15 research outputs found

    Recommendations, guidelines, and best practice for the use of human induced pluripotent stem cells for neuropharmacological studies of neuropsychiatric disorders

    Get PDF
    The number of individuals suffering from neuropsychiatric disorders (NPDs) has increased worldwide, with 3 million disability-adjusted life-years calculated in 2019. Though research using various approaches including genetics, imaging, clinical and animal models has advanced our knowledge regarding NPDs, we still lack basic knowledge regarding the underlying pathophysiological mechanisms. Moreover, there is an urgent need for highly effective therapeutics for NPDs. Human induced pluripotent stem cells (hiPSCs) generated from somatic cells enabled scientists to create brain cells in a patient-specific manner. However, there are challenges to the use of hiPSCs that need to be addressed. In the current paper, consideration of best practices for neuropharmacological and neuropsychiatric research using hiPSCs will be discussed. Specifically, we provide recommendations for best practice in patient recruitment, including collecting demographic, clinical, medical (before and after treatment and response), diagnostic (including scales) and genetic data from the donors. We highlight considerations regarding donor genetics and sex, in addition to discussing biological and technical replicates. Furthermore, we present our views on selecting control groups/lines, experimental designs, and considerations for conducting neuropharmacological studies using hiPSC-based models in the context of NPDs. In doing so, we explore key issues in the field concerning reproducibility, statistical analysis, and how to translate in vitro studies into clinically relevant observations. The aim of this article is to provide a key resource for hiPSC researchers to perform robust and reproducible neuropharmacological studies, with the ultimate aim of improving identification and clinical translation of novel therapeutic drugs for NPDs

    Recommendations, guidelines, and best practice for the use of human induced pluripotent stem cells for neuropharmacological studies of neuropsychiatric disorders

    Get PDF
    The number of individuals suffering from neuropsychiatric disorders (NPDs) has increased worldwide, with 3 million disability-adjusted life-years calculated in 2019. Though research using various approaches including genetics, imaging, clinical and animal models has advanced our knowledge regarding NPDs, we still lack basic knowledge regarding the underlying pathophysiological mechanisms. Moreover, there is an urgent need for highly effective therapeutics for NPDs i. Human induced pluripotent stem cells (hiPSCs) generated from somatic cells enabled scientists to create brain cells in a patient-specific manner. However, there are challenges to the use of hiPSCs that need to be addressed. In the current paper, consideration of best practices for neuropharmacological and neuropsychiatric research using hiPSCs will be discussed. Specifically, we provide recommendations for best practice in patient recruitment, including collecting demographic, clinical, medical (before and after treatment and response), diagnostic (incl. scales) and genetic data from the donors. We highlight considerations regarding donor genetics and sex, in addition to discussing biological and technical replicates. Furthermore, we present our views on selecting control groups/lines, experimental designs, and considerations for conducting neuropharmacological studies using hiPSC-based models in the context of NPDs. In doing so, we explore key issues in the field concerning reproducibility, statistical analysis, and how to translate in vitro studies into clinically relevant observations. The aim of this article is to provide a key resource for hiPSC researchers to perform robust and reproducible neuropharmacological studies, with the ultimate aim of improving identification and clinical translation of novel therapeutic drugs for NPDs

