Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
FEBS Lett ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997225

ABSTRACT

SSR128129E (SSR) is a unique small-molecule inhibitor of fibroblast growth factor receptors (FGFRs). SSR is a high-affinity allosteric binder that selectively blocks one of the two major FGFR-mediated pathways. The mechanisms of SSR activity were studied previously in much detail, allowing the identification of its binding site, located in the hydrophobic groove of the receptor D3 domain. The binding site overlaps with the position of an N-terminal helix, an element exclusive for the FGF8b growth factor, which could potentially convert SSR from an allosteric inhibitor into an orthosteric blocker for the particular FGFR/FGF8b system. In this regard, we report here on the structural and functional investigation of FGF8b/FGFR3c system and the effects imposed on it by SSR. We show that SSR is equally or more potent in inhibiting FGF8b-induced FGFR signaling compared to FGF2-induced activation. On the other hand, when studied in the context of separate extracellular domains of FGFR3c in solution with NMR spectroscopy, SSR is unable to displace the N-terminal helix of FGF8b from its binding site on FGFR3c and behaves as a weak orthosteric inhibitor. The substantial inconsistency between the results obtained with cell culture and for the individual water-soluble subdomains of the FGFR proteins points to the important role played by the cell membrane.

2.
Neuron ; 112(2): 175-177, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38237553

ABSTRACT

In this issue of Neuron, Bhat et al.1 unveil the temporary reawakening of an embryonic guidance program, which facilitates the alignment of blood neovessels, creating a supportive "bridge" microenvironment for axon regrowth and tissue regeneration after peripheral nervous system (PNS) injury.


Subject(s)
Axons , Nerve Regeneration , Axons/physiology , Nerve Regeneration/physiology , Neurons , Peripheral Nervous System/physiology
3.
Sci Rep ; 13(1): 20497, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37993550

ABSTRACT

Dendritic spines are considered a morphological proxy for excitatory synapses, rendering them a target of many different lines of research. Over recent years, it has become possible to simultaneously image large numbers of dendritic spines in 3D volumes of neural tissue. In contrast, currently no automated method for 3D spine detection exists that comes close to the detection performance reached by human experts. However, exploiting such datasets requires new tools for the fully automated detection and analysis of large numbers of spines. Here, we developed an efficient analysis pipeline to detect large numbers of dendritic spines in volumetric fluorescence imaging data acquired by two-photon imaging in vivo. The core of our pipeline is a deep convolutional neural network that was pretrained on a general-purpose image library and then optimized on the spine detection task. This transfer learning approach is data efficient while achieving a high detection precision. To train and validate the model we generated a labeled dataset using five human expert annotators to account for the variability in human spine detection. The pipeline enables fully automated dendritic spine detection reaching a performance slightly below that of the human experts. Our method for spine detection is fast, accurate and robust, and thus well suited for large-scale datasets with thousands of spines. The code is easily applicable to new datasets, achieving high detection performance, even without any retraining or adjustment of model parameters.


Subject(s)
Dendritic Spines , Nerve Tissue , Humans , Neural Networks, Computer , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods
4.
Neuron ; 110(22): 3656-3660, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36356578

ABSTRACT

In May, an interdisciplinary group gathered in Crete for the Molecular Neurobiology Workshop. Scientists shared data acquired by vastly diverse techniques to understand how the nervous system, with only a limited number of components, is assembled to respond to infinite stimuli. Ideas of molecular cues, timing, switching, and context emerged.


Subject(s)
Neurosciences , Neurobiology
5.
STAR Protoc ; 3(3): 101507, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35776645

ABSTRACT

Assessing protein distribution with super-resolution in tissue is often complicated and restrictive. Here, we describe a protocol for immunostaining and expansion microscopy imaging of mouse brain organotypic slice cultures. We detail an Imaris analysis workflow to analyze the surface vs intracellular distribution of AMPA receptors at super-resolution during homeostatic plasticity. We have optimized the protocol for brain organotypic slice culture and tested in acute brain slices. This protocol is suitable to study protein distribution under multiple plasticity paradigms. For complete details on the use and execution of this protocol, please refer to Bissen et al. (2021).


Subject(s)
Microscopy , Receptors, AMPA , Animals , Brain/diagnostic imaging , Mice , Organ Culture Techniques , Receptors, AMPA/metabolism
6.
Front Cell Dev Biol ; 10: 890852, 2022.
Article in English | MEDLINE | ID: mdl-35573692

ABSTRACT

Evolutionary studies indicate that the nervous system evolved prior to the vascular system, but the increasing complexity of organisms prompted the vascular system to emerge in order to meet the growing demand for oxygen and nutrient supply. In recent years, it has become apparent that the symbiotic communication between the nervous and the vascular systems goes beyond the exclusive covering of the demands on nutrients and oxygen carried by blood vessels. Indeed, this active interplay between both systems is crucial during the development of the central nervous system (CNS). Several neural-derived signals that initiate and regulate the vascularization of the CNS have been described, however less is known about the vascular signals that orchestrate the development of the CNS cytoarchitecture. Here, we focus on reviewing the effects of blood vessels in the process of neurogenesis during CNS development in vertebrates. In mammals, we describe the spatiotemporal features of vascular-driven neurogenesis in two brain regions that exhibit different neurogenic complexity in their germinal zone, the hindbrain and the forebrain.

7.
Cell Rep ; 36(7): 109522, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34407407

ABSTRACT

Neuro-vascular communication is essential to synchronize central nervous system development. Here, we identify angiopoietin/Tie2 as a neuro-vascular signaling axis involved in regulating dendritic morphogenesis of Purkinje cells (PCs). We show that in the developing cerebellum Tie2 expression is not restricted to blood vessels, but it is also present in PCs. Its ligands angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are expressed in neural cells and endothelial cells (ECs), respectively. PC-specific deletion of Tie2 results in reduced dendritic arborization, which is recapitulated in neural-specific Ang1-knockout and Ang2 full-knockout mice. Mechanistically, RNA sequencing reveals that Tie2-deficient PCs present alterations in gene expression of multiple genes involved in cytoskeleton organization, dendritic formation, growth, and branching. Functionally, mice with deletion of Tie2 in PCs present alterations in PC network functionality. Altogether, our data propose Ang/Tie2 signaling as a mediator of intercellular communication between neural cells, ECs, and PCs, required for proper PC dendritic morphogenesis and function.


Subject(s)
Angiopoietin-2/metabolism , Dendrites/metabolism , Morphogenesis , Purkinje Cells/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Angiopoietin-1/metabolism , Animals , Cerebellum/blood supply , Cerebellum/growth & development , Gene Deletion , Gene Expression Regulation , Integrases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Organ Specificity
8.
Curr Opin Neurobiol ; 69: 202-213, 2021 08.
Article in English | MEDLINE | ID: mdl-34077852

ABSTRACT

PURPOSE OF THE REVIEW: The synchronic development of vascular and nervous systems is orchestrated by common molecules that regulate the communication between both systems. The identification of these common guiding cues and the developmental processes regulated by neurovascular communication are slowly emerging. In this review, we describe the molecules modulating the neurovascular development and their impact in processes such as angiogenesis, neurogenesis, neuronal migration, and brain homeostasis. RECENT FINDINGS: Blood vessels not only are involved in nutrient and oxygen supply of the central nervous system (CNS) but also exert instrumental functions controlling developmental neurogenesis, CNS cytoarchitecture, and neuronal plasticity. Conversely, neurons modulate CNS vascularization and brain endothelial properties such as blood-brain barrier and vascular hyperemia. SUMMARY: The integration of the active role of endothelial cells in the development and maintenance of neuronal function is important to obtain a more holistic view of the CNS complexity and also to understand how the vasculature is involved in neuropathological conditions.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Brain , Central Nervous System , Neurons
9.
Cell Rep ; 34(13): 108923, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789115

