52 research outputs found

    The NKG2D Ligands RAE-1δ and RAE-1ε Differ with Respect to Their Receptor Affinity, Expression Profiles and Transcriptional Regulation

    Get PDF
    BACKGROUND: RAE-1 is a ligand of the activating receptor NKG2D expressed by NK cells, NKT, γδT and some CD8(+)T lymphocytes. RAE-1 is overexpressed in tumor cell lines and its expression is induced after viral infection and genotoxic stress. We have recently demonstrated that RAE-1 is expressed in the adult subventricular zone (SVZ) from C57BL/6 mice. RAE-1 is also expressed in vitro by neural stem/progenitor cells (NSPCs) and plays a non-immune role in cell proliferation. The C57BL/6 mouse genome contains two rae-1 genes, rae-1δ and rae-1ε encoding two different proteins. The goals of this study are first to characterize the in vivo and in vitro expression of each gene and secondly to elucidate the mechanisms underlying their respective expression, which are far from known. PRINCIPAL FINDINGS: We observed that Rae-1δ and Rae-1ε transcripts are differentially expressed according to tissues, pathological conditions and cell lines. Embryonic tissue and the adult SVZ mainly expressed Rae-1δ transcripts. The NSPCs derived from the SVZ also mainly expressed RAE-1δ. The interest of this result is especially related to the observation that RAE-1δ is a weak NKG2D ligand compared to RAE-1ε. On the contrary, cell lines expressed either similar levels of RAE-1δ and RAE-1ε proteins or only RAE-1ε. Since the protein expression correlated with the level of transcripts for each rae-1 gene, we postulated that transcriptional regulation is one of the main processes explaining the difference between RAE-1δ and RAE-1ε expression. We indeed identified two different promoter regions for each gene: one mainly involved in the control of rae-1δ gene expression and the other in the control of rae-1ε expression. CONCLUSIONS/SIGNIFICANCE: RAE-1δ and RAE-1ε differ with respect to their function and the control of their expression. Immune function would be mainly exerted by RAE-1ε and non-immune function by RAE-1δ

    Olfactory Stem Cells, a New Cellular Model for Studying Molecular Mechanisms Underlying Familial Dysautonomia

    Get PDF
    International audienceBackground: Familial dysautonomia (FD) is a hereditary neuropathy caused by mutations in the IKBKAP gene, the most common of which results in variable tissue-specific mRNA splicing with skipping of exon 20. Defective splicing is especially severe in nervous tissue, leading to incomplete development and progressive degeneration of sensory and autonomic neurons. The specificity of neuron loss in FD is poorly understood due to the lack of an appropriate model system. To better understand and modelize the molecular mechanisms of IKBKAP mRNA splicing, we collected human olfactory ecto-mesenchymal stem cells (hOE-MSC) from FD patients. hOE-MSCs have a pluripotent ability to differentiate into various cell lineages, including neurons and glial cells.Methodology/Principal Findings: We confirmed IKBKAP mRNA alternative splicing in FD hOE-MSCs and identified 2 novel spliced isoforms also present in control cells. We observed a significant lower expression of both IKBKAP transcript and IKAP/hELP1 protein in FD cells resulting from the degradation of the transcript isoform skipping exon 20. We localized IKAP/hELP1 in different cell compartments, including the nucleus, which supports multiple roles for that protein. We also investigated cellular pathways altered in FD, at the genome-wide level, and confirmed that cell migration and cytoskeleton reorganization were among the processes altered in FD. Indeed, FD hOE-MSCs exhibit impaired migration compared to control cells. Moreover, we showed that kinetin improved exon 20 inclusion and restores a normal level of IKAP/hELP1 in FD hOE-MSCs. Furthermore, we were able to modify the IKBKAP splicing ratio in FD hOE-MSCs, increasing or reducing the WT (exon 20 inclusion):MU (exon 20 skipping) ratio respectively, either by producing free-floating spheres, or by inducing cells into neural differentiation.Conclusions/Significance: hOE-MSCs isolated from FD patients represent a new approach for modeling FD to better understand genetic expression and possible therapeutic approaches. This model could also be applied to other neurological genetic diseases

    Mechanisms that determine the correct order of exon joining

    No full text

    Proteasome inhibitors to alleviate aberrant IKBKAP mRNA splicing and low IKAP/hELP1 synthesis in familial dysautonomia

    No full text
    International audienceFD is a rare neurodegenerative disorder caused by a mutation of the IKBKAP gene, which induces low expression levels of the Elongator subunit IKAP/hELP1 protein. A rational strategy for FD treatment could be to identify drugs increasing IKAP/hELP1 expression levels by blocking protein degradation pathways such as the 26S proteasome. Proteasome inhibitors are promising molecules emerging in cancer treatment and could thus constitute an enticing pharmaceutical strategy for FD treatment. Therefore, we tested three proteasome inhibitors on FD human olfactory ecto-mesenchymal stem cells (hOE-MSCs): two approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA), bortezomib and carfilzomib, as well as epoxomicin. Although all 3 inhibitors demonstrated activity in correcting IKBKAP mRNA aberrant splicing, carfilzomib was superior in enhancing IKAP/hELP1 quantity. Moreover, we observed a synergistic effect of suboptimal doses of carfilzomib on kinetin in improving IKBKAP isoforms ratio and IKAP/hELP1 expression levels allowing to counterbalance carfilzomib toxicity. Finally, we identified several dysregulated miRNAs after carfilzomib treatment that target proteasome-associated mRNAs and determined that IKAP/hELP1 deficiency in FD pathology is correlated to an overactivity of the 26S proteasome. Altogether, these results reinforce the rationale for using chemical compounds inhibiting the 26S proteasome as an innovative option for FD and a promising therapeutic pathway for many other neurodegenerative diseases

    MicroRNA screening identifies a link between NOVA1 expression and a low level of IKAP in familial dysautonomia

    Get PDF
    International audienceFamilial dysautonomia (FD) is a rare neurodegenerative disease caused by a mutation in intron 20 of the IKBKAP gene (c.2204+6T>C), leading to tissue-specific skipping of exon 20 and a decrease in the synthesis of the encoded protein IKAP (also known as ELP1). Small non-coding RNAs known as microRNAs (miRNAs) are important post-transcriptional regulators of gene expression and play an essential role in the nervous system development and function. To better understand the neuronal specificity of IKAP loss, we examined expression of miRNAs in human olfactory ecto-mesenchymal stem cells (hOE-MSCs) from five control individuals and five FD patients. We profiled the expression of 373 miRNAs using microfluidics and reverse transcription coupled to quantitative PCR (RT-qPCR) on two biological replicate series of hOE-MSC cultures from healthy controls and FD patients. This led to the total identification of 26 dysregulated miRNAs in FD, validating the existence of a miRNA signature in FD. We then selected the nine most discriminant miRNAs for further analysis. The signaling pathways affected by these dysregulated miRNAs were largely within the nervous system. In addition, many targets of these dysregulated miRNAs had been previously demonstrated to be affected in FD models. Moreover, we found that four of our nine candidate miRNAs target the neuron-specific splicing factor NOVA1. We demonstrated that overexpression of miR-203a-3p leads to a decrease of NOVA1, counter-balanced by an increase of IKAP, supporting a potential interaction between NOVA1 and IKAP. Taken together, these results reinforce the choice of miRNAs as potential therapeutic targets and suggest that NOVA1 could be a regulator of FD pathophysiology

    Les cellules souches olfactives humaines (un nouveau modèle d'étude des mécanismes à l'origine d'une maladie neurodégénérative, la dysautonomie familiale)

    No full text
    La dysautonomie familiale (FD) est une neuropathie héréditaire provoquée par des mutations au sein du gène IKBKAP, la plus commune d'entre elles induisant un épissage alternatif de l'exon 20 au sein de du pré-ARNm de façon tissu-spécifique. L'épissage aberrant est particulièrement prononcé dans les tissus nerveux, conduisant à la dégénerescence progressive des neurones sensoriels et autonomes. La spécificité de la perte des cellules nerveuses dans la FD est mal comprise, par manque d'un modèle approprié. Afin de mieux comprendre les mécanismes moléculaires de l'épissage des ARNm d'IKBKAP, nous avons utilisé un modèle original : les cellules souches olfactives ecto-mesenchymateuses (hOE-MSC) de patients FD. Les hOE-MSC sont pluripotentes et ont la capacité de se différencier en diverses lignées cellulaires, y compris les neurones et les cellules gliales.Nous avons confirmé la présence du transcrit exempt de l'exon 20 d'IKBKAP dans les hOE-MSC de FD et nous avons observé une expression significativement inférieure de la somme des transcrits IKBKAP chez ces patients, du fait de la dégradation d'une partie des isoforme aberrants. Cette réduction est correlée avec une réduction d'expression de la protéine traduite à partir du transcrit d IKBKAP possèdant l exon 20, IKAP/hELP1. Nous avons localisé IKAP/hELP1 dans différents compartiments cellulaires, y compris le noyau, ce qui soutient des rôles multiples de cette protéine. Nous avons confirmé que la kinétine, une cytokinine, améliorait le taux de transcrit incluant l'exon 20 et rétablissait des niveaux normaux d'IKAP/hELP1 dans les hOE-MSC de FD. Par ailleurs, nous avons pu modifier le rapport d'épissage d'IKBKAP en augmentant ou en réduisant le ratio WT (inclusion de l'exon 20) : MU (saut de l'exon 20) respectivement, en produisant des sphères flottantes, ou en engageant les cellules vers une différentiation neurale. Les sphères et les cellules différenciées ont été étudiées au niveau pan-génomique, ce qui a permis d'identifier le développement du système nerveux comme étant le processus le plus affecté chez les FD. De plus, nous soulignons le rôle de la kinétine comme un probable régulateur de facteurs d'épissage contribuant à la restauration d'un épissage correct d'IKBKAP.Les hOE-MSC isolées de patients FD représentent une nouvelle approche pour modéliser la pathologie et mieux comprendre l'expression génétique et les approches thérapeutiques possibles de la FD. En outre, elles offrent une application originale à la compréhension d'autres maladies génétiques neurologiques.Familial dysautonomia (FD) is a hereditary neuropathy caused by mutations in the IKBKAP gene, the most common of which results in variable tissue-specific mRNA splicing with skipping of exon 20. Defective splicing is especially severe in nervous tissue, leading to incomplete development and progressive degeneration of sensory and autonomic neurons. The specificity of neuron loss in FD is poorly understood due to the lack of an appropriate model system. To better understand and modelize the molecular mechanisms of IKBKAP mRNA splicing, we collected human olfactory ecto-mesenchymal stem cells (hOE-MSCs) from FD patients. hOE-MSCs have a pluripotent ability to differentiate into various cell lineages, including neurons and glial cells.We confirmed IKBKAP mRNA alternative splicing in FD hOE-MSCs and observed a significant lower expression of both IKBKAP transcripts and IKAP/hELP1 protein in FD cells resulting from the degradation of the transcript isoform skipping exon 20. We localized IKAP/hELP1 in different cell compartments, including the nucleus, which supports multiple roles for that protein. Moreover, we showed that kinetin improved exon 20 inclusion and restores a normal level of IKAP/hELP1 in FD hOE-MSCs. Furthermore, we were able to modify the IKBKAP splicing ratio in FD hOE-MSCs, increasing or reducing the WT (exon 20 inclusion):MU (exon 20 skipping) ratio respectively, either by producing free-floating spheres, or by inducing cells into neural differentiation. Spheres forming cells and lineage neuroglial progenitors were investigated at the genome-wide level, and we confirmed that nervous system development was the most altered process in FD. More, we highlight kinetin role as a putative regulator of splicing factors which contribute to restore a correct splicing of IKBKAP.hOE-MSCs isolated from FD patients represent a new approach for modeling FD to better understand genetic expression and possible therapeutic approaches. This model could also be applied to other neurological genetic diseases.AIX-MARSEILLE2-Bib.electronique (130559901) / SudocSudocFranceF

    REGULATION DE L'EXPRESSION DE GENE HLA-G LORS DE LA REPONSE AU STRESS ET ANALYSE DE L'ACTIVATION SELECTIVE DE CE GENE DANS LES MELANOMES ET LES CARCINOMES RENAUX

    No full text
    PARIS5-BU-Necker : Fermée (751152101) / SudocPARIS-BIUM (751062103) / SudocSudocFranceF

    Biomarker development: current issues and perspectives

    No full text
    International audienceWe have read the letter of Ho et al. in response to our work with great interest. We agree with their general comment regarding the interest of evaluating agreement between two multiplex immunoassays. Ho et al. reminds us that correlation analysis suffers from limitations in assessing agreement between methods. Such agreement should be assessed using Bland-Altman plots. This method is largely known as the original 1986 paper of Bland and Altman has been the most frequently cited article ever to appear in the Lancet and is one of the ten most frequently cited statistical articles ever (Bland and Altman, 2012). Nevertheless, we decided to use Spearman’s rank correlation, since our original intent was to compare methods and biological fluids, which is not exactly the same as assessing the degree of agreement. Ultimately, several conditions are required to evaluate agreement with Bland-Altman graphics
    • …
    corecore