Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36.284
Filter
1.
Mol Genet Genomic Med ; 12(8): e2500, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39101447

ABSTRACT

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.


Subject(s)
Homozygote , Phenotype , Spasms, Infantile , WW Domain-Containing Oxidoreductase , Humans , WW Domain-Containing Oxidoreductase/genetics , Female , Infant , Spasms, Infantile/genetics , Spasms, Infantile/pathology , Anticonvulsants/therapeutic use , Mutation , Vigabatrin/therapeutic use , Tumor Suppressor Proteins
2.
Zhonghua Bing Li Xue Za Zhi ; 53(8): 797-802, 2024 Aug 08.
Article in Chinese | MEDLINE | ID: mdl-39103260

ABSTRACT

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.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Mutation , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Carcinoma, Renal Cell/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Male , Female , Middle Aged , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Aged , Adult , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Diagnosis, Differential
3.
Epigenetics ; 19(1): 2381849, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39109527

ABSTRACT

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.


Subject(s)
Histones , Spermatogenesis , Ubiquitination , Male , Animals , Spermatogenesis/genetics , Histones/metabolism , Mice , Methylation , Mice, Knockout , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Cell Line
4.
Clin Exp Med ; 24(1): 187, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39136841

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Glioblastoma , Single-Cell Analysis , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Prognosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Tumor Microenvironment/genetics , Temozolomide/therapeutic use , Macrophages/metabolism , Transcriptome , Telomerase/genetics , Tumor Suppressor Proteins/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , DNA Modification Methylases/genetics , DNA Repair Enzymes
5.
Retina ; 44(9): 1580-1589, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39167579

ABSTRACT

PURPOSE: To determine the association between gene-expression profiling (GEP), next-generation sequencing (NGS), preferentially expressed antigen in melanoma (PRAME) features, and metastatic risk in patients with uveal melanoma (UM). METHODS: A retrospective analysis of patients with UM treated by brachytherapy or enucleation by a single ocular oncologist was conducted from November 2020 and July 2022. Clinicopathologic features, patient outcomes, GEP classification, NGS, and PRAME results were recorded. RESULTS: Comprehensive GEP, PRAME, and NGS testing was performed on 135 UMs. The presence of eukaryotic translation initiation factor 1A, X-chromosomal and splicing factor 3B subunit 1 mutations was significantly associated with GEP class 1A and GEP class 1B, respectively. The presence of BRCA- associated protein-1 mutation was significantly associated with GEP class 2. The average largest basal diameter for tumors with eukaryotic translation initiation factor 1A, X-chromosomal mutations was significantly smaller than those with splicing factor 3B subunit 1 mutations and BRCA1-associated protein-1 mutations. Class 2 tumors metastasized sooner than GEP class 1 tumors. Tumors with splicing factor 3B subunit 1 and/or BRCA1-associated protein-1 mutations metastasized sooner compared with tumors that had either no driver mutation or no mutations at all. Tumors with splicing factor 3B subunit 1 did not have a significantly different time to metastasis compared with tumors with BRCA1-associated protein-1 (P value = 0.97). Forty tumors (30%) were PRAME positive, and the remaining 95 tumors (70%) were PRAME negative. Tumors with PRAME-positive status did not have a significantly different time to metastasis compared with tumors without PRAME-positive status (P value = 0.11). CONCLUSION: GEP, NGS, and PRAME expression analysis help determine different levels of metastatic risk in UM. Although other prognostic tests exist, the following study reports on the use of NGS for metastatic prognostication in UM. However, limitations of NGS exist, especially with small lesions that are technically difficult to biopsy.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Melanoma , Uveal Neoplasms , Humans , Uveal Neoplasms/genetics , Uveal Neoplasms/diagnosis , Melanoma/genetics , Retrospective Studies , Male , Female , Middle Aged , Antigens, Neoplasm/genetics , Gene Expression Profiling/methods , Aged , Biomarkers, Tumor/genetics , Mutation , Adult , Gene Expression Regulation, Neoplastic , Aged, 80 and over , Eukaryotic Initiation Factor-1/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Brachytherapy , Phosphoproteins , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
6.
Biol Direct ; 19(1): 69, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39164777

ABSTRACT

A substantive body of evidence has demonstrated the significant roles of circular RNA (circRNA) in cancer. However, the contribution of dysregulated circRNAs to ovarian cancer (OC) remains elusive. We aim to elucidate the critical roles and mechanisms of hsa_circ_0020093, which was demonstrated to be downregulated in OC tissues in our previous study. In this study, we confirmed the decreased expression of hsa_circ_0020093 in OC tissues and cell lines and demonstrated the negative correlation between its expression and FIGO stage, abdominal implantation and CA125 level of OC patients. Through gain and loss of function studies, we confirmed the inhibitory role of hsa_circ_0020093 in ovarian tumor growth in vitro and in vivo. Mechanistically, based on the peri-nuclear accumulation of hsa_circ_0020093, we discovered the interaction between hsa_circ_0020093 and the mitochondrial protein LRPPRC by RNA pull-down, mass spectrometry, RNA Binding Protein Immunoprecipitation. As a result, qRT-PCR and transmission electron microscopy results showed that the mitochondria mRNA expression and mitochondria abundance were decreased upon hsa_circ_0020093-overexpression. Meanwhile, we also unearthed the hsa_circ_0020093/miR-107/LATS2 axis in OC according to RNA-sequencing, RIP and luciferase reporter assay data. Furthermore, LRPPRC and LATS2 are both reported as the upstream regulators of YAP, our study also studied the crosstalk between hsa_circ_0020093, LRPPRC and miR-107/LATS2, and unearthed the up-regulation of phosphorylated YAP in hsa_circ_0020093-overexpressing OC cells and xenograft tumors. Collectively, our study indicated the novel mechanism of hsa_circ_0020093 in suppressing OC progression through both hsa_circ_0020093/LRPPRC and hsa_circ_0020093/miR-107/LATS2 axes, providing a potential therapeutic target for OC patients.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Protein Serine-Threonine Kinases , RNA, Circular , Signal Transduction , Tumor Suppressor Proteins , Humans , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Mice , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Mice, Nude
7.
Life Sci Alliance ; 7(11)2024 Nov.
Article in English | MEDLINE | ID: mdl-39187374

ABSTRACT

The mTORC1-complex is negatively regulated by TSC1 and TSC2. Activation of Hedgehog signaling is strictly dependent on communication between Smoothened and the Hedgehog-signaling effector and transcription factor, GLI2, in the primary cilium. Details about this communication are not known, and we wanted to explore this further. Here we report that in Tsc2 -/- MEFs constitutively activated mTORC1 led to mis-localization of Smoothened to the plasma membrane, combined with increased concentration of GLI2 in the cilia and reduced Hedgehog signaling, measured by reduced expression of the Hedgehog target gene, Gli1 Inhibition of mTORC1 rescued the cellular localization of Smoothened to the cilia, reduced the cilia concentration of GLI2, and restored Hedgehog signaling. Our results reveal evidence for a two-step activation process of GLI2. The first step includes GLI2 stabilization and cilium localization, whereas the second step includes communication with cilia-localized Smoothened. We found that mTORC1 inhibits the second step. This is the first demonstration that mTORC1 is involved in the regulation of Hedgehog signaling.


Subject(s)
Cilia , Hedgehog Proteins , Mechanistic Target of Rapamycin Complex 1 , Signal Transduction , Smoothened Receptor , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins , Zinc Finger Protein Gli2 , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Cilia/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Fibroblasts/metabolism , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Cell Membrane/metabolism , Mice, Knockout
8.
Invest Ophthalmol Vis Sci ; 65(10): 26, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39163035

ABSTRACT

Purpose: Uveal melanoma (UM) is the most common primary intraocular malignancy with a high probability of metastatic disease. Although excellent treatment options for primary UM are available, therapy for metastatic disease remain limited. Drug discovery studies using mouse models have thus far failed to provide therapeutic solutions, highlighting the need for novel models. Here, we optimize zebrafish xenografts as a potential model for drug discovery by showcasing the behavior of multiple cell lines and novel findings on mutation-dependent compound synergism/antagonism using Z-Tada; an algorithm to objectively characterize output measurements. Methods: Prognostic relevant primary (N = 4) and metastatic UM (N = 1) cell lines or healthy melanocytes (N = 2) were inoculated at three distinct inoculation sites. Standardized quantifications independent of inoculation site were obtained using Z-Tada; an algorithm to measure tumor burden and the number, size, and distance of disseminated tumor cells. Sequentially, we utilized this model to validate combinatorial synergism or antagonism seen in vitro. Results: Detailed analysis of 691 zebrafish xenografts demonstrated perivitelline space inoculation provided robust data with high probability of cell dissemination. Cell lines with more invasive behavior (SF3B1mut and BAP1mut) behaved most aggressive in this model. Combinatorial drug treatment illustrated synergism or antagonism is mutation-dependent, which were confirmed in vivo. Combinatorial treatment differed per xenograft-model, as it either inhibited overall tumor burden or cell dissemination. Conclusions: Perivitelline space inoculation provides robust zebrafish xenografts with the ability for high-throughput drug screening and robust data acquisition using Z-Tada. This model demonstrates that drug discovery for uveal melanoma must take mutational subclasses into account, especially in combinatorial treatment discoveries.


Subject(s)
Melanoma , Mutation , Uveal Neoplasms , Zebrafish , Uveal Neoplasms/genetics , Uveal Neoplasms/drug therapy , Uveal Neoplasms/pathology , Animals , Melanoma/genetics , Melanoma/drug therapy , Melanoma/pathology , Humans , Cell Line, Tumor , Disease Models, Animal , Xenograft Model Antitumor Assays , Drug Synergism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Ubiquitin Thiolesterase/genetics , Tumor Suppressor Proteins
9.
Cell Commun Signal ; 22(1): 411, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39180088

ABSTRACT

BACKGROUND: p63 is a transcription factor with intrinsic pioneer factor activity and pleiotropic functions. Transforming growth factor ß (TGFß) signaling via activation and cooperative action of canonical, SMAD, and non-canonical, MAP-kinase (MAPK) pathways, elicits both anti- and pro-tumorigenic properties, including cell stemness and invasiveness. TGFß activates the ΔNp63 transcriptional program in cancer cells; however, the link between TGFß and p63 in unmasking the epigenetic landscape during tumor progression allowing chromatin accessibility and gene transcription, is not yet reported. METHODS: Small molecule inhibitors, including protein kinase inhibitors and RNA-silencing, provided loss of function analyses. Sphere formation assays in cancer cells, chromatin immunoprecipitation and mRNA expression assays were utilized in order to gain mechanistic evidence. Mass spectrometry analysis coupled to co-immunoprecipitation assays revealed novel p63 interactors and their involvement in p63-dependent transcription. RESULTS: The sphere-forming capacity of breast cancer cells was enhanced upon TGFß stimulation and significantly decreased upon ΔNp63 depletion. Activation of TGFß signaling via p38 MAPK signaling induced ΔNp63 phosphorylation at Ser 66/68 resulting in stabilized ΔNp63 protein with enhanced DNA binding properties. TGFß stimulation altered the ratio of H3K27ac and H3K27me3 histone modification marks, pointing towards higher H3K27ac and increased p300 acetyltransferase recruitment to chromatin. By silencing the expression of ΔNp63, the TGFß effect on chromatin remodeling was abrogated. Inhibition of H3K27me3, revealed the important role of TGFß as the upstream signal for guiding ΔNp63 to the TGFß/SMAD gene loci, as well as the indispensable role of ΔNp63 in recruiting histone modifying enzymes, such as p300, to these genomic regions, regulating chromatin accessibility and gene transcription. Mechanistically, TGFß through SMAD activation induced dissociation of ΔNp63 from NURD or NCOR/SMRT histone deacetylation complexes, while promoted the assembly of ΔNp63-p300 complexes, affecting the levels of histone acetylation and the outcome of ΔNp63-dependent transcription. CONCLUSIONS: ΔNp63, phosphorylated and recruited by TGFß to the TGFß/SMAD/ΔNp63 gene loci, promotes chromatin accessibility and transcription of target genes related to stemness and cell invasion.


Subject(s)
Epigenesis, Genetic , Neoplasm Invasiveness , Neoplastic Stem Cells , Transcription Factors , Transforming Growth Factor beta , Tumor Suppressor Proteins , Humans , Transforming Growth Factor beta/metabolism , Epigenesis, Genetic/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Phosphorylation , Gene Expression Regulation, Neoplastic , Signal Transduction
10.
Rev Med Liege ; 79(7-8): 467-470, 2024 Jul.
Article in French | MEDLINE | ID: mdl-39129541

ABSTRACT

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.


Subject(s)
Mutation , Humans , Animals , Tumor Suppressor Proteins/genetics , GTP-Binding Proteins/genetics , Male , Drosophila melanogaster/genetics , Female , Infant
11.
Sci Rep ; 14(1): 18413, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117733

ABSTRACT

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.


Subject(s)
Genome-Wide Association Study , Neoplasms , Polymorphism, Single Nucleotide , Tumor Suppressor Proteins , Humans , Tumor Suppressor Proteins/genetics , Neoplasms/genetics , Neoplasms/drug therapy , Female , Male , Genetic Predisposition to Disease , Transcription Factors/genetics , Case-Control Studies , DNA, Intergenic/genetics , Middle Aged , Aged , Chromosome Mapping
13.
Oncol Rep ; 52(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39155859

ABSTRACT

It is well known how the precise localization of glioblastoma multiforme (GBM) predicts the direction of tumor spread in the surrounding neuronal structures. The aim of the present review is to reveal the lateralization of GBM by evaluating the anatomical regions where it is frequently located as well as the main molecular alterations observed in different brain regions. According to the literature, the precise or most frequent lateralization of GBM has yet to be determined. However, it can be said that GBM is more frequently observed in the frontal lobe. Tractus and fascicles involved in GBM appear to be focused on the corticospinal tract, superior longitudinal I, II and III fascicles, arcuate fascicle long segment, frontal strait tract, and inferior fronto­occipital fasciculus. Considering the anatomical features of GBM and its brain involvement, it is logical that the main brain regions involved are the frontal­temporal­parietal­occipital lobes, respectively. Although tumor volumes are higher in the right hemisphere, it has been determined that the prognosis of patients diagnosed with cancer in the left hemisphere is worse, probably reflecting the anatomical distribution of some detrimental alterations such as TP53 mutations, PTEN loss, EGFR amplification, and MGMT promoter methylation. There are theories stating that the right hemisphere is less exposed to external influences in its development as it is responsible for the functions necessary for survival while tumors in the left hemisphere may be more aggressive. To shed light on specific anatomical and molecular features of GBM in different brain regions, the present review article is aimed at describing the main lateralization pathways as well as gene mutations or epigenetic modifications associated with the development of brain tumors.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Mutation , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Prognosis , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism
14.
Genes Dev ; 38(13-14): 675-691, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39137945

ABSTRACT

Tumor suppressor genes play critical roles in normal tissue homeostasis, and their dysregulation underlies human diseases including cancer. Besides human genetics, model organisms such as Drosophila have been instrumental in discovering tumor suppressor pathways that were subsequently shown to be highly relevant in human cancer. Here we show that hyperplastic disc (Hyd), one of the first tumor suppressors isolated genetically in Drosophila and encoding an E3 ubiquitin ligase with hitherto unknown substrates, and Lines (Lin), best known for its role in embryonic segmentation, define an obligatory tumor suppressor protein complex (Hyd-Lin) that targets the zinc finger-containing oncoprotein Bowl for ubiquitin-mediated degradation, with Lin functioning as a substrate adaptor to recruit Bowl to Hyd for ubiquitination. Interestingly, the activity of the Hyd-Lin complex is directly inhibited by a micropeptide encoded by another zinc finger gene, drumstick (drm), which functions as a pseudosubstrate by displacing Bowl from the Hyd-Lin complex, thus stabilizing Bowl. We further identify the epigenetic regulator Polycomb repressive complex1 (PRC1) as a critical upstream regulator of the Hyd-Lin-Bowl pathway by directly repressing the transcription of the micropeptide drm Consistent with these molecular studies, we show that genetic inactivation of Hyd, Lin, or PRC1 resulted in Bowl-dependent hyperplastic tissue overgrowth in vivo. We also provide evidence that the mammalian homologs of Hyd (UBR5, known to be recurrently dysregulated in various human cancers), Lin (LINS1), and Bowl (OSR1/2) constitute an analogous protein degradation pathway in human cells, and that OSR2 promotes prostate cancer tumorigenesis. Altogether, these findings define a previously unrecognized tumor suppressor pathway that links epigenetic program to regulated protein degradation in tissue growth control and tumorigenesis.


Subject(s)
Carcinogenesis , Drosophila Proteins , Proteolysis , Ubiquitin-Protein Ligases , Animals , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Carcinogenesis/genetics , Humans , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/embryology , Genes, Tumor Suppressor , Ubiquitination , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/genetics
15.
Nat Commun ; 15(1): 6953, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138205

ABSTRACT

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.


Subject(s)
Ebolavirus , Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Transcription, Genetic , Virus Release , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Phosphorylation , Ebolavirus/physiology , Ebolavirus/genetics , Ebolavirus/metabolism , HEK293 Cells , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , YAP-Signaling Proteins/metabolism , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism , Host-Pathogen Interactions , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics
16.
J Extracell Vesicles ; 13(8): e12488, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39104296

ABSTRACT

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.


Subject(s)
Drug Resistance, Neoplasm , Extracellular Vesicles , Hippo Signaling Pathway , MicroRNAs , Osteonectin , Pancreatic Neoplasms , Pancreatic Stellate Cells , Protein Serine-Threonine Kinases , Tumor Microenvironment , MicroRNAs/metabolism , MicroRNAs/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Humans , Pancreatic Stellate Cells/metabolism , Animals , Protein Serine-Threonine Kinases/metabolism , Mice , Osteonectin/metabolism , Osteonectin/genetics , Extracellular Vesicles/metabolism , Cell Line, Tumor , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Gene Expression Regulation, Neoplastic , Gemcitabine , Signal Transduction , Mice, Nude
18.
Cell Death Dis ; 15(8): 610, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174499

ABSTRACT

PARP1 is crucial in DNA damage repair, chromatin remodeling, and transcriptional regulation. The principle of synthetic lethality has effectively guided the application of PARP inhibitors in treating tumors carrying BRCA1/2 mutations. Meanwhile, PARP inhibitors have exhibited efficacy in BRCA-proficient patients, further highlighting the necessity for a deeper understanding of PARP1 function and its inhibition in cancer therapy. Here, we unveil PIN2/TRF1-interacting telomerase inhibitor 1 (PINX1) as an uncharacterized PARP1-interacting protein that synergizes with PARP inhibitors upon its depletion across various cancer cell lines. Loss of PINX1 compromises DNA damage repair capacity upon etoposide treatment. The vulnerability of PINX1-deficient cells to etoposide and PARP inhibitors could be effectively restored by introducing either a full-length or a mutant form of PINX1 lacking telomerase inhibitory activity. Mechanistically, PINX1 is recruited to DNA lesions through binding to the ZnF3-BRCT domain of PARP1, facilitating the downstream recruitment of the DNA repair factor XRCC1. In the absence of DNA damage, PINX1 constitutively binds to PARP1, promoting PARP1-chromatin association and transcription of specific DNA damage repair proteins, including XRCC1, and transcriptional regulators, including GLIS3. Collectively, our findings identify PINX1 as a multifaceted partner of PARP1, crucial for safeguarding cells against genotoxic stress and emerging as a potential candidate for targeted tumor therapy.


Subject(s)
Cell Cycle Proteins , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Cell Line, Tumor , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , DNA Damage , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , DNA Repair/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Etoposide/pharmacology , X-ray Repair Cross Complementing Protein 1
19.
Pathologie (Heidelb) ; 45(5): 316-323, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39110166

ABSTRACT

Diffuse mesotheliomas are characterized by recurrent genomic alterations involving tumor suppressors and epigenetic regulators such as BAP1, CDKN2A, MTAP, and NF2. Depending on the differential diagnosis as informed by histologic assessment, one can apply the appropriate immunohistochemical and/or molecular panels to reach the correct pathologic diagnosis, sometimes even in cases with limited tissues. Biomarkers aid in the diagnosis of mesothelioma in the following scenarios: 1) For a tumor that is overtly malignant, how can one distinguish mesothelioma from other tumors? 2) For a mesothelial proliferation, how can one distinguish mesothelioma from a reactive process? To distinguish mesotheliomas from carcinomas, at least two positive and two negative markers are currently recommended. To distinguish sarcomatoid mesothelioma from pleomorphic carcinoma, even more markers-and sometimes molecular testing-are needed. To distinguish mesothelioma from reactive mesothelial conditions, useful immunohistochemical biomarkers include BAP1, MTAP, and merlin, which serve as surrogates for the corresponding gene mutation status. In patients with unusual clinical history, for tumors with a peculiar microscopic appearance, and/or in cases with an equivocal immunophenotypic profile, molecular testing can help to exclude mimics and to confirm the pathologic diagnosis.


Subject(s)
Biomarkers, Tumor , Mesothelioma , Humans , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Mesothelioma/diagnosis , Mesothelioma/genetics , Mesothelioma/pathology , Mesothelioma/metabolism , Diagnosis, Differential , Pathology, Molecular/methods , Immunohistochemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
20.
BMC Genomics ; 25(1): 757, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095712

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Developmental , Matrix Attachment Region Binding Proteins , Neocortex , Transcription Factors , Transcriptome , Animals , Neocortex/metabolism , Neocortex/growth & development , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Profiling , Mice, Knockout , Repressor Proteins , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL