanova G
- Subjects
- Humans, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Proto-Oncogene Proteins c-mdm2 metabolism, Triple Negative Breast Neoplasms, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Summary: In this issue of Cancer Discovery, Adams and colleagues present the discovery of a potent PROTAC, MDM2 degrader, which activates wild-type p53 leading to cancer cell death. Importantly, in a number of in vitro and in vivo experiments, the authors show that the depletion of MDM2 by PROTAC kills p53-mutant or p53-null cancer cells. See related article by Adams et al., p. 1210 (5)., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
56. Mutant p53 gain of function mediates cancer immune escape that is counteracted by APR-246.
- Author
-
Zhou X, Santos GS, Zhan Y, Oliveira MMS, Rezaei S, Singh M, Peuget S, Westerberg LS, Johnsen JI, and Selivanova G
- Subjects
- Mice, Animals, Gain of Function Mutation, Interferons genetics, Interferons metabolism, Inflammation genetics, Tumor Microenvironment genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Neoplasms genetics
- Abstract
Background: p53 mutants contribute to the chronic inflammatory tumour microenvironment (TME). In this study, we address the mechanism of how p53 mutants lead to chronic inflammation in tumours and how to transform it to restore cancer immune surveillance., Methods: Our analysis of RNA-seq data from The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) project revealed that mutant p53 (mtp53) cancers correlated with chronic inflammation. We used cell-based assays and a mouse model to discover a novel gain of function of mtp53 and the effect of the mtp53 reactivating compound APR-246 on the anti-tumour immune response., Results: We found that tumour samples from patients with breast carcinoma carrying mtp53 showed elevated Interferon (IFN) signalling, Tumour Inflammation Signature (TIS) score and infiltration of CD8+ T cells compared to wild type p53 (wtp53) tumours. We showed that the expression of IFN and immune checkpoints were elevated in tumour cells in a mtp53-dependent manner, suggesting a novel gain of function. Restoration of wt function to mtp53 by APR-246 induced the expression of endogenous retroviruses, IFN signalling and repressed immune checkpoints. Moreover, APR-246 promoted CD4+ T cells infiltration and IFN signalling and prevented CD8+ T cells exhaustion within the TME in vivo., Conclusions: Breast carcinomas with mtp53 displayed enhanced inflammation. APR-246 boosted the interferon response or represses immune checkpoints in p53 mutant tumour cells, and restores cancer immune surveillance in vivo., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
57. Decreased DNA Damage and Improved p53 Specificity of RITA Analogs.
- Author
-
Zhan Y, Zhou X, Peuget S, Singh M, Peyser BD, Fan Z, and Selivanova G
- Subjects
- Apoptosis, Cell Line, Tumor, DNA Damage, Furans pharmacology, Humans, Sulfotransferases genetics, RNA Polymerase II, Tumor Suppressor Protein p53 genetics
- Abstract
Reactivation of p53 tumor-suppressor function by small molecules is an attractive strategy to defeat cancer. A potent p53-reactivating molecule RITA, which triggers p53-dependent apoptosis in human tumor cells in vitro and in vivo, exhibits p53-independent cytotoxicity due to modifications by detoxification enzyme Sulfotransferase 1A1 (SULT1A1), producing a reactive carbocation. Several synthetic modifications to RITA's heterocyclic scaffold lead to higher energy barriers for carbocation formation. In this study, we addressed the question whether RITA analogs NSC777196 and NSC782846 can induce p53-dependent apoptosis without SULT1A1-dependent DNA damage. We found that RITA analog NSC782846, but not NSC777196, induced p53-regulated genes, targeted oncogene addiction, and killed cancer cells upon p53 reactivation, but without induction of DNA damage and inhibition RNA pol II. Our results might demonstrate a method for designing more specific and potent RITA analogs to accelerate translation of p53-targeting compounds from laboratory bench to clinic., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
58. Novel Allosteric Mechanism of Dual p53/MDM2 and p53/MDM4 Inhibition by a Small Molecule.
- Author
-
Grinkevich VV, Vema A, Fawkner K, Issaeva N, Andreotti V, Dickinson ER, Hedström E, Spinnler C, Inga A, Larsson LG, Karlén A, Wilhelm M, Barran PE, Okorokov AL, Selivanova G, and Zawacka-Pankau JE
- Abstract
Restoration of the p53 tumor suppressor for personalised cancer therapy is a promising treatment strategy. However, several high-affinity MDM2 inhibitors have shown substantial side effects in clinical trials. Thus, elucidation of the molecular mechanisms of action of p53 reactivating molecules with alternative functional principle is of the utmost importance. Here, we report a discovery of a novel allosteric mechanism of p53 reactivation through targeting the p53 N-terminus which promotes inhibition of both p53/MDM2 (murine double minute 2) and p53/MDM4 interactions. Using biochemical assays and molecular docking, we identified the binding site of two p53 reactivating molecules, RITA (reactivation of p53 and induction of tumor cell apoptosis) and protoporphyrin IX (PpIX). Ion mobility-mass spectrometry revealed that the binding of RITA to serine 33 and serine 37 is responsible for inducing the allosteric shift in p53, which shields the MDM2 binding residues of p53 and prevents its interactions with MDM2 and MDM4. Our results point to an alternative mechanism of blocking p53 interaction with MDM2 and MDM4 and may pave the way for the development of novel allosteric inhibitors of p53/MDM2 and p53/MDM4 interactions., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Grinkevich, Vema, Fawkner, Issaeva, Andreotti, Dickinson, Hedström, Spinnler, Inga, Larsson, Karlén, Wilhelm, Barran, Okorokov, Selivanova and Zawacka-Pankau.)
- Published
- 2022
- Full Text
- View/download PDF
59. Enterobacteria impair host p53 tumor suppressor activity through mRNA destabilization.
- Author
-
Aschtgen MS, Fragkoulis K, Sanz G, Normark S, Selivanova G, Henriques-Normark B, and Peuget S
- Subjects
- Enterobacteriaceae genetics, Enterobacteriaceae metabolism, Humans, Lipopolysaccharides pharmacology, RNA, Messenger genetics, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism, Neoplasms, Tumor Suppressor Protein p53 metabolism
- Abstract
Increasing evidence highlights the role of bacteria in the physiopathology of cancer. However, the underlying molecular mechanisms remains poorly understood. Several cancer-associated bacteria have been shown to produce toxins which interfere with the host defense against tumorigenesis. Here, we show that lipopolysaccharides from Klebsiella pneumoniae and other Enterobacteria strongly inhibit the host tumor suppressor p53 pathway through a novel mechanism of p53 regulation. We found that lipopolysaccharides destabilize TP53 mRNA through a TLR4-NF-κB-mediated inhibition of the RNA-binding factor Wig-1. Importantly, we show that K. pneumoniae disables two major tumor barriers, oncogene-induced DNA damage signaling and senescence, by impairing p53 transcriptional activity upon DNA damage and oncogenic stress. Furthermore, we found an inverse correlation between the levels of TLR4 and p53 mutation in colorectal tumors. Hence, our data suggest that the repression of p53 by Enterobacteria via TLR4 alleviates the selection pressure for p53 oncogenic mutations and shapes the genomic evolution of cancer., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
60. Pharmacologic Activation of p53 Triggers Viral Mimicry Response Thereby Abolishing Tumor Immune Evasion and Promoting Antitumor Immunity.
- Author
-
Zhou X, Singh M, Sanz Santos G, Guerlavais V, Carvajal LA, Aivado M, Zhan Y, Oliveira MMS, Westerberg LS, Annis DA, Johnsen JI, and Selivanova G
- Subjects
- Humans, Immunotherapy, Tumor Escape, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Interferon Type I, Melanoma drug therapy, Melanoma genetics
- Abstract
The repression of repetitive elements is an important facet of p53's function as a guardian of the genome. Paradoxically, we found that p53 activated by MDM2 inhibitors induced the expression of endogenous retroviruses (ERV) via increased occupancy on ERV promoters and inhibition of two major ERV repressors, histone demethylase LSD1 and DNA methyltransferase DNMT1. Double-stranded RNA stress caused by ERVs triggered type I/III interferon expression and antigen processing and presentation. Pharmacologic activation of p53 in vivo unleashed the IFN program, promoted T-cell infiltration, and significantly enhanced the efficacy of checkpoint therapy in an allograft tumor model. Furthermore, the MDM2 inhibitor ALRN-6924 induced a viral mimicry pathway and tumor inflammation signature genes in patients with melanoma. Our results identify ERV expression as the central mechanism whereby p53 induction overcomes tumor immune evasion and transforms tumor microenvironment to a favorable phenotype, providing a rationale for the synergy of MDM2 inhibitors and immunotherapy., Significance: We found that p53 activated by MDM2 inhibitors induced the expression of ERVs, in part via epigenetic factors LSD1 and DNMT1. Induction of IFN response caused by ERV derepression upon p53-targeting therapies provides a possibility to overcome resistance to immune checkpoint blockade and potentially transform "cold" tumors into "hot." This article is highlighted in the In This Issue feature, p. 2945., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
61. p53-Dependent Repression: DREAM or Reality?
- Author
-
Peuget S and Selivanova G
- Abstract
p53 is a major tumor suppressor that integrates diverse types of signaling in mammalian cells. In response to a broad range of intra- or extra-cellular stimuli, p53 controls the expression of multiple target genes and elicits a vast repertoire of biological responses. The exact code by which p53 integrates the various stresses and translates them into an appropriate transcriptional response is still obscure. p53 is tightly regulated at multiple levels, leading to a wide diversity in p53 complexes on its target promoters and providing adaptability to its transcriptional program. As p53-targeted therapies are making their way into clinics, we need to understand how to direct p53 towards the desired outcome (i.e., cell death, senescence or other) selectively in cancer cells without affecting normal tissues or the immune system. While the core p53 transcriptional program has been proposed, the mechanisms conferring a cell type- and stimuli-dependent transcriptional outcome by p53 require further investigations. The mechanism by which p53 localizes to repressed promoters and manages its co-repressor interactions is controversial and remains an important gap in our understanding of the p53 cistrome. We hope that our review of the recent literature will help to stimulate the appreciation and investigation of largely unexplored p53-mediated repression.
- Published
- 2021
- Full Text
- View/download PDF
62. Editor's Note: Dual Targeting of Wild-Type and Mutant p53 by Small-molecule RITA Results in the Inhibition of N-Myc and Key Survival Oncogenes and Kills Neuroblastoma Cells In Vivo and In Vitro .
- Author
-
Burmakin M, Shi Y, Hedström E, Kogner P, and Selivanova G
- Published
- 2021
- Full Text
- View/download PDF
63. Class IV Lasso Peptides Synergistically Induce Proliferation of Cancer Cells and Sensitize Them to Doxorubicin.
- Author
-
Guerrero-Garzón JF, Madland E, Zehl M, Singh M, Rezaei S, Aachmann FL, Courtade G, Urban E, Rückert C, Busche T, Kalinowski J, Cao YR, Jiang Y, Jiang CL, Selivanova G, and Zotchev SB
- Abstract
Heterologous expression of a biosynthesis gene cluster from Amycolatopsis sp. resulted in the discovery of two unique class IV lasso peptides, felipeptins A1 and A2. A mixture of felipeptins stimulated proliferation of cancer cells, while having no such effect on the normal cells. Detailed investigation revealed, that pre-treatment of cancer cells with a mixture of felipeptins resulted in downregulation of the tumor suppressor Rb, making the cancer cells to proliferate faster. Pre-treatment with felipeptins made cancer cells considerably more sensitive to the anticancer agent doxorubicin and re-sensitized doxorubicin-resistant cells to this drug. Structural characterization and binding experiments showed an interaction between felipeptins resulting in complex formation, which explains their synergistic effect. This discovery may open an alternative avenue in cancer treatment, helping to eliminate quiescent cells that often lead to cancer relapse., Competing Interests: The authors declare no competing interest., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
64. Identification and targeting of selective vulnerability rendered by tamoxifen resistance.
- Author
-
Singh M, Zhou X, Chen X, Santos GS, Peuget S, Cheng Q, Rihani A, Arnér ESJ, Hartman J, and Selivanova G
- Subjects
- Antineoplastic Agents, Hormonal chemistry, Antineoplastic Agents, Hormonal pharmacology, Apoptosis, Arylsulfotransferase genetics, Arylsulfotransferase metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Female, Humans, Tamoxifen chemistry, Tumor Cells, Cultured, Breast Neoplasms drug therapy, Small Molecule Libraries pharmacology, Tamoxifen pharmacology
- Abstract
Background: The estrogen receptor (ER)-positive breast cancer represents over 80% of all breast cancer cases. Even though adjuvant hormone therapy with tamoxifen (TMX) is saving lives of patients with ER-positive breast cancer, the acquired resistance to TMX anti-estrogen therapy is the main hurdle for successful TMX therapy. Here we address the mechanism for TMX resistance and explore the ways to eradicate TMX-resistant breast cancer in both in vitro and ex vivo experiments., Experimental Design: To identify compounds able to overcome TMX resistance, we used short-term and long-term viability assays in cancer cells in vitro and in patient samples in 3D ex vivo, analysis of gene expression profiles and cell line pharmacology database, shRNA screen, CRISPR-Cas9 genome editing, real-time PCR, immunofluorescent analysis, western blot, measurement of oxidative stress using flow cytometry, and thioredoxin reductase 1 enzymatic activity., Results: Here, for the first time, we provide an ample evidence that a high level of the detoxifying enzyme SULT1A1 confers resistance to TMX therapy in both in vitro and ex vivo models and correlates with TMX resistance in metastatic samples in relapsed patients. Based on the data from different approaches, we identified three anticancer compounds, RITA (Reactivation of p53 and Induction of Tumor cell Apoptosis), aminoflavone (AF), and oncrasin-1 (ONC-1), whose tumor cell inhibition activity is dependent on SULT1A1. We discovered thioredoxin reductase 1 (TrxR1, encoded by TXNRD1) as a target of bio-activated RITA, AF, and ONC-1. SULT1A1 depletion prevented the inhibition of TrxR1, induction of oxidative stress, DNA damage signaling, and apoptosis triggered by the compounds. Notably, RITA efficiently suppressed TMX-unresponsive patient-derived breast cancer cells ex vivo., Conclusion: We have identified a mechanism of resistance to TMX via hyperactivated SULT1A1, which renders selective vulnerability to anticancer compounds RITA, AF, and ONC-1, and provide a rationale for a new combination therapy to overcome TMX resistance in breast cancer patients. Our novel findings may provide a strategy to circumvent TMX resistance and suggest that this approach could be developed further for the benefit of relapsed breast cancer patients.
- Published
- 2020
- Full Text
- View/download PDF
65. Thermal Proteome Profiling Identifies Oxidative-Dependent Inhibition of the Transcription of Major Oncogenes as a New Therapeutic Mechanism for Select Anticancer Compounds.
- Author
-
Peuget S, Zhu J, Sanz G, Singh M, Gaetani M, Chen X, Shi Y, Saei AA, Visnes T, Lindström MS, Rihani A, Moyano-Galceran L, Carlson JW, Hjerpe E, Joneborg U, Lehti K, Hartman J, Helleday T, Zubarev R, and Selivanova G
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Line, Tumor, Female, Flavonoids pharmacology, Flavonoids therapeutic use, Furans pharmacology, Furans therapeutic use, Gene Expression Profiling, Humans, Indoles pharmacology, Indoles therapeutic use, Oxidative Stress drug effects, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Proteome genetics, Proteomics methods, Recombinational DNA Repair drug effects, Signal Transduction drug effects, Signal Transduction genetics, Synthetic Lethal Mutations drug effects, Antineoplastic Combined Chemotherapy Protocols pharmacology, Gene Expression Regulation, Neoplastic drug effects, Oncogenes genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Transcription, Genetic drug effects
- Abstract
Identification of the molecular mechanism of action (MoA) of bioactive compounds is a crucial step for drug development but remains a challenging task despite recent advances in technology. In this study, we applied multidimensional proteomics, sensitivity correlation analysis, and transcriptomics to identify a common MoA for the anticancer compounds RITA, aminoflavone (AF), and oncrasin-1 (Onc-1). Global thermal proteome profiling revealed that the three compounds target mRNA processing and transcription, thereby attacking a cancer vulnerability, transcriptional addiction. This led to the preferential loss of expression of oncogenes involved in PDGF, EGFR, VEGF, insulin/IGF/MAPKK, FGF, Hedgehog, TGFβ, and PI3K signaling pathways. Increased reactive oxygen species level in cancer cells was a prerequisite for targeting the mRNA transcription machinery, thus conferring cancer selectivity to these compounds. Furthermore, DNA repair factors involved in homologous recombination were among the most prominently repressed proteins. In cancer patient samples, RITA, AF, and Onc-1 sensitized to poly(ADP-ribose) polymerase inhibitors both in vitro and ex vivo These findings might pave a way for new synthetic lethal combination therapies. Significance: These findings highlight agents that target transcriptional addiction in cancer cells and suggest combination treatments that target RNA processing and DNA repair pathways simultaneously as effective cancer therapies., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
66. p53 CRISPR Deletion Affects DNA Structure and Nuclear Architecture.
- Author
-
Rangel-Pozzo A, Booth S, Yu PLI, Singh M, Selivanova G, and Mai S
- Abstract
The TP53 gene is a key tumor suppressor. Although the tumor suppressor p53 was one of the first to be characterized as a transcription factor, with its main function potentiated by its interaction with DNA, there are still many unresolved questions about its mechanism of action. Here, we demonstrate a novel role for p53 in the maintenance of nuclear architecture of cells. Using three-dimensional (3D) imaging and spectral karyotyping, as well as super resolution microscopy of DNA structure, we observe significant differences in 3D telomere signatures, DNA structure and DNA-poor spaces as well gains or losses of chromosomes, between normal and tumor cells with CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)-deleted or wild-type TP53 . Additionally, treatment with Nutlin-3 results in differences in nuclear architecture of telomeres in wild-type but not in p53 knockout MCF-7 (Michigan Cancer Foundation-7) cells. Nutlin-3 binds to the p53-binding pocket of mouse double minute 2 (MDM2) and blocks the p53-MDM2 interaction. Moreover, we demonstrate that another p53 stabilizing small molecule, RITA (reactivation of p53 and induction of tumor cell apoptosis), also induces changes in 3D DNA structure, apparently in a p53 independent manner. These results implicate p53 activity in regulating nuclear organization and, additionally, highlight the divergent effects of the p53 targeting compounds Nutlin-3 and RITA.
- Published
- 2020
- Full Text
- View/download PDF
67. Pifithrin-α alters p53 post-translational modifications pattern and differentially inhibits p53 target genes.
- Author
-
Zhu J, Singh M, Selivanova G, and Peuget S
- Subjects
- Apoptosis drug effects, Basic Helix-Loop-Helix Transcription Factors drug effects, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, HCT116 Cells, Humans, MCF-7 Cells, NF-E2-Related Factor 2 drug effects, Protein Processing, Post-Translational genetics, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, RNA Interference, RNA, Small Interfering genetics, Reactive Oxygen Species metabolism, Receptors, Aryl Hydrocarbon drug effects, Toluene pharmacology, Transcription, Genetic genetics, Tumor Suppressor Protein p53 metabolism, Benzothiazoles pharmacology, Imidazoles metabolism, Piperazines metabolism, Protein Processing, Post-Translational drug effects, Toluene analogs & derivatives, Transcription, Genetic drug effects, Tumor Suppressor Protein p53 antagonists & inhibitors
- Abstract
Pifithrin-α (PFT-α) is a small molecule which has been widely used as a specific inhibitor of p53 transcription activity. However, its molecular mechanism of action remains unclear. PFT-α has also been described to display potent p53-independent activity in cells. In this study, we addressed the mechanism of action of PFT-α. We found that PFT-α failed to prevent the effects of Mdm2 inhibitor Nutlin-3 on cell cycle and apoptosis in several cancer cell lines. However, PFT-α rescued normal primary fibroblasts from growth inhibition by Nutlin-3. PFT-α displayed a very limited effect on p53-dependent transcription upon its activation by Nutlin-3. Moreover, PFT-α inhibitory effect on transcription was highly dependent on the nature of the p53 target gene. PFT-α attenuated post-translational modifications of p53 without affecting total p53 protein level. Finally, we found that PFT-α can decrease the level of intracellular reactive oxygen species through activation of an aryl hydrocarbon receptor (AHR)-Nrf2 axis in a p53-independent manner. In conclusion, PFT-α inhibits only some aspects of p53 function, therefore it should be used with extreme caution to study p53-dependent processes.
- Published
- 2020
- Full Text
- View/download PDF
68. Therapeutic targeting of mutant p53 in pediatric acute lymphoblastic leukemia.
- Author
-
Demir S, Boldrin E, Sun Q, Hampp S, Tausch E, Eckert C, Ebinger M, Handgretinger R, Kronnie GT, Wiesmüller L, Stilgenbauer S, Selivanova G, Debatin KM, and Meyer LH
- Subjects
- Adult, Apoptosis genetics, Child, Doxorubicin, Humans, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Alterations of the tumor suppressor gene TP53 are found in different cancers, in particular in carcinomas of adults. In pediatric acute lymphoblastic leukemia (ALL), TP53 mutations are infrequent but enriched at relapse. As in most cancers, mainly DNA-binding domain missense mutations are found, resulting in accumulation of mutant p53, poor therapy response, and inferior outcome. Different strategies to target mutant p53 have been developed including reactivation of p53's wildtype function by the small molecule APR-246. We investigated TP53 mutations in cell lines and 62 B-cell precursor ALL samples and evaluated the activity of APR-246 in TP53 -mutated or wildtype ALL. We identified cases with TP53 missense mutations, high (mutant) p53 expression and insensitivity to the DNA-damaging agent doxorubicin. In TP53 -mutated ALL, APR-246 induced apoptosis showing strong anti-leukemia activity. APR-246 restored mutant p53 to its wildtype conformation, leading to pathway activation with induction of transcriptional targets and re-sensitization to genotoxic therapy in vitro and in vivo In addition, induction of oxidative stress contributed to APR-246-mediated cell death. In a preclinical model of patient-derived TP53 -mutant ALL, APR-246 reduced leukemia burden and synergized strongly with the genotoxic agent doxorubicin, leading to superior leukemia-free survival in vivo Thus, targeting mutant p53 by APR-246, restoring its tumor suppressive function, seems to be an effective therapeutic strategy for this high-risk group of TP53 -mutant ALL., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
69. RITA requires eIF2α-dependent modulation of mRNA translation for its anti-cancer activity.
- Author
-
Ristau J, van Hoef V, Peuget S, Zhu J, Guan BJ, Liang S, Hatzoglou M, Topisirovic I, Selivanova G, and Larsson O
- Subjects
- Antineoplastic Agents, Apoptosis, Breast Neoplasms metabolism, Breast Neoplasms pathology, DNA-Binding Proteins genetics, Eukaryotic Initiation Factor-2 genetics, Female, Humans, MCF-7 Cells, Neoplasm Proteins genetics, Phosphorylation, RNA, Messenger genetics, Reactive Oxygen Species metabolism, Signal Transduction, Tumor Suppressor Protein p53 genetics, eIF-2 Kinase metabolism, Breast Neoplasms prevention & control, DNA-Binding Proteins metabolism, Eukaryotic Initiation Factor-2 metabolism, Gene Expression Regulation, Neoplastic, Neoplasm Proteins metabolism, Protein Biosynthesis, RNA, Messenger metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Tumor protein 53 (p53, encoded by the TP53 gene) is a key tumor suppressor regulating cell fates in response to internal and external stresses. As TP53 is mutated or silenced in a majority of tumors, reactivation of p53 by small molecules represents a promising strategy in cancer therapeutics. One such agent is RITA (reactivation of p53 and induction of tumor cell apoptosis), which restores p53 expression in cells with hyperactive HDM2 and induces apoptosis. Yet, mechanisms underlying the anticancer activity of RITA are incompletely understood. Here we show that RITA suppresses mRNA translation independently of p53 by inducing eIF2α phosphorylation. Surprisingly, reactivation of p53 following RITA treatment is critically dependent on eIF2α phosphorylation. Moreover, inhibition of eIF2α phosphorylation attenuates pro-apoptotic and anti-neoplastic effects of RITA, while inducing phosphorylation of eIF2α enhances the anticancer activity of RITA. Collectively, these findings demonstrate that the translational machinery plays a major role in determining the antineoplastic activity of RITA, and suggest that combining p53 activators and translation modulators may be beneficial.
- Published
- 2019
- Full Text
- View/download PDF
70. Inhibition of p53 inhibitors: progress, challenges and perspectives.
- Author
-
Sanz G, Singh M, Peuget S, and Selivanova G
- Subjects
- Animals, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Humans, Mice, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 is the major tumor suppressor and the most frequently inactivated gene in cancer. p53 could be disabled either by mutations or by upstream negative regulators, including, but not limited to MDM2 and MDMX. p53 activity is required for the prevention as well as for the eradication of cancers. Restoration of p53 activity in mouse models leads to the suppression of established tumors of different origin. These findings provide a strong support to the anti-cancer strategy aimed for p53 reactivation. In this review, we summarize recent progress in the development of small molecules, which restore the tumor suppressor function of wild-type p53 and discuss their clinical advance. We discuss different aspects of p53-mediated response, which contribute to suppression of tumors, including non-canonical p53 activities, such as regulation of immune response. While targeting p53 inhibitors is a very promising approach, there are certain limitations and concerns that the intensive research and clinical evaluation of compounds will hopefully help to overcome., (© The Author(s) (2019). Published by Oxford University Press on behalf of Journal of Molecular Cell Biology, IBCB, SIBS, CAS.)
- Published
- 2019
- Full Text
- View/download PDF
71. Prediction of response to anti-cancer drugs becomes robust via network integration of molecular data.
- Author
-
Franco M, Jeggari A, Peuget S, Böttger F, Selivanova G, and Alexeyenko A
- Subjects
- Cell Line, Drug Development methods, Antineoplastic Agents therapeutic use, Biomarkers metabolism, Computational Biology methods, Drug Screening Assays, Antitumor methods, Neoplasms drug therapy
- Abstract
Despite the widening range of high-throughput platforms and exponential growth of generated data volume, the validation of biomarkers discovered from large-scale data remains a challenging field. In order to tackle cancer heterogeneity and comply with the data dimensionality, a number of network and pathway approaches were invented but rarely systematically applied to this task. We propose a new method, called NEAmarker, for finding sensitive and robust biomarkers at the pathway level. scores from network enrichment analysis transform the original space of altered genes into a lower-dimensional space of pathways. These dimensions are then correlated with phenotype variables. The method was first tested using in vitro data from three anti-cancer drug screens and then on clinical data of The Cancer Genome Atlas. It proved superior to the single-gene and alternative enrichment analyses in terms of (1) universal applicability to different data types with a possibility of cross-platform integration, (2) consistency of the discovered correlates between independent drug screens, and (3) ability to explain differential survival of treated patients. Our new screen of anti-cancer compounds validated the performance of multivariate models of drug sensitivity. The previously proposed methods of enrichment analysis could achieve comparable levels of performance in certain tests. However, only our method could discover predictors of both in vitro response and patient survival given administration of the same drug.
- Published
- 2019
- Full Text
- View/download PDF
72. RITA downregulates Hedgehog-GLI in medulloblastoma and rhabdomyosarcoma via JNK-dependent but p53-independent mechanism.
- Author
-
Azatyan A, Gallo-Oller G, Diao Y, Selivanova G, Johnsen JI, and Zaphiropoulos PG
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cerebellar Neoplasms enzymology, Cerebellar Neoplasms genetics, Cerebellar Neoplasms pathology, Female, Hedgehog Proteins genetics, Humans, Medulloblastoma enzymology, Medulloblastoma genetics, Medulloblastoma pathology, Mice, Nude, Pyridines pharmacology, Pyrimidines pharmacology, Rhabdomyosarcoma enzymology, Rhabdomyosarcoma genetics, Rhabdomyosarcoma pathology, Signal Transduction drug effects, Tumor Burden drug effects, Tumor Suppressor Protein p53 genetics, Xenograft Model Antitumor Assays, Zinc Finger Protein GLI1 analysis, Zinc Finger Protein GLI1 genetics, Antineoplastic Agents pharmacology, Cerebellar Neoplasms drug therapy, Furans pharmacology, Hedgehog Proteins metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Medulloblastoma drug therapy, Rhabdomyosarcoma drug therapy, Tumor Suppressor Protein p53 metabolism, Zinc Finger Protein GLI1 metabolism
- Abstract
Overactivation of the Hedgehog (HH) signaling pathway is implicated in many cancers. In this study, we demonstrate that the small molecule RITA, a p53 activator, effectively downregulates HH signaling in human medulloblastoma and rhabdomyosarcoma cells irrespective of p53. This is mediated by a ROS-independent activation of the MAP kinase JNK. We also show that in vitro RITA sensitized cells to the GLI antagonist GANT61, as co-administration of the two drugs had more pronounced effects on cell proliferation and apoptosis. In vivo administration of RITA or GANT61 suppressed rhabdomyosarcoma xenograft growth in nude mice; however, co-administration did not further enhance tumor suppression, even though cell proliferation was decreased. RITA was more potent than GANT61 in downregulating HH target gene expression; surprisingly, this suppressive effect was almost completely eliminated when the two drugs were administered together. Notably, RNA-seq demonstrated a broader response of pathways involved in cancer cell growth in the combination treatment, providing a plausible interpretation for tumor reduction in the absence of HH signaling downregulation., (Copyright © 2018 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
73. MYC and RAS are unable to cooperate in overcoming cellular senescence and apoptosis in normal human fibroblasts.
- Author
-
Zhang F, Zakaria SM, Högqvist Tabor V, Singh M, Tronnersjö S, Goodwin J, Selivanova G, Bartek J, Castell A, and Larsson LG
- Subjects
- Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA Damage drug effects, Doxorubicin pharmacology, Fibroblasts cytology, Fibroblasts metabolism, Humans, RNA Interference, RNA, Small Interfering metabolism, Tamoxifen pharmacology, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, ras Proteins genetics, Apoptosis drug effects, Cellular Senescence drug effects, Proto-Oncogene Proteins c-myc metabolism, ras Proteins metabolism
- Abstract
The MYC and RAS oncogenes are sufficient for transformation of normal rodent cells. This cooperativity is at least in part based on suppression of RAS-induced cellular senescence by MYC and block of MYC-induced apoptosis by RAS - thereby canceling out two main barriers against tumor development. However, it remains unclear whether MYC and RAS cooperate in this way in human cells, where MYC and RAS are not sufficient for transformation. To address this question, we established a combined Tet-inducible H-RAS
V12 and hydroxytamoxifen-inducible MycER system in normal human BJ fibroblasts. We show here that activation of RAS alone induced senescence while activation of MYC alone or together with RAS triggered DNA damage, induction of p53 and massive apoptosis, suggesting that RAS cannot rescue MYC-induced apoptosis in this system. Although coexpression with MYC reduced certain RAS-induced senescence markers (histone H3 lysine 9 trimethylation and senescence-associated β-GAL activity), the induction of the senescence marker p16INK4A was further enhanced and the culture ceased to proliferate within a few days, revealing that MYC could not fully suppress RAS-induced senescence. Furthermore, depletion of p53, which enhanced proliferation and rescued the cells from RAS-induced senescence, did not abrogate MYC-induced apoptosis. We conclude that MYC and RAS are unable to cooperate in overcoming senescence and apoptosis in normal human fibroblasts even after depletion of p53, indicating that additional oncogenic events are required to abrogate these fail-safe mechanisms and pave the way for cellular transformation. These findings have implications for our understanding of the transformation process in human cells. Abbreviations and acronyms: CDK: Cyclin-dependent kinase; DDR: DNA damage response; DOX: Doxycycline; EdU: 5-ethynyl-2'-deoxyuridine; FACS: Fluorescence Activated Cell Sorting; MycER: MYC-estrogen receptor; OHT: 4-hydroxytamoxifen; OIS: Oncogene-induced senescence; PP2A: Protein phosphatase 2A; ROS: Reactive oxygen species; SA-β-GAL: Senescence-associated β-galactosidase; SAHF: Senescence-associated heterochromatin foci; shRNA: Short hairpin RNA; YFP: Yellow fluorescent protein.- Published
- 2018
- Full Text
- View/download PDF
74. Reactivation of mutant p53 and induction of apoptosis in human tumor cells by maleimide analogs.
- Author
-
Bykov VJN, Issaeva N, Zache N, Shilov A, Hultcrantz M, Bergman J, Selivanova G, and Wiman KG
- Published
- 2017
- Full Text
- View/download PDF
75. Ablation of Key Oncogenic Pathways by RITA-Reactivated p53 Is Required for Efficient Apoptosis.
- Author
-
Grinkevich VV, Nikulenkov F, Shi Y, Enge M, Bao W, Maljukova A, Gluch A, Kel A, Sangfelt O, and Selivanova G
- Published
- 2017
- Full Text
- View/download PDF
76. APR-246/PRIMA-1 MET inhibits thioredoxin reductase 1 and converts the enzyme to a dedicated NADPH oxidase.
- Author
-
Peng X, Zhang MQ, Conserva F, Hosny G, Selivanova G, Bykov VJ, Arnér ES, and Wiman KG
- Published
- 2017
- Full Text
- View/download PDF
77. The use of ion mobility mass spectrometry to probe modulation of the structure of p53 and of MDM2 by small molecule inhibitors.
- Author
-
Dickinson ER, Jurneczko E, Nicholson J, Hupp TR, Zawacka-Pankau J, Selivanova G, and Barran PE
- Abstract
Developing drug-like molecules to inhibit the interactions formed by disordered proteins is desirable due to the high correlation of disorder with protein implicated in disease, but is challenging due in part to the lack of atomistically resolved and resolvable structures from conformationally dynamic systems. Ion mobility mass spectrometry (IM-MS) is well-positioned to assess protein ligand interactions along with the effect of a given inhibitor on conformation. Here we demonstrate the use of IM-MS to characterize the effect of two inhibitors RITA and Nutlin-3 on their respective binding partners: p53 and MDM2. RITA binds N-terminal transactivation domain of p53 (Np53) weakly, preventing direct observation of the complex in the gas phase. Nonetheless, upon incubation with RITA, we observe an alteration in the charge state distribution and in the conformational distributions adopted by Np53 in the gas phase. This finding supports the hypothesis that RITAs mode of action proceeds via a conformational change in p53. Circular dichroism corroborates our gas phase findings, showing a slight increase in secondary structure content on ligand incubation, and HDX-MS experiments also highlight the dynamic properties of this protein. Using the same approach we present data to show the effect of Nutlin-3 binding to the N-terminal domain of MDM2 (N-MDM2), N-MDM2 presents as at least two conformational families in the absence of Nutlin-3. Upon Nutlin-3 binding, the protein undergoes a compaction event similar to that exhibited by RITA on Np53. This multi-technique approach highlights the inherent disorder in these systems; and in particular exemplifies the power of IM-MS as a technique to study transient interactions between small molecule inhibitors and intrinsically disordered proteins.
- Published
- 2015
- Full Text
- View/download PDF
78. Pharmacological reactivation of p53 as a strategy to treat cancer.
- Author
-
Zawacka-Pankau J and Selivanova G
- Subjects
- Alleles, Animals, Clinical Trials, Phase II as Topic, Evidence-Based Medicine, Humans, Mice, Neoplasms metabolism, Phenotype, Signal Transduction drug effects, Treatment Outcome, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents therapeutic use, Molecular Targeted Therapy methods, Mutation, Missense, Neoplasms drug therapy, Neoplasms genetics, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics
- Abstract
It has been confirmed through studies using the technique of unbiased sequencing that the TP53 tumour suppressor is the most frequently inactivated gene in cancer. This finding, together with results from earlier studies, provides compelling evidence for the idea that p53 ablation is required for the development and maintenance of tumours. Genetic reconstitution of the function of p53 leads to the suppression of established tumours as shown in mouse models. This strongly supports the notion that p53 reactivation by small molecules could provide an efficient strategy to treat cancer. In this review, we summarize recent advances in the development of small molecules that restore the function of mutant p53 by different mechanisms, including stabilization of its folding by Apr-246, which is currently being tested in a Phase II clinical trial. We discuss several classes of compounds that reactivate wild-type p53, such as Mdm2 inhibitors, which are currently undergoing clinical testing, MdmX inhibitors and molecules targeting factors upstream of Mdm2/X or p53 itself. Finally, we consider the clinical applications of compounds targeting p53 and the p53 pathway., (© 2014 The Association for the Publication of the Journal of Internal Medicine.)
- Published
- 2015
- Full Text
- View/download PDF
79. The conserved Trp114 residue of thioredoxin reductase 1 has a redox sensor-like function triggering oligomerization and crosslinking upon oxidative stress related to cell death.
- Author
-
Xu J, Eriksson SE, Cebula M, Sandalova T, Hedström E, Pader I, Cheng Q, Myers CR, Antholine WE, Nagy P, Hellman U, Selivanova G, Lindqvist Y, and Arnér ES
- Subjects
- Animals, Cell Death drug effects, Cell Line, Tumor, Flavin-Adenine Dinucleotide metabolism, Furans pharmacology, HCT116 Cells, Humans, Kinetics, Masoprocol pharmacology, Models, Molecular, Mutant Proteins metabolism, Oxidation-Reduction drug effects, Rats, Structure-Activity Relationship, Conserved Sequence, Cross-Linking Reagents pharmacology, Oxidative Stress drug effects, Protein Multimerization drug effects, Thioredoxin Reductase 1 metabolism, Tryptophan metabolism
- Abstract
The selenoprotein thioredoxin reductase 1 (TrxR1) has several key roles in cellular redox systems and reductive pathways. Here we discovered that an evolutionarily conserved and surface-exposed tryptophan residue of the enzyme (Trp114) is excessively reactive to oxidation and exerts regulatory functions. The results indicate that it serves as an electron relay communicating with the FAD moiety of the enzyme, and, when oxidized, it facilitates oligomerization of TrxR1 into tetramers and higher multimers of dimers. A covalent link can also be formed between two oxidized Trp114 residues of two subunits from two separate TrxR1 dimers, as found both in cell extracts and in a crystal structure of tetrameric TrxR1. Formation of covalently linked TrxR1 subunits became exaggerated in cells on treatment with the pro-oxidant p53-reactivating anticancer compound RITA, in direct correlation with triggering of a cell death that could be prevented by antioxidant treatment. These results collectively suggest that Trp114 of TrxR1 serves a function reminiscent of an irreversible sensor for excessive oxidation, thereby presenting a previously unrecognized level of regulation of TrxR1 function in relation to cellular redox state and cell death induction.
- Published
- 2015
- Full Text
- View/download PDF
80. Modulation of the poly (ADP-ribose) polymerase inhibitor response and DNA recombination in breast cancer cells by drugs affecting endogenous wild-type p53.
- Author
-
Ireno IC, Wiehe RS, Stahl AI, Hampp S, Aydin S, Troester MA, Selivanova G, and Wiesmüller L
- Subjects
- Benzothiazoles pharmacology, Cell Line, Tumor drug effects, Female, Gene Knockdown Techniques, Humans, Imidazoles pharmacology, Isoquinolines pharmacology, Molecular Weight, Piperazines pharmacology, Poly(ADP-ribose) Polymerases genetics, Poly(ADP-ribose) Polymerases metabolism, RNA, Small Interfering, Toluene analogs & derivatives, Toluene pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Enzyme Inhibitors pharmacology, Genes, p53, Poly(ADP-ribose) Polymerase Inhibitors, Recombinational DNA Repair drug effects
- Abstract
Synthetic lethal interactions between poly (ADP-ribose) polymerase (PARP) and homologous recombination (HR) repair pathways have been exploited for the development of novel mono- and combination cancer therapies. The tumor suppressor p53 was demonstrated to exhibit indirect and direct regulatory activities in DNA repair, particularly in DNA double-strand break (DSB)-induced and replication-associated HR. In this study, we tested a potential influence of the p53 status on the response to PARP inhibition, which is known to cause replication stress. Silencing endogenous or inducibly expressing p53 we found a protective effect of p53 on PARP inhibitor (PARPi)-mediated cytotoxicities. This effect was specific for wild-type versus mutant p53 and observed in cancer but not in non-transformed cell lines. Enhanced cytotoxicities after treatment with the p53-inhibitory drug Pifithrinα further supported p53-mediated resistance to PARP inhibition. Surprisingly, we equally observed increased PARPi sensitivity in the presence of the p53-activating compound Nutlin-3. As a common denominator, both drug responses correlated with decreased HR activities: Pifithrinα downregulated spontaneous HR resulting in damage accumulation. Nutlin-3 induced a decrease of DSB-induced HR, which was accompanied by a severe drop in RAD51 protein levels. Thus, we revealed a novel link between PARPi responsiveness and p53-controlled HR activities. These data expand the concept of cell and stress type-dependent healer and killer functions of wild-type p53 in response to cancer therapeutic treatment. Our findings have implications for the individualized design of cancer therapies using PARPi and the potentially combined use of p53-modulatory drugs., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
81. Integrated high-throughput analysis identifies Sp1 as a crucial determinant of p53-mediated apoptosis.
- Author
-
Li H, Zhang Y, Ströse A, Tedesco D, Gurova K, and Selivanova G
- Subjects
- HCT116 Cells, Humans, Apoptosis, High-Throughput Screening Assays, Sp1 Transcription Factor metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The restoration of p53 tumor suppressor function is a promising therapeutic strategy to combat cancer. However, the biological outcomes of p53 activation, ranging from the promotion of growth arrest to the induction of cell death, are hard to predict, which limits the clinical application of p53-based therapies. In the present study, we performed an integrated analysis of genome-wide short hairpin RNA screen and gene expression data and uncovered a previously unrecognized role of Sp1 as a central modulator of the transcriptional response induced by p53 that leads to robust induction of apoptosis. Sp1 is indispensable for the pro-apoptotic transcriptional repression by p53, but not for the induction of pro-apoptotic genes. Furthermore, the p53-dependent pro-apoptotic transcriptional repression required the co-binding of Sp1 to p53 target genes. Our results also highlight that Sp1 shares with p53 a common regulator, MDM2, which targets Sp1 for proteasomal degradation. This uncovers a new mechanism of the tight control of apoptosis in cells. Our study advances the understanding of the molecular basis of p53-mediated apoptosis and implicates Sp1 as one of its key modulators. We found that small molecules reactivating p53 can differentially modulate Sp1, thus providing insights into how to manipulate p53 response in a controlled way.
- Published
- 2014
- Full Text
- View/download PDF
82. Wild type p53 reactivation: from lab bench to clinic.
- Author
-
Selivanova G
- Subjects
- Animals, Humans, Neoplasms drug therapy, Neoplasms metabolism, Neoplasms pathology, Neoplasms prevention & control, Proto-Oncogene Proteins c-mdm2 metabolism, Small Molecule Libraries pharmacology, Small Molecule Libraries therapeutic use, Tumor Suppressor Protein p53 genetics, Laboratories, Tumor Suppressor Protein p53 metabolism
- Abstract
The p53 tumor suppressor is the most frequently inactivated gene in cancer. Several mouse models have demonstrated that the reconstitution of the p53 function suppresses the growth of established tumors. These facts, taken together, promote the idea of p53 reactivation as a strategy to combat cancer. This review will focus on recent advances in the development of small molecules which restore the function of wild type p53 by blocking its inhibitors Mdm2 and MdmX or their upstream regulators and discuss the impact of different p53 functions for tumor prevention and tumor eradication. Finally, the recent progress in p53 research will be analyzed concerning the role of p53 cofactors and cellular environment in the biological response upon p53 reactivation and how this can be applied in clinic., (Copyright © 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
83. ROS-dependent activation of JNK converts p53 into an efficient inhibitor of oncogenes leading to robust apoptosis.
- Author
-
Shi Y, Nikulenkov F, Zawacka-Pankau J, Li H, Gabdoulline R, Xu J, Eriksson S, Hedström E, Issaeva N, Kel A, Arnér ES, and Selivanova G
- Subjects
- Cell Cycle Proteins, Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases, DNA Damage drug effects, DNA Repair, HCT116 Cells, Humans, Hydrogen Peroxide pharmacology, MCF-7 Cells, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oxidants pharmacology, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases genetics, Phosphoprotein Phosphatases metabolism, Protein Phosphatase 2C, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, RNA, Small Interfering metabolism, Reactive Oxygen Species analysis, Reactive Oxygen Species metabolism, Thioredoxin Reductase 1 metabolism, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Apoptosis drug effects, JNK Mitogen-Activated Protein Kinases metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Rescue of the p53 tumor suppressor is an attractive cancer therapy approach. However, pharmacologically activated p53 can induce diverse responses ranging from cell death to growth arrest and DNA repair, which limits the efficient application of p53-reactivating drugs in clinic. Elucidation of the molecular mechanisms defining the biological outcome upon p53 activation remains a grand challenge in the p53 field. Here, we report that concurrent pharmacological activation of p53 and inhibition of thioredoxin reductase followed by generation of reactive oxygen species (ROS), result in the synthetic lethality in cancer cells. ROS promote the activation of c-Jun N-terminal kinase (JNK) and DNA damage response, which establishes a positive feedback loop with p53. This converts the p53-induced growth arrest/senescence to apoptosis. We identified several survival oncogenes inhibited by p53 in JNK-dependent manner, including Mcl1, PI3K, eIF4E, as well as p53 inhibitors Wip1 and MdmX. Further, we show that Wip1 is one of the crucial executors downstream of JNK whose ablation confers the enhanced and sustained p53 transcriptional response contributing to cell death. Our study provides novel insights for manipulating p53 response in a controlled way. Further, our results may enable new pharmacological strategy to exploit abnormally high ROS level, often linked with higher aggressiveness in cancer, to selectively kill cancer cells upon pharmacological reactivation of p53.
- Published
- 2014
- Full Text
- View/download PDF
84. APR-246/PRIMA-1MET inhibits thioredoxin reductase 1 and converts the enzyme to a dedicated NADPH oxidase.
- Author
-
Peng X, Zhang MQ, Conserva F, Hosny G, Selivanova G, Bykov VJ, Arnér ES, and Wiman KG
- Subjects
- Animals, Cell Death drug effects, Cell Line, Tumor, Cell Survival drug effects, Cytoprotection drug effects, Gene Knockdown Techniques, Humans, RNA, Small Interfering metabolism, Rats, Reactive Oxygen Species metabolism, Thioredoxin Reductase 1 metabolism, NADPH Oxidases metabolism, Quinuclidines pharmacology, Thioredoxin Reductase 1 antagonists & inhibitors
- Abstract
The low-molecular-weight compound APR-246 (PRIMA-1(MET)) restores wild-type conformation and function to mutant p53, and triggers apoptosis in tumor cells. We show here that APR-246 also targets the selenoprotein thioredoxin reductase 1 (TrxR1), a key regulator of cellular redox balance. APR-246 inhibited both recombinant TrxR1 in vitro and TrxR1 in cells. A Sec-to-Cys mutant of TrxR1 was not inhibited by APR-246, suggesting targeting of the selenocysteine residue in wild-type TrxR1. Preheated APR-246 and its conversion product methylene quinuclidinone (MQ) were much more efficient TrxR1 inhibitors than APR-246 itself, indicating that MQ is the active compound responsible for TrxR1 enzyme inhibition. TrxR1 inhibited by MQ was still functional as a pro-oxidant NADPH oxidase. Knockdown of TrxR1 caused a partial and reproducible attenuation of APR-246-induced tumor cell death independently of p53 status. Cellular TrxR1 activity was also inhibited by APR-246 irrespective of p53 status. We show that APR-246 can directly affect cellular redox status via targeting of TrxR1. Our findings provide an explanation for the previously observed effects of APR-246 on tumor cells lacking mutant p53.
- Published
- 2013
- Full Text
- View/download PDF
85. Dual targeting of wild-type and mutant p53 by small molecule RITA results in the inhibition of N-Myc and key survival oncogenes and kills neuroblastoma cells in vivo and in vitro.
- Author
-
Burmakin M, Shi Y, Hedström E, Kogner P, and Selivanova G
- Subjects
- Animals, Antineoplastic Agents pharmacology, Blotting, Western, Cell Proliferation drug effects, Female, Humans, Immunoprecipitation, In Vitro Techniques, Mice, Mice, SCID, Mutation genetics, Neuroblastoma metabolism, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Xenograft Model Antitumor Assays, Apoptosis drug effects, Neuroblastoma drug therapy, Neuroblastoma pathology, Oncogenes drug effects, Piperazines pharmacology, Proto-Oncogene Proteins c-myc antagonists & inhibitors, Tumor Suppressor Protein p53 antagonists & inhibitors
- Abstract
Purpose: Restoration of the p53 function in tumors is a promising therapeutic strategy due to the high potential of p53 as tumor suppressor and the fact that established tumors depend on p53 inactivation for their survival. Here, we addressed the question whether small molecule RITA can reactivate p53 in neuroblastoma and suppress the growth of neuroblastoma cells in vitro and in vivo., Experimental Design: The ability of RITA to inhibit growth and to induce apoptosis was shown in seven neuroblastoma cell lines. Mechanistic studies were carried out to determine the p53 dependence and the molecular mechanism of RITA-induced apoptosis in neuroblastoma, using cell viability assays, RNAi silencing, co-immunoprecipitation, qPCR, and Western blotting analysis. In vivo experiments were conducted to study the effect of RITA on human neuroblastoma xenografts in mice., Results: RITA induced p53-dependent apoptosis in a set of seven neuroblastoma cell lines, carrying wild-type or mutant p53; it activated p53 and triggered the expression of proapoptotic p53 target genes. Importantly, p53 activated by RITA inhibited several key oncogenes that are high-priority targets for pharmacologic anticancer strategies in neuroblastoma, including N-Myc, Aurora kinase, Mcl-1, Bcl-2, Wip-1, MDM2, and MDMX. Moreover, RITA had a strong antitumor effect in vivo., Conclusions: Reactivation of wild-type and mutant p53 resulting in the induction of proapoptotic factors along with ablation of key oncogenes by compounds such as RITA may be a highly effective strategy to treat neuroblastoma., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
86. Insights into p53 transcriptional function via genome-wide chromatin occupancy and gene expression analysis.
- Author
-
Nikulenkov F, Spinnler C, Li H, Tonelli C, Shi Y, Turunen M, Kivioja T, Ignatiev I, Kel A, Taipale J, and Selivanova G
- Subjects
- Aurora Kinase A, Aurora Kinases, Chromatin Immunoprecipitation, Chromosome Mapping, Furans pharmacology, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, Humans, Imidazoles pharmacology, MCF-7 Cells, Piperazines pharmacology, Promoter Regions, Genetic, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Response Elements, STAT3 Transcription Factor metabolism, Sp1 Transcription Factor metabolism, Transcription, Genetic, Tumor Suppressor Protein p53 genetics, Chromatin metabolism, Genome, Human, Tumor Suppressor Protein p53 metabolism
- Abstract
The tumor-suppressor p53 can induce various biological responses. Yet, it is not clear whether it is p53 in vivo promoter selectivity that triggers different transcription programs leading to different outcomes. Our analysis of genome-wide chromatin occupancy by p53 using chromatin immunoprecipitation (ChIP)-seq revealed 'p53 default program', that is, the pattern of major p53-bound sites that is similar upon p53 activation by nutlin3a, reactivation of p53 and induction of tumor cell apoptosis (RITA) or 5-fluorouracil in breast cancer cells, despite different biological outcomes. Parallel analysis of gene expression allowed identification of 280 novel p53 target genes, including p53-repressed AURKA. We identified Sp1 as one of the p53 modulators, which confer specificity to p53-mediated transcriptional response upon RITA. Further, we found that STAT3 antagonizes p53-mediated repression of a subset of genes, including AURKA.
- Published
- 2012
- Full Text
- View/download PDF
87. Protein kinase Cα (PKCα) regulates p53 localization and melanoma cell survival downstream of integrin αv in three-dimensional collagen and in vivo.
- Author
-
Smith SD, Enge M, Bao W, Thullberg M, Costa TD, Olofsson H, Gashi B, Selivanova G, and Strömblad S
- Subjects
- Active Transport, Cell Nucleus genetics, Cell Line, Tumor, Cell Nucleus genetics, Cell Nucleus pathology, Cell Survival genetics, Collagen chemistry, Collagen genetics, Humans, Integrin alphaV genetics, Melanoma genetics, Melanoma pathology, Protein Kinase C-alpha genetics, Tumor Suppressor Protein p53 genetics, Up-Regulation genetics, Cell Nucleus metabolism, Collagen metabolism, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Integrin alphaV metabolism, Melanoma metabolism, Protein Kinase C-alpha metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Protein kinase C α (PKCα) is overexpressed in numerous types of cancer. Importantly, PKCα has been linked to metastasis of malignant melanoma in patients. However, it has been unclear how PKCα may be regulated and how it exerts its role in melanoma. Here, we identified a role for PKCα in melanoma cell survival in a three-dimensional collagen model mimicking the in vivo pathophysiology of the dermis. A pathway was identified that involved integrin αv-mediated up-regulation of PKCα and PKCα-dependent regulation of p53 localization, which was connected to melanoma cell survival. Melanoma survival and growth in three-dimensional microenvironments requires the expression of integrin αv, which acts to suppress p53 activity. Interestingly, microarray analysis revealed that PKCα was up-regulated by integrin αv in a three-dimensional microenvironment-dependent manner. Integrin αv was observed to promote a relocalization of endogenous p53 from the nucleus to the cytoplasm upon growth in three-dimensional collagen as well as in vivo, whereas stable knockdown of PKCα inhibited the integrin αv-mediated relocalization of p53. Importantly, knockdown of PKCα also promoted apoptosis in three-dimensional collagen and in vivo, resulting in reduced tumor growth. This indicates that PKCα constitutes a crucial component of the integrin αv-mediated pathway(s) that promote p53 relocalization and melanoma survival.
- Published
- 2012
- Full Text
- View/download PDF
88. A novel facet of tumor suppression by p53: Induction of tumor immunogenicity.
- Author
-
Li H, Lakshmikanth T, Carbone E, and Selivanova G
- Abstract
Pharmacological reactivation of the p53 tumor suppressor is a promising strategy for anti-cancer therapy due to its high potential to elicit apoptosis or growth arrest in cancer cells. Recently we uncovered the mechanism of activation of the innate immune response by p53 upon its activation by small molecules.
- Published
- 2012
- Full Text
- View/download PDF
89. Inhibition of glycolytic enzymes mediated by pharmacologically activated p53: targeting Warburg effect to fight cancer.
- Author
-
Zawacka-Pankau J, Grinkevich VV, Hünten S, Nikulenkov F, Gluch A, Li H, Enge M, Kel A, and Selivanova G
- Subjects
- Cell Hypoxia genetics, Cell Line, Tumor, Glucose genetics, Glucose Transport Proteins, Facilitative biosynthesis, Glucose Transport Proteins, Facilitative genetics, Glucose Transporter Type 1 biosynthesis, Glucose Transporter Type 1 genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Neoplasms genetics, Neoplasms therapy, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Tumor Suppressor Protein p53 genetics, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Glucose metabolism, Glycolysis, Neoplasms enzymology, Response Elements, Tumor Suppressor Protein p53 metabolism
- Abstract
Unique sensitivity of tumor cells to the inhibition of glycolysis is a good target for anticancer therapy. Here, we demonstrate that the pharmacologically activated tumor suppressor p53 mediates the inhibition of glycolytic enzymes in cancer cells in vitro and in vivo. We showed that p53 binds to the promoters of metabolic genes and represses their expression, including glucose transporters SLC2A12 (GLUT12) and SLC2A1 (GLUT1). Furthermore, p53-mediated repression of transcription factors c-Myc and HIF1α, key drivers of ATP-generating pathways in tumors, contributed to ATP production block. Inhibition of c-Myc by p53 mediated the ablation of several glycolytic genes in normoxia, whereas in hypoxia down-regulation of HIF1α contributed to this effect. We identified Sp1 as a transcription cofactor cooperating with p53 in the ablation of metabolic genes. Using different approaches, we demonstrated that glycolysis block contributes to the robust induction of apoptosis by p53 in cancer cells. Taken together, our data suggest that tumor-specific reinstatement of p53 function targets the "Achilles heel" of cancer cells (i.e. their dependence on glycolysis), which could contribute to the tumor-selective killing of cancer cells by pharmacologically activated p53.
- Published
- 2011
- Full Text
- View/download PDF
90. Abrogation of Wip1 expression by RITA-activated p53 potentiates apoptosis induction via activation of ATM and inhibition of HdmX.
- Author
-
Spinnler C, Hedström E, Li H, de Lange J, Nikulenkov F, Teunisse AF, Verlaan-de Vries M, Grinkevich V, Jochemsen AG, and Selivanova G
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Cell Line, Tumor, Down-Regulation, Gene Expression Regulation drug effects, Humans, Nuclear Proteins metabolism, Phosphoprotein Phosphatases genetics, Proteasome Endopeptidase Complex metabolism, Protein Phosphatase 2C, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2 metabolism, Signal Transduction, Tumor Suppressor Protein p53 antagonists & inhibitors, Apoptosis drug effects, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, Furans pharmacology, Phosphoprotein Phosphatases metabolism, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Inactivation of the p53 tumour suppressor, either by mutation or by overexpression of its inhibitors Hdm2 and HdmX is the most frequent event in cancer. Reactivation of p53 by targeting Hdm2 and HdmX is therefore a promising strategy for therapy. However, Hdm2 inhibitors do not prevent inhibition of p53 by HdmX, which impedes p53-mediated apoptosis. Here, we show that p53 reactivation by the small molecule RITA leads to efficient HdmX degradation in tumour cell lines of different origin and in xenograft tumours in vivo. Notably, HdmX degradation occurs selectively in cancer cells, but not in non-transformed cells. We identified the inhibition of the wild-type p53-induced phosphatase 1 (Wip1) as the major mechanism important for full engagement of p53 activity accomplished by restoration of the ataxia telangiectasia mutated (ATM) kinase-signalling cascade, which leads to HdmX degradation. In contrast to previously reported transactivation of Wip1 by p53, we observed p53-dependent repression of Wip1 expression, which disrupts the negative feedback loop conferred by Wip1. Our study reveals that the depletion of both HdmX and Wip1 potentiates cell death due to sustained activation of p53. Thus, RITA is an example of a p53-reactivating drug that not only blocks Hdm2, but also inhibits two important negative regulators of p53 - HdmX and Wip1, leading to efficient elimination of tumour cells.
- Published
- 2011
- Full Text
- View/download PDF
91. Pharmacological activation of p53 triggers anticancer innate immune response through induction of ULBP2.
- Author
-
Li H, Lakshmikanth T, Garofalo C, Enge M, Spinnler C, Anichini A, Szekely L, Kärre K, Carbone E, and Selivanova G
- Subjects
- Cell Line, Tumor, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Methylation, Furans pharmacology, GPI-Linked Proteins metabolism, Humans, Killer Cells, Natural metabolism, Killer Cells, Natural physiology, NK Cell Lectin-Like Receptor Subfamily K antagonists & inhibitors, NK Cell Lectin-Like Receptor Subfamily K metabolism, Up-Regulation, Immunity, Innate immunology, Intercellular Signaling Peptides and Proteins metabolism, Neoplasms immunology, Tumor Suppressor Protein p53 metabolism
- Abstract
Escape of tumor cells from cell-intrinsic barrier mediated by tumor suppressors and cell-extrinsic barrier mediated by the immune system is crucial for tumorigenesis. Growing evidence suggests that reactivation of tumor suppressor function or restoration of anticancer immunity is promising strategy for anticancer therapy due to their high potential to combat cancer. p53, a key tumor suppressor, represses tumorigenesis by eliciting growth arrest, apoptosis or senescence in cancer cells. Here, we unravel that, apart from these cell-autonomous effects, p53 activates the innate immune response against cancer cells. Our results show that pharmacological reactivation of p53 can stimulate the expression of ULPB2, a ligand for NK cell activating receptor NKG2D in human tumor cells of different origin, which enhance the susceptibility of tumor cells to NK cell-mediated killing. The molecular mechanism controlling ULPB2 expression by p53 is neither ATM/ATR- nor caspase-dependent. Using several approaches, we identified p53 as a direct transcriptional regulator of ULBP2 and found a p53 response element within ULBP2 gene, which confers the p53 regulation. Furthermore, we demonstrated that demethylation of p53-binding region within ULBP2 gene was required for p53-dependent induction of ULPB2, which can be achieved via repression of DNA methyltransferases (DNMTs) by p53. This molecular evidence for the direct control of immunosurveillance by p53 links tumor suppressor activation to innate immune stimuli and provides a possibility to integrate cell-extrinsic and -intrinsic defenses against tumorigenesis by pharmacological activation of p53, which may increase the probability to achieve a durable therapeutic success., (© 2011 Landes Bioscience)
- Published
- 2011
- Full Text
- View/download PDF
92. PRIMA-1Met/APR-246 induces apoptosis and tumor growth delay in small cell lung cancer expressing mutant p53.
- Author
-
Zandi R, Selivanova G, Christensen CL, Gerds TA, Willumsen BM, and Poulsen HS
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Disease Progression, Genes, p53, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Mice, Mice, Nude, Mutant Proteins genetics, Mutant Proteins metabolism, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology, Time Factors, Xenograft Model Antitumor Assays, Apoptosis drug effects, Cell Proliferation drug effects, Lung Neoplasms drug therapy, Quinuclidines pharmacology, Quinuclidines therapeutic use, Small Cell Lung Carcinoma drug therapy
- Abstract
Purpose: Small cell lung cancer (SCLC) is a highly malignant disease with poor prognosis, necessitating the need to develop new and efficient treatment modalities. PRIMA-1(Met) (p53-dependent reactivation of massive apoptosis), also known as APR-246, is a small molecule, which restores tumor suppressor function to mutant p53 and induces cancer cell death in various cancer types. Since p53 is mutated in more than 90% of SCLC, we investigated the ability of PRIMA-1(Met) to induce apoptosis and inhibit tumor growth in SCLC with different p53 mutations., Experimental Design: The therapeutic effect of PRIMA-1(Met)/APR-246 was studied in SCLC cells in vitro using cell viability assay, fluorescence-activated cell-sorting analysis, p53 knockdown studies, and Western blot analyses. The antitumor potential of PRIMA-1(Met)/APR-246 was further evaluated in two different SCLC xenograft models., Results: PRIMA-1(Met)/APR-246 efficiently inhibited the growth of the SCLC cell lines expressing mutant p53 in vitro and induced apoptosis, associated with increased fraction of cells with fragmented DNA, caspase-3 activation, PARP cleavage, Bax and Noxa upregulation and Bcl-2 downregulation in the cells. The growth suppressive effect of PRIMA-1(Met)/APR-246 was markedly reduced in SCLC cell lines transfected with p53 siRNA, supporting the role of mutant p53 in PRIMA-1(Met)/APR-246-induced cell death. Moreover, in vivo studies showed significant antitumor effects of PRIMA-1(Met) after i.v. injection in SCLC mouse models with no apparent toxicity., Conclusion: This study is the first to show the potential use of p53-reactivating molecules such as PRIMA-1(Met)/APR-246 for the treatment of SCLC., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
93. PRIMA-1Met/APR-246 induces wild-type p53-dependent suppression of malignant melanoma tumor growth in 3D culture and in vivo.
- Author
-
Bao W, Chen M, Zhao X, Kumar R, Spinnler C, Thullberg M, Issaeva N, Selivanova G, and Strömblad S
- Subjects
- Animals, Apoptosis, Apoptosis Regulatory Proteins metabolism, Apoptotic Protease-Activating Factor 1 metabolism, Caspase 3 metabolism, Caspase 9 metabolism, Cell Culture Techniques, Cell Line, Tumor, Humans, Melanoma metabolism, Mice, Mice, Nude, Proto-Oncogene Proteins metabolism, RNA Interference, RNA, Small Interfering, Transplantation, Heterologous, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 physiology, Antineoplastic Agents therapeutic use, Melanoma drug therapy, Quinuclidines therapeutic use, Tumor Suppressor Protein p53 metabolism
- Abstract
Disseminating malignant melanoma is a lethal disease highly resistant to radio- and chemotherapy. Therefore, the development of new treatment strategies is strongly needed. Tumor suppressor p53-mediated apoptosis is essential for the response to radio- and chemotherapy. Although p53 is not frequently mutated in melanoma, it is inactivated by integrin αv-mediated signaling, as we previously demonstrated 1, which may account, at least partially, for increased apoptosis resistance of malignant melanoma. In this study we addressed the question whether functional restoration of p53 by APR-246 (PRIMA-1Met), which can reactivate mutant p53 and induce massive apoptosis in cancer cells, is able to restore the function of inactive p53 in melanoma. Using a three-dimensional collagen gel (3D-collagen) to culture melanoma cells carrying wild-type p53, we found that APR-246 treatment resulted in activation of p53, leading to increased expression of p53 pro-apoptotic targets Apaf1 and PUMA and activation of caspase- 9 and -3. Moreover, APR-246 triggered melanoma cell apoptosis that was mediated by p53 and caspase 9. Importantly, APR-246 treatment also suppressed human melanoma xenograft tumors in vivo in a p53-dependent manner. Thus, wild-type p53 reactivation may provide a novel approach for malignant melanoma treatment, with APR-246 as a candidate drug for such a development.
- Published
- 2011
- Full Text
- View/download PDF
94. Rescue of the apoptotic-inducing function of mutant p53 by small molecule RITA.
- Author
-
Zhao CY, Grinkevich VV, Nikulenkov F, Bao W, and Selivanova G
- Subjects
- Cell Line, Tumor, Cell Proliferation, Humans, Mutation, Neoplasms drug therapy, Neoplasms radiotherapy, Protein Binding, Proto-Oncogene Proteins c-mdm2 metabolism, Transcriptional Activation, Tumor Suppressor Protein p53 genetics, Apoptosis, Furans therapeutic use, Tumor Suppressor Protein p53 metabolism
- Abstract
Expression of mutant p53 correlates with poor prognosis in many tumors, therefore strategies aimed at reactivation of mutant p53 are likely to provide important benefits for treatment of tumors that are resistant to chemotherapy and radiotherapy. We have previously identified and characterized a small molecule RITA which binds p53 and induces a conformational change which prevents the binding of p53 to several inhibitors, including its own destructor MDM2. In this way, RITA rescues the tumor suppression function of wild type p53. Here, we demonstrate that RITA suppressed the growth and induced apoptosis in human tumor cell lines of a diverse origin carrying mutant p53 proteins. RITA restored transcriptional transactivation and transrepression function of several hot spot p53 mutants. The ability of RITA to rescue the activity of different p53 mutants suggests its generic mechanism of action. Thus, RITA is a promising lead for the development of anti-cancer drugs that reactivate the tumor suppressor function of p53 in cancer cells irrespective whether they express mutant or wild type p53.
- Published
- 2010
- Full Text
- View/download PDF
95. Rescue of p53 function by small-molecule RITA in cervical carcinoma by blocking E6-mediated degradation.
- Author
-
Zhao CY, Szekely L, Bao W, and Selivanova G
- Subjects
- Apoptosis, CDC2 Protein Kinase chemistry, Cell Proliferation, Cell Survival, Cyclin B1 metabolism, Female, HeLa Cells, Humans, Ubiquitin chemistry, cdc25 Phosphatases chemistry, Carcinoma drug therapy, Carcinoma metabolism, Furans pharmacology, Oncogene Proteins, Viral metabolism, Repressor Proteins metabolism, Tumor Suppressor Protein p53 metabolism, Uterine Cervical Neoplasms drug therapy, Uterine Cervical Neoplasms metabolism
- Abstract
Proteasomal degradation of p53 by human papilloma virus (HPV) E6 oncoprotein plays a pivotal role in the survival of cervical carcinoma cells. Abrogation of HPV-E6-dependent p53 destruction can therefore be a good strategy to combat cervical carcinomas. Here, we show that a small-molecule reactivation of p53 and induction of tumor cell apoptosis (RITA) is able to induce the accumulation of p53 and rescue its tumor suppressor function in cells containing high-risk HPV16 and HPV18 by inhibiting HPV-E6-mediated proteasomal degradation. RITA blocks p53 ubiquitination by preventing p53 interaction with E6-associated protein, required for HPV-E6-mediated degradation. RITA activates the transcription of proapoptotic p53 targets Noxa, PUMA, and BAX, and repressed the expression of pro-proliferative factors CyclinB1, CDC2, and CDC25C, resulting in p53-dependent apoptosis and cell cycle arrest. Importantly, RITA showed substantial suppression of cervical carcinoma xenografts in vivo. These results provide a proof of principle for the treatment of cervical cancer in a p53-dependent manner by using small molecules that target p53., ((c)2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
96. Therapeutic targeting of p53 by small molecules.
- Author
-
Selivanova G
- Subjects
- Animals, Humans, Neoplasms metabolism, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Small Molecule Libraries, Tumor Suppressor Protein p53 metabolism
- Abstract
Aberrant p53 function is one of the major requirements for tumor development. Reactivation of p53 function by small molecules is a promising strategy to combat cancer due to potent tumor suppressor activities of p53. Recent developments in p53 biology reveal that manipulation of p53 function might pave way to a long cancer-free life. A number of small molecules which rescue p53 function by different mechanisms, acting upstream of p53 or targeting the p53 protein itself have been identified. Notably, these molecules trigger different biological outcomes, suggesting that it might be feasible to direct p53-mediated response in a desired way. In this review I discuss the latest developments in the search for small molecules which rescue p53 function by targeting the p53 protein., (Copyright (c) 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
97. Integrins and mutant p53 on the road to metastasis.
- Author
-
Selivanova G and Ivaska J
- Subjects
- Animals, ErbB Receptors metabolism, Humans, Mice, Neoplasms pathology, Integrin alpha5beta1 metabolism, Neoplasm Metastasis, Tumor Suppressor Protein p53 metabolism
- Abstract
Understanding how tumor cells invade tissues is key to developing drugs to block metastasis. In this issue, Muller et al. (2009) report that a mutant form of the tumor suppressor p53 in cancer cells boosts the endocytic recycling of the adhesion molecule integrin alpha5beta1 and of epidermal growth factor receptor, promoting invasion and metastasis., (Copyright 2009 Elsevier Inc. All rights reserved.)
- Published
- 2009
- Full Text
- View/download PDF
98. p53-dependent inhibition of TrxR1 contributes to the tumor-specific induction of apoptosis by RITA.
- Author
-
Hedström E, Eriksson S, Zawacka-Pankau J, Arnér ES, and Selivanova G
- Subjects
- Auranofin pharmacology, Cell Line, Tumor, Down-Regulation genetics, Down-Regulation physiology, Humans, RNA, Small Interfering metabolism, Reactive Oxygen Species agonists, Reactive Oxygen Species metabolism, Thioredoxin Reductase 1 metabolism, Tumor Suppressor Protein p53 genetics, Up-Regulation genetics, Up-Regulation physiology, Apoptosis, Furans pharmacology, Neoplasms metabolism, Thioredoxin Reductase 1 antagonists & inhibitors, Tumor Suppressor Protein p53 metabolism
- Abstract
Thioredoxin reductase 1 (TrxR1) is a key regulator in many redox-dependent cellular pathways, and is often overexpressed in cancer. Several studies have identified TrxR1 as a potentially important target for anticancer therapy. The low molecular weight compound RITA (NSC 652287) binds p53 and induces p53-dependent apoptosis. Here we found that RITA also targets TrxR1 by non-covalent binding, followed by inhibition of its activity in vitro and by inhibition of TrxR activity in cancer cells. Interestingly, a novel approximately 130 kDa form of TrxR1, presumably representing a stable covalently linked dimer, and an increased generation of reactive oxygen species (ROS) were induced by RITA in cancer cells in a p53-dependent manner. Similarly, the gold-based TrxR inhibitor auranofin induced apoptosis related to oxidative stress, but independently of p53 and without apparent induction of the approximately 130 kDa form of TrxR1. In contrast to the effects observed in cancer cells, RITA did not inhibit TrxR or ROS formation in normal fibroblasts (NHDF). The inhibition of TrxR1 can sensitize tumor cells to agents that induce oxidative stress and may directly trigger cell death. Thus, our results suggest that a unique p53-dependent effect of RITA on TrxR1 in cancer cells might synergize with p53-dependent induction of pro-apoptotic genes and oxidative stress, thereby leading to a robust induction of cancer cell death, without affecting non-transformed cells.
- Published
- 2009
- Full Text
- View/download PDF
99. HIPK2 regulation by MDM2 determines tumor cell response to the p53-reactivating drugs nutlin-3 and RITA.
- Author
-
Rinaldo C, Prodosmo A, Siepi F, Moncada A, Sacchi A, Selivanova G, and Soddu S
- Subjects
- Apoptosis drug effects, Apoptosis physiology, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Line, Tumor, HCT116 Cells, Humans, Mitosis drug effects, Protein Serine-Threonine Kinases deficiency, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, RNA, Messenger biosynthesis, RNA, Messenger genetics, Up-Regulation drug effects, Carrier Proteins biosynthesis, Furans pharmacology, Imidazoles pharmacology, Piperazines pharmacology, Protein Serine-Threonine Kinases biosynthesis, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
In the past few years, much effort has been devoted to show the single-target specificity of nongenotoxic, p53 reactivating compounds. However, the divergent biological responses induced by the different compounds, even in the same tumor cells, demand additional mechanistic insights, whose knowledge may lead to improved drug design or selection of the most potent drug combinations. To address the molecular mechanism underlying induction of mitotic arrest versus clinically more desirable apoptosis, we took advantage of two MDM2 antagonists, Nutlin-3 and RITA, which respectively produce these two outcomes. We show that, along with p53 reactivation, the proapoptotic p53-activator HIPK2 is degraded by MDM2 in Nutlin-3-treated cells, but activated by transiently reduced MDM2 levels in RITA-treated ones. Gain- and loss-of-function experiments revealed the functional significance of MDM2-mediated HIPK2 regulation in cell decision between mitotic arrest and apoptosis in both types of p53 reactivation. These data indicate that strategies of p53 reactivation by MDM2 inhibition should also take into consideration MDM2 targets other than p53, such as the apoptosis activator HIPK2.
- Published
- 2009
- Full Text
- View/download PDF
100. Ablation of key oncogenic pathways by RITA-reactivated p53 is required for efficient apoptosis.
- Author
-
Grinkevich VV, Nikulenkov F, Shi Y, Enge M, Bao W, Maljukova A, Gluch A, Kel A, Sangfelt O, and Selivanova G
- Subjects
- Cell Line, Tumor, Down-Regulation, Furans pharmacology, Humans, Microtubule-Associated Proteins metabolism, Myeloid Cell Leukemia Sequence 1 Protein, Neoplasms pathology, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction, Small Molecule Libraries, beta Catenin metabolism, Apoptosis, Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Targeting "oncogene addiction" is a promising strategy for anticancer therapy. We report a potent inhibition of crucial oncogenes by p53 upon reactivation by small-molecule RITA in vitro and in vivo. RITA-activated p53 unleashes the transcriptional repression of antiapoptotic proteins Mcl-1, Bcl-2, MAP4, and survivin; blocks the Akt pathway on several levels; and downregulates c-Myc, cyclin E, and beta-catenin. p53 ablates c-Myc expression via several mechanisms at the transcriptional and posttranscriptional level. We show that the threshold for p53-mediated transrepression of survival genes is higher than for transactivation of proapoptotic targets. Inhibition of oncogenes by p53 reduces the cell's ability to buffer proapoptotic signals and elicits robust apoptosis. Our study highlights the role of transcriptional repression for p53-mediated tumor suppression.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.