Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Proc Natl Acad Sci U S A ; 116(16): 8000-8009, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926666

ABSTRACT

Neural stem cells continuously generate newborn neurons that integrate into and modify neural circuitry in the adult hippocampus. The molecular mechanisms that regulate or perturb neural stem cell proliferation and differentiation, however, remain poorly understood. Here, we have found that mouse hippocampal radial glia-like (RGL) neural stem cells express the synaptic cochaperone cysteine string protein-α (CSP-α). Remarkably, in CSP-α knockout mice, RGL stem cells lose quiescence postnatally and enter into a high-proliferation regime that increases the production of neural intermediate progenitor cells, thereby exhausting the hippocampal neural stem cell pool. In cell culture, stem cells in hippocampal neurospheres display alterations in proliferation for which hyperactivation of the mechanistic target of rapamycin (mTOR) signaling pathway is the primary cause of neurogenesis deregulation in the absence of CSP-α. In addition, RGL cells lose quiescence upon specific conditional targeting of CSP-α in adult neural stem cells. Our findings demonstrate an unanticipated cell-autonomic and circuit-independent disruption of postnatal neurogenesis in the absence of CSP-α and highlight a direct or indirect CSP-α/mTOR signaling interaction that may underlie molecular mechanisms of brain dysfunction and neurodegeneration.


Subject(s)
HSP40 Heat-Shock Proteins , Membrane Proteins , Neural Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Hippocampus/cytology , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neurogenesis/genetics , Neuronal Ceroid-Lipofuscinoses , Signal Transduction/genetics
3.
Nature ; 560(7719): 441-446, 2018 08.
Article in English | MEDLINE | ID: mdl-30111840

ABSTRACT

Common genetic contributions to autism spectrum disorder (ASD) reside in risk gene variants that individually have minimal effect sizes. As environmental factors that perturb neurodevelopment also underlie idiopathic ASD, it is crucial to identify altered regulators that can orchestrate multiple ASD risk genes during neurodevelopment. Cytoplasmic polyadenylation element binding proteins 1-4 (CPEB1-4) regulate the translation of specific mRNAs by modulating their poly(A)-tails and thereby participate in embryonic development and synaptic plasticity. Here we find that CPEB4 binds transcripts of most high-confidence ASD risk genes. The brains of individuals with idiopathic ASD show imbalances in CPEB4 transcript isoforms that result from decreased inclusion of a neuron-specific microexon. In addition, 9% of the transcriptome shows reduced poly(A)-tail length. Notably, this percentage is much higher for high-confidence ASD risk genes, correlating with reduced expression of the protein products of ASD risk genes. An equivalent imbalance in CPEB4 transcript isoforms in mice mimics the changes in mRNA polyadenylation and protein expression of ASD risk genes and induces ASD-like neuroanatomical, electrophysiological and behavioural phenotypes. Together, these data identify CPEB4 as a regulator of ASD risk genes.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Genetic Predisposition to Disease/genetics , Polyadenylation , RNA Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Exons/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Neurons/metabolism , Phenotype , Protein Binding , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transcriptome
4.
Aging Cell ; 17(5): e12821, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30058223

ABSTRACT

The striatum integrates motor behavior using a well-defined microcircuit whose individual components are independently affected in several neurological diseases. The glial cell line-derived neurotrophic factor (GDNF), synthesized by striatal interneurons, and Sonic hedgehog (Shh), produced by the dopaminergic neurons of the substantia nigra (DA SNpc), are both involved in the nigrostriatal maintenance but the reciprocal neurotrophic relationships among these neurons are only partially understood. To define the postnatal neurotrophic connections among fast-spiking GABAergic interneurons (FS), cholinergic interneurons (ACh), and DA SNpc, we used a genetically induced mouse model of postnatal DA SNpc neurodegeneration and separately eliminated Smoothened (Smo), the obligatory transducer of Shh signaling, in striatal interneurons. We show that FS postnatal survival relies on DA SNpc and is independent of Shh signaling. On the contrary, Shh signaling but not dopaminergic striatal innervation is required to maintain ACh in the postnatal striatum. ACh are required for DA SNpc survival in a GDNF-independent manner. These data demonstrate the existence of three parallel but interdependent neurotrophic relationships between SN and striatal interneurons, partially defined by Shh and GDNF. The definition of these new neurotrophic interactions opens the search for new molecules involved in the striatal modulatory circuit maintenance with potential therapeutic value.


Subject(s)
Corpus Striatum/physiology , Dopaminergic Neurons/physiology , Interneurons/physiology , Nerve Net/physiology , Substantia Nigra/physiology , Acetylcholine/metabolism , Action Potentials , Animals , Animals, Newborn , Cell Survival , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hedgehog Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/pathology , Signal Transduction
5.
ACS Nano ; 11(4): 3429-3432, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28388033

ABSTRACT

The release of chemical mediators is an essential element of cell-to-cell communication. Signaling molecules such as neurotransmitters and hormones are stored in membrane-bound organelles called secretory vesicles. Some of these organelles can store molecules at high concentrations, overcoming the osmotic shock that could burst the organelle. These organelles contain a proteinaceous matrix that traps the molecules and avoids high intravesicular osmotic pressure. The functional nanostructure and internal organization of the matrix is not well understood. A report by Lovric et al. in this issue of ACS Nano provides insight into the storage of a small molecule-dopamine-within the intraluminal compartments of a secretory vesicle. Lovric et al. used a powerful combination of high spatial resolution mass spectrometry and transmission electron microscopy in conjunction with amperometric measurements of exocytotic release to delineate the temporal and spatial fate of intravesicular dopamine and its interaction with the matrix.


Subject(s)
Dopamine , Secretory Vesicles , Exocytosis , Nanostructures , Spectrometry, Mass, Secondary Ion
6.
EMBO Mol Med ; 8(11): 1289-1309, 2016 11.
Article in English | MEDLINE | ID: mdl-27807076

ABSTRACT

Skeletal muscle regeneration by muscle satellite cells is a physiological mechanism activated upon muscle damage and regulated by Notch signaling. In a family with autosomal recessive limb-girdle muscular dystrophy, we identified a missense mutation in POGLUT1 (protein O-glucosyltransferase 1), an enzyme involved in Notch posttranslational modification and function. In vitro and in vivo experiments demonstrated that the mutation reduces O-glucosyltransferase activity on Notch and impairs muscle development. Muscles from patients revealed decreased Notch signaling, dramatic reduction in satellite cell pool and a muscle-specific α-dystroglycan hypoglycosylation not present in patients' fibroblasts. Primary myoblasts from patients showed slow proliferation, facilitated differentiation, and a decreased pool of quiescent PAX7+ cells. A robust rescue of the myogenesis was demonstrated by increasing Notch signaling. None of these alterations were found in muscles from secondary dystroglycanopathy patients. These data suggest that a key pathomechanism for this novel form of muscular dystrophy is Notch-dependent loss of satellite cells.


Subject(s)
Glucosyltransferases/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Biopsy , Glycosylation , Glycosyltransferases/metabolism , Humans , Muscles/pathology , Sequence Analysis, DNA , Spain
7.
PLoS Biol ; 14(7): e1002522, 2016 07.
Article in English | MEDLINE | ID: mdl-27454736

ABSTRACT

One of the most fascinating properties of the brain is the ability to function smoothly across decades of a lifespan. Neurons are nondividing mature cells specialized in fast electrical and chemical communication at synapses. Often, neurons and synapses operate at high levels of activity through sophisticated arborizations of long axons and dendrites that nevertheless stay healthy throughout years. On the other hand, aging and activity-dependent stress strike onto the protein machineries turning proteins unfolded and prone to form pathological aggregates associated with neurodegeneration. How do neurons protect from those insults and remain healthy for their whole life? Ali and colleagues now present a molecular mechanism by which the enzyme nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) acts not only as a NAD synthase involved in axonal maintenance but as a molecular chaperone helping neurons to overcome protein unfolding and protein aggregation.


Subject(s)
Axons , Nicotinamide-Nucleotide Adenylyltransferase , Dendrites , Molecular Chaperones , NAD , Neurons
8.
J Physiol ; 593(13): 2867-88, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25981717

ABSTRACT

KEY POINTS: Neurotransmitter release requires a tight coupling between synaptic vesicle exocytosis and endocytosis with dynamin being a key protein in that process. We used imaging techniques to examine the time course of endocytosis at mouse motor nerve terminals expressing synaptopHluorin, a genetically encoded reporter of the synaptic vesicle cycle. We separated two sequential phases of endocytosis taking place during the stimulation train: early and late endocytosis. Freshly released synaptic vesicle proteins are preferentially retrieved during the early phase, which is very sensitive to dynasore, an inhibitor of dynamin GTPase activity. Synaptic vesicle proteins pre-existing at the plasma membrane before the stimulation are preferentially retrieved during the late phase, which is very sensitive to myristyl trimethyl ammonium bromide (MitMAB), an inhibitor of the dynamin-phospholipid interaction. ABSTRACT: Synaptic endocytosis is essential at nerve terminals to maintain neurotransmitter release by exocytosis. Here, at the neuromuscular junction of synaptopHluorin (spH) transgenic mice, we have used imaging to study exo- and endocytosis occurring simultaneously during nerve stimulation. We observed two endocytosis components, which occur sequentially during stimulation. The early component of endocytosis apparently internalizes spH molecules freshly exocytosed. This component was sensitive to dynasore, a blocker of dynamin 1 GTPase activity. In contrast, this early component was resistant to myristyl trimethyl ammonium bromide (MiTMAB), a competitive agent that blocks dynamin binding to phospholipid membranes. The late component of endocytosis is likely to internalize spH molecules that pre-exist at the plasma membrane before stimulation starts. This component was blocked by MiTMAB, perhaps by impairing the binding of dynamin or other key endocytic proteins to phospholipid membranes. Our study suggests the co-existence of two sequential synaptic endocytosis steps taking place during stimulation that are susceptible to pharmacological dissection: an initial step, preferentially sensitive to dynasore, that internalizes vesicular components immediately after they are released, and a MiTMAB-sensitive step that internalizes vesicular components pre-existing at the plasma membrane surface. In addition, we report that post-stimulus endocytosis also has several components with different sensitivities to dynasore and MiTMAB.


Subject(s)
Dynamins/antagonists & inhibitors , Endocytosis , Hydrazones/pharmacology , Motor Neurons/drug effects , Neuromuscular Junction/drug effects , Animals , Mice , Motor Neurons/metabolism , Motor Neurons/physiology , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiology
9.
ACS Nano ; 7(8): 6605-18, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23826767

ABSTRACT

Polyelectrolyte multilayer (PEM) capsules are carrier vehicles with great potential for biomedical applications. With the future aim of designing biocompatible, effective therapeutic delivery systems (e.g., for cancer), the pathway of internalization (uptake and fate) of PEM capsules was investigated. In particular the following experiments were performed: (i) the study of capsule co-localization with established endocytic markers, (ii) switching-off endocytotic pathways with pharmaceutical/chemical inhibitors, and (iii) characterization and quantification of capsule uptake with confocal and electron microscopy. As result, capsules co-localized with lipid rafts and with phagolysosomes, but not with other endocytic vesicles. Chemical interference of endocytosis with chemical blockers indicated that PEM capsules enter the investigated cell lines through a mechanism slightly sensitive to electrostatic interactions, independent of clathrin and caveolae, and strongly dependent on cholesterol-rich domains and organelle acidification. Microscopic characterization of cells during capsule uptake showed the formation of phagocytic cups (vesicles) to engulf the capsules, an increased number of mitochondria, and a final localization in the perinuclear cytoplasma. Combining all these indicators we conclude that PEM capsule internalization in general occurs as a combination of different sequential mechanisms. Initially, an adsorptive mechanism due to strong electrostatic interactions governs the stabilization of the capsules at the cell surface. Membrane ruffling and filopodia extensions are responsible for capsule engulfing through the formation of a phagocytic cup. Co-localization with lipid raft domains activates the cell to initiate a lipid-raft-mediated macropinocytosis. Internalization vesicles are very acidic and co-localize only with phagolysosome markers, excluding caveolin-mediated pathways and indicating that upon phagocytosis the capsules are sorted to heterophagolysosomes.


Subject(s)
Biocompatible Materials/chemistry , Capsules/chemistry , Electrolytes/chemistry , Adsorption , Animals , Caveolae/chemistry , Cell Line, Tumor , Clathrin/chemistry , Cytoplasm/metabolism , Drug Delivery Systems , Endocytosis , Humans , Membrane Microdomains/chemistry , Mice , Microscopy, Confocal , Microscopy, Electron , Mitochondria/metabolism , Nanotechnology/methods , Phagocytosis , Phagosomes/chemistry , Static Electricity
10.
Neuron ; 74(1): 151-65, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22500637

ABSTRACT

Cysteine string protein-α (CSP-α) is a synaptic vesicle protein that prevents activity-dependent neurodegeneration by poorly understood mechanisms. We have studied the synaptic vesicle cycle at the motor nerve terminals of CSP-α knock-out mice expressing the synaptopHluorin transgene. Mutant nerve terminals fail to sustain prolonged release and the number of vesicles available to be released decreases. Strikingly, the SNARE protein SNAP-25 is dramatically reduced. In addition, endocytosis during the stimulus fails to maintain the size of the recycling synaptic vesicle pool during prolonged stimulation. Upon depolarization, the styryl dye FM 2-10 becomes trapped and poorly releasable. Consistently with the functional results, electron microscopy analysis revealed characteristic features of impaired synaptic vesicle recycling. The unexpected defect in vesicle recycling in CSP-α knock-out mice provides insights into understanding molecular mechanisms of degeneration in motor nerve terminals.


Subject(s)
Exocytosis/physiology , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Motor Neurons/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Animals , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Motor Neurons/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Recombinant Fusion Proteins/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/metabolism
11.
J Neurosci ; 31(3): 1106-13, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21248135

ABSTRACT

In Huntington's disease (HD), the expansion of polyglutamine (polyQ) repeats at the N terminus of the ubiquitous protein huntingtin (htt) leads to neurodegeneration in specific brain areas. Neurons degenerating in HD develop synaptic dysfunctions. However, it is unknown whether mutant htt impacts synaptic function in general. To investigate that, we have focused on the nerve terminals of motor neurons that typically do not degenerate in HD. Here, we have studied synaptic transmission at the neuromuscular junction of transgenic mice expressing a mutant form of htt (R6/1 mice). We have found that the size and frequency of miniature endplate potentials are similar in R6/1 and control mice. In contrast, the amplitude of evoked endplate potentials in R6/1 mice is increased compared to controls. Consistent with a presynaptic increase of release probability, synaptic depression under high-frequency stimulation is higher in R6/1 mice. In addition, no changes were detected in the size and dynamics of the recycling synaptic vesicle pool. Moreover, we have found increased amounts of the synaptic vesicle proteins synaptobrevin 1,2/VAMP 1,2 and cysteine string protein-α, and the SNARE protein SNAP-25, concomitant with normal levels of other synaptic vesicle markers. Our results reveal that the transgenic expression of a mutant form of htt leads to an unexpected gain of synaptic function. That phenotype is likely not secondary to neurodegeneration and might be due to a primary deregulation in synaptic protein levels. Our findings could be relevant to understand synaptic toxic effects of proteins with abnormal polyQ repeats.


Subject(s)
Huntington Disease/physiopathology , Neuromuscular Junction/physiopathology , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Action Potentials/physiology , Animals , Blotting, Western , Disease Models, Animal , Electrophysiology , Evoked Potentials/physiology , HSP40 Heat-Shock Proteins/metabolism , Huntington Disease/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Neuromuscular Junction/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Peptides , R-SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Vesicle-Associated Membrane Protein 1/metabolism
12.
J Neurosci ; 30(21): 7377-91, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20505105

ABSTRACT

The continuous release of neurotransmitter could be seen to place a persistent burden on presynaptic proteins, one that could compromise nerve terminal function. This supposition and the molecular mechanisms that might protect highly active synapses merit investigation. In hippocampal cultures from knock-out mice lacking the presynaptic cochaperone cysteine string protein-alpha (CSP-alpha), we observe progressive degeneration of highly active synaptotagmin 2 (Syt2)-expressing GABAergic synapses, but surprisingly not of glutamatergic terminals. In CSP-alpha knock-out mice, synaptic degeneration of basket cell terminals occurs in vivo in the presence of normal glutamatergic synapses onto dentate gyrus granule cells. Consistent with this, in hippocampal cultures from these mice, the frequency of miniature IPSCs, caused by spontaneous GABA release, progressively declines, whereas the frequency of miniature excitatory AMPA receptor-mediated currents (mEPSCs), caused by spontaneous release of glutamate, is normal. However, the mEPSC amplitude progressively decreases. Remarkably, long-term block of glutamatergic transmission in cultures lacking CSP-alpha substantially rescues Syt2-expressing GABAergic synapses from neurodegeneration. These findings demonstrate that elevated neural activity increases synapse vulnerability and that CSP-alpha is essential to maintain presynaptic function under a physiologically high-activity regimen.


Subject(s)
Inhibitory Postsynaptic Potentials/physiology , Nerve Degeneration/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Age Factors , Animals , Animals, Newborn , Astrocytes/physiology , Bicuculline/pharmacology , Cells, Cultured , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Agents/pharmacology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Glutamic Acid/metabolism , HSP40 Heat-Shock Proteins/deficiency , Hippocampus/cytology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Membrane Proteins/deficiency , Mice , Mice, Knockout , Microscopy, Confocal/methods , Microscopy, Electron, Transmission/methods , Mutation/genetics , Nerve Degeneration/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Synapses/genetics , Synapses/ultrastructure
13.
Biochem Soc Trans ; 38(2): 488-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20298208

ABSTRACT

HD (Huntington's disease) is produced by the expression of mutant forms of the protein htt (huntingtin) containing a pathologically expanded poly-glutamine repeat. For unknown reasons, in HD patients and HD mouse models, neurons from the striatum and cerebral cortex degenerate and lead to motor dysfunction and dementia. Synaptic transmission in those neurons becomes progressively altered during the course of the disease. However, the relationship between synaptic dysfunction and neurodegeneration in HD is not yet clear. Are there early specific functional synaptic changes preceding symptoms and neurodegeneration? What is the role of those changes in neuronal damage? Recent experiments in a Drosophila model of HD have showed that abnormally increased neurotransmitter release might be a leading cause of neurodegeneration. In the present review, we summarize recently described synaptic alterations in HD animal models and discuss potential underlying molecular mechanisms.


Subject(s)
Huntington Disease/physiopathology , Presynaptic Terminals/physiology , Animals , Disease Models, Animal , Drosophila/physiology , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Nerve Net/physiopathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/physiology , Ubiquitin/metabolism
14.
J Neurosci ; 27(20): 5422-30, 2007 May 16.
Article in English | MEDLINE | ID: mdl-17507564

ABSTRACT

We monitored presynaptic exocytosis and vesicle recycling at neuromuscular junctions of transgenic mice expressing synaptopHluorin (spH), using simultaneous optical and electrophysiological recordings. Synaptic transmission was indistinguishable from that in wild-type controls. Fluorescence rose during and decayed monotonically after stimulus trains to the nerve, with amplitudes and decay times increasing with the amount of stimulation. The relatively large size of synaptic terminals allowed us to examine the spatial profile of fluorescence changes. We identified hot spots of exocytosis, which were stable with repeated trains. Photobleach experiments showed that spH freshly exposed by nerve stimulation was not preferentially retrieved by compensatory endocytosis; instead, most retrieved spH preexisted in the surface membrane. Finally, we compared fluorescence and electrical [summed end-plate potentials (EPPs)] estimates of exocytosis, which diverged during repeated trains, as fluorescence exceeded summed EPPs, although the average amplitude of miniature EPPs was unchanged. This might reflect exocytosis of spH-containing, acetylcholine-free ("empty") vesicles or other organelles during intense stimulation.


Subject(s)
Gene Expression Regulation/physiology , Green Fluorescent Proteins/biosynthesis , Neuromuscular Junction/physiology , Presynaptic Terminals/physiology , Recombinant Fusion Proteins/biosynthesis , Synaptic Transmission/physiology , Animals , Green Fluorescent Proteins/analysis , Mice , Mice, Transgenic , Neuromuscular Junction/chemistry , Presynaptic Terminals/chemistry , Recombinant Fusion Proteins/analysis , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism
15.
Proc Natl Acad Sci U S A ; 104(7): 2525-30, 2007 Feb 13.
Article in English | MEDLINE | ID: mdl-17287346

ABSTRACT

CASK is an evolutionarily conserved multidomain protein composed of an N-terminal Ca2+/calmodulin-kinase domain, central PDZ and SH3 domains, and a C-terminal guanylate kinase domain. Many potential activities for CASK have been suggested, including functions in scaffolding the synapse, in organizing ion channels, and in regulating neuronal gene transcription. To better define the physiological importance of CASK, we have now analyzed CASK "knockdown" mice in which CASK expression was suppressed by approximately 70%, and CASK knockout (KO) mice, in which CASK expression was abolished. CASK knockdown mice are viable but smaller than WT mice, whereas CASK KO mice die at first day after birth. CASK KO mice exhibit no major developmental abnormalities apart from a partially penetrant cleft palate syndrome. In CASK-deficient neurons, the levels of the CASK-interacting proteins Mints, Veli/Mals, and neurexins are decreased, whereas the level of neuroligin 1 (which binds to neurexins that in turn bind to CASK) is increased. Neurons lacking CASK display overall normal electrical properties and form ultrastructurally normal synapses. However, glutamatergic spontaneous synaptic release events are increased, and GABAergic synaptic release events are decreased in CASK-deficient neurons. In contrast to spontaneous neurotransmitter release, evoked release exhibited no major changes. Our data suggest that CASK, the only member of the membrane-associated guanylate kinase protein family that contains a Ca2+/calmodulin-dependent kinase domain, is required for mouse survival and performs a selectively essential function without being in itself required for core activities of neurons, such as membrane excitability, Ca2+-triggered presynaptic release, or postsynaptic receptor functions.


Subject(s)
Guanylate Kinases/physiology , Synapses/physiology , Animals , Gene Deletion , Glutamic Acid/metabolism , Guanylate Kinases/deficiency , Guanylate Kinases/genetics , Mice , Mice, Knockout , Neurons/cytology , Neurons/physiology , Survival , Synapses/metabolism
16.
Proc Natl Acad Sci U S A ; 103(8): 2926-31, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16477021

ABSTRACT

Cysteine string protein (CSP) alpha is an abundant synaptic vesicle protein that contains a DNA-J domain characteristic of Hsp40-type cochaperones. Previous studies showed that deletion of CSPalpha in mice leads to massive lethal neurodegeneration but did not clarify how the neurodegeneration affects specific subpopulations of neurons. Here, we analyzed the effects of the CSPalpha deficiency on tonically active ribbon synapses of the retina and the inner ear. We show that CSPalpha-deficient photoreceptor terminals undergo dramatic and rapidly progressive neurodegeneration that starts before eye opening and initially does not affect other retinal synapses. These changes are associated with progressive blindness. In contrast, ribbon synapses of auditory hair cells did not exhibit presynaptic impairments in CSPalpha-deficient mice. Hair cells, but not photoreceptor cells or central neurons, express CSPbeta, thereby accounting for the lack of a hair-cell phenotype in CSPalpha knockout mice. Our data demonstrate that tonically active ribbon synapses in retina are particularly sensitive to the deletion of CSPalpha and that expression of at least one CSP isoform is essential to protect such tonically active synapses from neurodegeneration.


Subject(s)
Blindness/pathology , Carrier Proteins/physiology , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/pathology , Photoreceptor Cells, Vertebrate/ultrastructure , Synapses/ultrastructure , Animals , Blindness/genetics , Blindness/metabolism , Carrier Proteins/genetics , HSP40 Heat-Shock Proteins/deficiency , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/physiology , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retina/pathology
17.
Cell ; 123(3): 383-96, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16269331

ABSTRACT

Alpha-synuclein and cysteine-string protein-alpha (CSPalpha) are abundant synaptic vesicle proteins independently linked to neurodegeneration. Dominantly inherited mutations in alpha-synuclein cause Parkinson's disease, but the physiological role of alpha-synuclein remains unknown. Deletion of CSPalpha produces rapidly progressive neurodegeneration in mice, presumably because the cochaperone function of CSPalpha is essential for neuronal survival. Here, we report the surprising finding that transgenic expression of alpha-synuclein abolishes the lethality and neurodegeneration caused by deletion of CSPalpha. Conversely, ablation of endogenous synucleins exacerbates these phenotypes. Deletion of CSPalpha inhibits SNARE complex assembly; transgenic alpha-synuclein ameliorates this inhibition. In preventing neurodegeneration in CSPalpha-deficient mice, alpha-synuclein does not simply substitute for CSPalpha but acts by a downstream mechanism that requires phospholipid binding by alpha-synuclein. These observations reveal a powerful in vivo activity of alpha-synuclein in protecting nerve terminals against injury and suggest that this activity operates in conjunction with CSPalpha and SNARE proteins on the presynaptic membrane interface.


Subject(s)
HSP40 Heat-Shock Proteins/metabolism , Nerve Degeneration/genetics , Synaptic Membranes/physiology , alpha-Synuclein/physiology , Animals , Brain/metabolism , Brain/pathology , HSP40 Heat-Shock Proteins/genetics , Mice , Mice, Knockout , Nerve Degeneration/pathology , Phospholipids/metabolism , SNARE Proteins/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Synaptic Membranes/genetics , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics , beta-Synuclein/genetics
18.
Brain Res Brain Res Rev ; 49(2): 416-28, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16054223

ABSTRACT

In neurons, a network of endocytic proteins accomplishes highly regulated processes such as synaptic vesicle cycling and the timely internalization of intracellular signaling molecules. In this review, we discuss recent advances on molecular networks created through interactions between proteins bearing the Eps15 homology (EH) domain and partner proteins containing the Asn-Pro-Phe (NPF) motif, which participate in important aspects of neuronal function as the synaptic vesicle cycle, the internalization of nerve growth factor (NGF), the determination of neuronal cell fate, the development of synapses and the trafficking of postsynaptic receptors. We discuss novel functional findings on the role of intersectin and synaptojanin and then we focus on the features of an emerging family of EH domain proteins termed EHDs (EH domain proteins), which are important for endocytic recycling of membrane proteins.


Subject(s)
EF Hand Motifs/physiology , Neurons/cytology , Phosphoproteins/metabolism , Sequence Homology, Amino Acid , Synapses/metabolism , Amino Acid Sequence , Animals , Conservation of Natural Resources , Endocytosis/physiology , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphoproteins/genetics
19.
Neuron ; 42(2): 237-51, 2004 Apr 22.
Article in English | MEDLINE | ID: mdl-15091340

ABSTRACT

Cysteine string protein alpha (CSPalpha)--an abundant synaptic vesicle protein that contains a DNA-J domain characteristic of Hsp40 chaperones--is thought to regulate Ca2+ channels and/or synaptic vesicle exocytosis. We now show that, in young mice, deletion of CSPalpha does not impair survival and causes no significant changes in presynaptic Ca2+ currents or synaptic vesicle exocytosis as measured in the Calyx of Held synapse. At 2-4 weeks of age, however, CSPalpha-deficient mice develop a progressive, fatal sensorimotor disorder. The neuromuscular junctions and Calyx synapses of CSPalpha-deficient mice exhibit increasing neurodegenerative changes, synaptic transmission becomes severely impaired, and the mutant mice die at approximately 2 months of age. Our data suggest that CSPalpha is not essential for the normal operation of Ca2+ channels or exocytosis but acts as a presynaptic chaperone that maintains continued synaptic function, raising the possibility that enhanced CSPalpha function could attenuate neurodegenerative diseases.


Subject(s)
Membrane Proteins/biosynthesis , Nerve Degeneration/metabolism , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/ultrastructure , HSP40 Heat-Shock Proteins , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Degeneration/genetics , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Presynaptic Terminals/ultrastructure , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure
20.
J Gen Physiol ; 122(3): 265-76, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12939392

ABSTRACT

We tested the long-standing hypothesis that synaptotagmin 1 is the Ca2+ sensor for fast neurosecretion by analyzing the intracellular Ca2+ dependence of large dense-core vesicle exocytosis in a mouse strain carrying a mutated synaptotagmin C2A domain. The mutation (R233Q) causes a twofold increase in the KD of Ca2+-dependent phospholipid binding to the double C2A-C2B domain of synaptotagmin. Using photolysis of caged calcium and capacitance measurements we found that secretion from mutant cells had lower secretory rates, longer secretory delays, and a higher intracellular Ca2+-threshold for secretion due to a twofold increase in the apparent KD of the Ca2+ sensor for fast exocytosis. Single amperometric fusion events were unchanged. We conclude that Ca2+-dependent phospholipid binding to synaptotagmin 1 mirrors the intracellular Ca2+ dependence of exocytosis.


Subject(s)
Calcium-Binding Proteins , Calcium/physiology , Exocytosis/physiology , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Secretory Vesicles/physiology , Animals , Arginine/genetics , Chromaffin Cells/metabolism , Differential Threshold , Electric Capacitance , Glutamine/genetics , Intracellular Membranes/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Phospholipids/metabolism , Point Mutation/genetics , Synaptotagmin I , Synaptotagmins , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...