22 research outputs found

    HID-1 controls formation of large dense core vesicles by influencing cargo sorting and trans-Golgi network acidification

    Get PDF
    Large dense core vesicles (LDCVs) mediate the regulated release of neuropeptides and peptide hormones. They form at the trans-Golgi network (TGN), where their soluble content aggregates to form a dense core, but the mechanisms controlling biogenesis are still not completely understood. Recent studies have implicated the peripheral membrane protein HID-1 in neuropeptide sorting and insulin secretion. Using CRISPR/Cas9, we generated HID-1 KO rat neuroendocrine cells, and we show that the absence of HID-1 results in specific defects in peptide hormone and monoamine storage and regulated secretion. Loss of HID-1 causes a reduction in the number of LDCVs and affects their morphology and biochemical properties, due to impaired cargo sorting and dense core formation. HID-1 KO cells also exhibit defects in TGN acidification together with mislocalization of the Golgi-enriched vacuolar H+-ATPase subunit isoform a2. We propose that HID-1 influences early steps in LDCV formation by controlling dense core formation at the TGN.</jats:p

    The CaMKII/NMDA receptor complex controls hippocampal synaptic transmission by kinase-dependent and independent mechanisms.

    Get PDF
    CaMKII is one of the most studied synaptic proteins, but many critical issues regarding its role in synaptic function remain unresolved. Using a CRISPR-based system to delete CaMKII and replace it with mutated forms in single neurons, we have rigorously addressed its various synaptic roles. In brief, basal AMPAR and NMDAR synaptic transmission both require CaMKIIα, but not CaMKIIβ, indicating that, even in the adult, synaptic transmission is determined by the ongoing action of CaMKIIα. While AMPAR transmission requires kinase activity, NMDAR transmission does not, implying a scaffolding role for the CaMKII protein instead. LTP is abolished in the absence of CaMKIIα and/or CaMKIIβ and with an autophosphorylation impaired CaMKIIα (T286A). With the exception of NMDAR synaptic currents, all aspects of CaMKIIα signaling examined require binding to the NMDAR, emphasizing the essential role of this receptor as a master synaptic signaling hub

    Efficient, Complete Deletion of Synaptic Proteins using CRISPR

    Get PDF
    One of the most powerful ways to test the function of a protein is to characterize the consequences of its deletion. In the past, this has involved inactivation of the gene by homologous recombination either in the germline or later through conditional deletion. RNA interference (RNAi) provides an alternative way to knock down proteins, but both of these approaches have their limitations. Recently, the CRISPR/Cas9 system has suggested another way to selectively inactivate genes. We have now tested this system in postmitotic neurons by targeting two well-characterized synaptic proteins, the obligatory GluN1 subunit of the NMDA receptor and the GluA2&nbsp;subunit of the AMPA receptor. Expression of CRISPR/Cas9 in hippocampal slice cultures completely eliminated NMDA receptor and GluA2 function. CRISPR/Cas9 thus provides a powerful tool to study the function of synaptic proteins

    Artificial tethering of the HID-1 C-terminus domain to the TGN is sufficient to rescue the HID-1 KO phenotype.

    No full text
    (A-C) HID-1 KO PC12 cells were transiently co-transfected with GFP-TGN38, GFP-TGN38-HID-1-C-terminus or HID-1-GFP and NPY-sfCherry3. (A) Anti-GFP immunoblot showing expression level of the fusion proteins. (B-C) Cells were washed and incubated in Tyrode’s buffer containing 2.5 mM K+ (basal) or 90 mM K+ (stimulated) for 30 min at 37°C. The amount of cellular and secreted NPY-sfCherry3 was determined using a plate reader (Tecan). *, p D) HID-1 KO PC12 cells were transiently co-transfected with GFP, GFP-TGN38-HID-1-C-terminus, immunostained for SgII and the amount of fluorescence in GFP positive cells was quantified and normalized to WT PC12 cells. ***, p<0.001 relative to HID-1-KO by one-way ANOVA (n = 226 cells for WT, n = 217 cells for KO, n = 186 cells for GFP, n = 185 cells for GFP-TGN38-Cterm from three independent experiments). The bar graphs indicate mean ± s.e.</p

    HID-1 myristoylation is not required for function.

    No full text
    (A-C) HID-1 KO PC12 cells transduced with indicated HID-1-HA lentivirus were washed and incubated for 30 minutes in Tyrode’s solution containing 2.5 mM K+ (basal) or 90 mM K+ (stimulated). Cellular and secreted SgII were measured by quantitative fluorescent immunoblotting (A), with the secreted SgII normalized to tubulin and expressed as percent of basal secretion in the KO (B), and the cellular SgII normalized to tubulin (C). *, p < 0.05 relative to HID-1 KO by one-way ANOVA (n = 4). The bar graphs indicate mean ± s.e.</p

    Protein sequence alignment of HID-1 C-terminus (103 amino acids) for the indicated species and for the human mutation.

    No full text
    Protein sequence alignment of HID-1 C-terminus (103 amino acids) for the indicated species and for the human mutation.</p
    corecore