13 research outputs found

    Role of aberrant glycosylation in ovarian cancer dissemination

    Get PDF
    Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, and understanding the molecular changes associated with EOC etiology could lead to the identification of novel targets for more effective therapeutic interventions. Glycosylation represents a post-translational modification (PTM) of proteins playing a major role in various cellular functions. Moreover, glycosylation participates in major pathobiological events during tumor progression, as aberrant expression of glycan structures has been shown to contribute in alterations of specific cellular onco-phenotypes, including tumor cell proliferation, migration and invasion. This review aims to describe what is currently known about aberrant glycosylation in EOC, and more specifically, the contribution of aberrant O-linked glycosylation in EOC progression. We also discuss our findings about the altered GALNT3 overexpression in EOC and its involvement in disease dissemination through aberrant mucin O-glycosylation, as well as the potential to exploit the role of GALNT3 in understanding the general mechanisms of abnormal glycosylation implicated in EOC spreading. Further analyses in cancer glycobiology could significantly enhance our understanding of the molecular mechanisms of cancer progression, including EOC dissemination, and could lead to the identification of novel biomarkers/therapeutic targets for better management of this deadly disease.Biomedical Reviews 2014; 25: 83-92

    Elucidating the role of the family of GalNAc-Transferases in aberrant protein O-glycosylation in the progression of epithelial ovarian cancer