ABSTRACT

Despite decades of work, much remains elusive about molecular events at the interplay between physiological and structural changes underlying neuronal plasticity. Here, we combined repetitive live imaging and expansion microscopy in organotypic brain slice cultures to quantitatively characterize the dynamic changes of the intracellular versus surface pools of GluA2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) across the different dendritic spine types and the shaft during hippocampal homeostatic plasticity. Mechanistically, we identify ephrinB2 and glutamate receptor interacting protein (GRIP) 1 as mediating AMPAR relocation to the mushroom spine surface following lesion-induced denervation. Moreover, stimulation with the ephrinB2 specific receptor EphB4 not only prevents the lesion-induced disappearance of mushroom spines but is also sufficient to shift AMPARs to the surface and rescue spine recovery in a GRIP1 dominant-negative background. Thus, our results unravel a crucial role for ephrinB2 during homeostatic plasticity and identify a potential pharmacological target to improve dendritic spine plasticity upon injury.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dendritic Spines/metabolism , Ephrin-B2/metabolism , Homeostasis , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Animals , Cell Membrane/metabolism , Denervation , Mice, Inbred C57BL , Receptor, EphB4/metabolism , Receptors, AMPA/metabolism
10.
Trends Neurosci ; 44(5): 393-405, 2021 05.
Article in English | MEDLINE | ID: mdl-33423792

ABSTRACT

The blood-brain barrier (BBB) is a dynamic platform for exchange of substances between the blood and the brain parenchyma, and it is an essential functional gatekeeper for the central nervous system (CNS). While it is widely recognized that BBB disruption is a hallmark of several neurovascular pathologies, an aspect of the BBB that has received somewhat less attention is the dynamic modulation of BBB tightness to maintain brain homeostasis in response to extrinsic environmental factors and physiological changes. In this review, we summarize how BBB integrity adjusts in critical stages along the life span, as well as how BBB permeability can be altered by common stressors derived from nutritional habits, environmental factors and psychological stress.


Subject(s)
Blood-Brain Barrier , Brain , Biological Transport , Central Nervous System , Homeostasis , Humans
11.
EMBO Rep ; 22(2): e48961, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33512764

ABSTRACT

Endothelial tip cells are essential for VEGF-induced angiogenesis, but underlying mechanisms are elusive. The Ena/VASP protein family, consisting of EVL, VASP, and Mena, plays a pivotal role in axon guidance. Given that axonal growth cones and endothelial tip cells share many common features, from the morphological to the molecular level, we investigated the role of Ena/VASP proteins in angiogenesis. EVL and VASP, but not Mena, are expressed in endothelial cells of the postnatal mouse retina. Global deletion of EVL (but not VASP) compromises the radial sprouting of the vascular plexus in mice. Similarly, endothelial-specific EVL deletion compromises the radial sprouting of the vascular plexus and reduces the endothelial tip cell density and filopodia formation. Gene sets involved in blood vessel development and angiogenesis are down-regulated in EVL-deficient P5-retinal endothelial cells. Consistently, EVL deletion impairs VEGF-induced endothelial cell proliferation and sprouting, and reduces the internalization and phosphorylation of VEGF receptor 2 and its downstream signaling via the MAPK/ERK pathway. Together, we show that endothelial EVL regulates sprouting angiogenesis via VEGF receptor-2 internalization and signaling.


Subject(s)
Cell Adhesion Molecules/physiology , Endothelial Cells , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2 , Animals , Endothelial Cells/metabolism , Mice , Morphogenesis , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Cell Rep ; 31(4): 107568, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32348765

ABSTRACT

Anti-angiogenic treatment targeting vascular endothelial growth factor (VEGF)-VEGFR2 signaling has shown limited efficacy in lung cancer patients. Here, we demonstrate that inhibition of VEGFR2 in tumor cells, expressed in ∼20% of non-squamous non-small cell lung cancer (NSCLC) patients, leads to a pro-invasive phenotype. Drug-induced inhibition of tumor VEGFR2 interferes with the formation of the EphA2/VEGFR2 heterocomplex, thereby allowing RSK to interact with Serine 897 of EphA2. Inhibition of RSK decreases phosphorylation of Serine 897 EphA2. Selective genetic modeling of Serine 897 of EphA2 or inhibition of EphA2 abrogates the formation of metastases in vivo upon VEGFR2 inhibition. In summary, these findings demonstrate that VEGFR2-targeted therapy conditions VEGFR2-positive NSCLC to Serine 897 EphA2-dependent aggressive tumor growth and metastasis. These data shed light on the molecular mechanisms explaining the limited efficacy of VEGFR2-targeted anti-angiogenic treatment in lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Receptor, EphA2/metabolism , Serine/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Vascular Endothelial Growth Factor Receptor-2/genetics
13.
Am J Physiol Cell Physiol ; 318(4): C719-C731, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31967857

