Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36.267
Filtrer
1.
Rev Med Liege ; 79(7-8): 467-470, 2024 Jul.
Article de Français | MEDLINE | ID: mdl-39129541

RÉSUMÉ

RHOBTB2 was first described as epileptogenic when it presents a missense variant in 2016 and studied more specifically in 2018. It is a gene that causes rare, but potentially severe childhood epileptic encephalopathy. In 2021, research confirmed that heterozygous mutations of RHOBTB2 included other clinical signs besides these encephalopathies. Thus, these infantile epilepsies are mainly associated with highly variable phenotypes, with developmental delay, post-traumatic encephalitis, paroxysmal movement disorders and iconographic brain damage. In this work, after presenting a clinical case, we will recall the role of RhoGTPases on neuronal development. We will then discuss a study which highlighted the neurodevelopmental impact of mutations on the RHOBTB2 gene by carrying out work on Drosophila melanogaster flies. Finally, we will compare the presented clinical case with a literature review.


Le gène RHOBTB2 est décrit pour la première fois comme épileptogène alors qu'il présente un variant faux-sens en 2016, puis est étudié plus précisément en 2018. Il s'agit d'un gène qui est à l'origine d'encéphalopathies épileptiques infantiles rares, mais pouvant être sévères. En 2021, des recherches ont confirmé que les mutations hétérozygotes de RHOBTB2 englobaient d'autres signes cliniques que ces encéphalopathies. Ainsi, ces épilepsies infantiles sont associées, principalement, avec des phénotypes fortement variables, à un retard développemental, à des encéphalites post-traumatiques, à des troubles paroxystiques des mouvements et à des atteintes iconographiques de l'encéphale. Dans ce travail, après avoir présenté un cas clinique, nous rappellerons le rôle des RhoGTPases sur le développement neuronal. Nous discuterons ensuite d'une étude qui a mis en évidence l'impact neurodéveloppemental de mutations sur le gène RHOBTB2 en réalisant des travaux sur des mouches Drosophila melanogaster. Pour terminer, nous mettrons le cas clinique présenté en parallèle avec une revue de la littérature réalisée par rapport à ce gène.


Sujet(s)
Mutation , Humains , Animaux , Protéines suppresseurs de tumeurs/génétique , Protéines G/génétique , Mâle , Drosophila melanogaster/génétique , Femelle , Nourrisson
2.
Sci Rep ; 14(1): 18413, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39117733

RÉSUMÉ

Cancer therapy-related cardiac dysfunction (CTRCD), which commonly includes left ventricular dysfunction and heart failure, is the main adverse effect of anticancer therapy. In recent years several candidate genes studies and genome-wide association studies have identified common genetic variants associated with CTRCD, but evidence remains limited and few genetic variants are robust. A genome-wide meta-analysis of CTRCD was performed with 852 oncology patients receiving cancer therapy. DNA samples were genotyped and imputed to perform a GWAS meta-analysis for case-control (N = 852 (380 cases and 472 controls) and extreme phenotypes (N = 618 (78 cases and 472 controls) looking for genetic variants that predispose to CTRCD. The results were validated in a replicate cohort of 1,191 oncology patients (245 cases and 946 controls). Functional mapping of the replicated loci was then performed. The meta-analysis showed 9 and 17 loci suggestively associated (P-value < 1 × 10-5) with CTRCD in case-control and extreme phenotypes analyses, respectively. The 3q28 locus (rs rs7652759, P = 5.64 × 10-6) in the case-control analysis was the strongest signal, with up to 64 SNPs above the suggestive significance threshold. The rs7652759, an intergenic variant between TPRG1 and TP63 genes, was the only variant validated in the replication cohort (P-value = 0.01). Functional mapping of this significant locus revealed up to 5 new genes potentially involved in the CTRCD. We identified the intergenic region near TP63 as a novel CTRCD susceptibility locus. In the future, the genotyping of these markers could be considered in new CTRCD risk scores to improve preventive strategies in cardio-oncology.


Sujet(s)
Étude d'association pangénomique , Tumeurs , Polymorphisme de nucléotide simple , Protéines suppresseurs de tumeurs , Humains , Protéines suppresseurs de tumeurs/génétique , Tumeurs/génétique , Tumeurs/traitement médicamenteux , Femelle , Mâle , Prédisposition génétique à une maladie , Facteurs de transcription/génétique , Études cas-témoins , ADN intergénique/génétique , Adulte d'âge moyen , Sujet âgé , Cartographie chromosomique
3.
Nat Commun ; 15(1): 6953, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138205

RÉSUMÉ

Filovirus-host interactions play important roles in all stages of the virus lifecycle. Here, we identify LATS1/2 kinases and YAP, key components of the Hippo pathway, as critical regulators of EBOV transcription and egress. Specifically, we find that when YAP is phosphorylated by LATS1/2, it localizes to the cytoplasm (Hippo "ON") where it sequesters VP40 to prevent egress. In contrast, when the Hippo pathway is "OFF", unphosphorylated YAP translocates to the nucleus where it transcriptionally activates host genes and promotes viral egress. Our data reveal that LATS2 indirectly modulates filoviral VP40-mediated egress through phosphorylation of AMOTp130, a positive regulator of viral egress, but more surprisingly that LATS1/2 kinases directly modulate EBOV transcription by phosphorylating VP30, an essential regulator of viral transcription. In sum, our findings highlight the potential to exploit the Hippo pathway/filovirus axis for the development of host-oriented countermeasures targeting EBOV and related filoviruses.


Sujet(s)
Ebolavirus , Voie de signalisation Hippo , Protein-Serine-Threonine Kinases , Transduction du signal , Facteurs de transcription , Transcription génétique , Libération de particules virales , Humains , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Phosphorylation , Ebolavirus/physiologie , Ebolavirus/génétique , Ebolavirus/métabolisme , Cellules HEK293 , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines de signalisation YAP/métabolisme , Protéines de la matrice virale/métabolisme , Protéines de la matrice virale/génétique , Fièvre hémorragique à virus Ebola/virologie , Fièvre hémorragique à virus Ebola/métabolisme , Interactions hôte-pathogène , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique
4.
Clin Exp Med ; 24(1): 187, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39136841

RÉSUMÉ

Glioblastoma (GBM) is a highly heterogeneous disease with poor clinical outcomes. To comprehensively dissect the molecular landscape of GBM and heterogeneous macrophage clusters in the progression of GBM, this study integrates single-cell and bulk transcriptome data to recognize a distinct pro-tumor macrophage cluster significantly associated with the prognosis of GBM and develop a GBM prognostic signature to facilitate prior subtypes. Leveraging glioma single-cell sequencing data, we identified a novel pro-tumor macrophage subgroup, marked by S100A9, which might interact with endothelial cells to facilitate tumor progression via angiogenesis. To further benefit clinical application, a prognostic signature was established with the genes associated with pro-tumor macrophages. Patients classified within the high-risk group characterized with enrichment in functions related to tumor progression, including epithelial-mesenchymal transition and hypoxia, displays elevated mutations in the TERT promoter region, reduced methylation in the MGMT promoter region, poorer prognoses, and diminished responses to temozolomide therapy, thus effectively discriminating between the prognostic outcomes of GBM patients. Our research sheds light on the intricate microenvironment of gliomas and identifies potential molecular targets for the development of novel therapeutic approaches.


Sujet(s)
Analyse de profil d'expression de gènes , Glioblastome , Analyse sur cellule unique , Humains , Glioblastome/génétique , Glioblastome/anatomopathologie , Pronostic , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Microenvironnement tumoral/génétique , Témozolomide/usage thérapeutique , Macrophages/métabolisme , Transcriptome , Telomerase/génétique , Protéines suppresseurs de tumeurs/génétique , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/métabolisme , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , DNA modification methylases/génétique , Enzymes de réparation de l'ADN
6.
BMC Genomics ; 25(1): 757, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095712

RÉSUMÉ

BACKGROUND: It is known that the neurodevelopmental disorder associated gene, Satb2, plays important roles in determining the upper layer neuron specification. However, it is not well known how this gene regulates other neocortical regions during the development. It is also lack of comprehensive delineation of its spatially regulatory pathways in neocortical development. RESULTS: In this work, we utilized spatial transcriptomics and immuno-staining to systematically investigate the region-specific gene regulation of Satb2 by comparing the Satb2+/+ and Satb2-/- mice at embryonic stages, including the ventricle zone (VZ) or subventricle zone (SVZ), intermediate zone (IZ) and cortical plate (CP) respectively. The staining result reveals that these three regions become moderately or significantly thinner in the Satb2-/- mice. In the cellular level, the cell number increases in the VZ/SVZ, whereas the cell number decreases in the CP. The spatial transcriptomics data show that many important genes and relevant pathways are dysregulated in Satb2-/- mice in a region-specific manner. In the VZ/SVZ, the key genes involved in neural precursor cell proliferation, including the intermediate progenitor marker Tbr2 and the lactate production related gene Ldha, are up-regulated in Satb2-/- mice. In the IZ, the key genes in regulating neuronal differentiation and migration, such as Rnd2, exhibit ectopic expressions in the Satb2-/- mice. In the CP, the lineage-specific genes, Tbr1 and Bcl11b, are abnormally expressed. The neuropeptide related gene Npy is down-regulated in Satb2-/- mice. Finally, we validated the abnormal expressions of key regulators by using immunofluorescence or qPCR. CONCLUSIONS: In summary, our work provides insights on the region-specific genes and pathways which are regulated by Satb2 in neocortical development.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Protéines de liaison aux séquences d'ADN MAR , Néocortex , Facteurs de transcription , Transcriptome , Animaux , Néocortex/métabolisme , Néocortex/croissance et développement , Protéines de liaison aux séquences d'ADN MAR/génétique , Protéines de liaison aux séquences d'ADN MAR/métabolisme , Souris , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Analyse de profil d'expression de gènes , Souris knockout , Protéines de répression , Protéines suppresseurs de tumeurs
7.
Mol Genet Genomic Med ; 12(8): e2500, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39101447

RÉSUMÉ

BACKGROUND: Variations in the WWOX gene have been identified as the leading cause of several central nervous system disorders. However, most previous reports have focused on the description of clinical phenotype, neglecting functional verification. Herein, we presented a case of a patient with developmental epileptic encephalopathy (DEE) caused by WWOX gene variation. CASE PRESENTATION: Our patient was a 13-month-old girl with abnormal facial features, including facial hypotonia, arched eyebrows, a broad nose, and a depressed nasal bridge. She also had sparse and yellow hair, a low anterior hairline, and a short neck. Before the age of 8 months, she was suffering from mild seizures. Her developmental delay gradually worsened, and she suffered infantile spasms. After treatment with vigabatrin, seizures subsided. WWOX gene homozygous variation c.172+1G>C was identified using whole exome sequencing. Further minigene assay confirmed that the variation site affected splicing, causing protein truncation and affecting its function. CONCLUSION: Clinical phenotype and minigene results suggest that WWOX gene homozygous variation c.172+1G>C can cause severe DEE. We also concluded that vigabatrin can effectively treat seizures.


Sujet(s)
Homozygote , Phénotype , Spasmes infantiles , Oxydoréductase contenant des domaines WW , Humains , Oxydoréductase contenant des domaines WW/génétique , Femelle , Nourrisson , Spasmes infantiles/génétique , Spasmes infantiles/anatomopathologie , Anticonvulsivants/usage thérapeutique , Mutation , Vigabatrine/usage thérapeutique , Protéines suppresseurs de tumeurs
8.
Zhonghua Bing Li Xue Za Zhi ; 53(8): 797-802, 2024 Aug 08.
Article de Chinois | MEDLINE | ID: mdl-39103260

RÉSUMÉ

Objective: To investigate the clinicopathological characteristics, immunophenotypes, molecular features, and differential diagnosis of BAP1 mutated clear cell renal cell carcinoma (CCRCC) for better understanding this entity. Methods: Clinical data, histological morphology, immunophenotypes and molecular characteristics of 18 BAP1 mutated CCRCC cases diagnosed at the Department of Pathology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China from January 2020 to December 2022 were analyzed. The patients were followed up. Results: There were 17 males and 1 female patients, aged from 39 to 72 years, with an average age of 56.3 years. Sixteen patients with primary CCRCC were followed up for an average of 24 months, 7 patients had metastases occurred from 4 to 22 months postoperatively. Thirteen of the 16 patients were alive at the time of the last follow-up while 3 patients died 12, 15, and 20 months after the surgery, respectively. One patient underwent retroperitoneal mass resection, but had lung metastasis 32 months after surgery. One case received cervical tumor resection and died at 22 months after the surgery. Characteristic CCRCC regions were identified in 11 of the 18 cases. The tumor cells were arranged in papillary, alveolar, and large nest patterns. Abundant lymphoid tissue, necrosis, and psammoma bodies were seen. Tumor cells showed abundant eosinophilic cytoplasm, and sometimes exhibited rhabdoid differentiation. Round eosinophilic globules were located in the cytoplasm and extracellular matrix. There were 9 cases with WHO/International Society of Urological Pathology grade 3, and 9 cases with grade 4. PAX8 (18/18), carbonic anhydrase 9 (CA9, 16/18), CD10 (18/18), and vimentin (18/18) were positive in the vast majority of tumors.TFE3 was expressed in 5 cases, with strong expression in only 1 case. Eighteen cases were all positive for P504s. Twelve cases harbored a BAP1 mutation combined with von Hippel-Lindau (VHL) mutation, and 2 cases had mutations in BAP1, VHL and PBRM1 simultaneously. SETD2 mutation was not found in any of the cases. Conclusions: BAP1 mutated CCRCC contained papillary, alveolar, and large nest patterns, eosinophilic cytoplasm, high-grade nucleoli, and collagen globules, with P504s positivity. In practical work, when encountering CCRCC containing these features, pathologists should consider the possibility of BAP1 mutations and conduct related molecular tests.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Mutation , Protéines suppresseurs de tumeurs , Ubiquitin thiolesterase , Humains , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Néphrocarcinome/chirurgie , Néphrocarcinome/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitin thiolesterase/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/chirurgie , Sujet âgé , Adulte , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Facteur de transcription PAX-8/génétique , Facteur de transcription PAX-8/métabolisme , Diagnostic différentiel
9.
Epigenetics ; 19(1): 2381849, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39109527

RÉSUMÉ

Gametogenetin binding protein 2 (GGNBP2) was indispensable in normal spermatids for transformation into mature spermatozoa in mice, and when Gametogenetin binding protein 2 is bound to BRCC36 and RAD51, the complex participates in repairing DNA double-strand breaks (DSB) during the meiotic progression of spermatocytes. Ggnbp2 knockout resulted in the up-regulation of H2AK119ubi and down-regulation of H2BK120ubi in GC-2 cells (mouse spermatogonia-derived cell line) and postnatal day 18 testis lysate. Our results also demonstrated that Gametogenetin binding protein 2 inducedASXL1 to activate the deubiquitinating enzyme BAP1 in deubiquitinating H2A, while Gametogenetin binding protein 2 knockout disrupted the interaction between ASXL1 and BAP1, resulting in BAP1 localization change. Furthermore, the Gametogenetin binding protein 2 deletion reduced H2B ubiquitination by affecting E2 enzymes and E3 ligase binding. Gametogenetin binding protein 2 regulated H2A and H2B ubiquitination levels and controlled H3K27 and H3K79 methylation by PRC2 subunits and histone H3K79 methyltransferase. Altogether, our results suggest that Ggnbp2 knockout increased DNA damage response by promoting H2A ubiquitination and H3K27trimethylation (H3K27me3) and reduced nucleosome stability by decreasing H2B ubiquitination and H3K79 dimethylation (H3K79me2), revealing new mechanisms of epigenetic phenomenon during spermatogenesis. Gametogenetin binding protein 2 seems critical in regulating histone modification and chromatin structure in spermatogenesis.


Sujet(s)
Histone , Spermatogenèse , Ubiquitination , Mâle , Animaux , Spermatogenèse/génétique , Histone/métabolisme , Souris , Méthylation , Souris knockout , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Lignée cellulaire
10.
Clin Transl Med ; 14(8): e1804, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39118288
11.
Cancer Med ; 13(15): e7456, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39118481

RÉSUMÉ

OBJECTIVES: High-grade gliomas (HGGs) are highly malignant, aggressive, and have a high incidence and mortality rate. The aim of this study was to investigate survival outcomes and prognostic factors in patients with HGGs. METHODS: In this retrospective study, a total of 159 patients with histologically confirmed HGGs were included. The recruitment period was from January 2011 to December 2019. We evaluated patient demographic data, tumor characteristics, treatment methods, immunocytochemistry results, overall survival (OS) time, and progression-free survival (PFS) time using Kaplan-<>Meier survival analysis with log-rank testing. Additionally, we employed Cox regression analysis to identify independent factors associated with survival outcomes. RESULTS: Kaplan-Meier survival analysis revealed that the 1-, 2-, and 5-years OS rates were 81.8%, 50.3%, and 12.6%, respectively. Similarly, the 1-, 2-, and 5-years PFS rates were 50.9%, 22.4%, and 3.1%, respectively. The median OS duration was 35.0 months. The univariate analysis indicated that postoperative pathological classification, grade, and age were significantly associated with patient outcomes (p < 0.01). Among the patients, 147 received concurrent chemoradiotherapy, while 12 did not. The immunohistochemical markers of ki-67, MGMT, IDH1R132H, and p53 demonstrated statistically significant differences in their prognostic impact (p = 0.001, p = 0.020, p = 0.003, and p = 0.021, respectively). In conclusion, we found that grades, age, pathological classification, ki-67, MGMT, and IDH1R132H expression were statistically significantly associated with PFS (p < 0.01, p = 0.004, p = 0.003, p = 0.001, p = 0.036, and p = 0.028). Additionally, immunohistochemical expressions of TRIB3 and AURKA were significantly higher in patients with shorter survival (p = 0.015 and p = 0.023). CONCLUSIONS: Tumor grade and the use of concurrent chemoradiotherapy after surgery were independent prognostic factors that significantly influenced patient survival. Additionally, tumor grade and MGMT expression were found to be independent factors affecting progression-free survival (PFS). Notably, the expression of TRIB3 and AURKA was higher in patients with poor survival outcomes.


Sujet(s)
Tumeurs du cerveau , Gliome , Grading des tumeurs , Humains , Femelle , Mâle , Gliome/mortalité , Gliome/anatomopathologie , Gliome/thérapie , Gliome/métabolisme , Études rétrospectives , Adulte d'âge moyen , Adulte , Pronostic , Sujet âgé , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Chimioradiothérapie , Jeune adulte , Estimation de Kaplan-Meier , Marqueurs biologiques tumoraux/métabolisme , Survie sans progression , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , DNA modification methylases/génétique , DNA modification methylases/métabolisme , Analyse multifactorielle , Protéines suppresseurs de tumeurs/métabolisme , Taux de survie , Adolescent , Enzymes de réparation de l'ADN/métabolisme , Enzymes de réparation de l'ADN/génétique , Enzymes de réparation de l'ADN/analyse
12.
J Extracell Vesicles ; 13(8): e12488, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39104296

RÉSUMÉ

Pancreatic cancer remains one of the most lethal malignant diseases. Gemcitabine-based chemotherapy is still one of the first-line systemic treatments, but chemoresistance occurs in the majority of patients. Recently, accumulated evidence has demonstrated the role of the tumour microenvironment in promoting chemoresistance. In the tumour microenvironment, pancreatic stellate cells (PSCs) are among the main cellular components, and extracellular vesicles (EVs) are common mediators of cell‒cell communication. In this study, we showed that SP1-transcribed miR-31-5p not only targeted LATS2 in pancreatic cancer cells but also regulated the Hippo pathway in PSCs through EV transfer. Consequently, PSCs synthesized and secreted protein acidic and rich in cysteins (SPARC), which was preferentially expressed in stromal cells, stimulating Extracellular Signal regulated kinase (ERK) signalling in pancreatic cancer cells. Therefore, pancreatic cancer cell survival and chemoresistance were improved due to both the intrinsic Hippo pathway regulated by miR-31-5p and external SPARC-induced ERK signalling. In mouse models, miR-31-5p overexpression in pancreatic cancer cells promoted the chemoresistance of coinjected xenografts. In a tissue microarray, pancreatic cancer patients with higher miR-31-5p expression had shorter overall survival. Therefore, miR-31-5p regulates the Hippo pathway in multiple cell types within the tumour microenvironment via EVs, ultimately contributing to the chemoresistance of pancreatic cancer cells.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Vésicules extracellulaires , Voie de signalisation Hippo , microARN , Ostéonectine , Tumeurs du pancréas , Cellules stellaires pancréatiques , Protein-Serine-Threonine Kinases , Microenvironnement tumoral , microARN/métabolisme , microARN/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Humains , Cellules stellaires pancréatiques/métabolisme , Animaux , Protein-Serine-Threonine Kinases/métabolisme , Souris , Ostéonectine/métabolisme , Ostéonectine/génétique , Vésicules extracellulaires/métabolisme , Lignée cellulaire tumorale , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Régulation de l'expression des gènes tumoraux , , Transduction du signal , Souris nude
13.
Invest Ophthalmol Vis Sci ; 65(8): 37, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39042403

RÉSUMÉ

Purpose: Cell lines are being used in preclinical uveal melanoma (UM) research. Because not all cell lines harbor typical GNAQ or GNA11 hotspot mutations, we aimed at better classifying them and determining whether we could find genetic causes to explain the protein and mRNA expression profiles of the cell lines. Methods: We studied protein and mRNA expression of 14 UM cell lines and determined the presence of single nucleotide variants and small insertions and deletions with next-generation sequencing and copy number alterations with a single nucleotide polymorphism array. The lists of differentially expressed proteins and genes were merged, and shared lists were created, keeping only terms with concordant mRNA and protein expression. Enrichment analyses were performed on the shared lists. Results: Cell lines Mel285 and Mel290 are separate from GNA-mutated cell lines and show downregulation of melanosome-related markers. Both lack typical UM mutations but each harbors four putatively deleterious variants in CTNNB1, PPP1R10, LIMCH1, and APC in Mel285 and ARID1A, PPP1R10, SPG11, and RNF43 in Mel290. The upregulated terms in Mel285 and Mel290 did not point to a convincing alternative origin. Mel285 shows loss of chromosomes 1p, 3p, partial 3q, 6, and partial 8p, whereas Mel290 shows loss of 1p and 6. Expression in the other 12 cell lines was related to BAP1 expression. Conclusions: Although Mel285 and Mel290 have copy number alterations that fit UM, multi-omics analyses show that they belong to a separate group compared to the other analyzed UM cell lines. Therefore, they may not be representative models to test potential therapeutic targets for UM.


Sujet(s)
Sous-unités alpha Gq-G11 des protéines G , Sous-unités alpha des protéines G , Régulation de l'expression des gènes tumoraux , Mélanome , Mutation , ARN messager , Protéines suppresseurs de tumeurs , Ubiquitin thiolesterase , Tumeurs de l'uvée , Tumeurs de l'uvée/génétique , Tumeurs de l'uvée/métabolisme , Tumeurs de l'uvée/anatomopathologie , Mélanome/génétique , Mélanome/métabolisme , Mélanome/anatomopathologie , Humains , Ubiquitin thiolesterase/génétique , ARN messager/génétique , Sous-unités alpha des protéines G/génétique , Protéines suppresseurs de tumeurs/génétique , Sous-unités alpha Gq-G11 des protéines G/génétique , Sous-unités alpha Gq-G11 des protéines G/métabolisme , Lignée cellulaire tumorale , Variations de nombre de copies de segment d'ADN , Polymorphisme de nucléotide simple , Analyse de mutations d'ADN
14.
Clin Epigenetics ; 16(1): 97, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044262

RÉSUMÉ

BACKGROUND: Esophagogastric junction cancer (EJC) refers to malignant tumors that develop at the junction between the stomach and the esophagus. TUSC1 is a recently identified tumor suppressor gene known for its involvement in various types of cancer. The objective of this investigation was to elucidate the regulatory influence of DNA methylation on TUSC1 expression and its role in the progression of EJC. METHODS: Bioinformatics software was utilized to analyze the expression of TUSC1, enriched pathways, and highly methylated sites in the promoter region. TUSC1 expression in EJC was assessed using quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blot (WB), and immunohistochemistry. Methylation-specific PCR was employed to detect the methylation level of TUSC1. To analyze the effects of TUSC1 and 5-AZA-2 on tumor cell proliferation, migration, invasion, cell cycle, and apoptosis, several assays including CCK-8, colony formation, transwell, and flow cytometry were conducted. The expression of MDM2 was assessed using qRT-PCR and WB. WB detected the expression of p53, and p-p53, markers for EJC cell proliferation, epithelial-mesenchymal transition, and apoptosis. The role of TUSC1 in tumor occurrence in vivo was examined using a xenograft mouse model. RESULTS: TUSC1 expression was significantly downregulated in EJC. Overexpression of TUSC1 and treatment with 5-AZA-2 inhibited the malignant progression of EJC cells. In EJC, low methylation levels promoted the expression of TUSC1. Upregulation of TUSC1 suppressed the expression of MDM2 and activated the p53 signaling pathway. Inactivation of this pathway attenuated the inhibitory effect of TUSC1 overexpression on EJC cell proliferation, migration, invasion, and other behaviors. Animal experiments demonstrated that TUSC1 overexpression inhibited EJC tumor growth and metastasis in vivo. CONCLUSION: TUSC1 was commonly downregulated in EJC and regulated by methylation. It repressed the malignant progression of EJC tumors by mediating the p53 pathway, suggesting its potential as a diagnostic and therapeutic target for EJC.


Sujet(s)
Prolifération cellulaire , Méthylation de l'ADN , Tumeurs de l'oesophage , Régulation de l'expression des gènes tumoraux , Protéines suppresseurs de tumeurs , Humains , Méthylation de l'ADN/génétique , Animaux , Protéines suppresseurs de tumeurs/génétique , Souris , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/génétique , Prolifération cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Évolution de la maladie , Jonction oesogastrique/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Apoptose/génétique , Régions promotrices (génétique)/génétique , Protéine p53 suppresseur de tumeur/génétique , Mâle , Mouvement cellulaire/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Régulation négative/génétique , Femelle , Souris nude
15.
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 , Animaux , Femelle , Humains , Mâle , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Facteurs de transcription ARNTL/métabolisme , Facteurs de transcription ARNTL/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Cancer du nasopharynx/génétique , Cancer du nasopharynx/anatomopathologie , Cancer du nasopharynx/métabolisme , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/anatomopathologie , Tumeurs du rhinopharynx/métabolisme , Métastase tumorale , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Transduction du signal , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines de signalisation YAP/métabolisme
16.
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
17.
PLoS Biol ; 22(7): e3002696, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38959200

RÉSUMÉ

Sterile alpha motif domain-containing proteins 9 and 9-like (SAMD9/9L) are associated with life-threatening genetic diseases in humans and are restriction factors of poxviruses. Yet, their cellular function and the extent of their antiviral role are poorly known. Here, we found that interferon-stimulated human SAMD9L restricts HIV-1 in the late phases of replication, at the posttranscriptional and prematuration steps, impacting viral translation and, possibly, endosomal trafficking. Surprisingly, the paralog SAMD9 exerted an opposite effect, enhancing HIV-1. More broadly, we showed that SAMD9L restricts primate lentiviruses, but not a gammaretrovirus (MLV), nor 2 RNA viruses (arenavirus MOPV and rhabdovirus VSV). Using structural modeling and mutagenesis of SAMD9L, we identified a conserved Schlafen-like active site necessary for HIV-1 restriction by human and a rodent SAMD9L. By testing a gain-of-function constitutively active variant from patients with SAMD9L-associated autoinflammatory disease, we determined that SAMD9L pathogenic functions also depend on the Schlafen-like active site. Finally, we found that the constitutively active SAMD9L strongly inhibited HIV, MLV, and, to a lesser extent, MOPV. This suggests that the virus-specific effect of SAMD9L may involve its differential activation/sensing and the virus ability to evade from SAMD9L restriction. Overall, our study identifies SAMD9L as an HIV-1 antiviral factor from the cell autonomous immunity and deciphers host determinants underlying the translational repression. This provides novel links and therapeutic avenues against viral infections and genetic diseases.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Lentivirus des primates , Réplication virale , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Animaux , Lentivirus des primates/génétique , Lentivirus des primates/métabolisme , Cellules HEK293 , Biosynthèse des protéines , Facteurs de restriction antiviraux , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Infections à VIH/virologie , Infections à VIH/traitement médicamenteux , Protéines suppresseurs de tumeurs
18.
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
19.
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
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE