Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.596
Filter
1.
J Neurosci ; 44(37)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39147589

ABSTRACT

The cerebellum plays an important role in diverse brain functions, ranging from motor learning to cognition. Recent studies have suggested that molecular and cellular heterogeneity within cerebellar lobules contributes to functional differences across the cerebellum. However, the specific relationship between molecular and cellular heterogeneity and diverse functional outputs of different regions of the cerebellum remains unclear. Here, we describe a previously unappreciated form of synaptic heterogeneity at parallel fiber synapses to Purkinje cells in the mouse cerebellum (both sexes). In contrast to uniform fast synaptic transmission, we found that the properties of slow synaptic transmission varied by up to threefold across different lobules of the mouse cerebellum, resulting in surprising heterogeneity. Depending on the location of a Purkinje cell, the time of peak of slow synaptic currents varied by hundreds of milliseconds. The duration and decay time of these currents also spanned hundreds of milliseconds, based on lobule. We found that, as a consequence of the heterogeneous synaptic dynamics, the same brief input stimulus was transformed into prolonged firing patterns over a range of timescales that depended on Purkinje cell location.


Subject(s)
Mice, Inbred C57BL , Purkinje Cells , Synaptic Transmission , Animals , Purkinje Cells/physiology , Mice , Synaptic Transmission/physiology , Male , Female , Cerebellum/physiology , Cerebellum/cytology , Excitatory Postsynaptic Potentials/physiology , Synapses/physiology , Time Factors , Action Potentials/physiology
2.
Sci Rep ; 14(1): 18226, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107382

ABSTRACT

Theory predicts that nonlinear summation of synaptic potentials within dendrites allows neurons to perform linearly non-separable computations (LNSCs). Using Boolean analysis approaches, we predicted that both supralinear and sublinear synaptic summation could allow single neurons to implement a type of LNSC, the feature binding problem (FBP), which does not require inhibition contrary to the exclusive-or function (XOR). Notably, sublinear dendritic operations enable LNSCs when scattered synaptic activation generates increased somatic spike output. However, experimental demonstrations of scatter-sensitive neuronal computations have not yet been described. Using glutamate uncaging onto cerebellar molecular layer interneurons, we show that scattered synaptic-like activation of dendrites evoked larger compound EPSPs than clustered synaptic activation, generating a higher output spiking probability. Moreover, we also demonstrate that single interneurons can indeed implement the FBP. Using a biophysical model to explore the conditions in which a neuron might be expected to implement the FBP, we establish that sublinear summation is necessary but not sufficient. Other parameters such as the relative sublinearity, the EPSP size, depolarization amplitude relative to action potential threshold, and voltage fluctuations all influence whether the FBP can be performed. Since sublinear synaptic summation is a property of passive dendrites, we expect that many different neuron types can implement LNSCs.


Subject(s)
Dendrites , Interneurons , Models, Neurological , Dendrites/physiology , Animals , Interneurons/physiology , Action Potentials/physiology , Excitatory Postsynaptic Potentials/physiology , Synapses/physiology , Cerebellum/physiology , Cerebellum/cytology , Neurons/physiology , Mice
3.
J Cell Biol ; 223(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39115447

ABSTRACT

Nuclear migration is critical for the proper positioning of neurons in the developing brain. It is known that bidirectional microtubule motors are required for nuclear transport, yet the mechanism of the coordination of opposing motors is still under debate. Using mouse cerebellar granule cells, we demonstrate that Nesprin-2 serves as a nucleus-motor adaptor, coordinating the interplay of kinesin-1 and dynein. Nesprin-2 recruits dynein-dynactin-BicD2 independently of the nearby kinesin-binding LEWD motif. Both motor binding sites are required to rescue nuclear migration defects caused by the loss of function of Nesprin-2. In an intracellular cargo transport assay, the Nesprin-2 fragment encompassing the motor binding sites generates persistent movements toward both microtubule minus and plus ends. Nesprin-2 drives bidirectional cargo movements over a prolonged period along perinuclear microtubules, which advance during the migration of neurons. We propose that Nesprin-2 keeps the nucleus mobile by coordinating opposing motors, enabling continuous nuclear transport along advancing microtubules in migrating cells.


Subject(s)
Cell Nucleus , Dyneins , Kinesins , Microtubule-Associated Proteins , Microtubules , Nerve Tissue Proteins , Neurons , Animals , Microtubules/metabolism , Neurons/metabolism , Kinesins/metabolism , Kinesins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Dyneins/metabolism , Cell Nucleus/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Active Transport, Cell Nucleus , Dynactin Complex/metabolism , Dynactin Complex/genetics , Cell Movement , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cerebellum/metabolism , Cerebellum/cytology , Binding Sites , Humans
4.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39137043

ABSTRACT

Primary cilia on granule cell neuron progenitors in the developing cerebellum detect sonic hedgehog to facilitate proliferation. Following differentiation, cerebellar granule cells become the most abundant neuronal cell type in the brain. While granule cell cilia are essential during early developmental stages, they become infrequent upon maturation. Here, we provide nanoscopic resolution of cilia in situ using large-scale electron microscopy volumes and immunostaining of mouse cerebella. In many granule cells, we found intracellular cilia, concealed from the external environment. Cilia were disassembled in differentiating granule cell neurons-in a process we call cilia deconstruction-distinct from premitotic cilia resorption in proliferating progenitors. In differentiating granule cells, cilia deconstruction involved unique disassembly intermediates, and, as maturation progressed, mother centriolar docking at the plasma membrane. Unlike ciliated neurons in other brain regions, our results show the deconstruction of concealed cilia in differentiating granule cells, which might prevent mitogenic hedgehog responsiveness. Ciliary deconstruction could be paradigmatic of cilia removal during differentiation in other tissues.


Subject(s)
Cell Differentiation , Cerebellum , Cilia , Hedgehog Proteins , Neurons , Cilia/metabolism , Cilia/ultrastructure , Animals , Neurons/metabolism , Neurons/cytology , Neurons/ultrastructure , Mice , Cerebellum/metabolism , Cerebellum/cytology , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Neurogenesis , Centrioles/metabolism , Centrioles/ultrastructure , Mice, Inbred C57BL
5.
Cell Rep Methods ; 4(7): 100816, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38981474

ABSTRACT

We developed a method that utilizes fluorescent labeling of nuclear envelopes alongside cytometry sorting for the selective isolation of Purkinje cell (PC) nuclei. Beginning with SUN1 reporter mice, we GFP-tagged envelopes to confirm that PC nuclei could be accurately separated from other cell types. We then developed an antibody-based protocol to make PC nuclear isolation more robust and adaptable to cerebellar tissues of any genotypic background. Immunofluorescent labeling of the nuclear membrane protein RanBP2 enabled the isolation of PC nuclei from C57BL/6 cerebellum. By analyzing the expression of PC markers, nuclear size, and nucleoli number, we confirmed that our method delivers a pure fraction of PC nuclei. To demonstrate its applicability, we isolated PC nuclei from spinocerebellar ataxia type 7 (SCA7) mice and identified transcriptional changes in known and new disease-associated genes. Access to pure PC nuclei offers insights into PC biology and pathology, including the nature of selective neuronal vulnerability.


Subject(s)
Mice, Inbred C57BL , Purkinje Cells , Animals , Purkinje Cells/metabolism , Mice , Cell Nucleus/metabolism , Cerebellum/metabolism , Cerebellum/cytology , Antibodies , GTP-Binding Proteins , Serine-Type D-Ala-D-Ala Carboxypeptidase
6.
Commun Biol ; 7(1): 806, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961250

ABSTRACT

Developmental synapse elimination is crucial for shaping mature neural circuits. In the neonatal mouse cerebellum, Purkinje cells (PCs) receive excitatory synaptic inputs from multiple climbing fibers (CFs) and synapses from all but one CF are eliminated by around postnatal day 20. Heterosynaptic interaction between CFs and parallel fibers (PFs), the axons of cerebellar granule cells (GCs) forming excitatory synapses onto PCs and molecular layer interneurons (MLIs), is crucial for CF synapse elimination. However, mechanisms for this heterosynaptic interaction are largely unknown. Here we show that deletion of AMPA-type glutamate receptor functions in GCs impairs CF synapse elimination mediated by metabotropic glutamate receptor 1 (mGlu1) signaling in PCs. Furthermore, CF synapse elimination is impaired by deleting NMDA-type glutamate receptors from MLIs. We propose that PF activity is crucial for CF synapse elimination by directly activating mGlu1 in PCs and indirectly enhancing the inhibition of PCs through activating NMDA receptors in MLIs.


Subject(s)
Cerebellum , Receptors, Metabotropic Glutamate , Synapses , Animals , Cerebellum/metabolism , Cerebellum/physiology , Cerebellum/cytology , Synapses/physiology , Synapses/metabolism , Mice , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/genetics , Purkinje Cells/metabolism , Purkinje Cells/physiology , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Interneurons/metabolism , Interneurons/physiology , Mice, Knockout , Mice, Inbred C57BL
7.
Cells ; 13(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38994990

ABSTRACT

In zebrafish, like in mammals, radial glial cells (RGCs) can act as neural progenitors during development and regeneration in adults. However, the heterogeneity of glia subpopulations entails the need for different specific markers of zebrafish glia. Currently, fluorescent protein expression mediated by a regulatory element from the glial fibrillary acidic protein (gfap) gene is used as a prominent glia reporter. We now expand this tool by demonstrating that a regulatory element from the mouse Fatty acid binding protein 7 (Fabp7) gene drives reliable expression in fabp7-expressing zebrafish glial cells. By using three different Fabp7 regulatory element-mediated fluorescent protein reporter strains, we reveal in double transgenic zebrafish that progenitor cells expressing fluorescent proteins driven by the Fabp7 regulatory element give rise to radial glia, oligodendrocyte progenitors, and some neuronal precursors. Furthermore, Bergmann glia represent the almost only glial population of the zebrafish cerebellum (besides a few oligodendrocytes), and the radial glia also remain in the mature cerebellum. Fabp7 regulatory element-mediated reporter protein expression in Bergmann glia progenitors suggests their origin from the ventral cerebellar proliferation zone, the ventricular zone, but not from the dorsally positioned upper rhombic lip. These new Fabp7 reporters will be valuable for functional studies during development and regeneration.


Subject(s)
Animals, Genetically Modified , Fatty Acid-Binding Protein 7 , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Fatty Acid-Binding Protein 7/metabolism , Fatty Acid-Binding Protein 7/genetics , Neuroglia/metabolism , Cerebellum/metabolism , Cerebellum/cytology , Oligodendroglia/metabolism , Oligodendroglia/cytology , Mice , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
8.
Nature ; 632(8027): 1092-1100, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048016

ABSTRACT

Placebo effects are notable demonstrations of mind-body interactions1,2. During pain perception, in the absence of any treatment, an expectation of pain relief can reduce the experience of pain-a phenomenon known as placebo analgesia3-6. However, despite the strength of placebo effects and their impact on everyday human experience and the failure of clinical trials for new therapeutics7, the neural circuit basis of placebo effects has remained unclear. Here we show that analgesia from the expectation of pain relief is mediated by rostral anterior cingulate cortex (rACC) neurons that project to the pontine nucleus (rACC→Pn)-a precerebellar nucleus with no established function in pain. We created a behavioural assay that generates placebo-like anticipatory pain relief in mice. In vivo calcium imaging of neural activity and electrophysiological recordings in brain slices showed that expectations of pain relief boost the activity of rACC→Pn neurons and potentiate neurotransmission in this pathway. Transcriptomic studies of Pn neurons revealed an abundance of opioid receptors, further suggesting a role in pain modulation. Inhibition of the rACC→Pn pathway disrupted placebo analgesia and decreased pain thresholds, whereas activation elicited analgesia in the absence of placebo conditioning. Finally, Purkinje cells exhibited activity patterns resembling those of rACC→Pn neurons during pain-relief expectation, providing cellular-level evidence for a role of the cerebellum in cognitive pain modulation. These findings open the possibility of targeting this prefrontal cortico-ponto-cerebellar pathway with drugs or neurostimulation to treat pain.


Subject(s)
Neural Pathways , Pain Perception , Pain , Placebo Effect , Animals , Female , Male , Mice , Analgesia , Anticipation, Psychological/physiology , Calcium Signaling , Cerebellum/cytology , Cerebellum/physiology , Cognition/physiology , Electrophysiology , Gene Expression Profiling , Gyrus Cinguli/cytology , Gyrus Cinguli/physiology , Mice, Inbred C57BL , Neurons/physiology , Pain/physiopathology , Pain/prevention & control , Pain/psychology , Pain Management/methods , Pain Management/psychology , Pain Management/trends , Pain Perception/physiology , Pain Threshold/physiology , Pain Threshold/psychology , Pons/cytology , Pons/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Purkinje Cells/physiology , Receptors, Opioid/metabolism , Synaptic Transmission
10.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38912572

ABSTRACT

The neurons of the three cerebellar nuclei (CN) are the primary output neurons of the cerebellum. The excitatory neurons (e) of the medial (m) CN (eCNm) were recently divided into molecularly defined subdomains in the adult; however, how they are established during development is not known. We define molecular subdomains of the mouse embryonic eCNm using single-cell RNA-sequencing and spatial expression analysis, showing that they evolve during embryogenesis to prefigure the adult. Furthermore, eCNm are transcriptionally divergent from cells in the other nuclei by embryonic day 14.5. We previously showed that loss of the homeobox genes En1 and En2 leads to loss of approximately half of the embryonic eCNm. We demonstrate that mutation of En1/2 in the embryonic eCNm results in death of specific posterior eCNm molecular subdomains and downregulation of TBR2 (EOMES) in an anterior embryonic subdomain, as well as reduced synaptic gene expression. We further reveal a similar function for EN1/2 in mediating TBR2 expression, neuron differentiation and survival in the other excitatory neurons (granule and unipolar brush cells). Thus, our work defines embryonic eCNm molecular diversity and reveals conserved roles for EN1/2 in the cerebellar excitatory neuron lineage.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins , Neurons , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Neurons/metabolism , Neurons/cytology , Cell Survival/genetics , Cell Differentiation/genetics , Cerebellum/embryology , Cerebellum/metabolism , Cerebellum/cytology , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Cerebellar Nuclei/metabolism , Cerebellar Nuclei/embryology , Cerebellar Nuclei/cytology , Single-Cell Analysis , Nerve Tissue Proteins
11.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891784

ABSTRACT

The central nervous system of Pacific salmon retains signs of embryonic structure throughout life and a large number of neuroepithelial neural stem cells (NSCs) in the proliferative areas of the brain, in particular. However, the adult nervous system and neurogenesis studies on rainbow trout, Oncorhynchus mykiss, are limited. Here, we studied the localization of glutamine synthetase (GS), vimentin (Vim), and nestin (Nes), as well as the neurons formed in the postembryonic period, labeled with doublecortin (DC), under conditions of homeostatic growth in adult cerebellum and brainstem of Oncorhynchus mykiss using immunohistochemical methods and Western Immunoblotting. We observed that the distribution of vimentin (Vim), nestin (Nes), and glutamine synthetase (GS), which are found in the aNSPCs of both embryonic types (neuroepithelial cells) and in the adult type (radial glia) in the cerebellum and the brainstem of trout, has certain features. Populations of the adult neural stem/progenitor cells (aNSPCs) expressing GS, Vim, and Nes have different morphologies, localizations, and patterns of cluster formation in the trout cerebellum and brainstem, which indicates the morphological and, obviously, functional heterogeneity of these cells. Immunolabeling of PCNA revealed areas in the cerebellum and brainstem of rainbow trout containing proliferating cells which coincide with areas expressing Vim, Nes, and GS. Double immunolabeling revealed the PCNA/GS PCNA/Vim coexpression patterns in the neuroepithelial-type cells in the PVZ of the brainstem. PCNA/GS coexpression in the RG was detected in the submarginal zone of the brainstem. The results of immunohistochemical study of the DC distribution in the cerebellum and brainstem of trout have showed a high level of expression of this marker in various cell populations. This may indicate: (i) high production of the adult-born neurons in the cerebellum and brainstem of adult trout, (ii) high plasticity of neurons in the cerebellum and brainstem of trout. We assume that the source of new cells in the trout brain, along with PVZ and SMZ, containing proliferating cells, may be local neurogenic niches containing the PCNA-positive and silent (PCNA-negative), but expressing NSC markers, cells. The identification of cells expressing DC, Vim, and Nes in the IX-X cranial nerve nuclei of trout was carried out.


Subject(s)
Brain Stem , Cerebellum , Neural Stem Cells , Neurogenesis , Neuronal Plasticity , Oncorhynchus mykiss , Animals , Oncorhynchus mykiss/metabolism , Oncorhynchus mykiss/growth & development , Cerebellum/metabolism , Cerebellum/cytology , Cerebellum/growth & development , Neurogenesis/physiology , Neuronal Plasticity/physiology , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Brain Stem/metabolism , Brain Stem/cytology , Vimentin/metabolism , Neurons/metabolism , Neurons/cytology , Proliferating Cell Nuclear Antigen/metabolism , Glutamate-Ammonia Ligase/metabolism
12.
J Neurosci ; 44(27)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38839301

ABSTRACT

Phospholipids (PLs) are asymmetrically distributed at the plasma membrane. This asymmetric lipid distribution is transiently altered during calcium-regulated exocytosis, but the impact of this transient remodeling on presynaptic function is currently unknown. As phospholipid scramblase 1 (PLSCR1) randomizes PL distribution between the two leaflets of the plasma membrane in response to calcium activation, we set out to determine its role in neurotransmission. We report here that PLSCR1 is expressed in cerebellar granule cells (GrCs) and that PLSCR1-dependent phosphatidylserine egress occurred at synapses in response to neuron stimulation. Synaptic transmission is impaired at GrC Plscr1 -/- synapses, and both PS egress and synaptic vesicle (SV) endocytosis are inhibited in Plscr1 -/- cultured neurons from male and female mice, demonstrating that PLSCR1 controls PL asymmetry remodeling and SV retrieval following neurotransmitter release. Altogether, our data reveal a novel key role for PLSCR1 in SV recycling and provide the first evidence that PL scrambling at the plasma membrane is a prerequisite for optimal presynaptic performance.


Subject(s)
Cerebellum , Phospholipid Transfer Proteins , Synapses , Synaptic Transmission , Synaptic Vesicles , Animals , Synaptic Vesicles/metabolism , Synaptic Transmission/physiology , Mice , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/genetics , Female , Male , Cerebellum/cytology , Synapses/metabolism , Synapses/physiology , Cells, Cultured , Mice, Knockout , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Endocytosis/physiology
13.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38860486

ABSTRACT

Cerebellar granule neuron progenitors (GNPs) originate from the upper rhombic lip (URL), a germinative niche in which developmental defects produce human diseases. T-cell factor (TCF) responsiveness and Notch dependence are hallmarks of self-renewal in neural stem cells. TCF activity, together with transcripts encoding proneural gene repressors hairy and enhancer of split (Hes/Hey), are detected in the URL; however, their functions and regulatory modes are undeciphered. Here, we established amphibian as a pertinent model for studying vertebrate URL development. The amphibian long-lived URL is TCF active, whereas the external granular layer (EGL) is non-proliferative and expresses hes4 and hes5 genes. Using functional and transcriptomic approaches, we show that TCF activity is necessary for URL emergence and maintenance. We establish that the transcription factor Barhl1 controls GNP exit from the URL, acting partly through direct TCF inhibition. Identification of Barhl1 target genes suggests that, besides TCF, Barhl1 inhibits transcription of hes5 genes independently of Notch signaling. Observations in amniotes suggest a conserved role for Barhl in maintenance of the URL and/or EGL via co-regulation of TCF, Hes and Hey genes.


Subject(s)
Cerebellum , Neural Stem Cells , Animals , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Cerebellum/cytology , Cerebellum/metabolism , Gene Expression Regulation, Developmental , Neurons/metabolism , Neurons/cytology , Receptors, Notch/metabolism , Receptors, Notch/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Rhombencephalon/metabolism , Rhombencephalon/cytology , Stem Cell Niche , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics
14.
Neuron ; 112(16): 2749-2764.e7, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-38870929

ABSTRACT

In classical cerebellar learning, Purkinje cells (PkCs) associate climbing fiber (CF) error signals with predictive granule cells (GrCs) that were active just prior (∼150 ms). The cerebellum also contributes to behaviors characterized by longer timescales. To investigate how GrC-CF-PkC circuits might learn seconds-long predictions, we imaged simultaneous GrC-CF activity over days of forelimb operant conditioning for delayed water reward. As mice learned reward timing, numerous GrCs developed anticipatory activity ramping at different rates until reward delivery, followed by widespread time-locked CF spiking. Relearning longer delays further lengthened GrC activations. We computed CF-dependent GrC→PkC plasticity rules, demonstrating that reward-evoked CF spikes sufficed to grade many GrC synapses by anticipatory timing. We predicted and confirmed that PkCs could thereby continuously ramp across seconds-long intervals from movement to reward. Learning thus leads to new GrC temporal bases linking predictors to remote CF reward signals-a strategy well suited for learning to track the long intervals common in cognitive domains.


Subject(s)
Cerebellum , Learning , Purkinje Cells , Reward , Animals , Cerebellum/physiology , Cerebellum/cytology , Mice , Purkinje Cells/physiology , Learning/physiology , Conditioning, Operant/physiology , Male , Mice, Inbred C57BL , Nerve Fibers/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Time Factors , Action Potentials/physiology
15.
Nat Commun ; 15(1): 4645, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821918

ABSTRACT

Non-synaptic (intrinsic) plasticity of membrane excitability contributes to aspects of memory formation, but it remains unclear whether it merely facilitates synaptic long-term potentiation or plays a permissive role in determining the impact of synaptic weight increase. We use tactile stimulation and electrical activation of parallel fibers to probe intrinsic and synaptic contributions to receptive field plasticity in awake mice during two-photon calcium imaging of cerebellar Purkinje cells. Repetitive activation of both stimuli induced response potentiation that is impaired in mice with selective deficits in either synaptic or intrinsic plasticity. Spatial analysis of calcium signals demonstrated that intrinsic, but not synaptic plasticity, enhances the spread of dendritic parallel fiber response potentiation. Simultaneous dendrite and axon initial segment recordings confirm these dendritic events affect axonal output. Our findings support the hypothesis that intrinsic plasticity provides an amplification mechanism that exerts a permissive control over the impact of long-term potentiation on neuronal responsiveness.


Subject(s)
Cerebellum , Dendrites , Long-Term Potentiation , Neuronal Plasticity , Purkinje Cells , Synapses , Animals , Purkinje Cells/physiology , Mice , Neuronal Plasticity/physiology , Cerebellum/physiology , Cerebellum/cytology , Long-Term Potentiation/physiology , Dendrites/physiology , Synapses/physiology , Calcium/metabolism , Male , Axons/physiology , Mice, Inbred C57BL , Electric Stimulation , Female
16.
Nat Commun ; 15(1): 4003, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734715

ABSTRACT

Accurate perception and behavior rely on distinguishing sensory signals arising from unexpected events from those originating from our own voluntary actions. In the vestibular system, sensory input that is the consequence of active self-motion is canceled early at the first central stage of processing to ensure postural and perceptual stability. However, the source of the required cancellation signal was unknown. Here, we show that the cerebellum combines sensory and motor-related information to predict the sensory consequences of active self-motion. Recordings during attempted but unrealized head movements in two male rhesus monkeys, revealed that the motor-related signals encoded by anterior vermis Purkinje cells explain their altered sensitivity to active versus passive self-motion. Further, a model combining responses from ~40 Purkinje cells accounted for the cancellation observed in early vestibular pathways. These findings establish how cerebellar Purkinje cells predict sensory outcomes of self-movements, resolving a long-standing issue of sensory signal suppression during self-motion.


Subject(s)
Macaca mulatta , Purkinje Cells , Animals , Purkinje Cells/physiology , Male , Head Movements/physiology , Cerebellum/physiology , Cerebellum/cytology , Vestibule, Labyrinth/physiology , Motion Perception/physiology
17.
STAR Protoc ; 5(2): 102936, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38735042

ABSTRACT

GABAergic interneurons are inhibitory neurons of the CNS, playing a fundamental role in neural circuitry and activity. Here, we provide a robust protocol for the successful enrichment of human cerebellar GABAergic interneurons from human induced pluripotent stem cells (iPSCs) and measuring intracellular calcium transients. We describe in detail steps for culturing iPSCs; generating embryoid bodies; and differentiating and enriching for cerebellar GABAergic neurons (cGNs), with precise steps for their molecular characterization. We then detail the procedure for adeno-associated virus-mediated transduction of cGNs with genetically encoded calcium indicators, followed by intracellular calcium imaging and analyses. For complete details on the use and execution of this protocol, please refer to Pilotto et al.1.


Subject(s)
Calcium , Cell Differentiation , Cerebellum , GABAergic Neurons , Induced Pluripotent Stem Cells , Interneurons , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Humans , Calcium/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/cytology , Interneurons/metabolism , Interneurons/cytology , Cell Differentiation/physiology , Cerebellum/cytology , Cerebellum/metabolism , Cell Culture Techniques/methods , Cells, Cultured
18.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38733989

ABSTRACT

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Subject(s)
Cerebellum , Neurons , Retina , Animals , Female , Male , Mice , Cerebellum/metabolism , Cerebellum/blood supply , Cerebellum/cytology , Ion Channels/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/metabolism , Retinal Vessels/metabolism
19.
Neuron ; 112(14): 2333-2348.e6, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38692278

ABSTRACT

Molecular layer interneurons (MLIs) account for approximately 80% of the inhibitory interneurons in the cerebellar cortex and are vital to cerebellar processing. MLIs are thought to primarily inhibit Purkinje cells (PCs) and suppress the plasticity of synapses onto PCs. MLIs also inhibit, and are electrically coupled to, other MLIs, but the functional significance of these connections is not known. Here, we find that two recently recognized MLI subtypes, MLI1 and MLI2, have a highly specialized connectivity that allows them to serve distinct functional roles. MLI1s primarily inhibit PCs, are electrically coupled to each other, fire synchronously with other MLI1s on the millisecond timescale in vivo, and synchronously pause PC firing. MLI2s are not electrically coupled, primarily inhibit MLI1s and disinhibit PCs, and are well suited to gating cerebellar-dependent behavior and learning. The synchronous firing of electrically coupled MLI1s and disinhibition provided by MLI2s require a major re-evaluation of cerebellar processing.


Subject(s)
Interneurons , Neural Inhibition , Purkinje Cells , Animals , Purkinje Cells/physiology , Interneurons/physiology , Neural Inhibition/physiology , Mice , Cerebellum/cytology , Cerebellum/physiology , Mice, Transgenic , Action Potentials/physiology , Mice, Inbred C57BL , Cerebellar Cortex/physiology , Cerebellar Cortex/cytology
20.
J Mol Neurosci ; 74(2): 44, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630337

ABSTRACT

Plants are a valuable source of information for pharmacological research and new drug discovery. The present study aimed to evaluate the neuroprotective potential of the leaves of the medicinal plant Sterculia setigera. In vitro, the effect of Sterculia setigera leaves dry hydroethanolic extract (SSE) was tested on cultured cerebellar granule neurons (CGN) survival when exposed to hydrogen peroxide (H2O2) or 6-hydroxydopamine (6-OHDA), using the viability probe fluorescein diacetate (FDA), a lactate dehydrogenase (LDH) activity assay, an immunocytochemical staining against Gap 43, and the quantification of the expression of genes involved in apoptosis, necrosis, or oxidative stress. In vivo, the effect of intraperitoneal (ip) injection of SSE was assessed on the developing brain of 8-day-old Wistar rats exposed to ethanol neurotoxicity by measuring caspase-3 activity on cerebellum homogenates, the expression of some genes in tissue extracts, the thickness of cerebellar cortical layers and motor coordination. In vitro, SSE protected CGN against H2O2 and 6-OHDA-induced cell death at a dose of 10 µg/mL, inhibited the expression of genes Casp3 and Bad, and upregulated the expression of Cat and Gpx7. In vivo, SSE significantly blocked the deleterious effect of ethanol by reducing the activity of caspase-3, inhibiting the expression of Bax and Tp53, preventing the reduction of the thickness of the internal granule cell layer of the cerebellar cortex, and restoring motor functions. Sterculia setigera exerts neuroactive functions as claimed by traditional medicine and should be a good candidate for the development of a neuroprotective treatment against neurodegenerative diseases.


Subject(s)
Cell Death , Ethanol , Neurons , Neuroprotective Agents , Plant Extracts , Plant Leaves , Sterculia , Animals , Rats , Caspase 3/metabolism , Ethanol/administration & dosage , Ethanol/chemistry , Ethanol/toxicity , Hydrogen Peroxide/toxicity , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Rats, Wistar , Sterculia/chemistry , Plant Leaves/chemistry , Plants, Medicinal/chemistry , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Lactate Dehydrogenases/metabolism , GAP-43 Protein/analysis , Apoptosis/genetics , Oxidative Stress/genetics , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/pathology , Cerebellum/physiology , Male , Female , Cells, Cultured , Cell Death/drug effects , Gene Expression Regulation/drug effects , Phytochemicals/administration & dosage , Phytochemicals/analysis , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/pharmacology , Antioxidants/analysis , Antioxidants/chemistry , Antioxidants/pharmacology , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Liquid Chromatography-Mass Spectrometry , Secondary Metabolism
SELECTION OF CITATIONS
SEARCH DETAIL