    La protéine RAB6-GTPase : un régulateur général de la sécrétion post-Golgienne

    No full text
    Le trafic intracellulaire est un processus fondamental qui maintient l'homéostasie cellulaire. Les RAB GTPases sont des régulateurs clés du trafic intracellulaire. RAB6 est la RAB résidente la plus abondante du Golgi. RAB6 est un régulateur clé de l'homéostasie Golgienne. Mon projet de thèse s'est intéressé à l'étude de la fonction de RAB6 dans la sécrétion post-Golgienne. Des études précédentes ont montré que la déplétion de RAB6 inhibe l'arrivée à la membrane plasmique de différents cargos : dans des cellules HeLa, NPY et VSV-G, et TNFα dans les macrophages. Nous avons donc émis l'hypothèse que RAB6 pourrait être un régulateur général de la sécrétion post-Golgienne. A l'aide de cellules MEFs RAB6 KO, nous avons d'abord montré que la sécrétion de toutes les protéines nouvellement synthétisées est inhibée. Pour comprendre les mécanismes entraînant cet effet, nous avons étudié le rôle de RAB6 dans le transport post-Golgien de trois types différents de cargos : GPI-APs (PLAP et CD59), collagen X, une protéine soluble, et une protéine transmembranaire TNFα. Afin de synchroniser le transport de cargos, nous avons utilisé le système RUSH. Ainsi, nous avons montré que RAB6 est présent sur les vésicules post-Golgiennes contenant les 3 types de cargos et que la déplétion de RAB6 affecte leur sécrétion. Les effecteurs de RAB6 sont aussi impliqués: Myosine II dans leur fission du Golgi, KIF5B dans leur transport vers la périphérie cellulaire, ELKS dans leur arrimage à la membrane plasmique. Finalement, nous avons pu montrer que les 3 cargos sont présents dans les mêmes vésicules post-Golgiennes avec RAB6. Ces résultats montrent que RAB6 régule la sécrétion de différents cargos.Intracellular trafficking is a fundamental process which ensures cell homeostasis. RAB GTPases are key regulators of intracellular trafficking. RAB6 is the most abundant Golgi resident RAB and is a key regulator of Golgi homeostasis. My Ph.D project focused on understanding the function of RAB6 in post-Golgi secretion.Previous reports have shown that RAB6 depletion impairs the arrival at the plasma membrane of different cargoes: in HeLa cells, NPY and VSV-G and TNFα in macrophages. We thus hypothesized that RAB6 could be a general regulator of post-Golgi transport steps. Using MEF cells from RAB6 KO mice, we first showed that the secretion of all newly synthesized proteins is affected. To decipher the mechanisms leading to this inhibition, we have then investigated the role of RAB6 in the post-Golgi transport of three different classes of proteins, GPI-anchored proteins (such as Placental Alkaline phosphatase or PLAP and CD59), collagen X, a soluble protein, and the transmembrane protein TNFα. In order to synchronize transport of newly-synthetized cargoes along the secretory pathway, we used the RUSH system. Here, we show that RAB6 is present on post Golgi vesicles containing the three types of cargo and that RAB6 depletion affects their secretion to the plasma membrane. RAB6 effectors are also implicated: Myosin II for their fission from the Golgi, KIF5B for their transport to the cell periphery, ELKS/RAB2IP2 for their docking with the plasma membrane. Finally, we could show that these three cargoes are present in the same post-Golgi transport carriers with RAB6. Altogether, these results show that RAB6 regulates the secretion of a wide number of cargo proteins

    Ultrastructural and dynamic studies of the endosomal compartment in Down syndrome

    No full text
    International audienceEnlarged early endosomes have been visualized in Alzheimer's disease (AD) and Down syndrome (DS) using conventional confocal microscopy at a resolution corresponding to endosomal size (hundreds of nm). In order to overtake the diffraction limit, we used super-resolution structured illumination microscopy (SR-SIM) and transmission electron microscopies (TEM) to analyze the early endosomal compartment in DS.By immunofluorescence and confocal microscopy, we confirmed that the volume of Early Endosome Antigen 1 (EEA1)-positive puncta was 13-19% larger in fibroblasts and iPSC-derived neurons from individuals with DS, and in basal forebrain cholinergic neurons (BFCN) of the Ts65Dn mice modelling DS. However, EEA1-positive structures imaged by TEM or SR-SIM after chemical fixation had a normal size but appeared clustered. In order to disentangle these discrepancies, we imaged optimally preserved High Pressure Freezing (HPF)-vitrified DS fibroblasts by TEM and found that early endosomes were 75% denser but remained normal-sized.RNA sequencing of DS and euploid fibroblasts revealed a subgroup of differentially-expressed genes related to cargo sorting at multivesicular bodies (MVBs). We thus studied the dynamics of endocytosis, recycling and MVB-dependent degradation in DS fibroblasts. We found no change in endocytosis, increased recycling and delayed degradation, suggesting a "traffic jam" in the endosomal compartment.Finally, we show that the phosphoinositide PI (3) P, involved in early endosome fusion, is decreased in DS fibroblasts, unveiling a new mechanism for endosomal dysfunctions in DS and a target for pharmacotherapy

    Specific Mutations in the Cholesterol-Binding Site of APP Alter Its Processing and Favor the Production of Shorter, Less Toxic Aβ Peptides

    Get PDF
    International audienceAbstract Excess brain cholesterol is strongly implicated in the pathogenesis of Alzheimer’s disease (AD). Here we evaluated how the presence of a cholesterol-binding site (CBS) in the transmembrane and juxtamembrane regions of the amyloid precursor protein (APP) regulates its processing. We generated nine point mutations in the APP gene, changing the charge and/or hydrophobicity of the amino-acids which were previously shown as part of the CBS. Most mutations triggered a reduction of amyloid-β peptides Aβ40 and Aβ42 secretion from transiently transfected HEK293T cells. Only the mutations at position 28 of Aβ in the APP sequence resulted in a concomitant significant increase in the production of shorter Aβ peptides. Mass spectrometry (MS) confirmed the predominance of Aβx-33 and Aβx-34 with the APP K28A mutant. The enzymatic activity of α-, β-, and γ-secretases remained unchanged in cells expressing all mutants. Similarly, subcellular localization of the mutants in early endosomes did not differ from the APP WT protein. A transient increase of plasma membrane cholesterol enhanced the production of Aβ40 and Aβ42 by APP WT , an effect absent in APP K28A mutant. Finally, WT but not CBS mutant Aβ derived peptides bound to cholesterol-rich exosomes. Collectively, the present data revealed a major role of juxtamembrane amino acids of the APP CBS in modulating the production of toxic Aβ species. More generally, they underpin the role of cholesterol in the pathophysiology of AD

    C11ORF24 Is a Novel Type I Membrane Protein That Cycles between the Golgi Apparatus and the Plasma Membrane in Rab6-Positive Vesicles

    Get PDF
    <div><p>The Golgi apparatus is an intracellular compartment necessary for post-translational modification, sorting and transport of proteins. It plays a key role in mitotic entry through the Golgi mitotic checkpoint. In order to identify new proteins involved in the Golgi mitotic checkpoint, we combine the results of a knockdown screen for mitotic phenotypes and a localization screen. Using this approach, we identify a new Golgi protein C11ORF24 (NP_071733.1). We show that C11ORF24 has a signal peptide at the N-terminus and a transmembrane domain in the C-terminal region. C11ORF24 is localized on the Golgi apparatus and on the <i>trans-</i>Golgi network. A large part of the protein is present in the lumen of the Golgi apparatus whereas only a short tail extends into the cytosol. This cytosolic tail is well conserved in evolution. By FRAP experiments we show that the dynamics of C11ORF24 in the Golgi membrane are coherent with the presence of a transmembrane domain in the protein. C11ORF24 is not only present on the Golgi apparatus but also cycles to the plasma membrane <i>via</i> endosomes in a pH sensitive manner. Moreover, via video-microscopy studies we show that C11ORF24 is found on transport intermediates and is colocalized with the small GTPase RAB6, a GTPase involved in anterograde transport from the Golgi to the plasma membrane. Knocking down C11ORF24 does not lead to a mitotic phenotype or an intracellular transport defect in our hands. All together, these data suggest that C11ORF24 is present on the Golgi apparatus, transported to the plasma membrane and cycles back through the endosomes by way of RAB6 positive carriers.</p> </div

    C11ORF24 has a long luminal domain and a short cytosolic tail.

    No full text
    <p>(A) Cartoon of a cell expressing GFP-tagged GalT or C11ORF24 and stained with an anti-GFP antibody and an anti-GM130 antibody. When the cells are not permeabilized (left) the antibodies don’t bind their targets. After saponin permeabilization (middle) all antibodies are able to reach their targets since all membranes are permeabilized. After digitonin permeabilization (right) only the cytosolic epitopes (GM130) are labeled whereas the luminal epitopes (GFP of GalT or C11ORF24) are protected by the intact Golgi membranes. Protocol modified from [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0082223#B18" target="_blank">18</a>] (B) HeLa cells expressing GFP-GalT (a-c) or GFP-C11ORF24 (d-f) were stained with GFP and GM130 antibodies prior to fixation (a, d) or after fixation and permeabilization with saponin (b,e) or after fixation and permeabilization with digitonin (c,f). The GFP of C11ORF24 is accessible for the anti GFP antibody only after permeabilization of the Golgi membrane. (C) HeLa cells expressing GFP-GalT were stained with GFP and C11ORF24 antibodies prior to fixation (a) or after fixation and permeabilization with saponin (b) or after fixation and permeabilization with digitonin (c). The epitope of the C11ORF24 antibody is accessible only after permeabilization of the Golgi membrane. (D) HeLa cells were either transfected with GFP-RAB6 (line 1) or GFP-C11ORF24 (line 2) 18h prior to the incubation with the anti-GFP and internalization was performed at 37°C for 90 minutes. Cells were then fixed and stained with a secondary antibody and the localization of the anti-GFP antibody was compared with the GFP signal.</p

    C11ORF24 is not necessary for the formation of RAB6 positive transport carriers.

    No full text
    <p>(A) After a 48 hours knockdown by shRNA HeLa cells were transfected with GFP-RAB6 and the next day they were imaged every second for 30 seconds by spinning disk microscopy. Snap shots from the Movies S1 and S2 are presented for the control shRNA (upper lines) compared to the C11ORF24 shRNA (lower lines). (B) The number of RAB6 positive transport carrier per cell and the speed of theses carriers was then quantified for both treatments from 3 independent experiments and expressed as a percentage of the control. The number of cells is indicated for each treatment on the graphs (n). The error bars represent the SEM.</p
    corecore