Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 116
Filtrer
1.
BMC Cancer ; 24(1): 334, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38475740

RÉSUMÉ

BACKGROUND: Ribosomal RNA processing protein 15 (RRP15) has been found to regulate the progression of hepatocellular carcinoma (HCC). Nevertheless, the extent to which it contributes to the spread of HCC cells remains uncertain. Thus, the objective of this research was to assess the biological function of RRP15 in the migration of HCC. METHODS: The expression of RRP15 in HCC tissue microarray (TMA), tumor tissues and cell lines were determined. In vitro, the effects of RRP15 knockdown on the migration, invasion and adhesion ability of HCC cells were assessed by wound healing assay, transwell and adhesion assay, respectively. The effect of RRP15 knockdown on HCC migration was also evaluated in vivo in a mouse model. RESULTS: Bioinformatics analysis showed that high expression of RRP15 was significantly associated with low survival rate of HCC. The expression level of RRP15 was strikingly upregulated in HCC tissues and cell lines compared with the corresponding controls, and TMA data also indicated that RRP15 was a pivotal prognostic factor for HCC. RRP15 knockdown in HCC cells reduced epithelial-to-mesenchymal transition (EMT) and inhibited migration in vitro and in vivo, independent of P53 expression. Mechanistically, blockade of RRP15 reduced the protein level of the transcription factor POZ/BTB and AT hook containing zinc finger 1 (PATZ1), resulting in decreased expression of the downstream genes encoding laminin 5 subunits, LAMC2 and LAMB3, eventually suppressing the integrin ß4 (ITGB4)/focal adhesion kinase (FAK)/nuclear factor κB kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway. CONCLUSIONS: RRP15 promotes HCC migration by activating the LAMC2/ITGB4/FAK pathway, providing a new target for future HCC treatment.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Maturation post-transcriptionnelle des ARN , Protéines ribosomiques , Animaux , Souris , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire , Transition épithélio-mésenchymateuse/génétique , Focal adhesion protein-tyrosine kinases/génétique , Focal adhesion protein-tyrosine kinases/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Facteurs de transcription/génétique , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme
2.
Clin Res Hepatol Gastroenterol ; 48(3): 102289, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38307254

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide. Dysregulation of ribosome biogenesis increases the risk of cancer. RPF2 (ribosome production factor 2 homolog), a member of the BRIX family, is involved in ribosome biogenesis. However, the biological functions of RPF2 in HCC remain unclear. This study aims to evaluate the function of RPF2 and its clinical significance in HCC. We collected 45 pairs of HCC/adjacent samples and 291 HCC samples. These samples were used to perform immunohistochemical analysis and western blot. Six cell lines were used to perform western blot, and two of cell lines, SMCC-7721 and SNU449, were subjected to CCK-8, wound healing and transwell assays. Immunofluorescence staining was executed in SMCC-7721 cells. The protein levels of RPF2 were higher in HCC tissues than in adjacent tissues. Immunofluorescence staining showed that the RPF2 protein was located in the nucleuses, especially the nucleolus. Furthermore, the immunohistochemical analysis showed that high expression levels of nuclear RPF2 correlated with poor prognosis, vascular invasion, liver cirrhosis and tumor size. Cell experiments showed that overexpression of RPF2 promoted cell proliferation, migration and invasion, while knockdown of RPF2 tended to show the opposite effect. This is the first report that RPF2 is involved in HCC progression. The levels of RPF2 were significantly high in HCC tumors and had a side effect on prognosis in HCC patients. RPF2 has the potential to be a useful marker for HCC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Humains , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/anatomopathologie , Pertinence clinique , Pronostic , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Prolifération cellulaire , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux
3.
Neuro Oncol ; 25(12): 2191-2206, 2023 12 08.
Article de Anglais | MEDLINE | ID: mdl-37531290

RÉSUMÉ

BACKGROUND: High-grade adult-type diffuse gliomas (HGGs) constitute a heterogeneous group of aggressive tumors that are mostly incurable. Recent advances highlighting the contribution of ribosomes to cancer development have offered new clinical perspectives. Here, we uncovered that isocitrate dehydrogenase (IDH)wt and IDHmut HGGs display distinct alterations of ribosome biology, in terms of rRNA epitranscriptomics and ribosome biogenesis, which could constitute novel hallmarks that can be exploited for the management of these pathologies. METHODS: We analyzed (1) the ribosomal RNA 2'O-ribose methylation (rRNA 2'Ome) using RiboMethSeq and in-house developed bioinformatics tools (https://github.com/RibosomeCRCL/ribomethseq-nfandrRMSAnalyzer) on 3 independent cohorts compiling 71 HGGs (IDHwt n = 30, IDHmut n = 41) and 9 non-neoplastic samples, (2) the expression of ribosome biogenesis factors using medium throughput RT-qPCR as a readout of ribosome biogenesis, and (3) the sensitivity of 5 HGG cell lines to RNA Pol I inhibitors (CX5461, BMH-21). RESULTS: Unsupervised analysis demonstrated that HGGs could be distinguished based on their rRNA 2'Ome epitranscriptomic profile, with IDHwt glioblastomas displaying the most significant alterations of rRNA 2'Ome at specific sites. In contrast, IDHmut HGGs are largely characterized by an overexpression of ribosome biogenesis factors compared to non-neoplastic tissues or IDHwt glioblastomas. Finally, IDHmut HGG-derived spheroids display higher cytotoxicity to CX5461 than IDHwt glioblastoma, while all HGG spheroids display a similar cytotoxicity to BMH-21. CONCLUSIONS: In HGGs, IDH mutational status is associated with specific alterations of the ribosome biology and with distinct sensitivities to RNA Pol I inhibitors.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Gliome , Adulte , Humains , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , Tumeurs du cerveau/anatomopathologie , Glioblastome/génétique , Glioblastome/métabolisme , ARN ribosomique/génétique , ARN ribosomique/métabolisme , Gliome/anatomopathologie , Méthylation , Ribosomes/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Mutation
4.
Adv Sci (Weinh) ; 10(19): e2206098, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37142882