ABSTRACT

Carotid body (CB) type I cells sense the blood Po2 and generate a nervous signal for stimulating ventilation and circulation when blood oxygen levels decline. Three oxygen-sensing enzyme complexes may be used for this purpose: 1) mitochondrial electron transport chain metabolism, 2) heme oxygenase 2 (HO-2)-generating CO, and/or 3) an NAD(P)H oxidase (NOX). We hypothesize that intracellular redox changes are the link between the sensor and nervous signals. To test this hypothesis type I cell autofluorescence of flavoproteins (Fp) and NAD(P)H within the mouse CB ex vivo was recorded as Fp/(Fp+NAD(P)H) redox ratio. CB type I cell redox ratio transiently declined with the onset of hypoxia. Upon reoxygenation, CB type I cells showed a significantly increased redox ratio. As a control organ, the non-oxygen-sensing sympathetic superior cervical ganglion (SCG) showed a continuously reduced redox ratio upon hypoxia. CN-, diphenyleneiodonium, or reactive oxygen species influenced chemoreceptor discharge (CND) with subsequent loss of O2 sensitivity and inhibited hypoxic Fp reduction only in the CB but not in SCG Fp, indicating a specific role of Fp in the oxygen-sensing process. Hypoxia-induced changes in CB type I cell redox ratio affected peptidyl prolyl isomerase Pin1, which is believed to colocalize with the NADPH oxidase subunit p47phox in the cell membrane to trigger the opening of potassium channels. We postulate that hypoxia-induced changes in the Fp-mediated redox ratio of the CB regulate the Pin1/p47phox tandem to alter type I cell potassium channels and therewith CND.


Subject(s)
Carotid Body/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Chemoreceptor Cells/metabolism , Flavoproteins/metabolism , Hypoxia/metabolism , Lung/metabolism , Mice , Mitochondria/metabolism , Potassium Channels/metabolism
14.
Nat Cell Biol ; 22(1): 97-107, 2020 01.
Article in English | MEDLINE | ID: mdl-31907411

ABSTRACT

Diffuse brain infiltration by glioma cells causes detrimental disease progression, but its multicellular coordination is poorly understood. We show here that glioma cells infiltrate the brain collectively as multicellular networks. Contacts between moving glioma cells are adaptive epithelial-like or filamentous junctions stabilized by N-cadherin, ß-catenin and p120-catenin, which undergo kinetic turnover, transmit intercellular calcium transients and mediate directional persistence. Downregulation of p120-catenin compromises cell-cell interaction and communication, disrupts collective networks, and both the cadherin and RhoA binding domains of p120-catenin are required for network formation and migration. Deregulating p120-catenin further prevents diffuse glioma cell infiltration of the mouse brain with marginalized microlesions as the outcome. Transcriptomics analysis has identified p120-catenin as an upstream regulator of neurogenesis and cell cycle pathways and a predictor of poor clinical outcome in glioma patients. Collective glioma networks infiltrating the brain thus depend on adherens junctions dynamics, the targeting of which may offer an unanticipated strategy to halt glioma progression.


Subject(s)
Adherens Junctions/metabolism , Catenins/metabolism , Cell Adhesion/physiology , Glioma/pathology , Animals , Brain/metabolism , Brain/pathology , Cadherins/metabolism , Cell Line, Tumor , Down-Regulation/physiology , Glioma/metabolism , Phosphoproteins/metabolism , Phosphorylation , Delta Catenin
15.
Elife ; 82019 12 23.
Article in English | MEDLINE | ID: mdl-31868583

ABSTRACT

Axon branching is crucial for proper formation of neuronal networks. Although originally identified as an angiogenic factor, VEGF also signals directly to neurons to regulate their development and function. Here we show that VEGF and its receptor VEGFR2 (also known as KDR or FLK1) are expressed in mouse hippocampal neurons during development, with VEGFR2 locally expressed in the CA3 region. Activation of VEGF/VEGFR2 signaling in isolated hippocampal neurons results in increased axon branching. Remarkably, inactivation of VEGFR2 also results in increased axon branching in vitro and in vivo. The increased CA3 axon branching is not productive as these axons are less mature and form less functional synapses with CA1 neurons. Mechanistically, while VEGF promotes the growth of formed branches without affecting filopodia formation, loss of VEGFR2 increases the number of filopodia and enhances the growth rate of new branches. Thus, a controlled VEGF/VEGFR2 signaling is required for proper CA3 hippocampal axon branching during mouse hippocampus development.


Subject(s)
Axons/physiology , Hippocampus/growth & development , Hippocampus/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Ephrin-B2/genetics , Gene Expression Regulation, Developmental , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Pseudopodia/metabolism , Signal Transduction/genetics , Synapses/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
16.
Elife ; 82019 12 23.
Article in English | MEDLINE | ID: mdl-31868584

ABSTRACT

Vascular endothelial growth factor (VEGF) is an angiogenic factor that play important roles in the nervous system, although it is still unclear which receptors transduce those signals in neurons. Here, we show that in the developing hippocampus VEGFR2 (also known as KDR or FLK1) is expressed specifically in the CA3 region and it is required for dendritic arborization and spine morphogenesis in hippocampal neurons. Mice lacking VEGFR2 in neurons (Nes-cre Kdrlox/-) show decreased dendritic arbors and spines as well as a reduction in long-term potentiation (LTP) at the associational-commissural - CA3 synapses. Mechanistically, VEGFR2 internalization is required for VEGF-induced spine maturation. In analogy to endothelial cells, ephrinB2 controls VEGFR2 internalization in neurons. VEGFR2-ephrinB2 compound mice (Nes-cre Kdrlox/+ Efnb2lox/+) show reduced dendritic branching, reduced spine head size and impaired LTP. Our results demonstrate the functional crosstalk of VEGFR2 and ephrinB2 in vivo to control dendritic arborization, spine morphogenesis and hippocampal circuitry development.


Subject(s)
Dendrites/metabolism , Ephrin-B2/metabolism , Hippocampus/metabolism , Neurogenesis/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , CA3 Region, Hippocampal , Dendritic Spines/metabolism , Endothelial Cells/metabolism , Ephrin-B2/genetics , Gene Expression Regulation, Developmental , Long-Term Potentiation/physiology , Mice , Neurogenesis/genetics , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/physiology , Transcriptome , Vascular Endothelial Growth Factor Receptor-2/genetics
17.
Cell ; 179(7): 1661-1676.e19, 2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31835038

ABSTRACT

Reliable detection of disseminated tumor cells and of the biodistribution of tumor-targeting therapeutic antibodies within the entire body has long been needed to better understand and treat cancer metastasis. Here, we developed an integrated pipeline for automated quantification of cancer metastases and therapeutic antibody targeting, named DeepMACT. First, we enhanced the fluorescent signal of cancer cells more than 100-fold by applying the vDISCO method to image metastasis in transparent mice. Second, we developed deep learning algorithms for automated quantification of metastases with an accuracy matching human expert manual annotation. Deep learning-based quantification in 5 different metastatic cancer models including breast, lung, and pancreatic cancer with distinct organotropisms allowed us to systematically analyze features such as size, shape, spatial distribution, and the degree to which metastases are targeted by a therapeutic monoclonal antibody in entire mice. DeepMACT can thus considerably improve the discovery of effective antibody-based therapeutics at the pre-clinical stage. VIDEO ABSTRACT.


Subject(s)
Antibodies/therapeutic use , Deep Learning , Diagnosis, Computer-Assisted/methods , Drug Therapy, Computer-Assisted/methods , Neoplasms/pathology , Animals , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Neoplasm Metastasis , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Software , Tumor Microenvironment
18.
Annu Rev Cell Dev Biol ; 35: 615-635, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31590587

ABSTRACT

Molecular cross talk between the nervous and vascular systems is necessary to maintain the correct coupling of organ structure and function. Molecular pathways shared by both systems are emerging as major players in the communication of the neuronal compartment with the endothelium. Here we review different aspects of this cross talk and how vessels influence the development and homeostasis of the nervous system. Beyond the classical role of the vasculature as a conduit to deliver oxygen and metabolites needed for the energy-demanding neuronal compartment, vessels emerge as powerful signaling systems that control and instruct a variety of cellular processes during the development of neurons and glia, such as migration, differentiation, and structural connectivity. Moreover, a broad spectrum of mild to severe vascular dysfunctions occur in various pathologies of the nervous system, suggesting that mild structural and functional changes at the neurovascular interface may underlie cognitive decline in many of these pathological conditions.


Subject(s)
Central Nervous System/blood supply , Neuroglia/cytology , Neurons/cytology , Neurovascular Coupling/physiology , Peripheral Nervous System/blood supply , Animals , Blood Vessels/cytology , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Differentiation , Cell Movement , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/metabolism , Homeostasis/physiology , Humans , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Neuroglia/physiology , Neurons/physiology , Peripheral Nervous System/cytology , Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism
19.
Cell Mol Life Sci ; 76(11): 2133-2169, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30937469

ABSTRACT

To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.


Subject(s)
Carrier Proteins/metabolism , Nerve Net/physiology , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Nuclear Proteins/metabolism , Receptors, AMPA/metabolism , Animals , Carrier Proteins/genetics , Egg Proteins/genetics , Egg Proteins/metabolism , Gene Expression Regulation , Glutamic Acid/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Neural Networks, Computer , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/genetics , Protein Transport , Receptors, AMPA/genetics , Synapses/metabolism , Synaptic Transmission
20.
Mol Autism ; 9: 56, 2018.
Article in English | MEDLINE | ID: mdl-30443311

ABSTRACT

Background: Altered neuronal development is discussed as the underlying pathogenic mechanism of autism spectrum disorders (ASD). Copy number variations of 16p11.2 have recurrently been identified in individuals with ASD. Of the 29 genes within this region, quinolinate phosphoribosyltransferase (QPRT) showed the strongest regulation during neuronal differentiation of SH-SY5Y neuroblastoma cells. We hypothesized a causal relation between this tryptophan metabolism-related enzyme and neuronal differentiation. We thus analyzed the effect of QPRT on the differentiation of SH-SY5Y and specifically focused on neuronal morphology, metabolites of the tryptophan pathway, and the neurodevelopmental transcriptome. Methods: The gene dosage-dependent change of QPRT expression following Chr16p11.2 deletion was investigated in a lymphoblastoid cell line (LCL) of a deletion carrier and compared to his non-carrier parents. Expression of QPRT was tested for correlation with neuromorphology in SH-SY5Y cells. QPRT function was inhibited in SH-SY5Y neuroblastoma cells using (i) siRNA knockdown (KD), (ii) chemical mimicking of loss of QPRT, and (iii) complete CRISPR/Cas9-mediated knock out (KO). QPRT-KD cells underwent morphological analysis. Chemically inhibited and QPRT-KO cells were characterized using viability assays. Additionally, QPRT-KO cells underwent metabolite and whole transcriptome analyses. Genes differentially expressed upon KO of QPRT were tested for enrichment in biological processes and co-regulated gene-networks of the human brain. Results: QPRT expression was reduced in the LCL of the deletion carrier and significantly correlated with the neuritic complexity of SH-SY5Y. The reduction of QPRT altered neuronal morphology of differentiated SH-SY5Y cells. Chemical inhibition as well as complete KO of the gene were lethal upon induction of neuronal differentiation, but not proliferation. The QPRT-associated tryptophan pathway was not affected by KO. At the transcriptome level, genes linked to neurodevelopmental processes and synaptic structures were affected. Differentially regulated genes were enriched for ASD candidates, and co-regulated gene networks were implicated in the development of the dorsolateral prefrontal cortex, the hippocampus, and the amygdala. Conclusions: In this study, QPRT was causally related to in vitro neuronal differentiation of SH-SY5Y cells and affected the regulation of genes and gene networks previously implicated in ASD. Thus, our data suggest that QPRT may play an important role in the pathogenesis of ASD in Chr16p11.2 deletion carriers.


Subject(s)
Autism Spectrum Disorder/genetics , Cell Differentiation/genetics , Neurons/cytology , Pentosyltransferases/genetics , Cell Line, Tumor , Chromosome Deletion , Chromosomes, Human, Pair 16 , DNA Copy Number Variations , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...