Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.931
Filtrer
2.
Proc Natl Acad Sci U S A ; 121(29): e2405231121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38990952

RÉSUMÉ

We report that ~1.8% of all mesothelioma patients and 4.9% of those younger than 55, carry rare germline variants of the BRCA1 associated RING domain 1 (BARD1) gene that were predicted to be damaging by computational analyses. We conducted functional assays, essential for accurate interpretation of missense variants, in primary fibroblasts that we established in tissue culture from a patient carrying the heterozygous BARD1V523A mutation. We found that these cells had genomic instability, reduced DNA repair, and impaired apoptosis. Investigating the underlying signaling pathways, we found that BARD1 forms a trimeric protein complex with p53 and SERCA2 that regulates calcium signaling and apoptosis. We validated these findings in BARD1-silenced primary human mesothelial cells exposed to asbestos. Our study elucidated mechanisms of BARD1 activity and revealed that heterozygous germline BARD1 mutations favor the development of mesothelioma and increase the susceptibility to asbestos carcinogenesis. These mesotheliomas are significantly less aggressive compared to mesotheliomas in asbestos workers.


Sujet(s)
Signalisation calcique , Réparation de l'ADN , Prédisposition génétique à une maladie , Mutation germinale , Mésothéliome , Protéines suppresseurs de tumeurs , Ubiquitin-protein ligases , Humains , Réparation de l'ADN/génétique , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Mésothéliome/génétique , Signalisation calcique/génétique , Femelle , Mâle , Adulte d'âge moyen , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Apoptose/génétique , Fibroblastes/métabolisme , Amiante/toxicité , Instabilité du génome
3.
Nat Genet ; 56(7): 1434-1445, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38969833

RÉSUMÉ

Many variants that we inherit from our parents or acquire de novo or somatically are rare, limiting the precision with which we can associate them with disease. We performed exhaustive saturation genome editing (SGE) of BAP1, the disruption of which is linked to tumorigenesis and altered neurodevelopment. We experimentally characterized 18,108 unique variants, of which 6,196 were found to have abnormal functions, and then used these data to evaluate phenotypic associations in the UK Biobank. We also characterized variants in a large population-ascertained tumor collection, in cancer pedigrees and ClinVar, and explored the behavior of cancer-associated variants compared to that of variants linked to neurodevelopmental phenotypes. Our analyses demonstrated that disruptive germline BAP1 variants were significantly associated with higher circulating levels of the mitogen IGF-1, suggesting a possible pathological mechanism and therapeutic target. Furthermore, we built a variant classifier with >98% sensitivity and specificity and quantify evidence strengths to aid precision variant interpretation.


Sujet(s)
Édition de gène , Mutation germinale , Protéines suppresseurs de tumeurs , Ubiquitin thiolesterase , Humains , Mutation germinale/génétique , Ubiquitin thiolesterase/génétique , Protéines suppresseurs de tumeurs/génétique , Édition de gène/méthodes , Tumeurs/génétique , Prédisposition génétique à une maladie , Pedigree , Femelle , Mâle
4.
Elife ; 132024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38995680

RÉSUMÉ

Truncation of the protein-protein interaction SH3 domain of the membrane remodeling Bridging Integrator 1 (BIN1, Amphiphysin 2) protein leads to centronuclear myopathy. Here, we assessed the impact of a set of naturally observed, previously uncharacterized BIN1 SH3 domain variants using conventional in vitro and cell-based assays monitoring the BIN1 interaction with dynamin 2 (DNM2) and identified potentially harmful ones that can be also tentatively connected to neuromuscular disorders. However, SH3 domains are typically promiscuous and it is expected that other, so far unknown partners of BIN1 exist besides DNM2, that also participate in the development of centronuclear myopathy. In order to shed light on these other relevant interaction partners and to get a holistic picture of the pathomechanism behind BIN1 SH3 domain variants, we used affinity interactomics. We identified hundreds of new BIN1 interaction partners proteome-wide, among which many appear to participate in cell division, suggesting a critical role of BIN1 in the regulation of mitosis. Finally, we show that the identified BIN1 mutations indeed cause proteome-wide affinity perturbation, signifying the importance of employing unbiased affinity interactomic approaches.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Myopathies congénitales structurales , Protéines nucléaires , Protéines suppresseurs de tumeurs , Domaine d'homologie SRC , Myopathies congénitales structurales/métabolisme , Myopathies congénitales structurales/génétique , Humains , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Liaison aux protéines , Dynamine-II/métabolisme , Dynamine-II/génétique , Mutation
5.
Medicine (Baltimore) ; 103(28): e38867, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996143

RÉSUMÉ

BACKGROUND: Lung adenocarcinoma (LUAD) represents the most prevalent type of lung cancer. SHOX2 and RASSF1A methylation have been identified as important biomarkers for diagnosis and prognosis of lung cancer. Bronchoalveolar lavage fluid (BALF) exhibits good specificity and sensitivity in diagnosing pulmonary diseases, but its acquisition is challenging and may cause discomfort to patients. In clinical, plasma samples are more convenient to obtain than BALF; however, there is little research on the concurrent detection of SHOX2 and RASSF1A methylation in plasma. This study aims to assess the diagnostic value of a combined promoter methylation assay for SHOX2 and RASSF1A in early-stage LUAD using plasma samples. METHODS: BALF and blood samples were obtained from 36 early-stage LUAD patients, with a control group of nineteen non-tumor individuals. The promoter methylation levels of SHOX2 and RASSF1A in all subjects were assessed using the human SHOX2 and RASSF1A gene methylation kit. RESULTS: The methylation detection rate of SHOX2 and RASSF1A in plasma was 61.11%, slightly lower than that in BALF (66.7%). The Chi-square test revealed no significant difference in the methylation rate between BALF and plasma (P > 0.05). The area under the receiver operating characteristic (ROC) curve analysis for blood was 0.806 (95% CI, 0.677 to 0.900), while for BALF it was 0.781 (95% CI, 0.649 to 0.881). Additionally, we conducted an analysis on the correlation between SHOX2 and RASSF1A methylation levels in plasma with gender, age, tumor differentiation, pathologic classification, and other clinicopathological variables; however, no significant correlations were observed. CONCLUSIONS: Measurement of SHOX2 and RASSF1A methylation for early diagnosis of LUAD can be achieved with high sensitivity and specificity by using plasma as a substitute for BALF samples.


Sujet(s)
Adénocarcinome pulmonaire , Marqueurs biologiques tumoraux , Méthylation de l'ADN , Dépistage précoce du cancer , Protéines à homéodomaine , Tumeurs du poumon , Régions promotrices (génétique) , Protéines suppresseurs de tumeurs , Humains , Mâle , Femelle , Adulte d'âge moyen , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/sang , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/diagnostic , Adénocarcinome pulmonaire/sang , Tumeurs du poumon/diagnostic , Tumeurs du poumon/génétique , Tumeurs du poumon/sang , Dépistage précoce du cancer/méthodes , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/génétique , Sujet âgé , Protéines à homéodomaine/génétique , Protéines à homéodomaine/sang , Liquide de lavage bronchoalvéolaire/composition chimique , Courbe ROC , Adulte , Sensibilité et spécificité , Études cas-témoins
6.
Cell Death Dis ; 15(7): 466, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956029

RÉSUMÉ

Metastasis is the major culprit of treatment failure in nasopharyngeal carcinoma (NPC). Aryl hydrocarbon receptor nuclear translocator like 2 (ARNTL2), a core circadian gene, plays a crucial role in the development of various tumors. Nevertheless, the biological role and mechanism of ARNTL2 are not fully elucidated in NPC. In this study, ARNTL2 expression was significantly upregulated in NPC tissues and cells. Overexpression of ARNTL2 facilitated NPC cell migration and invasion abilities, while inhibition of ARNTL2 in similarly treated cells blunted migration and invasion abilities in vitro. Consistently, in vivo xenograft tumor models revealed that ARNTL2 silencing reduced nude mice inguinal lymph node and lung metastases, as well as tumor growth. Mechanistically, ARNTL2 negatively regulated the transcription expression of AMOTL2 by directly binding to the AMOTL2 promoter, thus reducing the recruitment and stabilization of AMOTL2 to LATS1/2 kinases, which strengthened YAP nuclear translocation by suppressing LATS-dependent YAP phosphorylation. Inhibition of AMOTL2 counteracted the effects of ARNTL2 knockdown on NPC cell migration and invasion abilities. These findings suggest that ARNTL2 may be a promising therapeutic target to combat NPC metastasis and further supports the crucial roles of circadian genes in cancer development.


Sujet(s)
Facteurs de transcription ARNTL , Protéines adaptatrices de la transduction du signal , Angiomotines , Mouvement cellulaire , Souris nude , Cancer du nasopharynx , Tumeurs du rhinopharynx , Invasion tumorale , Facteurs de transcription , Protéines de signalisation YAP , Humains , Animaux , Cancer du nasopharynx/génétique , Cancer du nasopharynx/anatomopathologie , Cancer du nasopharynx/métabolisme , Lignée cellulaire tumorale , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines de signalisation YAP/métabolisme , Mouvement cellulaire/génétique , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription ARNTL/métabolisme , Facteurs de transcription ARNTL/génétique , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/anatomopathologie , Tumeurs du rhinopharynx/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Transduction du signal , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Mâle , Métastase tumorale , Femelle , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique
7.
BMC Med Genomics ; 17(1): 176, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956580

RÉSUMÉ

BACKGROUND: Intellectual disability (ID) is a neurodevelopmental condition affecting around 2% of children and young adults worldwide, characterized by deficits in intellectual functioning and adaptive behavior. Genetic factors contribute to the development of ID phenotypes, including mutations and structural changes in chromosomes. Pathogenic variants in the HCFC1 gene cause X-linked mental retardation syndrome, also known as Siderius type X-linked mental retardation. The MN1 gene is necessary for palate development, and mutations in this gene result in a genetic condition called CEBALID syndrome. METHODS: Exome sequencing was used to identify the disease-causing variants in two affected families, A and B, from various regions of Pakistan. Affected individuals in these two families presented ID, developmental delay, and behavioral abnormalities. The validation and co-segregation analysis of the filtered variant was carried out using Sanger sequencing. RESULTS: In an X-linked family A, a novel hemizygous missense variant (c.5705G > A; p.Ser1902Asn) in the HCFC1 gene (NM_005334.3) was identified, while in family B exome sequencing revealed a heterozygous nonsense variant (c.3680 G > A; p. Trp1227Ter) in exon-1 of the MN1 gene (NM_032581.4). Sanger sequencing confirmed the segregation of these variants with ID in each family. CONCLUSIONS: The investigation of two Pakistani families revealed pathogenic genetic variants in the HCFC1 and MN1 genes, which cause ID and expand the mutational spectrum of these genes.


Sujet(s)
Facteur de prolifération cellulaire HCF , Déficience intellectuelle , Pedigree , Humains , Pakistan , Mâle , Déficience intellectuelle/génétique , Femelle , Facteur de prolifération cellulaire HCF/génétique , Protéines suppresseurs de tumeurs/génétique , Transactivateurs/génétique , Enfant , , Enfant d'âge préscolaire
8.
Sci Adv ; 10(27): eadk8958, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959315

RÉSUMÉ

The luminal-to-basal transition in mammary epithelial cells (MECs) is accompanied by changes in epithelial cell lineage plasticity; however, the underlying mechanism remains elusive. Here, we report that deficiency of Frmd3 inhibits mammary gland lineage development and induces stemness of MECs, subsequently leading to the occurrence of triple-negative breast cancer. Loss of Frmd3 in PyMT mice results in a luminal-to-basal transition phenotype. Single-cell RNA sequencing of MECs indicated that knockout of Frmd3 inhibits the Notch signaling pathway. Mechanistically, FERM domain-containing protein 3 (FRMD3) promotes the degradation of Disheveled-2 by disrupting its interaction with deubiquitinase USP9x. FRMD3 also interrupts the interaction of Disheveled-2 with CK1, FOXK1/2, and NICD and decreases Disheveled-2 phosphorylation and nuclear localization, thereby impairing Notch-dependent luminal epithelial lineage plasticity in MECs. A low level of FRMD3 predicts poor outcomes for breast cancer patients. Together, we demonstrated that FRMD3 is a tumor suppressor that functions as an endogenous activator of the Notch signaling pathway, facilitating the basal-to-luminal transformation in MECs.


Sujet(s)
Cellules épithéliales , Récepteurs Notch , Transduction du signal , Animaux , Cellules épithéliales/métabolisme , Femelle , Récepteurs Notch/métabolisme , Humains , Souris , Lignage cellulaire , Glandes mammaires animales/métabolisme , Glandes mammaires animales/cytologie , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Différenciation cellulaire , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/génétique
10.
PLoS One ; 19(7): e0304194, 2024.
Article de Anglais | MEDLINE | ID: mdl-38968186

RÉSUMÉ

Deficiencies in the ING4 tumor suppressor are associated with advanced stage tumors and poor patient survival in cancer. ING4 was shown to inhibit NF-kB in several cancers. As NF-kB is a key mediator of immune response, the ING4/NF-kB axis is likely to manifest in tumor-immune modulation but has not been investigated. To characterize the tumor immune microenvironment associated with ING4-deficient tumors, three approaches were employed in this study: First, tissue microarrays composed of 246 primary breast tumors including 97 ING4-deficient tumors were evaluated for the presence of selective immune markers, CD68, CD4, CD8, and PD-1, using immunohistochemical staining. Second, an immune-competent mouse model of ING4-deficient breast cancer was devised utilizing CRISPR-mediated deletion of Ing4 in a Tp53 deletion-derived mammary tumor cell line; mammary tumors were evaluated for immune markers using flow cytometry. Lastly, the METABRIC gene expression dataset was evaluated for patient survival related to the immune markers associated with Ing4-deleted tumors. The results showed that CD68, CD4, CD8, or PD-1, was not significantly associated with ING4-deficient breast tumors, indicating no enrichment of macrophages, T cells, or exhausted T cell types. In mice, Ing4-deleted mammary tumors had a growth rate comparable to Ing4-intact tumors but showed increased tumor penetrance and metastasis. Immune marker analyses of Ing4-deleted tumors revealed a significant increase in tumor-associated macrophages (Gr-1loCD11b+F4/80+) and a decrease in granzyme B-positive (GzmB+) CD4+ T cells, indicating a suppressive and/or less tumoricidal immune microenvironment. The METABRIC data analyses showed that low expression of GZMB was significantly associated with poor patient survival, as was ING4-low expression, in the basal subtype of breast cancer. Patients with GZMB-low/ING4-low tumors had the worst survival outcomes (HR = 2.80, 95% CI 1.36-5.75, p = 0.0004), supportive of the idea that the GZMB-low immune environment contributes to ING4-deficient tumor progression. Collectively, the study results demonstrate that ING4-deficient tumors harbor a microenvironment that contributes to immune evasion and metastasis.


Sujet(s)
Tumeurs du sein , Protéines du cycle cellulaire , Protéines à homéodomaine , Microenvironnement tumoral , Protéines suppresseurs de tumeurs , Microenvironnement tumoral/immunologie , Animaux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Tumeurs du sein/génétique , Femelle , Humains , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/déficit , Protéines suppresseurs de tumeurs/métabolisme , Souris , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Lignée cellulaire tumorale , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/déficit , Évolution de la maladie , Métastase tumorale
11.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38991726

RÉSUMÉ

PPTC7 is a mitochondrial-localized phosphatase that suppresses BNIP3- and NIX-mediated mitophagy, but the mechanisms underlying this regulation remain ill-defined. Here, we demonstrate that loss of PPTC7 upregulates BNIP3 and NIX post-transcriptionally and independent of HIF-1α stabilization. Loss of PPTC7 prolongs the half-life of BNIP3 and NIX while blunting their accumulation in response to proteasomal inhibition, suggesting that PPTC7 promotes the ubiquitin-mediated turnover of BNIP3 and NIX. Consistently, overexpression of PPTC7 limits the accumulation of BNIP3 and NIX protein levels, which requires an intact catalytic motif but is surprisingly independent of its targeting to mitochondria. Consistently, we find that PPTC7 is dual-localized to the outer mitochondrial membrane and the matrix. Importantly, anchoring PPTC7 to the outer mitochondrial membrane is sufficient to blunt BNIP3 and NIX accumulation, and proximity labeling and fluorescence co-localization experiments demonstrate that PPTC7 dynamically associates with BNIP3 and NIX within the native cellular environment. Collectively, these data reveal that a fraction of PPTC7 localizes to the outer mitochondrial membrane to promote the proteasomal turnover of BNIP3 and NIX, limiting basal mitophagy.


Sujet(s)
Protéines membranaires , Mitochondries , Membranes mitochondriales , Protéines mitochondriales , Mitophagie , Protéines proto-oncogènes , Mitophagie/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Humains , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Mitochondries/métabolisme , Membranes mitochondriales/métabolisme , Phosphoprotein Phosphatases/métabolisme , Phosphoprotein Phosphatases/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Cellules HeLa , Animaux
12.
Sci Rep ; 14(1): 16031, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992201

RÉSUMÉ

O6-methylguanine-DNA methyltransferase (MGMT) has been demonstrated to be an important prognostic and predictive marker in glioblastoma (GBM). To establish a reliable radiomics model based on MRI data to predict the MGMT promoter methylation status of GBM. A total of 183 patients with glioblastoma were included in this retrospective study. The visually accessible Rembrandt images (VASARI) features were extracted for each patient, and a total of 14676 multi-region features were extracted from enhanced, necrotic, "non-enhanced, and edematous" areas on their multiparametric MRI. Twelve individual radiomics models were constructed based on the radiomics features from different subregions and different sequences. Four single-sequence models, three single-region models and the combined radiomics model combining all individual models were constructed. Finally, the predictive performance of adding clinical factors and VASARI characteristics was evaluated. The ComRad model combining all individual radiomics models exhibited the best performance in test set 1 and test set 2, with the area under the receiver operating characteristic curve (AUC) of 0.839 (0.709-0.963) and 0.739 (0.581-0.897), respectively. The results indicated that the radiomics model combining multi-region and multi-parametric MRI features has exhibited promising performance in predicting MGMT methylation status in GBM. The Modeling scheme that combining all individual radiomics models showed best performance among all constructed moels.


Sujet(s)
Tumeurs du cerveau , Méthylation de l'ADN , DNA modification methylases , Enzymes de réparation de l'ADN , Glioblastome , Imagerie par résonance magnétique , Régions promotrices (génétique) , Protéines suppresseurs de tumeurs , Humains , Glioblastome/génétique , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Enzymes de réparation de l'ADN/génétique , DNA modification methylases/génétique , Protéines suppresseurs de tumeurs/génétique , Imagerie par résonance magnétique/méthodes , Femelle , Mâle , Adulte d'âge moyen , Tumeurs du cerveau/génétique , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/anatomopathologie , Études rétrospectives , Adulte , Sujet âgé , Pronostic , Courbe ROC ,
13.
BMC Oral Health ; 24(1): 780, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992585

RÉSUMÉ

BACKGROUND: This study delves into the intricate landscape of oral cancer, a global concern with a high incidence in Asian countries. We focus on oral squamous cell carcinoma (OSCC), primarily driven by the consumption of betel nut and its derivatives. OSCC often arises from premalignant lesions like oral submucous fibrosis (OSF). In Pakistan, OSCC is prevalent among men due to various addictive substances, including smokeless tobacco and chewing materials. Mutations in tumor suppressor genes, such as TP53 and p21, play crucial roles in this malignancy's development. We also explore the involvement of TUSC3 gene deletion in OSCC and OSF. METHODS: In this study we investigated demographics, TUSC3 gene expression, deletion analysis, and TP53 and p21 genetic alterations in OSCC and OSF patients (blood and tissue of 50 samples in each condition) who had tobacco derivates usage history. The association analysis was carried out mainly through PCR based genotyping. RESULTS: The study's patient cohort (OSCC and OSF) displayed a wide age range from 13 to 65 years (Mean = 32.96 years). Both conditions were more prevalent in males, with a male-female ratio of approximately 2.5:1. Chewing habits analysis revealed high frequencies of gutka use in both OSF and OSCC patients. TUSC3 expression analysis in OSCC cell lines indicated significant downregulation. Genotyping showed no TUSC3 deletion in OSF cases, but a deletion rate of over 22% in OSCC tissue samples. Analysis supported a significant association of TUSC3 deletion with OSCC development but not with OSF. Polymorphism in p53 exon 4 and p21 (rs1801270) were significantly associated with both OSCC and OSF, adding to their pathogenesis. Our findings further revealed a strong correlation between TUSC3 deletion and the excessive use of tobacco and related products, shedding light on the genetic underpinnings of OSCC development. CONCLUSIONS: Notably, our study provides a crucial insight into genetic aspects underlying OSCC and OSF in response of addictive consumption of areca nut, betel quid, and tobacco derivatives. A significant association between TUSC3 deletion and OSCC development, along with polymorphisms in TP53 and p21, underscores the importance of further research into the molecular mechanisms driving oral cancer progression for improved diagnosis and treatment outcomes.


Sujet(s)
Carcinome épidermoïde , Inhibiteur p21 de kinase cycline-dépendante , Protéines membranaires , Tumeurs de la bouche , Fibrose buccale sous-muqueuse , Tabac sans fumée , Protéine p53 suppresseur de tumeur , Humains , Mâle , Fibrose buccale sous-muqueuse/génétique , Tumeurs de la bouche/génétique , Tumeurs de la bouche/anatomopathologie , Femelle , Adulte , Adulte d'âge moyen , Carcinome épidermoïde/génétique , Pakistan , Sujet âgé , Tabac sans fumée/effets indésirables , Jeune adulte , Inhibiteur p21 de kinase cycline-dépendante/génétique , Adolescent , Protéines membranaires/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéines suppresseurs de tumeurs/génétique , Areca/effets indésirables , Délétion de gène , Facteurs sexuels
14.
Sci Rep ; 14(1): 15800, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982123

RÉSUMÉ

Although sex differences have been reported in patients with clear cell renal cell carcinoma (ccRCC), biological sex has not received clinical attention and genetic differences between sexes are poorly understood. This study aims to identify sex-specific gene mutations and explore their clinical significance in ccRCC. We used data from The Cancer Genome Atlas-Kidney Renal Clear Cell Carcinoma (TCGA-KIRC), The Renal Cell Cancer-European Union (RECA-EU) and Korean-KIRC. A total of 68 sex-related genes were selected from TCGA-KIRC through machine learning, and 23 sex-specific genes were identified through verification using the three databases. Survival differences according to sex were identified in nine genes (ACSS3, ALG13, ASXL3, BAP1, JADE3, KDM5C, KDM6A, NCOR1P1, and ZNF449). Female-specific survival differences were found in BAP1 in overall survival (OS) (TCGA-KIRC, p = 0.004; RECA-EU, p = 0.002; and Korean-KIRC, p = 0.003) and disease-free survival (DFS) (TCGA-KIRC, p = 0.001 and Korean-KIRC, p = 0.000004), and NCOR1P1 in DFS (TCGA-KIRC, p = 0.046 and RECA-EU, p = 0.00003). Male-specific survival differences were found in ASXL3 (OS, p = 0.017 in TCGA-KIRC; and OS, p = 0.005 in RECA-EU) and KDM5C (OS, p = 0.009 in RECA-EU; and DFS, p = 0.016 in Korean-KIRC). These results suggest that biological sex may be an important predictor and sex-specific tailored treatment may improve patient care in ccRCC.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Mutation , Humains , Néphrocarcinome/génétique , Néphrocarcinome/mortalité , Femelle , Mâle , Tumeurs du rein/génétique , Tumeurs du rein/mortalité , Adulte d'âge moyen , Protéines suppresseurs de tumeurs/génétique , Facteurs sexuels , Pronostic , Ubiquitin thiolesterase/génétique , Marqueurs biologiques tumoraux/génétique , Histone Demethylases/génétique , Survie sans rechute , Sujet âgé
15.
Mol Genet Genomic Med ; 12(7): e2488, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38963008

RÉSUMÉ

BACKGROUND: This study aimed to identify disease-causing variants within a Chinese family affected by Birt-Hogg-Dubé syndrome (BHDS), which arises from an autosomal dominant inheritance pattern attributed to variants in the folliculin (FLCN) gene, recognized as a tumor suppressor gene. METHODS: A Chinese proband diagnosed with BHDS due to renal tumors underwent next-generation sequencing (NGS), revealing a novel variant in the FLCN gene. Sanger sequencing was subsequently performed on blood samples obtained from family members to confirm the presence of this variant. RESULTS: A novel germline frameshift variant (NM_144997.5:c.977dup) was identified in five individuals among the screened family members, marking the first report of this variant. Additionally, a somatic frameshift variant (NM_144997.5:c.1252del) was detected in the renal tumors of the proband. No variant was detected in unaffected family members. CONCLUSIONS: A novel heterozygous variant was identified in exon 9 of the FLCN gene, which broadens the spectrum of FLCN variants. We recommend that molecular analysis of the FLCN gene be performed in patients with suspected BHDS and their families.


Sujet(s)
Syndrome de Birt-Hogg-Dubé , Mutation avec décalage du cadre de lecture , Pedigree , Protéines proto-oncogènes , Protéines suppresseurs de tumeurs , Humains , Syndrome de Birt-Hogg-Dubé/génétique , Syndrome de Birt-Hogg-Dubé/anatomopathologie , Protéines suppresseurs de tumeurs/génétique , Protéines proto-oncogènes/génétique , Mâle , Femelle , Adulte , Adulte d'âge moyen , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Mutation germinale , Hétérozygote , Peuples d'Asie de l'Est
16.
Biochem J ; 481(14): 923-944, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38985307

RÉSUMÉ

Maintenance of genome stability is of paramount importance for the survival of an organism. However, genomic integrity is constantly being challenged by various endogenous and exogenous processes that damage DNA. Therefore, cells are heavily reliant on DNA repair pathways that have evolved to deal with every type of genotoxic insult that threatens to compromise genome stability. Notably, inherited mutations in genes encoding proteins involved in these protective pathways trigger the onset of disease that is driven by chromosome instability e.g. neurodevelopmental abnormalities, neurodegeneration, premature ageing, immunodeficiency and cancer development. The ability of cells to regulate the recruitment of specific DNA repair proteins to sites of DNA damage is extremely complex but is primarily mediated by protein post-translational modifications (PTMs). Ubiquitylation is one such PTM, which controls genome stability by regulating protein localisation, protein turnover, protein-protein interactions and intra-cellular signalling. Over the past two decades, numerous ubiquitin (Ub) E3 ligases have been identified to play a crucial role not only in the initiation of DNA replication and DNA damage repair but also in the efficient termination of these processes. In this review, we discuss our current understanding of how different Ub E3 ligases (RNF168, TRAIP, HUWE1, TRIP12, FANCL, BRCA1, RFWD3) function to regulate DNA repair and replication and the pathological consequences arising from inheriting deleterious mutations that compromise the Ub-dependent DNA damage response.


Sujet(s)
Altération de l'ADN , Réparation de l'ADN , Réplication de l'ADN , Ubiquitin-protein ligases , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitination , Tumeurs/génétique , Tumeurs/métabolisme , Instabilité du génome , Maturation post-traductionnelle des protéines , Animaux , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme
17.
Toxicol Appl Pharmacol ; 489: 117005, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38880190

RÉSUMÉ

A remarkable cancer-related role of zinc finger protein 367 (ZNF367) has been demonstrated in multiple malignancies. However, whether ZNF367 has a role in small-cell lung cancer (SCLC) remains unexplored. The purpose of this work was to explore the potential role and mechanism of ZNF367 in SCLC. In silico analysis using the Gene Expression Omnibus (GEO) dataset revealed high levels of the ZNF367 transcript in SCLC. Examination of clinical tissues confirmed the significant abundance of ZNF367 in SCLC tissues compared with adjacent non-malignant tissues. The genetic depletion of ZNF367 in SCLC cells led to remarkable alterations in cell proliferation, the cell cycle, colony formation and chemosensitivity. Mechanistically, ZNF367 was shown to regulate the activation of yes-associated protein (YAP) associated with the up-regulation of phosphorylated large tumour suppressor kinase 2 (LATS2). Further investigation revealed that ZNF367 affected the LATS2-YAP cascade by regulating the expression of citron kinase (CIT). Re-expression of constitutively active YAP diminished the tumour-inhibiting function of ZNF367 depletion. Xenograft experiments confirmed the tumour-inhibiting effect of ZNF367 depletion in vivo. In summary, our results demonstrate that the inhibition of ZNF367 displays anticancer effects in SCLC by inhibiting YAP activation, suggesting it as a potential druggable oncogenic target.


Sujet(s)
Tumeurs du poumon , Souris nude , Protein-Serine-Threonine Kinases , Transduction du signal , Carcinome pulmonaire à petites cellules , Facteurs de transcription , Protéines suppresseurs de tumeurs , Protéines de signalisation YAP , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/anatomopathologie , Carcinome pulmonaire à petites cellules/métabolisme , Animaux , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Lignée cellulaire tumorale , Protéines de signalisation YAP/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Souris , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Mâle , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Nat Genet ; 56(7): 1377-1385, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38886586

RÉSUMÉ

The presence of basal lineage characteristics signifies hyperaggressive human adenocarcinomas of the breast, bladder and pancreas. However, the biochemical mechanisms that maintain this aberrant cell state are poorly understood. Here we performed marker-based genetic screens in search of factors needed to maintain basal identity in pancreatic ductal adenocarcinoma (PDAC). This approach revealed MED12 as a powerful regulator of the basal cell state in this disease. Using biochemical reconstitution and epigenomics, we show that MED12 carries out this function by bridging the transcription factor ΔNp63, a known master regulator of the basal lineage, with the Mediator complex to activate lineage-specific enhancer elements. Consistent with this finding, the growth of basal-like PDAC is hypersensitive to MED12 loss when compared to PDAC cells lacking basal characteristics. Taken together, our genetic screens have revealed a biochemical interaction that sustains basal identity in human cancer, which could serve as a target for tumor lineage-directed therapeutics.


Sujet(s)
Carcinome du canal pancréatique , Complexe médiateur , Tumeurs du pancréas , Facteurs de transcription , Protéines suppresseurs de tumeurs , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Complexe médiateur/génétique , Complexe médiateur/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Lignage cellulaire/génétique , Éléments activateurs (génétique)
19.
Cancer Lett ; 595: 216999, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-38823762

RÉSUMÉ

Tumor protein p63 isoform ΔNp63 plays roles in the squamous epithelium and squamous cell carcinomas (SCCs), including esophageal SCC (ESCC). By integrating data from cell lines and our latest patient-derived organoid cultures, derived xenograft models, and clinical sample transcriptomic analyses, we identified a novel and robust oncogenic role of ΔNp63 in ESCC. We showed that ΔNp63 maintains the repression of cancer cell endogenous retrotransposon expression and cellular double-stranded RNA sensing. These subsequently lead to a restricted cancer cell viral mimicry response and suppressed induction of tumor-suppressive type I interferon (IFN-I) signaling through the regulations of Signal transducer and activator of transcription 1, Interferon regulatory factor 1, and cGAS-STING pathway. The cancer cell ΔNp63/IFN-I signaling axis affects both the cancer cell and tumor-infiltrating immune cell (TIIC) compartments. In cancer cells, depletion of ΔNp63 resulted in reduced cell viability. ΔNp63 expression is negatively associated with the anticancer responses to viral mimicry booster treatments targeting cancer cells. In the tumor microenvironment, cancer cell TP63 expression negatively correlates with multiple TIIC signatures in ESCC clinical samples. ΔNp63 depletion leads to increased cancer cell antigen presentation molecule expression and enhanced recruitment and reprogramming of tumor-infiltrating myeloid cells. Similar IFN-I signaling and TIIC signature association with ΔNp63 were also observed in lung SCC. These results support the potential application of ΔNp63 as a therapeutic target and a biomarker to guide candidate anticancer treatments exploring viral mimicry responses.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Facteurs de transcription , Microenvironnement tumoral , Protéines suppresseurs de tumeurs , Humains , Microenvironnement tumoral/immunologie , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/immunologie , Carcinome épidermoïde de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/virologie , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/immunologie , Tumeurs de l'oesophage/virologie , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Lignée cellulaire tumorale , Animaux , Survie cellulaire , Régulation de l'expression des gènes tumoraux , Souris , Transduction du signal , Interféron de type I/métabolisme
20.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892042

RÉSUMÉ

Esophageal Cancer-Related Gene 2 (ECRG2), also known as Serine Peptidase Inhibitor Kazal type 7 (SPINK7), is a novel tumor suppressor gene from the SPINK family of genes that exhibits anticancer potential. ECRG2 was originally identified during efforts to discover genes involved in esophageal tumorigenesis. ECRG2 was one of those genes whose expression was absent or reduced in primary human esophageal cancers. Additionally, absent or reduced ECRG2 expression was also noted in several other types of human malignancies. ECRG2 missense mutations were identified in various primary human cancers. It was reported that a cancer-derived ECRG2 mutant (valine to glutamic acid at position 30) failed to induce cell death and caspase activation triggered by DNA-damaging anticancer drugs. Furthermore, ECRG2 suppressed cancer cell proliferation in cultured cells and grafted tumors in animals and inhibited cancer cell migration/invasion and metastasis. ECRG2 also was identified as a negative regulator of Hu-antigen R (HuR), an oncogenic RNA-binding protein that is known to regulate mRNA stability and the expression of transcripts corresponding to many cancer-related genes. ECRG2 function is important also for the regulation of inflammatory responses and the maintenance of epithelial barrier integrity in the esophagus. More recently, ECRG2 was discovered as one of the newest members of the pro-apoptotic transcriptional targets of p53. Two p53-binding sites (BS-1 and BS-2) were found within the proximal region of the ECRG2 gene promoter; the treatment of DNA-damaging agents in cancer cells significantly increased p53 binding to the ECRG2 promoter and triggered a strong ECRG2 promoter induction following DNA damage. Further, the genetic depletion of ECRG2 expression significantly impeded apoptotic cell death induced by DNA damage and wild-type p53 in cancer cells. These findings suggest that the loss of ECRG2 expression, commonly observed in human cancers, could play important roles in conferring anticancer drug resistance in human cancers. Thus, ECRG2 is a novel regulator in DNA damage-induced cell death that may also be a potential target for anticancer therapeutics.


Sujet(s)
Altération de l'ADN , Inhibiteurs de serine peptidase de type Kazal , Humains , Altération de l'ADN/génétique , Animaux , Inhibiteurs de serine peptidase de type Kazal/génétique , Inhibiteurs de serine peptidase de type Kazal/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...