    Get PDF
    Le cancer épithélial de l’ovaire (CEO) est la forme de cancer gynécologique la plus létale. Ainsi, la compréhension des changements moléculaires associés à ce cancer métastatique ovarien peut mener à l’identification de nouvelles cibles thérapeutiques essentielles. La glycosylation, une modification post-traductionnelle, joue un rôle important dans de nombreuses fonctions cellulaires. Cette glycosylation participe à des événements physiopathologiques majeurs durant la progression tumorale. De plus, il a été prouvé que l’expression aberrante des structures glycanes interfère avec des mécanismes cellulaires comme l’adhésion, la migration et la prolifération des cellules. Dans ce contexte, notre laboratoire a récemment montré que le gène codant pour la protéine N-acétylgalactosaminyltransférase 3 (GALNT3), membre de la famille des GalNAcTransférases (GalNAc-Ts), est hypométhylé et que la protéine GALNT3 est plus fortement exprimée dans les tumeurs CEO dont la sévérité est de grade élevé (“high-grade (HG) serous”), en comparaison avec des tumeurs à potentiel malin faible (“low malignant potential (LMP) ”) et des tissus ovariens normaux. Ces observations indiquent un fort potentiel oncogénique pour le gène GALNT3 dans les stades avancés du CEO. Ces premières constatations suggèrent également que la surexpression de GALNT3 peut jouer un rôle important dans la tumorigenèse du CEO en augmentant sa dissémination via une O-glycosylation de type mucine aberrante. Ces glycosylations anormales peuvent donc être impliquées dans la carcinogenèse ovarienne et nécessitent une étude approfondie. Dans ce projet de recherche, nous proposons d’approfondir les observations déjà obtenues in vitro en utilisant un modèle in vivo chez la souris, afin d’élucider le rôle fonctionnel de la GALNT3 et d’autres membres de cette famille dans la progression du CEO. A partir d’une étude de glycoprotéomique indépendante de la masse, qui a permis d’identifier des glycopeptides intacts ou métaboliquement marqués, ce projet de recherche a rendu possible la définition précise du rôle de GALNT3 dans la O-glycosylation des cibles de type mucine au sein des cellules CEO. Ainsi, via une recherche ciblée dans la base de données « SwissProt » du protéome humain, nous avons trouvé plusieurs centaines de glycoprotéines et glycopeptides uniques, différemment exprimés dans les clones cellulaires dépourvus en iv GALNT3 KD. Par la suite, nous avons identifié les gènes codant pour ces glycoprotéines et glycopeptides. Nous avons notamment trouvé, parmi la liste, un groupe de gènes impliqués dans le métabolisme cellulaire dont les modifications post-traductionnelles sont, de manière intéressante, principalement supprimées dans les clones GALNT3 KD. De plus, nous nous sommes intéressés aux autres membres de la famille des GalNAc-Ts dans le CEO et nous avons montré que de multiples membres et pas uniquement GALNT3 peuvent jouer un rôle important dans la dissémination et la progression du CEO. De plus, une découverte très intéressante fut la redondance possible des rôles joués par certains membres de la famille des GalNAc-Ts dans le CEO. Ainsi, nous avons identifié GALNT6 qui serait, à l’image de GALNT3, impliquée dans la dissémination et la progression du CEO. Cette implication du GALNT6 est supportée par le fait que cette protéine a les mêmes fonctions que GALNT3, suggérant un effet compensatoire de GALNT6 en absence de GALNT3. Pour tester cette hypothèse, nous avons abolie l’expression des deux protéines GALNT3 et GALNT6, in vivo, et nous avons observé une effet significatif sur la formation des tumeurs et la survie des animaux. Pour la suite de ce projet, nous proposons d’analyser la structure glycane des différentes glycoprotéines identifiées dans les cellules cancéreuses, afin de déterminer les altérations des modifications O-glycanes suite à la perte d’expression de GALNT3 et d’autres membres de la famille des GalNAc-Ts. En conclusion, notre étude contribue à comprendre la participation du glycoprotéome dans la tumorigenèse du CEO et à identifier d’autres cibles de type mucine ou des O-glycoprotéines dont l’expression aberrante serait modulée dans le CEO. Ainsi, pris dans son ensemble, ce projet de recherche montre la possibilité de discriminer entre des cellules cancéreuses et des cellules contrôles via les glycosylations de leurs protéines et permet d’entrevoir la glycobiologie comme une voie prometteuse pour l’identifier de nouveaux biomarqueurs pour le diagnostic du CEO.Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, thus understanding the molecular changes associated with ovarian cancer metastasis could lead to the identification of essential therapeutic targets. Glycosylation is a post-translational modification (PTM) of proteins playing a major role in various cell properties. Glycosylation participates in major pathophysiology events during tumor progressions, and the aberrant expression of glycan structures was shown to interfere with cell properties such as cell adhesion, migration, and proliferation. The lab has previously identified the polypeptide N-acetylgalactosaminyltransferase 3 (GALNT3) gene, a member of the GalNAc-Transferases (GalNAc-Ts) gene family, as hypomethylated and overexpressed in high-grade (HG) serous EOC tumors, compared to low malignant potential (LMP) EOC tumors and normal ovarian tissues. Taken together, the data obtained were indicative of a strong oncogenic potential of the GALNT3 gene in advanced EOC and suggest that GALNT3 overexpression might contribute to EOC dissemination through aberrant mucin O-glycosylation, thus specifying some of the putative mechanisms of abnormal glycosylation implicated in ovarian carcinogenesis, which warrant further investigation. The current research project focused on expanding the in vitro observations obtained by using animal models to investigate in vivo the functional significance of GALNT3 and other close members of the GalNAc-Ts gene family in serous EOC progression. Moreover, by applying a mass-independent chemical glycoproteomics platform to characterize intact, metabolically labeled glycopeptides, this project more profoundly characterized the role of GALNT3 in aberrant O-glycosylation of mucin-like targets in EOC cells. Isotopically recorded ions were searched against the Swiss-Prot human proteome; and data obtained were indicative of hundreds of unique glycoproteins and glycopeptides that were differentially expressed upon GALNT3 KD. Related gene groups were identified, and interestingly, genes implicated in mechanisms of cellular metabolic functions, and PTMs were found to be predominantly suppressed in GALNT3 KD clones. In accordance, we also investigated the role of other members of the GalNAc-T family in EOC and we showed that multiple members and not only GALNT3 can play an important role in EOC cancer dissemination and progression. One very interesting finding was the redundant role some members of the GalNAc-T family members play in EOC. We investigated the compensatory functions of GALNT3 and GALNT6, and we were able to demonstrate these two genes can impose that synthetic backup. Furthermore, we found that and their ablation can affect animal survival and tumor formation as observed both in vivo and in vitro. In continuation of this work, this project will focus on analyzing the glycan structures of those differentially expressed glycoproteins, to further examine the specific O-glycans alterations associated with the GALNT3 and other members of the GalNAc-Ts upon gene knockout (KO). Fully elaborated glycopeptides can reveal structural details of the glycoproteome, thus our results could give important information on the glycome in EOC cells, and the identification of other O-glycoproteins/mucin-like targets whose aberrant expression may be modulated by these in EOC. Taken together, the ability to mark differences in the glycosylation of proteins between cancer cells and control cells can emphasize glycobiology as a promising field for potential biomarker identification

    Optimized protocol for the generation of functional human induced-pluripotent-stem-cell-derived dopaminergic neurons

    No full text
    Summary: Generation of functional human dopaminergic (DA) neurons from human induced pluripotent stem cells (hiPSCs) is a crucial tool for modeling dopamine-related human diseases and cell replacement therapies. Here, we present a protocol to combine neuralizing transcription factor (NGN2) programming and DA patterning to differentiate hiPSCs into mature and functional induced DA (iDA) neurons. We describe steps from transduction of hiPSCs and neural induction through to differentiation and maturation of near-pure, fully functional iDA neurons within 3 weeks.For complete details on the use and execution of this protocol, please refer to Sheta et al. (2022).1 : Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics

    Alpha-Synuclein and the Endolysosomal System in Parkinson’s Disease: Guilty by Association

    No full text
    Abnormal accumulation of the protein α- synuclein (α-syn) into proteinaceous inclusions called Lewy bodies (LB) is the neuropathological hallmark of Parkinson’s disease (PD) and related disorders. Interestingly, a growing body of evidence suggests that LB are also composed of other cellular components such as cellular membrane fragments and vesicular structures, suggesting that dysfunction of the endolysosomal system might also play a role in LB formation and neuronal degeneration. Yet the link between α-syn aggregation and the endolysosomal system disruption is not fully elucidated. In this review, we discuss the potential interaction between α-syn and the endolysosomal system and its impact on PD pathogenesis. We propose that the accumulation of monomeric and aggregated α-syn disrupt vesicles trafficking, docking, and recycling, leading to the impairment of the endolysosomal system, notably the autophagy-lysosomal degradation pathway. Reciprocally, PD-linked mutations in key endosomal/lysosomal machinery genes (LRRK2, GBA, ATP13A2) also contribute to increasing α-syn aggregation and LB formation. Altogether, these observations suggest a potential synergistic role of α-syn and the endolysosomal system in PD pathogenesis and represent a viable target for the development of disease-modifying treatment for PD and related disorders

    Optogenetic-mediated induction and monitoring of α-synuclein aggregation in cellular models of Parkinson’s disease

    No full text
    Summary: Studying Parkinson’s disease (PD) is complex due to a lack of cellular models mimicking key aspects of protein pathology. Here, we present a protocol for inducing and monitoring α-synuclein aggregation in living cells using optogenetics. We describe steps for plasmid transduction, biochemical validation, immunocytochemistry, and live-cell confocal imaging. These induced aggregates fulfill the cardinal features of authentic protein inclusions observed in PD-diseased brains and offer a tool to study the role of protein aggregation in neurodegeneration.For complete details on the use and execution of this protocol, please refer to Bérard et al.1 : Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics

    The polypeptide GALNT6 Displays Redundant Functions upon Suppression of its Closest Homolog GALNT3 in Mediating Aberrant O-Glycosylation, Associated with Ovarian Cancer Progression

    No full text
    Epithelial ovarian cancer (EOC) represents the most lethal gynecologic malignancy; a better understanding of the molecular mechanisms associated with EOC etiology could substantially improve EOC management. Aberrant O-glycosylation in cancer is attributed to alteration of N-acetylgalactosaminyltransferases (GalNAc-Ts). Reports suggest a genetic and functional redundancy between GalNAc-Ts, and our previous data are indicative of an induction of GALNT6 expression upon GALNT3 suppression in EOC cells. We performed single GALNT3 and double GALNT3/T6 suppression in EOC cells, using a combination of the CRISPR-Cas9 system and shRNA-mediated gene silencing. The effect of single GALNT3 and double GALNT3/T6 inhibition was monitored both in vitro (on EOC cells roliferation, migration, and invasion) and in vivo (on tumor formation and survival of experimental animals). We confirmed that GALNT3 gene ablation leads to strong and rather compensatory GALNT6 upregulation in EOC cells. Moreover, double GALNT3/T6 suppression was significantly associated with stronger inhibitory effects on EOC cell proliferation, migration, and invasion, and accordingly displayed a significant increase in animal survival rates compared with GALNT3-ablated and control (Ctrl) EOC cells. Our data suggest a possible functional redundancy of GalNAc-Ts (GALNT3 and T6) in EOC, with the perspective of using both these enzymes as novel EOC biomarkers and/or therapeutic targets

    Non-invasive systemic viral delivery of human alpha-synuclein mimics selective and progressive neuropathology of Parkinson’s disease in rodent brains

    No full text
    Abstract Background Alpha-synuclein (α-syn) aggregation into proteinaceous intraneuronal inclusions, called Lewy bodies (LBs), is the neuropathological hallmark of Parkinson’s disease (PD) and related synucleinopathies. However, the exact role of α-syn inclusions in PD pathogenesis remains elusive. This lack of knowledge is mainly due to the absence of optimal α-syn-based animal models that recapitulate the different stages of neurodegeneration. Methods Here we describe a novel approach for a systemic delivery of viral particles carrying human α-syn allowing for a large-scale overexpression of this protein in the mouse brain. This approach is based on the use of a new generation of adeno-associated virus (AAV), AAV-PHP.eB, with an increased capacity to cross the blood-brain barrier, thus offering a viable tool for a non-invasive and large-scale gene delivery in the central nervous system. Results Using this model, we report that widespread overexpression of human α-syn induced selective degeneration of dopaminergic (DA) neurons, an exacerbated neuroinflammatory response in the substantia nigra and a progressive manifestation of PD-like motor impairments. Interestingly, biochemical analysis revealed the presence of insoluble α-syn oligomers in the midbrain. Together, our data demonstrate that a single non-invasive systemic delivery of viral particles overexpressing α-syn prompted selective and progressive neuropathology resembling the early stages of PD. Conclusions Our new in vivo model represents a valuable tool to study the role of α-syn in PD pathogenesis and in the selective vulnerability of nigral DA neurons; and offers the opportunity to test new strategies targeting α-syn toxicity for the development of disease-modifying therapies for PD and related disorders

    Proteomic dataset for altered glycoprotein expression upon GALNT3 knockdown in ovarian cancer cells

    Get PDF
    This article contains raw and processed data related to research published in “Role of the polypeptide N-acetylgalactosaminyltransferase 3 in ovarian cancer progression: possible implications in abnormal mucin O-glycosylation” [1]. The data presented here was obtained with the application of a bioorthogonal chemical reporter strategy analyzing differential glycoprotein expression following the knock-down (KD) of the GALNT3 gene in the epithelial ovarian cancer (EOC) cell line A2780s. LC-MS/MS mass spectrometry analysis was then performed and the processed data related to the identified glycoproteins show that several hundred proteins are differentially expressed between control and GALNT3 KD A2780s cells. The obtained data also uncover numerous novel glycoproteins; some of which could represent new potential EOC biomarkers and/or therapeutic targets. Keywords: GALNT3, Glycosylation, Ac4GalNAz labeling, Label free quantification, NetOGlyc and NetNGlyc prediction analysis, Glycoproteomic
    corecore