RÉSUMÉ

KMT2C and KMT2D are the most frequently mutated epigenetic genes in human cancers. While KMT2C is identified as a tumor suppressor in acute myeloid leukemia (AML), the role of KMT2D remains unclear in this disease, though its loss promotes B cell lymphoma and various solid cancers. Here, it is reported that KMT2D is downregulated or mutated in AML and its deficiency, through shRNA knockdown or CRISPR/Cas9 editing, accelerates leukemogenesis in mice. Hematopoietic stem and progenitor cells and AML cells with Kmt2d loss have significantly enhanced ribosome biogenesis and consistently, enlarged nucleolus, increased rRNA and protein synthesis rates. Mechanistically, it is found that KMT2D deficiency leads to the activation of the mTOR pathway in both mouse and human AML cells. Kmt2d directly regulates the expression of Ddit4, a negative regulator of the mTOR pathway. Consistent with the abnormal ribosome biogenesis, it is shown that CX-5461, an inhibitor of RNA polymerase I, significantly restrains the growth of AML with Kmt2d loss in vivo and extends the survival of leukemic mice. These studies validate KMT2D as a de facto tumor suppressor in AML and reveal an unprecedented vulnerability to ribosome biogenesis inhibition.


Sujet(s)
Leucémie aigüe myéloïde , Humains , Animaux , Souris , Leucémie aigüe myéloïde/métabolisme , Gènes suppresseurs de tumeur , Sérine-thréonine kinases TOR/métabolisme , Petit ARN interférent/métabolisme , Ribosomes/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie
5.
Gastroenterology ; 165(3): 629-646, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37247644

RÉSUMÉ

BACKGROUND & AIMS: Hyperactivation of ribosome biogenesis leads to hepatocyte transformation and plays pivotal roles in hepatocellular carcinoma (HCC) development. We aimed to identify critical ribosome biogenesis proteins that are overexpressed and crucial in HCC progression. METHODS: HEAT repeat containing 1 (HEATR1) expression and clinical correlations were analyzed using The Cancer Genome Atlas and Gene Expression Omnibus databases and further evaluated by immunohistochemical analysis of an HCC tissue microarray. Gene expression was knocked down by small interfering RNA. HEATR1-knockdown cells were subjected to viability, cell cycle, and apoptosis assays and used to establish subcutaneous and orthotopic tumor models. Chromatin immunoprecipitation and quantitative polymerase chain reaction were performed to detect the association of candidate proteins with specific DNA sequences. Endogenous coimmunoprecipitation combined with mass spectrometry was used to identify protein interactions. We performed immunoblot and immunofluorescence assays to detect and localize proteins in cells. The nucleolus ultrastructure was detected by transmission electron microscopy. Click-iT (Thermo Fisher Scientific) RNA imaging and puromycin incorporation assays were used to measure nascent ribosomal RNA and protein synthesis, respectively. Proteasome activity, 20S proteasome foci formation, and protein stability were evaluated in HEATR1-knockdown HCC cells. RESULTS: HEATR1 was the most up-regulated gene in a set of ribosome biogenesis mediators in HCC samples. High expression of HEATR1 was associated with poor survival and malignant clinicopathologic features in patients with HCC and contributed to HCC growth in vitro and in vivo. HEATR1 expression was regulated by the transcription factor specificity protein 1, which can be activated by insulin-like growth factor 1-mammalian target of rapamycin complex 1 signaling in HCC cells. HEATR1 localized predominantly in the nucleolus, bound to ribosomal DNA, and was associated with RNA polymerase I transcription/processing factors. Knockdown of HEATR1 disrupted ribosomal RNA biogenesis and impaired nascent protein synthesis, leading to reduced cytoplasmic proteasome activity and inhibitory-κB/nuclear factor-κB signaling. Moreover, HEATR1 knockdown induced nucleolar stress with increased nuclear proteasome activity and inactivation of the nucleophosmin 1-MYC axis. CONCLUSIONS: Our study revealed that HEATR1 is up-regulated by insulin-like growth factor 1-mammalian target of rapamycin complex 1-specificity protein 1 signaling in HCC and functions as a crucial regulator of ribosome biogenesis and proteome homeostasis to promote HCC development.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Humains , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Homéostasie , Température élevée , Facteur de croissance IGF-I/génétique , Tumeurs du foie/anatomopathologie , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Proteasome endopeptidase complex/génétique , Protéome/métabolisme , Ribosomes/métabolisme , Ribosomes/anatomopathologie , ARN ribosomique/génétique , ARN ribosomique/métabolisme
6.
J Cell Mol Med ; 27(3): 365-378, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36625087

RÉSUMÉ

Growth is crucially controlled by the functional ribosomes available in cells. To meet the enhanced energy demand, cancer cells re-wire and increase their ribosome biogenesis. The RNA-binding protein PNO1, a ribosome assembly factor, plays an essential role in ribosome biogenesis. The purpose of this study was to examine whether PNO1 can be used as a biomarker for lung adenocarcinoma and also examine the molecular mechanisms by which PNO1 knockdown by CRISPR/Cas9 inhibited growth and epithelial-mesenchymal transition (EMT). The expression of PNO1 was significantly higher in lung adenocarcinoma compared to normal lung tissues. PNO1 expression in lung adenocarcinoma patients increased with stage, nodal metastasis, and smoking. Lung adenocarcinoma tissues from males expressed higher PNO1 than those from females. Furthermore, lung adenocarcinoma tissues with mutant Tp53 expressed higher PNO1 than those with wild-type Tp53, suggesting the influence of Tp53 status on PNO1 expression. PNO1 knockdown inhibited cell viability, colony formation, and EMT, and induced apoptosis. Since dysregulated signalling through the Notch receptors promotes lung adenocarcinoma, we measured the effects of PNO1 inhibition on the Notch pathway. PNO1 knockdown inhibited Notch signalling by suppressing the expression of Notch receptors, their ligands, and downstream targets. PNO1 knockdown also suppressed CCND1, p21, PTGS-2, IL-1α, IL-8, and CXCL-8 genes. Overall, our data suggest that PNO1 can be used as a diagnostic biomarker, and also can be an attractive therapeutic target for the treatment of lung adenocarcinoma.


Sujet(s)
Adénocarcinome pulmonaire , Adénocarcinome , Tumeurs du poumon , Mâle , Femelle , Humains , Systèmes CRISPR-Cas/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome/anatomopathologie , Récepteurs Notch/génétique , Récepteurs Notch/métabolisme , Tumeurs du poumon/anatomopathologie , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Transition épithélio-mésenchymateuse/génétique , Lignée cellulaire tumorale , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme
7.
Phytomedicine ; 108: 154478, 2023 Jan.
Article de Anglais | MEDLINE | ID: mdl-36265255

RÉSUMÉ

Activation of mitogen-activated protein kinase (MAPK) and PI3K signaling confers resistance against sorafenib, a mainstay treatment for advanced hepatocellular carcinoma (HCC). Antrocin and ovatodiolide constitute as the most potent secondary metabolites isolated from Antrodia camphorata and Anisomeles indica, respectively. Both natural compounds have recently gained a lot of attention due to their putative inhibition of MAPK and PI3K signaling in various solid cancers. However, whether their combination is effective in HCC remains unknown. Here, we investigated their effect, alone or in various combinations, on MAPK and PI3K signaling pathways in HCC cells. An array of in vitro study were used to investigate anticancer and stemness effects to treat HCC, such as cytotoxicity, drug combination index, migration, invasion, colony formation, and tumor sphere formation. Drug effect in vivo was evaluated using mouse xenograft models. In this study, antrocin and ovatodiolide synergistically inhibited the SNU387, Hep3B, Mahlavu, and Huh7 cell lines. Sequential combination treatment of Huh7 and Mahlavu with ovatodiolide followed by antrocin resulted stronger cytotoxic effect than did treatment with antrocin followed by ovatodiolide, their simultaneous administration, antrocin alone, or ovatodiolide alone. In the Huh7 and Mahlavu cell lines, ovatodiolide→antrocin significantly suppressed colony formation and proliferation as well as markedly downregulated ERK1/2, Akt, and mTOR expression. Inhibition of ERK1/2 and Akt/mTOR signaling by ovatodiolide→antrocin suppressed ribosomal biogenesis, autophagy, and cancer stem cell-like phenotypes and promoted apoptosis in Huh7 and Mahlavu cells. The sorafenib-resistant clone of Huh7 was effectively inhibited by synergistic combination of both compound in vitro. Eventually, the ovatodiolide→antrocin combination synergistically suppressed the growth of HCC xenografts. Taken together, our findings suggested that ovatodiolide→antrocin combination may represent potential therapeutic approach for patients with advanced HCC.


Sujet(s)
Carcinome hépatocellulaire , Diterpènes , Tumeurs du foie , Animaux , Humains , Souris , Apoptose , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Sorafénib , Sérine-thréonine kinases TOR/métabolisme , Lactones/pharmacologie , Diterpènes/pharmacologie , Sesquiterpènes/pharmacologie , Cellules souches tumorales/effets des médicaments et des substances chimiques
8.
Neuro Oncol ; 25(6): 1058-1072, 2023 06 02.
Article de Anglais | MEDLINE | ID: mdl-36583853

RÉSUMÉ

BACKGROUND: High-grade gliomas are malignant brain tumors characterized by aggressiveness and resistance to chemotherapy. Prognosis remains dismal, highlighting the need to identify novel molecular dependencies and targets. Ribosome biogenesis (RiBi), taking place in the nucleolus, represents a promising target as several cancer types rely on high RiBi rates to sustain proliferation. Publicly available transcriptomics data of glioma patients revealed a positive correlation between RiBi rates and histological grades. We, therefore, hypothesized that glioma cells could be susceptible to RiBi inhibition. METHODS: Transcriptomics data from glioma patients were analyzed for RiBi-related processes. BMH-21, a small molecule inhibitor of RNA pol I transcription, was tested in adult and pediatric high-grade glioma cell lines and a zebrafish transplant model. Cellular phenotypes were evaluated by transcriptomics, cell cycle analysis, and viability assays. A chemical synergy screen was performed to identify drugs potentiating BMH-21-mediated effects. RESULTS: BMH-21 reduced glioma cell viability, induced apoptosis, and impaired the growth of transplanted glioma cells in zebrafish. Combining BMH-21 with TMZ potentiated cytotoxic effects. Moreover, BMH-21 synergized with Fibroblast Growth Factor Receptor (FGFR) inhibitor (FGFRi) Erdafitinib, a top hit in the chemical synergy screen. RiBi inhibition using BMH-21, POLR1A siRNA, or Actinomycin D revealed engagement of the FGFR-FGF2 pathway. BMH-21 downregulated FGFR1 and SOX2 levels, whereas FGF2 was induced and released from the nucleolus. CONCLUSIONS: This study conceptualizes the implementation of RiBi inhibition as a viable future therapeutic strategy for glioma and reveals an FGFR connection to the cellular response upon RiBi inhibition with potential translational value.


Sujet(s)
Gliome , Danio zébré , Animaux , Facteur de croissance fibroblastique de type 2/pharmacologie , Facteur de croissance fibroblastique de type 2/usage thérapeutique , Lignée cellulaire tumorale , Gliome/génétique , Prolifération cellulaire , Cycle cellulaire , Inhibiteurs de protéines kinases/pharmacologie , Ribosomes/métabolisme , Ribosomes/anatomopathologie
9.
Expert Rev Clin Pharmacol ; 15(6): 729-746, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35787725

RÉSUMÉ

INTRODUCTION: The ribosome is a ribonucleoprotein organelle responsible for protein synthesis, and its biogenesis is a highly coordinated process that involves many macromolecular components. Any acquired or inherited impairment in ribosome biogenesis or ribosomopathies is associated with the development of different cancers and rare genetic diseases. Interference with multiple steps of protein synthesis has been shown to promote tumor cell death. AREAS COVERED: We discuss the current insights about impaired ribosome biogenesis and their secondary consequences on protein synthesis, transcriptional and translational responses, proteotoxic stress, and other metabolic pathways associated with cancer and rare diseases. The modulation of different therapeutic chemical entities targeting cancer in in vitro and in vivo models have also been detailed. EXPERT OPINION: Despite the association between inherited mutations affecting ribosome biogenesis and cancer biology, the development of therapeutics targeting the essential cellular machinery has only started to emerge. New chemical entities should be designed to modulate different checkpoints (translating oncoproteins, dysregulation of specific ribosome-assembly machinery, ribosomal stress, and rewiring ribosomal functions). Although safe and effective therapies are lacking, consideration should be given to using existing drugs alone or in combination for long-term safety, with known risks for feasibility in clinical trials and synergistic effects.


Sujet(s)
Tumeurs , Protéines ribosomiques , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Biosynthèse des protéines , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme , Ribosomes/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie
10.
Int J Oncol ; 60(6)2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35543151

RÉSUMÉ

Metabolic rewiring fuels rapid cancer cell proliferation by promoting adjustments in energetic resources, and increasing glucose uptake and its conversion into lactate, even in the presence of oxygen. Furthermore, solid tumors often contain hypoxic areas and can rapidly adapt to low oxygen conditions by activating hypoxia inducible factor (HIF)­1α and several downstream pathways, thus sustaining cell survival and metabolic reprogramming. Since TNF receptor­associated protein 1 (TRAP1) is a HSP90 molecular chaperone upregulated in several human malignancies and is involved in cancer cell adaptation to unfavorable environments and metabolic reprogramming, in the present study, its role was investigated in the adaptive response to hypoxia in human colorectal cancer (CRC) cells and organoids. In the present study, glucose uptake, lactate production and the expression of key metabolic genes were evaluated in TRAP1­silenced CRC cell models under conditions of hypoxia/normoxia. Whole genome gene expression profiling was performed in TRAP1­silenced HCT116 cells exposed to hypoxia to establish the role of TRAP1 in adaptive responses to oxygen deprivation. The results revealed that TRAP1 was involved in regulating hypoxia­induced HIF­1α stabilization and glycolytic metabolism and that glucose transporter 1 expression, glucose uptake and lactate production were partially impaired in TRAP1­silenced CRC cells under hypoxic conditions. At the transcriptional level, the gene expression reprogramming of cancer cells driven by HIF­1α was partially inhibited in TRAP1­silenced CRC cells and organoids exposed to hypoxia. Moreover, Gene Set Enrichment Analysis of TRAP1­silenced HCT116 cells exposed to hypoxia demonstrated that TRAP1 was involved in the regulation of ribosome biogenesis and this occurred with the inhibition of the mTOR pathway. Therefore, as demonstrated herein, TRAP1 is a key factor in maintaining HIF­1α­induced genetic/metabolic program under hypoxic conditions and may represent a promising target for novel metabolic therapies.


Sujet(s)
Tumeurs colorectales , Oxygène , Hypoxie cellulaire , Tumeurs colorectales/anatomopathologie , Glucose/métabolisme , Glycolyse , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , Humains , Hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Lactates , Oxygène/métabolisme , Ribosomes/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Facteur-1 associé aux récepteurs de TNF/métabolisme
11.
Brain Tumor Pathol ; 39(2): 51-56, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-35508789

RÉSUMÉ

The characteristic features of plasticity and heterogeneity in glioblastoma (GB) cells cause therapeutic difficulties. GB cells are exposed to various stimuli from the tumor microenvironment and acquire the potential to resist chemoradiotherapy. To investigate how GB cells acquire stem cell-like phenotypes, we focused on ribosomal proteins, because ribosome incorporation has been reported to induce stem cell-like phenotypes in somatic cells. Furthermore, dysregulation of ribosome biogenesis has been reported in several types of cancer. We focused on ribosomal protein S6, which promotes sphere-forming ability and stem cell marker expression in GB cells. We expect that investigation of dysregulation of ribosome biogenesis and extra-ribosomal function in GB will provide new insights about the plasticity, heterogeneity, and therapeutic resistance of GB cells, which can potentially lead to revolutionary therapeutic strategies.


Sujet(s)
Glioblastome , Gliome , Glioblastome/traitement médicamenteux , Glioblastome/thérapie , Gliome/anatomopathologie , Humains , Cellules souches tumorales/anatomopathologie , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme , Protéines ribosomiques/usage thérapeutique , Ribosomes/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Microenvironnement tumoral
12.
Oncol Rep ; 47(6)2022 06.
Article de Anglais | MEDLINE | ID: mdl-35445733

RÉSUMÉ

The aim of the present study was to investigate the expression of ribosome assembly factor partner of NOB1 homolog (PNO1) and its association with the progression of breast cancer (BC) in patients, as well as its biological function and underlying mechanism of action in BC cells. Bioinformatics and immunohistochemical analyses revealed that PNO1 expression was significantly increased in BC tissues and its high mRNA expression was associated with shorter overall survival (OS) and relapse­free survival (RFS) of patients with BC, as well as multiple clinical characteristics (including advanced stage of NPI and SBR, etc.) of patients with BC. Biological functional studies revealed that transduction of lentivirus encoding sh­PNO1 significantly downregulated PNO1 expression, reduced cell confluency and the number of BC cells in vitro and inhibited tumor growth in vivo. Moreover, PNO1 knockdown decreased the cell viability and arrested cell cycle progression at the G2/M phase, as well as downregulated cyclin B1 (CCNB1) and cyclin­dependent kinase 1 (CDK1) protein expression in BC cells. Correlation analysis demonstrated that PNO1 expression was positively correlated with both CDK1 and CCNB1 expression in BC samples. Collectively, PNO1 was upregulated in BC and associated with BC patient survival, and PNO1 knockdown suppressed tumor growth in vitro and in vivo. In addition, positive regulation of CCNB1 and CDK1 may be one of the underlying mechanisms.


Sujet(s)
Tumeurs du sein , Tumeurs du sein triple-négatives , Tumeurs du sein/génétique , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Transformation cellulaire néoplasique/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Récidive tumorale locale/génétique , Protéines nucléaires/génétique , Protéines de liaison à l'ARN/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie
13.
Nat Commun ; 13(1): 134, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-35013307

RÉSUMÉ

Combined methylmalonic acidemia and homocystinuria (cblC) is the most common inborn error of intracellular cobalamin metabolism and due to mutations in Methylmalonic Aciduria type C and Homocystinuria (MMACHC). Recently, mutations in the transcriptional regulators HCFC1 and RONIN (THAP11) were shown to result in cellular phenocopies of cblC. Since HCFC1/RONIN jointly regulate MMACHC, patients with mutations in these factors suffer from reduced MMACHC expression and exhibit a cblC-like disease. However, additional de-regulated genes and the resulting pathophysiology is unknown. Therefore, we have generated mouse models of this disease. In addition to exhibiting loss of Mmachc, metabolic perturbations, and developmental defects previously observed in cblC, we uncovered reduced expression of target genes that encode ribosome protein subunits. We also identified specific phenotypes that we ascribe to deregulation of ribosome biogenesis impacting normal translation during development. These findings identify HCFC1/RONIN as transcriptional regulators of ribosome biogenesis during development and their mutation results in complex syndromes exhibiting aspects of both cblC and ribosomopathies.


Sujet(s)
Aminoacidopathies congénitales/génétique , Homocystinurie/génétique , Facteur de prolifération cellulaire HCF/génétique , Oxidoreductases/génétique , Protéines de répression/génétique , Ribosomes/génétique , Carence en vitamine B12/génétique , Aminoacidopathies congénitales/métabolisme , Aminoacidopathies congénitales/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Embryon de mammifère , Femelle , Régulation de l'expression des gènes au cours du développement , Homocystinurie/métabolisme , Homocystinurie/anatomopathologie , Facteur de prolifération cellulaire HCF/déficit , Humains , Mâle , Souris , Souris knockout , Mutation , Biogenèse des organelles , Oxidoreductases/déficit , Biosynthèse des protéines , Sous-unités de protéines/génétique , Sous-unités de protéines/métabolisme , Protéines de répression/déficit , Protéines ribosomiques/génétique , Protéines ribosomiques/métabolisme , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Vitamine B12/métabolisme , Carence en vitamine B12/métabolisme , Carence en vitamine B12/anatomopathologie
14.
Hum Mutat ; 43(3): 389-402, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34961992

RÉSUMÉ

Diamond-Blackfan anemia is a rare genetic disease characterized by erythroblastopenia and a large spectrum of developmental anomalies. The vast majority of the cases genetically described are linked to heterozygous pathogenic variants in more than 20 ribosomal protein genes. Here we report an atypical clinical case of DBA associated with a missense variant in RPL8, which encodes RPL8/uL2, a protein of the 60S large ribosomal subunit. RPL8 has been previously implicated as a candidate disease gene in one patient with DBA bearing another type of missense variant; however, evidence for pathogenicity was limited to computational tools. Using functional studies in lymphoblastoid cells as well as yeast models, we show that the RPL8 variants detected in these two patients encode functionally deficient proteins that affect ribosome production and are therefore likely pathogenic. We propose to include RPL8 in the list of DBA-associated genes.


Sujet(s)
Anémie de Blackfan-Diamond , Protéines ribosomiques , Anémie de Blackfan-Diamond/génétique , Humains , Mutation , Phénotype , Protéines ribosomiques/génétique , Ribosomes/génétique , Ribosomes/métabolisme , Ribosomes/anatomopathologie
15.
Nat Commun ; 12(1): 5044, 2021 08 19.
Article de Anglais | MEDLINE | ID: mdl-34413298

RÉSUMÉ

Indirect somatic genetic rescue (SGR) of a germline mutation is thought to be rare in inherited Mendelian disorders. Here, we establish that acquired mutations in the EIF6 gene are a frequent mechanism of SGR in Shwachman-Diamond syndrome (SDS), a leukemia predisposition disorder caused by a germline defect in ribosome assembly. Biallelic mutations in the SBDS or EFL1 genes in SDS impair release of the anti-association factor eIF6 from the 60S ribosomal subunit, a key step in the translational activation of ribosomes. Here, we identify diverse mosaic somatic genetic events (point mutations, interstitial deletion, reciprocal chromosomal translocation) in SDS hematopoietic cells that reduce eIF6 expression or disrupt its interaction with the 60S subunit, thereby conferring a selective advantage over non-modified cells. SDS-related somatic EIF6 missense mutations that reduce eIF6 dosage or eIF6 binding to the 60S subunit suppress the defects in ribosome assembly and protein synthesis across multiple SBDS-deficient species including yeast, Dictyostelium and Drosophila. Our data suggest that SGR is a universal phenomenon that may influence the clinical evolution of diverse Mendelian disorders and support eIF6 suppressor mimics as a therapeutic strategy in SDS.


Sujet(s)
Mutation , Grande sous-unité du ribosome des eucaryotes/métabolisme , Ribosomes/génétique , Ribosomes/anatomopathologie , Maladie de Shwachman/génétique , Maladie de Shwachman/anatomopathologie , Adolescent , Adulte , Animaux , Phénomènes biologiques , Cellules cultivées , Enfant , Enfant d'âge préscolaire , Dictyostelium , Drosophila , Facteurs d'initiation eucaryotes/génétique , Facteurs d'initiation eucaryotes/métabolisme , Cellules germinales , Humains , Nourrisson , Simulation de dynamique moléculaire , Facteurs élongation chaîne peptidique/génétique , Facteurs élongation chaîne peptidique/métabolisme , Liaison aux protéines , Biosynthèse des protéines , Protéines/génétique , Protéines/métabolisme , Petites particules nucléaires ribonucléoprotéiques U5/génétique , Petites particules nucléaires ribonucléoprotéiques U5/métabolisme , Ribosomes/métabolisme , Saccharomyces cerevisiae , Similitude de séquences d'acides aminés , Maladie de Shwachman/métabolisme , Jeune adulte
16.
Acta Neuropathol Commun ; 9(1): 110, 2021 06 19.
Article de Anglais | MEDLINE | ID: mdl-34147135

RÉSUMÉ

The synthesis of new proteins is a fundamental aspect of cellular life and is required for many neurological processes, including the formation, updating and extinction of long-term memories. Protein synthesis is impaired in neurodegenerative diseases including tauopathies, in which pathology is caused by aberrant changes to the microtubule-associated protein tau. We recently showed that both global de novo protein synthesis and the synthesis of select ribosomal proteins (RPs) are decreased in mouse models of frontotemporal dementia (FTD) which express mutant forms of tau. However, a comprehensive analysis of the effect of FTD-mutant tau on ribosomes is lacking. Here we used polysome profiling, de novo protein labelling and mass spectrometry-based proteomics to examine how ribosomes are altered in models of FTD. We identified 10 RPs which were decreased in abundance in primary neurons taken from the K3 mouse model of FTD. We further demonstrate that expression of human tau (hTau) decreases both protein synthesis and biogenesis of the 60S ribosomal subunit, with these effects being exacerbated in the presence of FTD-associated tau mutations. Lastly, we demonstrate that expression of the amino-terminal projection domain of hTau is sufficient to reduce protein synthesis and ribosomal biogenesis. Together, these data reinforce a role for tau in impairing ribosomal function.


Sujet(s)
Démence frontotemporale , Biosynthèse des protéines/physiologie , Protéines ribosomiques/métabolisme , Ribosomes/anatomopathologie , Protéines tau/métabolisme , Animaux , Femelle , Démence frontotemporale/génétique , Démence frontotemporale/métabolisme , Démence frontotemporale/anatomopathologie , Humains , Mâle , Souris , Ribosomes/métabolisme
17.
Int J Mol Sci ; 22(11)2021 May 30.
Article de Anglais | MEDLINE | ID: mdl-34070742

RÉSUMÉ

Nephrogenesis is driven by complex signaling pathways that control cell growth and differentiation. The endoplasmic reticulum chaperone calreticulin (Calr) is well known for its function in calcium storage and in the folding of glycoproteins. Its role in kidney development is still not understood. We provide evidence for a pivotal role of Calr in nephrogenesis in this investigation. We show that Calr deficiency results in the disrupted formation of an intact nephrogenic zone and in retardation of nephrogenesis, as evidenced by the disturbance in the formation of comma-shaped and s-shaped bodies. Using proteomics and transcriptomics approaches, we demonstrated that in addition to an alteration in Wnt-signaling key proteins, embryonic kidneys from Calr-/- showed an overall impairment in expression of ribosomal proteins which reveals disturbances in protein synthesis and nephrogenesis. CRISPR/cas9 mediated knockout confirmed that Calr deficiency is associated with a deficiency of several ribosomal proteins and key proteins in ribosome biogenesis. Our data highlights a direct link between Calr expression and the ribosome biogenesis.


Sujet(s)
Calcium/métabolisme , Calréticuline/génétique , Rein/métabolisme , Biogenèse des organelles , Protéines ribosomiques/génétique , Ribosomes/génétique , Animaux , Signalisation calcique , Calréticuline/déficit , Embryon de mammifère , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/anatomopathologie , Femelle , Régulation de l'expression des gènes au cours du développement , Glycoprotéines/classification , Glycoprotéines/génétique , Glycoprotéines/métabolisme , Rein/croissance et développement , Rein/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Organogenèse/génétique , Pliage des protéines , Protéomique/méthodes , Protéines ribosomiques/déficit , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Voie de signalisation Wnt
18.
Genome Biol ; 22(1): 191, 2021 06 28.
Article de Anglais | MEDLINE | ID: mdl-34183069

RÉSUMÉ

BACKGROUND: Little is known about the impact of trans-acting genetic variation on the rates with which proteins are synthesized by ribosomes. Here, we investigate the influence of such distant genetic loci on the efficiency of mRNA translation and define their contribution to the development of complex disease phenotypes within a panel of rat recombinant inbred lines. RESULTS: We identify several tissue-specific master regulatory hotspots that each control the translation rates of multiple proteins. One of these loci is restricted to hypertrophic hearts, where it drives a translatome-wide and protein length-dependent change in translational efficiency, altering the stoichiometric translation rates of sarcomere proteins. Mechanistic dissection of this locus across multiple congenic lines points to a translation machinery defect, characterized by marked differences in polysome profiles and misregulation of the small nucleolar RNA SNORA48. Strikingly, from yeast to humans, we observe reproducible protein length-dependent shifts in translational efficiency as a conserved hallmark of translation machinery mutants, including those that cause ribosomopathies. Depending on the factor mutated, a pre-existing negative correlation between protein length and translation rates could either be enhanced or reduced, which we propose to result from mRNA-specific imbalances in canonical translation initiation and reinitiation rates. CONCLUSIONS: We show that distant genetic control of mRNA translation is abundant in mammalian tissues, exemplified by a single genomic locus that triggers a translation-driven molecular mechanism. Our work illustrates the complexity through which genetic variation can drive phenotypic variability between individuals and thereby contribute to complex disease.


Sujet(s)
Cardiomégalie/génétique , Initiation de la traduction , Locus de caractère quantitatif , ARN messager/génétique , Petit ARN nucléolaire/génétique , Protéines ribosomiques/génétique , Ribosomes/génétique , Animaux , Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Variation génétique , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Myocarde/métabolisme , Myocarde/anatomopathologie , Biogenèse des organelles , ARN messager/métabolisme , Petit ARN nucléolaire/métabolisme , Rats , Rats de lignée SHR , Rats transgéniques , Protéines ribosomiques/métabolisme , Ribosomes/métabolisme , Ribosomes/anatomopathologie , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Sarcomères/métabolisme , Sarcomères/anatomopathologie
19.
Commun Biol ; 4(1): 543, 2021 05 10.
Article de Anglais | MEDLINE | ID: mdl-33972671

RÉSUMÉ

The majorities of colorectal cancer (CRC) cases are sporadic in origin and a large proportion of etiologies are associated with environmental stress responses. In response to external and internal stress, the ribosome stands sentinel and stress-driven ribosomal dysfunction triggers the cellular decision pathways via transcriptional reprogramming. In the present study, PR domain zinc finger protein (PRDM) 1, a master transcriptional regulator, was found to be closely associated with ribosomal actions in patients with CRC and the murine models. Stress-driven ribosomal dysfunction enhanced PRDM1 levels in intestinal cancer cells, which contributed to their survival and enhanced cancer cell stemness against cancer treatment. Mechanistically, PRDM1 facilitated clustering modulation of insulin-like growth factor (IGF) receptor-associated genes, which supported cancer cell growth and stemness-linked features. Ribosomal dysfunction-responsive PRDM1 facilitated signaling remodeling for the survival of tumor progenitors, providing compelling evidence for the progression of sporadic CRC.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Tumeurs colorectales/traitement médicamenteux , Résistance aux médicaments antinéoplasiques , Cellules souches tumorales/anatomopathologie , Facteur-1 liant le domaine de régulation positive I/métabolisme , Ribosomes/anatomopathologie , Stress physiologique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Muqueuse intestinale/anatomopathologie , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris nude , Adulte d'âge moyen , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Facteur-1 liant le domaine de régulation positive I/génétique , Pronostic , Transduction du signal , Taux de survie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Nat Cell Biol ; 23(2): 127-135, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33495632

RÉSUMÉ

Ribosomes are multicomponent molecular machines that synthesize all of the proteins of living cells. Most of the genes that encode the protein components of ribosomes are therefore essential. A reduction in gene dosage is often viable albeit deleterious and is associated with human syndromes, which are collectively known as ribosomopathies1-3. The cell biological basis of these pathologies has remained unclear. Here, we model human ribosomopathies in Drosophila and find widespread apoptosis and cellular stress in the resulting animals. This is not caused by insufficient protein synthesis, as reasonably expected. Instead, ribosomal protein deficiency elicits proteotoxic stress, which we suggest is caused by the accumulation of misfolded proteins that overwhelm the protein degradation machinery. We find that dampening the integrated stress response4 or autophagy increases the harm inflicted by ribosomal protein deficiency, suggesting that these activities could be cytoprotective. Inhibition of TOR activity-which decreases ribosomal protein production, slows down protein synthesis and stimulates autophagy5-reduces proteotoxic stress in our ribosomopathy model. Interventions that stimulate autophagy, combined with means of boosting protein quality control, could form the basis of a therapeutic strategy for this class of diseases.


Sujet(s)
Mutation/génétique , Protéines/toxicité , Ribosomes/génétique , Ribosomes/anatomopathologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Allèles , Animaux , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Drosophila melanogaster/effets des médicaments et des substances chimiques , Drosophila melanogaster/métabolisme , Cellules HEK293 , Hétérozygote , Humains , Disques imaginaux/effets des médicaments et des substances chimiques , Disques imaginaux/métabolisme , Agrégats de protéines/effets des médicaments et des substances chimiques , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Protéomique , Protéines ribosomiques/biosynthèse , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Ailes d'animaux/effets des médicaments et des substances chimiques , Ailes d'animaux/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE