Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.283
Filtrer
1.
Anal Chim Acta ; 1321: 343048, 2024 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-39155100

RÉSUMÉ

BACKGROUND: It is estimated that over 50 % of human cancers are caused by mutations in the p53 gene. Early sensitive and accurate detection of the p53 gene is important for diagnosis of cancers in the early stage. However, conventional detection techniques often suffer from strict reaction conditions, or unsatisfied sensitivity, so we need to develop a new strategy for accurate detection of p53 gene with smart designability, multiple signal amplification in mild reaction conditions. RESULTS: In this study, CRISPR/Cas system is exploited in entropy-driven catalysis (EDC) and hybridization chain reaction (CHA) dual signal amplification sensing strategies. The products of both reactions can efficiently and separately activate CRISPR/Cas12a which greatly amplifies the fluorescent signal. The method has good linearity in p53 detection with the concentration ranged from 0.1 fM to 0.5 pM with ultra-low detection limit of 0.096 fM. It also showed good performance in serum, offering potentials for early disease detection. SIGNIFICANCE: The designed dual amplification dynamic DNA network system exhibits an ultra-sensitive fluorescence biosensing for p53 gene identification. The method is simple to operate and requires only one buffer for the experiment, and meanwhile shows smart designability which could be used for a wide range of markers. Thus, we believe the present work will provide a potential tool for the construction and development of sensitive fluorescent biosensors for diseases.


Sujet(s)
Systèmes CRISPR-Cas , Protéine p53 suppresseur de tumeur , Protéine p53 suppresseur de tumeur/génétique , Systèmes CRISPR-Cas/génétique , Humains , Techniques d'amplification d'acides nucléiques , Techniques de biocapteur/méthodes , ADN/composition chimique , ADN/génétique , Limite de détection , Gènes p53 , Hybridation d'acides nucléiques
2.
J Toxicol Environ Health A ; 87(18): 752-761, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-38922576

RÉSUMÉ

Although the last pandemic created an urgency for development of vaccines, there was a continuous and concerted effort to search for therapeutic medications among existing drugs with different indications. One of the medications of interest that underwent this change was infliximab (IFM). This drug is used as an anti-inflammatory, predominantly in patients with Crohn 's disease, colitis ulcerative, and rheumatoid arthritis. In addition to these patients, individuals infected with Coronavirus Disease (COVID-19) were administered this chimeric monoclonal antibody (IMF) to act as an immunomodulator for patients in the absence of comprehensive research. Consequently, the present study aimed to examine the genotoxic effects attributed to IFM treatment employing different assays in vivo using mouse Mus musculus. Therefore, IFM was found to induce genotoxic effects as evidenced by the comet assay but did not demonstrate genotoxic potential utilizing mouse bone marrow MN test. The results of evaluating the expression of the P53 and BCL-2 genes using RT-qPCR showed stimulation of expression of these genes at 24 hr followed by a decline at 48 hr. Although the comet assay provided positive results, it is noteworthy that based upon negative findings in the micronucleus test, the data did not demonstrate significant changes in the genetic material that might affect the therapeutic use of IFM. The stimulation of expression of P53 and BCL-2 genes at 24 hr followed by a decline at 48 hr suggest a transient, if any, effect on genetic material. However, there is still a need for more research to more comprehensively understand the genotoxic profile of this medication.


Sujet(s)
Infliximab , Protéine p53 suppresseur de tumeur , Animaux , Souris , Protéine p53 suppresseur de tumeur/génétique , Altération de l'ADN/effets des médicaments et des substances chimiques , Test des comètes , Tests de micronucleus , Protéines proto-oncogènes c-bcl-2/génétique , Mâle , Gènes p53/effets des médicaments et des substances chimiques , Gènes bcl-2/effets des médicaments et des substances chimiques
3.
J Cardiothorac Surg ; 19(1): 376, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926874

RÉSUMÉ

PURPOSE: The purpose of this study is to investigate whether gene mutations can lead to the growth of malignant pulmonary nodules. METHODS: Retrospective analysis was conducted on patients with pulmonary nodules at Hebei Provincial People's Hospital, collecting basic clinical information such as gender, age, BMI, and hematological indicators. According to the inclusion and exclusion criteria, 85 patients with malignant pulmonary nodules were selected for screening, and gene mutation testing was performed on all patient tissues to explore the relationship between gene mutations and the growth of malignant pulmonary nodules. RESULTS: There is a correlation between KRAS and TP53 gene mutations and the growth of pulmonary nodules (P < 0.05), while there is a correlation between KRAS and TP53 gene mutations and the growth of pulmonary nodules in the subgroup of invasive malignant pulmonary nodules (P < 0.05). CONCLUSION: Mutations in the TP53 gene can lead to the growth of malignant pulmonary nodules and are correlated with the degree of invasion of malignant pulmonary nodules.


Sujet(s)
Tumeurs du poumon , Mutation , Protéines proto-oncogènes p21(ras) , Protéine p53 suppresseur de tumeur , Humains , Mâle , Femelle , Études rétrospectives , Adulte d'âge moyen , Protéines proto-oncogènes p21(ras)/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Sujet âgé , Nodules pulmonaires multiples/génétique , Nodules pulmonaires multiples/imagerie diagnostique , Nodules pulmonaires multiples/anatomopathologie , Adulte , Analyse de mutations d'ADN , Gènes p53/génétique
4.
J Clin Oncol ; 42(18): 2186-2195, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38569124

RÉSUMÉ

PURPOSE: There exists a barrier between developing and disseminating risk prediction models in clinical settings. We hypothesize that this barrier may be lifted by demonstrating the utility of these models using incomplete data that are collected in real clinical sessions, as compared with the commonly used research cohorts that are meticulously collected. MATERIALS AND METHODS: Genetic counselors (GCs) collect family history when patients (ie, probands) come to MD Anderson Cancer Center for risk assessment of Li-Fraumeni syndrome, a genetic disorder characterized by deleterious germline mutations in the TP53 gene. Our clinical counseling-based (CCB) cohort consists of 3,297 individuals across 124 families (522 cases of single primary cancer and 125 cases of multiple primary cancers). We applied our software suite LFSPRO to make risk predictions and assessed performance in discrimination using AUC and in calibration using observed/expected (O/E) ratio. RESULTS: For prediction of deleterious TP53 mutations, we achieved an AUC of 0.78 (95% CI, 0.71 to 0.85) and an O/E ratio of 1.66 (95% CI, 1.53 to 1.80). Using the LFSPRO.MPC model to predict the onset of the second cancer, we obtained an AUC of 0.70 (95% CI, 0.58 to 0.82). Using the LFSPRO.CS model to predict the onset of different cancer types as the first primary, we achieved AUCs between 0.70 and 0.83 for sarcoma, breast cancer, or other cancers combined. CONCLUSION: We describe a study that fills in the critical gap in knowledge for the utility of risk prediction models. Using a CCB cohort, our previously validated models have demonstrated good performance and outperformed the standard clinical criteria. Our study suggests that better risk counseling may be achieved by GCs using these already-developed mathematical models.


Sujet(s)
Syndrome de Li-Fraumeni , Humains , Syndrome de Li-Fraumeni/génétique , Appréciation des risques , Femelle , Mâle , Tumeurs primitives multiples/génétique , Protéine p53 suppresseur de tumeur/génétique , Mutation germinale , Conseil génétique , Adulte , Prédisposition génétique à une maladie , Gènes p53 , Adulte d'âge moyen
5.
Arch Pediatr ; 31(4): 238-244, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38679547

RÉSUMÉ

BACKGROUND: Recurrent genetic abnormalities affecting pivotal signaling pathways are the hallmark of childhood acute lymphoblastic leukemia (ALL). The identification of these aberrations remains clinically important. Therefore, we sought to determine the cytogenetic profile and the mutational status of TP53 and RAS genes among Moroccan childhood cases of ALL. METHODS: In total, 35 patients with childhood ALL were enrolled in the study. The diagnosis and treatment were established in the Pediatric Hematology and Oncology Center at the Children's Hospital of Rabat. Chromosome banding analysis and fluorescence in situ hybridization were used to detect genetic aberrations. Blood samples were screened for TP53 and RAS mutations using Sanger sequencing. RESULTS: Of the 35 cases, 30 were B-lineage ALL (85.7 %). Moreover, a male predominance was observed. Cytogenetic analysis revealed chromosomal anomalies in 27 cases (77.1 %). The most frequent aberrations were high hyperdiploidy and BCR/ABL rearrangement. Interestingly, we found the rare t(15;16) and the t(8;14), which are uncommon translocations in pediatric B-ALL. The mutational analysis revealed Pro72Arg (rs1042522:C > G) and Arg213Arg (rs1800372:A > G) in TP53. In correlation with cytogenetic data, rs1042522:C > G showed a significant association with the occurrence of chromosomal translocations (p = 0.04). However, no variant was detected in NRAS and KRAS genes. CONCLUSION: Our findings emphasize the significance of detecting chromosomal abnormalities as relevant prognostic markers. We also suggest a low occurrence of genetic variants among Moroccan children with ALL.


Sujet(s)
Aberrations des chromosomes , Leucémie-lymphome lymphoblastique à précurseurs B et T , Protéine p53 suppresseur de tumeur , Humains , Mâle , Maroc , Femelle , Enfant , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Enfant d'âge préscolaire , Nourrisson , Protéine p53 suppresseur de tumeur/génétique , Adolescent , Gènes ras/génétique , Mutation , Gènes p53/génétique
6.
Acta Neuropathol Commun ; 12(1): 57, 2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38605367

RÉSUMÉ

Li-Fraumeni syndrome (LFS) is an autosomal dominant tumor predisposition syndrome caused by heterozygous germline mutations or deletions in the TP53 tumor suppressor gene. Central nervous system tumors, such as choroid plexus tumors, medulloblastomas, and diffuse gliomas, are frequently found in patients with LFS. Although molecular profiles of diffuse gliomas that develop in pediatric patients with LFS have been elucidated, those in adults are limited. Recently, diffuse gliomas have been divided into pediatric- and adult-type gliomas, based on their distinct molecular profiles. In the present study, we investigated the molecular profiles of high-grade gliomas in three adults with LFS. These tumors revealed characteristic histopathological findings of high-grade glioma or glioblastoma and harbored wild-type IDH1/2 according to whole exome sequencing (WES). However, these tumors did not exhibit the key molecular alterations of glioblastoma, IDH-wildtype such as TERT promoter mutation, EGFR amplification, or chromosome 7 gain and 10 loss. Although WES revealed no other characteristic gene mutations or copy number alterations in high-grade gliomas, such as those in histone H3 genes, PDGFRA amplification was found in all three cases together with uniparental disomy of chromosome 17p, where the TP53 gene is located. DNA methylation analyses revealed that all tumors exhibited DNA methylation profiles similar to those of pediatric-type high-grade glioma H3-wildtype and IDH-wildtype (pHGG H3-/IDH-wt), RTK1 subtype. These data suggest that high-grade gliomas developed in adult patients with LFS may be involved in pHGG H3-/IDH-wt. PDGFRA and homozygous alterations in TP53 may play pivotal roles in the development of this type of glioma in adult patients with LFS.


Sujet(s)
Tumeurs du cerveau , Gliome , Syndrome de Li-Fraumeni , Adulte , Humains , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Gènes p53 , Glioblastome/génétique , Glioblastome/anatomopathologie , Gliome/génétique , Gliome/anatomopathologie , Isocitrate dehydrogenases/génétique , Syndrome de Li-Fraumeni/génétique , Mutation/génétique
7.
Genes Chromosomes Cancer ; 63(3): e23229, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38481055

RÉSUMÉ

A close relationship has been demonstrated between genomic complexity and clinical outcome in uterine smooth muscle tumors. We studied the genomic profiles by array-CGH of 28 fumarate hydratase deficient leiomyomas and 37 leiomyomas with bizarre nuclei (LMBN) from 64 patients. Follow-up was available for 46 patients (from three to 249 months, mean 87.3 months). All patients were alive without evidence of disease. For 51 array-CGH interpretable tumors the mean Genomic Index (GI) was 16.4 (median: 9.8; from 1 to 57.8), significantly lower than the mean GI in LMS (mean GI 51.8, p < 0.001). We described three groups: (1) a group with FH deletion (24/58) with low GI (mean GI: 11 vs. 22,4, p = 0.02), (2) a group with TP53 deletion (17/58) with higher GI (22.4 vs. 11 p = 0.02), and (3) a group without genomic events on FH or TP53 genes (17/58) (mean GI:18.3; from 1 to 57.8). Because none of these tumors recurred and none showed morphological features of LMS we concluded that GI at the cut-off of 10 was not applicable in these subtypes of LM. By integration of all those findings, a GI <10 in LMBN remains a valuable argument for benignity. Conversely, in LMBN a GI >10 or alteration in tumor suppressor genes, should not alone warrant a diagnosis of malignancy. Nine tumors were tested with Nanocind CINSARC® signature and all were classified in low risk of recurrence. We propose, based on our observations, a diagnostic approach of these challenging lesions.


Sujet(s)
Léiomyome , Tumeurs de l'utérus , Femelle , Humains , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Fumarate hydratase/génétique , Léiomyome/génétique , Léiomyome/anatomopathologie , Gènes p53 , Génomique
8.
Biochem Biophys Res Commun ; 709: 149834, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38547608

RÉSUMÉ

BACKGROUND: Cadmium exposure induces dermatotoxicity and epidermal barrier disruption and leads to the development of various pathologies. HaCaT cells are immortalized human keratinocytes that are widely used as alternatives to primary human keratinocytes, particularly for evaluating cadmium toxicity. HaCaT cells bear two gain-of-function (GOF) mutations in the TP53 gene, which strongly affect p53 function. Mutant forms of p53 are known to correlate with increased resistance to various stimuli, including exposure to cytotoxic substances. In addition, keratin 17 (KRT17) was recently shown to be highly expressed in HaCaT cells in response to genotoxic stress. Moreover, p53 is a direct transcriptional repressor of KRT17. However, the impact of TP53 mutations in HaCaT cells on the regulation of cell death and keratin 17 expression is unclear. In this study, we aimed to evaluate the impact of p53 on the response to Cd-induced cytotoxicity. METHODS AND RESULTS: Employing the MTT assay and Annexin V/propidium iodide staining, we demonstrated that knockout of TP53 leads to a decrease in the sensitivity of HaCaT cells to the cytotoxic effects of cadmium. Specifically, HaCaT cells with TP53 knockout (TP53 KO HaCaT) exhibited cell death at a cadmium concentration of 10 µM or higher, whereas wild-type cells displayed cell death at a concentration of 30 µM. Furthermore, apoptotic cells were consistently detected in TP53 KO HaCaT cells upon exposure to low concentrations of cadmium (10 and 20 µM) but not in wild-type cells. Our findings also indicate that cadmium cytotoxicity is mediated by reactive oxygen species (ROS), which were significantly increased only in TP53 knockout cells treated with 30 µM cadmium. An examination of proteomic data revealed that TP53 knockout in HaCaT cells resulted in the upregulation of proteins involved in the regulation of apoptosis, redox systems, and DNA repair. Moreover, RT‒qPCR and immunoblotting showed that cadmium toxicity leads to dose-dependent induction of keratin 17 in p53-deficient cells but not in wild-type cells. CONCLUSIONS: The connection between mutant p53 in HaCaT keratinocytes and increased resistance to cadmium toxicity was demonstrated for the first time. Proteomic profiling revealed that TP53 knockout in HaCaT cells led to the activation of apoptosis regulatory circuits, redox systems, and DNA repair. In addition, our data support the involvement of keratin 17 in the regulation of DNA repair and cell death. Apparently, the induction of keratin 17 is p53-independent but may be inhibited by mutant p53.


Sujet(s)
Gènes p53 , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Cadmium/métabolisme , Kératine-17/génétique , Kératine-17/métabolisme , Protéomique , Lignée cellulaire , Mort cellulaire , Kératinocytes/métabolisme , Apoptose/génétique
9.
Cancer Commun (Lond) ; 44(3): 297-360, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38311377

RÉSUMÉ

Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.


Sujet(s)
Tumeurs , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Amis , Gènes p53 , Mutation , Tumeurs/génétique , Tumeurs/thérapie , Microenvironnement tumoral
10.
JCO Precis Oncol ; 8: e2300453, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38412388

RÉSUMÉ

PURPOSE: Establishing accurate age-related penetrance figures for the broad range of cancer types that occur in individuals harboring a pathogenic germline variant in the TP53 gene is essential to determine the most effective clinical management strategies. These figures also permit optimal use of cosegregation data for classification of TP53 variants of unknown significance. Penetrance estimation can easily be affected by bias from ascertainment criteria, an issue not commonly addressed by previous studies. MATERIALS AND METHODS: We performed a maximum likelihood penetrance estimation using full pedigree data from a multicenter study of 146 TP53-positive families, incorporating adjustment for the effect of ascertainment and population-specific background cancer risks. The analysis included pedigrees from Australia, Spain, and United States, with phenotypic information for 4,028 individuals. RESULTS: Core Li-Fraumeni syndrome (LFS) cancers (breast cancer, adrenocortical carcinoma, brain cancer, osteosarcoma, and soft tissue sarcoma) had the highest hazard ratios of all cancers analyzed in this study. The analysis also detected a significantly increased lifetime risk for a range of cancers not previously formally associated with TP53 pathogenic variant status, including colorectal, gastric, lung, pancreatic, and ovarian cancers. The cumulative risk of any cancer type by age 50 years was 92.4% (95% CI, 82.2 to 98.3) for females and 59.7% (95% CI, 39.9 to 81.3) for males. Females had a 63.3% (95% CI, 35.6 to 90.1) cumulative risk of developing breast cancer by age 50 years. CONCLUSION: The results from maximum likelihood analysis confirm the known high lifetime risk for the core LFS-associated cancer types providing new risk estimates and indicate significantly increased lifetime risks for several additional cancer types. Accurate cancer risk estimates will help refine clinical recommendations for TP53 pathogenic variant carriers and improve TP53 variant classification.


Sujet(s)
Tumeurs du sein , Syndrome de Li-Fraumeni , Mâle , Femelle , Humains , États-Unis , Adulte d'âge moyen , Syndrome de Li-Fraumeni/diagnostic , Syndrome de Li-Fraumeni/génétique , Gènes p53/génétique , Pedigree , Protéine p53 suppresseur de tumeur/génétique , Prédisposition génétique à une maladie/génétique , Tumeurs du sein/génétique , Facteurs de risque
11.
Cancer Discov ; 14(2): 211-213, 2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-38327191

RÉSUMÉ

Mutant p53 proteins are often highly expressed in human cancers and have been thought to have oncogenic driver gain-of-function (GOF) properties. Wang and colleagues show, surprisingly, that this is not the case because removing the TP53-mutant gene from human and mouse cancer cells using CRISPR technology has no effect on cancer cell growth in vitro or in vivo. See related article by Wang et al., p. 362 (10) .


Sujet(s)
Mutation gain de fonction , Protéine p53 suppresseur de tumeur , Humains , Souris , Animaux , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Délires , Lignée cellulaire tumorale , Gènes p53 , Mutation
12.
Cancer Lett ; 584: 216608, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38199587

RÉSUMÉ

Poly (ADP-ribose) polymerase inhibitors (PARPi) are used for patients with BRCA1/2 mutations, but patients with other mutations may benefit from PARPi treatment. Another mutation that is present in more cancers than BRCA1/2 is mutation to the TP53 gene. In 2D breast cancer cell lines, mutant p53 (mtp53) proteins tightly associate with replicating DNA and Poly (ADP-ribose) polymerase (PARP) protein. Combination drug treatment with the alkylating agent temozolomide and the PARPi talazoparib kills mtp53 expressing 2D grown breast cancer cell lines. We evaluated the sensitivity to the combination of temozolomide plus PARPi talazoparib treatment to breast and lung cancer patient-derived tumor organoids (PDTOs). The combination of the two drugs was synergistic for a cytotoxic response in PDTOs with mtp53 but not for PDTOs with wtp53. The combination of talazoparib and temozolomide induced more DNA double-strand breaks in mtp53 expressing organoids than in wild-type p53 expressing organoids as shown by increased γ-H2AX protein expression. Moreover, breast cancer tissue microarrays (TMAs) showed a positive correlation between stable p53 and high PARP1 expression in sub-groups of breast cancers, which may indicate sub-classes of breast cancers sensitive to PARPi therapy. These results suggest that mtp53 could be a biomarker to predict response to the combination of PARPi talazoparib-temozolomide treatment.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Tumeurs du poumon , Femelle , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Protéine BRCA1/génétique , Protéine BRCA1/métabolisme , Protéine BRCA2/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , ADN , Gènes p53 , Tumeurs du poumon/génétique , Mutation , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Poly(ADP-ribose) polymerases/métabolisme , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
13.
Cancer Lett ; 585: 216645, 2024 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-38280477

RÉSUMÉ

The TP53 gene, encoding the p53 protein, has been a focal point of research since its 1979 discovery, playing a crucial role in tumor suppression. Ferroptosis, a distinct form of cell death characterized by lipid peroxide accumulation, has gained prominence since its recognition in 2012. Recent studies have unveiled an intriguing connection between p53 and ferroptosis, with implications for cancer therapy. Recent research underscores p53 as a novel target for cancer therapy, influencing key metabolic processes in ferroptosis. Notably, p53 represses the expression of the cystine-glutamate antiporter SLC7A11, supporting p53-mediated tumor growth suppression. Furthermore, under metabolic stress, p53 mitigates ferroptosis sensitivity, aiding cancer cells in coping and delaying cell death. This dynamic interplay between p53 and ferroptosis has far-reaching implications for various diseases, particularly cancer. This review provides a comprehensive overview of ferroptosis in cancer cells, elucidating p53's role in regulating ferroptosis, and explores the potential of targeting p53 to induce ferroptosis for cancer therapy. Understanding this complex relationship between p53 and ferroptosis offers a promising avenue for developing innovative cancer treatments.


Sujet(s)
Ferroptose , Tumeurs , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Gènes p53 , Médecine de précision , Ferroptose/génétique , Espèces réactives de l'oxygène/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/génétique
15.
Mol Biol Rep ; 51(1): 205, 2024 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-38270700

RÉSUMÉ

Increasing evidence suggests that key cancer-causing driver genes continue to exert a sustained influence on the tumor microenvironment (TME), highlighting the importance of immunotherapeutic targeting of gene mutations in governing tumor progression. TP53 is a prominent tumor suppressor that encodes the p53 protein, which controls the initiation and progression of different tumor types. Wild-type p53 maintains cell homeostasis and genomic instability through complex pathways, and mutant p53 (Mut p53) promotes tumor occurrence and development by regulating the TME. To date, it has been wildly considered that TP53 is able to mediate tumor immune escape. Herein, we summarized the relationship between TP53 gene and tumors, discussed the mechanism of Mut p53 mediated tumor immune escape, and summarized the progress of applying p53 protein in immunotherapy. This study will provide a basic basis for further exploration of therapeutic strategies targeting p53 protein.


Sujet(s)
Tumeurs , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Gènes p53 , Tumeurs/génétique , Cognition , Instabilité du génome , Microenvironnement tumoral/génétique
17.
Article de Anglais | MEDLINE | ID: mdl-37801337

RÉSUMÉ

INTRODUCTION: In 1969, Li-Fraumeni syndrome (LFS), which is a rare cancer predisposition syndrome, was reported for the first time. The main problem in LFS is the mutation in the TP53 gene, which is a crucial tumor suppressor gene in the cell cycle. A hereditary syndrome is inherited in an autosomal dominant pattern. There is a significant correlation between this syndrome and various cancers such as sarcoma, breast cancer, brain tumors, and different other types of malignancies. This study aimed to identify the possibility of LFS in cancer patients in the East Azarbaijan, Iran. METHODS: In this experimental study, 45 children with cancer in the Northwest of Iran were investigated for LFS. DNA was extracted from the whole blood cells using the salting-out method. The region within the exons 5-8 of the TP53 gene has been replicated via Polymerase Chain Reaction (PCR) method. The PCR products were sent for Sanger sequencing, and finally, the data were analyzed by Chromas software. RESULTS: In the studied probands, in 12 (26.67%) cases, polymorphisms in Exon 6 and Introns 6 and Intron 7 were identified, and no mutation was observed in exons 5-8 of the TP53 gene. CONCLUSION: Our results show that there were no mutations in exons 5-8 of the TP53 gene as an indication of LFS possibility in these families. Further studies are needed to be done in a bigger population, and Next-Generation Sequencing (NGS) needs to be done to evaluate the whole genome of these patients to complete our data.


Sujet(s)
Syndrome de Li-Fraumeni , Enfant , Humains , Syndrome de Li-Fraumeni/génétique , Gènes p53 , Iran , Protéine p53 suppresseur de tumeur/génétique , Mutation germinale , Prédisposition génétique à une maladie
18.
Can J Physiol Pharmacol ; 102(1): 55-68, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37818839

RÉSUMÉ

This study concerned with assessing the effect of restoring p53 using PRIMA-1 on the anti-cancer activity of olaparib against TP53-mutant triple negative breast cancer (TNBC) cells and exploring the optimum synergistic concentrations and the underlying mechanism. Human BC cell lines, MDA-MB-231 with mutated TP53 gene, and MCF-7 with wild-type TP53 gene were treated with olaparib and/or PRIMA-1. The IC50 value for olaparib was significantly decreased by PRIMA-1 in MDA-MB-231 cells compared to MCF-7 cells. Contrary to MCF-7 cells, co-treatment with olaparib and PRIMA-1 had a synergistic anti-proliferative effect in MDA-MB-231 at all tested concentrations with the best synergistic combination at 45 and 8.5 µM, respectively, and furthermore PRIMA-1 enhanced olaparib-induced apoptosis. This synergistic apoptotic effect was associated with a significant boost in mRNA expression of TP53 gene, cell cycle arrest at G2/M phase, modulation of BRCA-1, BAX and Bcl2 proteins expressions, and induction of active caspase-3. These results present a clue for the utility of combined olaparib and PRIMA-1 in treatment of TP53-mutant TNBC invitro. PRIMA-1 triggers olaparib-induced MDA-MB-231 cell death in a synergistic manner via restoring TP53, decreasing BRCA-1 expression, cell cycle arrest, and enhancement of apoptosis via p53/BAX/Bcl2/caspase 3 pathway.


Sujet(s)
Tumeurs du sein triple-négatives , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Gènes p53 , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Protéine Bax/métabolisme , Lignée cellulaire tumorale , Apoptose , Mort cellulaire , Division cellulaire , Cycle cellulaire , Prolifération cellulaire
19.
Cancer Discov ; 14(1): 104-119, 2024 01 12.
Article de Anglais | MEDLINE | ID: mdl-37874259

RÉSUMÉ

People with Li-Fraumeni syndrome (LFS) harbor a germline pathogenic variant in the TP53 tumor suppressor gene, face a near 100% lifetime risk of cancer, and routinely undergo intensive surveillance protocols. Liquid biopsy has become an attractive tool for a range of clinical applications, including early cancer detection. Here, we provide a proof-of-principle for a multimodal liquid biopsy assay that integrates a targeted gene panel, shallow whole-genome, and cell-free methylated DNA immunoprecipitation sequencing for the early detection of cancer in a longitudinal cohort of 89 LFS patients. Multimodal analysis increased our detection rate in patients with an active cancer diagnosis over uni-modal analysis and was able to detect cancer-associated signal(s) in carriers prior to diagnosis with conventional screening (positive predictive value = 67.6%, negative predictive value = 96.5%). Although adoption of liquid biopsy into current surveillance will require further clinical validation, this study provides a framework for individuals with LFS. SIGNIFICANCE: By utilizing an integrated cell-free DNA approach, liquid biopsy shows earlier detection of cancer in patients with LFS compared with current clinical surveillance methods such as imaging. Liquid biopsy provides improved accessibility and sensitivity, complementing current clinical surveillance methods to provide better care for these patients. See related commentary by Latham et al., p. 23. This article is featured in Selected Articles from This Issue, p. 5.


Sujet(s)
Acides nucléiques acellulaires , Syndrome de Li-Fraumeni , Humains , Syndrome de Li-Fraumeni/diagnostic , Syndrome de Li-Fraumeni/génétique , Syndrome de Li-Fraumeni/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Dépistage précoce du cancer , Acides nucléiques acellulaires/génétique , Gènes p53 , Mutation germinale , Prédisposition génétique à une maladie
20.
Cell Death Differ ; 31(1): 1-8, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-38001255

RÉSUMÉ

Multiple Myeloma is a typical example of a neoplasm that shows significant differences in incidence, age of onset, type, and frequency of genetic alterations between patients of African and European ancestry. This perspective explores the hypothesis that both genetic polymorphisms and spontaneous somatic mutations in the TP53 tumor suppressor gene are determinants of these differences. In the US, the rates of occurrence of MM are at least twice as high in African Americans (AA) as in Caucasian Americans (CA). Strikingly, somatic TP53 mutations occur in large excess (at least 4-6-fold) in CA versus AA. On the other hand, TP53 contains polymorphisms specifying amino-acid differences that are under natural selection by the latitude of a population and have evolved during the migrations of humans over several hundred thousand years. The p53 protein plays important roles in DNA strand break repair and, therefore, in the surveillance of aberrant DNA recombination, leading to the B-cell translocations that are causal in the pathogenesis of MM. We posit that polymorphisms in one region of the TP53 gene (introns 2 and 3, and the proline-rich domain) specify a concentration of the p53 protein with a higher capacity to repress translocations in CA than AA patients. This, in turn, results in a higher risk of acquiring inactivating, somatic mutations in a different region of the TP53 gene (DNA binding domain) in CA than in AA patients. Such a mechanism, by which the polymorphic status of a gene influencing its own "spontaneous" mutation frequency, may provide a genetic basis to address ethnicity-related differences in the incidence and phenotypes of many different forms of cancer.


Sujet(s)
Myélome multiple , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/génétique , Myélome multiple/génétique , Mutation , Gènes p53 , Translocation génétique , ADN
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE