Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
Science ; 383(6687): eadg6757, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38452088

ABSTRACT

The hippocampal mossy fiber synapse, formed between axons of dentate gyrus granule cells and dendrites of CA3 pyramidal neurons, is a key synapse in the trisynaptic circuitry of the hippocampus. Because of its comparatively large size, this synapse is accessible to direct presynaptic recording, allowing a rigorous investigation of the biophysical mechanisms of synaptic transmission and plasticity. Furthermore, because of its placement in the very center of the hippocampal memory circuit, this synapse seems to be critically involved in several higher network functions, such as learning, memory, pattern separation, and pattern completion. Recent work based on new technologies in both nanoanatomy and nanophysiology, including presynaptic patch-clamp recording, paired recording, super-resolution light microscopy, and freeze-fracture and "flash-and-freeze" electron microscopy, has provided new insights into the structure, biophysics, and network function of this intriguing synapse. This brings us one step closer to answering a fundamental question in neuroscience: how basic synaptic properties shape higher network computations.


Subject(s)
Mossy Fibers, Hippocampal , Presynaptic Terminals , Mossy Fibers, Hippocampal/physiology , Mossy Fibers, Hippocampal/ultrastructure , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Synaptic Transmission , CA3 Region, Hippocampal , Pyramidal Cells , Humans , Animals
2.
Cell Rep ; 40(12): 111382, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130490

ABSTRACT

Rab3A-interacting molecule (RIM) is crucial for fast Ca2+-triggered synaptic vesicle (SV) release in presynaptic active zones (AZs). We investigated hippocampal giant mossy fiber bouton (MFB) AZ architecture in 3D using electron tomography of rapid cryo-immobilized acute brain slices in RIM1α-/- and wild-type mice. In RIM1α-/-, AZs are larger with increased synaptic cleft widths and a 3-fold reduced number of tightly docked SVs (0-2 nm). The distance of tightly docked SVs to the AZ center is increased from 110 to 195 nm, and the width of their electron-dense material between outer SV membrane and AZ membrane is reduced. Furthermore, the SV pool in RIM1α-/- is more heterogeneous. Thus, RIM1α, besides its role in tight SV docking, is crucial for synaptic architecture and vesicle pool organization in MFBs.


Subject(s)
Synapses , Synaptic Vesicles , Animals , Mice , Mossy Fibers, Hippocampal/ultrastructure , Presynaptic Terminals/ultrastructure , Synapses/ultrastructure , Synaptic Transmission , Synaptic Vesicles/ultrastructure
3.
PLoS Biol ; 19(6): e3001149, 2021 06.
Article in English | MEDLINE | ID: mdl-34153028

ABSTRACT

Synaptic plasticity is a cellular model for learning and memory. However, the expression mechanisms underlying presynaptic forms of plasticity are not well understood. Here, we investigate functional and structural correlates of presynaptic potentiation at large hippocampal mossy fiber boutons induced by the adenylyl cyclase activator forskolin. We performed 2-photon imaging of the genetically encoded glutamate sensor iGluu that revealed an increase in the surface area used for glutamate release at potentiated terminals. Time-gated stimulated emission depletion microscopy revealed no change in the coupling distance between P/Q-type calcium channels and release sites mapped by Munc13-1 cluster position. Finally, by high-pressure freezing and transmission electron microscopy analysis, we found a fast remodeling of synaptic ultrastructure at potentiated boutons: Synaptic vesicles dispersed in the terminal and accumulated at the active zones, while active zone density and synaptic complexity increased. We suggest that these rapid and early structural rearrangements might enable long-term increase in synaptic strength.


Subject(s)
Mossy Fibers, Hippocampal/metabolism , Presynaptic Terminals/metabolism , Animals , Colforsin/pharmacology , Glutamic Acid/metabolism , Male , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/ultrastructure , Neurotransmitter Agents/metabolism , Presynaptic Terminals/drug effects , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism
4.
Elife ; 102021 06 01.
Article in English | MEDLINE | ID: mdl-34061025

ABSTRACT

Neurotransmitter release is a highly controlled process by which synapses can critically regulate information transfer within neural circuits. While presynaptic receptors - typically activated by neurotransmitters and modulated by neuromodulators - provide a powerful way of fine-tuning synaptic function, their contribution to activity-dependent changes in transmitter release remains poorly understood. Here, we report that presynaptic NMDA receptors (preNMDARs) at mossy fiber boutons in the rodent hippocampus can be activated by physiologically relevant patterns of activity and selectively enhance short-term synaptic plasticity at mossy fiber inputs onto CA3 pyramidal cells and mossy cells, but not onto inhibitory interneurons. Moreover, preNMDARs facilitate brain-derived neurotrophic factor release and contribute to presynaptic calcium rise. Taken together, our results indicate that by increasing presynaptic calcium, preNMDARs fine-tune mossy fiber neurotransmission and can control information transfer during dentate granule cell burst activity that normally occur in vivo.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Mossy Fibers, Hippocampal/metabolism , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , Animals , CA3 Region, Hippocampal/metabolism , Calcium/metabolism , Calcium Signaling , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Mossy Fibers, Hippocampal/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/metabolism , Pyramidal Cells/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/ultrastructure , Time Factors
5.
Science ; 372(6539)2021 04 16.
Article in English | MEDLINE | ID: mdl-33859005

ABSTRACT

Protocadherin-19 (PCDH19) mutations cause early-onset seizures and cognitive impairment. The PCDH19 gene is on the X-chromosome. Unlike most X-linked disorders, PCDH19 mutations affect heterozygous females (PCDH19HET♀ ) but not hemizygous males (PCDH19HEMI♂ ); however, the reason why remains to be elucidated. We demonstrate that PCDH19, a cell-adhesion molecule, is enriched at hippocampal mossy fiber synapses. Pcdh19HET♀ but not Pcdh19HEMI♂ mice show impaired mossy fiber synaptic structure and physiology. Consistently, Pcdh19HET♀ but not Pcdh19HEMI♂ mice exhibit reduced pattern completion and separation abilities, which require mossy fiber synaptic function. Furthermore, PCDH19 appears to interact with N-cadherin at mossy fiber synapses. In Pcdh19HET♀ conditions, mismatch between PCDH19 and N-cadherin diminishes N-cadherin-dependent signaling and impairs mossy fiber synapse development; N-cadherin overexpression rescues Pcdh19HET♀ phenotypes. These results reveal previously unknown molecular and cellular mechanisms underlying the female-specific PCDH19 disorder phenotype.


Subject(s)
Cadherins/metabolism , Cognitive Dysfunction/physiopathology , Genetic Diseases, X-Linked/physiopathology , Mossy Fibers, Hippocampal/physiopathology , Synapses/physiology , Animals , CA3 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/ultrastructure , Cadherins/genetics , Cognitive Dysfunction/genetics , Disease Models, Animal , Epilepsy/genetics , Epilepsy/physiopathology , Female , Genes, X-Linked , Genetic Diseases, X-Linked/genetics , Long-Term Potentiation , Male , Mice , Mossy Fibers, Hippocampal/ultrastructure , Mutation , Protocadherins , Sex Characteristics , Synapses/ultrastructure , beta Catenin/metabolism
6.
PLoS One ; 15(10): e0240610, 2020.
Article in English | MEDLINE | ID: mdl-33049001

ABSTRACT

Presynaptic mitochondrial Ca2+ plays a critical role in the regulation of synaptic transmission and plasticity. The presynaptic bouton of the hippocampal mossy fiber (MF) is much larger in size than that of the Schaffer collateral (SC) synapse. Here we compare the structural and physiological characteristics of MF and SC presynaptic boutons to reveal functional and mechanistic differences between these two synapses. Our quantitative ultrastructural analysis using electron microscopy show many more mitochondria in MF presynaptic bouton cross-section profiles compared to SC boutons. Consistent with these results, post-tetanic potentiation (PTP), a form of presynaptic short-term plasticity dependent on mitochondrial Ca2+, is reduced by inhibition of mitochondrial Ca2+ release at MF synapses but not at SC synapses. However, blockade of mitochondrial Ca2+ release results in reduction of PTP at SC synapses by disynaptic MF stimulation. Furthermore, inhibition of mitochondrial Ca2+ release selectively decreases frequency facilitation evoked by short trains of presynaptic stimulation at MF synapses, while having no effect at SC synapses. Moreover, depletion of ER Ca2+ stores leads to reduction of PTP at MF synapses, but PTP is unaffected by ER Ca2+ depletion at SC synapses. These findings show that MF and SC synapses differ in presynaptic mitochondrial content as well as mitochondrial Ca2+ dependent synaptic plasticity, highlighting differential regulatory mechanisms of presynaptic plasticity at MF and SC synapses.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Mossy Fibers, Hippocampal/metabolism , Neuronal Plasticity/physiology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/ultrastructure , Male , Mice , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/metabolism , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/ultrastructure , Neuronal Plasticity/drug effects , Onium Compounds/pharmacology , Organophosphorus Compounds/pharmacology , Patch-Clamp Techniques , Thiazepines/pharmacology
7.
Neuron ; 107(3): 509-521.e7, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32492366

ABSTRACT

Post-tetanic potentiation (PTP) is an attractive candidate mechanism for hippocampus-dependent short-term memory. Although PTP has a uniquely large magnitude at hippocampal mossy fiber-CA3 pyramidal neuron synapses, it is unclear whether it can be induced by natural activity and whether its lifetime is sufficient to support short-term memory. We combined in vivo recordings from granule cells (GCs), in vitro paired recordings from mossy fiber terminals and postsynaptic CA3 neurons, and "flash and freeze" electron microscopy. PTP was induced at single synapses and showed a low induction threshold adapted to sparse GC activity in vivo. PTP was mainly generated by enlargement of the readily releasable pool of synaptic vesicles, allowing multiplicative interaction with other plasticity forms. PTP was associated with an increase in the docked vesicle pool, suggesting formation of structural "pool engrams." Absence of presynaptic activity extended the lifetime of the potentiation, enabling prolonged information storage in the hippocampal network.


Subject(s)
Memory, Short-Term/physiology , Mossy Fibers, Hippocampal/metabolism , Neuronal Plasticity/physiology , Pyramidal Cells/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Action Potentials/physiology , Animals , CA3 Region, Hippocampal/cytology , Dentate Gyrus/cytology , Mice , Microscopy, Electron , Mossy Fibers, Hippocampal/physiology , Mossy Fibers, Hippocampal/ultrastructure , Patch-Clamp Techniques , Pyramidal Cells/physiology , Pyramidal Cells/ultrastructure , Rats , Synapses/physiology , Synaptic Potentials/physiology
8.
Cell Rep ; 30(11): 3632-3643.e8, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187536

ABSTRACT

Although similar in molecular composition, synapses can exhibit strikingly distinct functional transmitter release and plasticity characteristics. To determine whether ultrastructural differences co-define this functional heterogeneity, we combine hippocampal organotypic slice cultures, high-pressure freezing, freeze substitution, and 3D-electron tomography to compare two functionally distinct synapses: hippocampal Schaffer collateral and mossy fiber synapses. We find that mossy fiber synapses, which exhibit a lower release probability and stronger short-term facilitation than Schaffer collateral synapses, harbor lower numbers of docked synaptic vesicles at active zones and a second pool of possibly tethered vesicles in their vicinity. Our data indicate that differences in the ratio of docked versus tethered vesicles at active zones contribute to distinct functional characteristics of synapses.


Subject(s)
Hippocampus/physiology , Hippocampus/ultrastructure , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Synapses/physiology , Synapses/ultrastructure , Animals , Cyclic AMP/metabolism , Excitatory Postsynaptic Potentials , Mice, Inbred C57BL , Mice, Knockout , Mossy Fibers, Hippocampal/physiology , Mossy Fibers, Hippocampal/ultrastructure , Neurotransmitter Agents/metabolism , Organ Culture Techniques , Secretory Vesicles/physiology , Secretory Vesicles/ultrastructure , Synaptic Vesicles/ultrastructure , Tissue Fixation
9.
eNeuro ; 6(2)2019.
Article in English | MEDLINE | ID: mdl-30937358

ABSTRACT

Hilar mossy cells in the dentate gyrus (DG) shape the firing and function of the hippocampal circuit. However, the neural circuitry providing afferent input to mossy cells is incompletely understood, and little is known about the development of these inputs. Thus, we used whole-cell recording and laser scanning photostimulation (LSPS) to characterize the developmental trajectory of local excitatory and inhibitory synaptic inputs to mossy cells in the mouse hippocampus. Hilar mossy cells were targeted by visualizing non-red fluorescent cells in the dentate hilus of GAD2-Cre; Ai9 mice that expressed tdTomato in GAD+ neurons, and were confirmed by post hoc morphological characterization. Our results show that at postnatal day (P)6-P7, mossy cells received more excitatory input from neurons in the proximal CA3 versus those in the DG. In contrast, at P13-P14 and P21-P28, the largest source of excitatory input originated in DG cells, while the strength of CA3 and hilar inputs declined. A developmental trend was also evident for inhibitory inputs. Overall inhibitory input at P6-P7 was weak, while inhibitory inputs from the DG cell layer and the hilus predominated at P13-P14 and P21-P28. The strength of local DG excitation and inhibition to mossy cells peaked at P13-P14 and decreased slightly in older P21-P28 mice. Together, these data provide new detailed information on the development of local synaptic connectivity of mossy cells, and suggests mechanisms through which developmental changes in local circuit inputs to hilar mossy cells shape their physiology and vulnerability to injury during postnatal periods.


Subject(s)
Mossy Fibers, Hippocampal/physiology , Mossy Fibers, Hippocampal/ultrastructure , Neural Pathways/physiology , Neural Pathways/ultrastructure , Neurogenesis/physiology , Animals , Female , Male , Mice , Synapses/physiology , Synapses/ultrastructure , Synaptic Transmission/physiology
10.
Radiología (Madr., Ed. impr.) ; 60(5): 404-412, sept.-oct. 2018. ilus, tab
Article in Spanish | IBECS | ID: ibc-175301

ABSTRACT

Objetivo: Evaluar las subestructuras hipocampales utilizando resonancia magnética en pacientes con esclerosis hipocampal (EH), comparando los resultados con el análisis morfológico y la volumetría global del hipocampo. Método: Se incluyeron 25 controles y 25 pacientes con EH, cuyo diagnóstico fue extraído del informe de la junta institucional de epilepsia. Se utilizó FreeSurfer para el procesamiento de los estudios y la obtención de los datos volumétricos. El volumen fue valorado de manera global y por subestructura: fimbria, subiculum, presubiculum, fisura hipocampal, CA1, CA2-CA3, CA4 y giro dentado (GD). Se consideró p <0,05 como estadísticamente significativo. Resultados: Se observó una disminución estadísticamente significativa en el hipocampo homolateral al foco epileptógeno en 19 de los 25 casos (76,0%). A excepción de la fisura hipocampal, se observó una disminución en todas las subestructuras hipocampales homolaterales en la EH derecha (CA1, p = 0,0223; CA2-CA3, p = 0,0066; CA4-GD, p = 0,0066; fimbria, p = 0,0046; presubiculum, p = 0,0087; subiculum, p = 0,0017) y la EH izquierda (CA1, p <0,0001; CA2-CA3, p <0,0001; CA4-GD, p <0,0001; fimbria, p = 0,0183; presubiculum, p <0,0001; subiculum, p <0,0001). En cuatro casos de EH izquierda, ninguna de las subestructuras presentó alteración estadísticamente significativa; sin embargo, se observó una tendencia de atrofia, principalmente en CA2-CA3 y CA4-GD. Conclusión: Los hallazgos sugieren la utilidad de la evaluación de las subestructuras hipocampales para mejorar el desempeño de la imagen en el diagnóstico de la EH


Objective: The pathological classification of hippocampal sclerosis is based on the loss of neurons in the substructures of the hippocampus. This study aimed to evaluate these substructures in patients with hippocampal sclerosis by magnetic resonance imaging and to compare the usefulness of this morphological analysis compared to that of volumetric analysis of the entire hippocampus. Material and methods: We included 25 controls and 25 patients with hippocampal sclerosis whose diagnosis was extracted from the institutional epilepsy board. We used FreeSurfer to process the studies and obtain the volumetric data. We evaluated overall volume and volume by substructure: fimbria, subiculum, presubiculum, hippocampal sulcus, CA1, CA2-CA3, CA4, and dentate gyrus (DG). We considered p < 0.05 statistically significant. Results: We observed statistically significant decreases in the volume of the hippocampus ipsilateral to the epileptogenic focus in 19 (76.0%) of the 25 cases. With the exception of the hippocampal sulcus, we observed a decrease in all ipsilateral hippocampal substructures in patients with right hippocampal sclerosis (CA1, p=0.0223; CA2-CA3, p=0.0066; CA4-GD, p=0.0066; fimbria, p=0.0046; presubiculum, p=0.0087; subiculum, p=0.0017) and in those with left hippocampal sclerosis (CA1, p<0.0001; CA2-CA3, p<0. 0001; CA4-GD, p<0. 0001; fimbria, p=0.0183; presubiculum, p<0. 0001; subiculum, p<0. 0001). In four patients with left hippocampal sclerosis, none of the substructures had statistically significant alterations, although a trend toward atrophy was observed, mainly in CA2-CA3 and CA4-GD. Conclusion: The findings suggest that it can be useful to assess the substructures of the hippocampus to improve the performance of diagnostic imaging in patients with hippocampal sclerosis


Subject(s)
Humans , Male , Female , Young Adult , Adult , Middle Aged , Hippocampus/ultrastructure , Sclerosis/diagnostic imaging , Epilepsy/diagnostic imaging , Case-Control Studies , Mossy Fibers, Hippocampal/ultrastructure , Parahippocampal Gyrus/ultrastructure , Magnetic Resonance Imaging/methods , Retrospective Studies
11.
Biochem Biophys Res Commun ; 503(4): 2312-2318, 2018 09 18.
Article in English | MEDLINE | ID: mdl-29964011

ABSTRACT

Mossy fiber sprouting (MFS) is a pathological phenomenon that is commonly observed in epilepsy, and plentiful data reveal that abnormal phosphorylated modification of tau protein plays a critical role in MSF by the regulation of microtubule dynamics and axonal transport. Ubiquitin C-terminal hydrolase L1 (UCH-L1), a proteasomal deubiquitinating enzyme, has been proved to be associated with tau aggregation through mediating degradation of ubiquitinated and hyperphosphorylated tau. Thus, this study aimed to determine the expression of UCH-L1 in the rat hippocampus during the pentylenetetrazole (PTZ)-induced process and to demonstrate the possible correlation with MFS in epileptogenesis. Seizures were established by intraperitoneal injection of PTZ and LDN-57444 was used to inhibit the hydrolase activity of UCH-L1. We used western blot, immunofluorescence, immunoprecipitation, and timm staining to detect phosphorylated modification of tau and MSF. The results presented that LDN-57444 induced the deteriorated severity of seizures, increased phosphorylation of tau and increased distribution of Timm granules in both the supragranular region of the dentate gyrus (DG) and the stratum pyramidale of CA3 subfield. Our results suggest that UCH-L1 may be associated with hippocampal MSF followed the epileptogenesis through mediating phosphorylation of tau. UCH-L1 may be a potential and novel therapeutic target to limit epileptogenesis.


Subject(s)
Kindling, Neurologic/physiology , Mossy Fibers, Hippocampal/ultrastructure , Pentylenetetrazole/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Animals , Epilepsy/chemically induced , Phosphorylation , Rats , tau Proteins/metabolism
12.
Neurosci Lett ; 677: 65-71, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29689341

ABSTRACT

Hippocampal mossy fibers (MFs) project from dentate gyrus granule cells onto the CA2-CA3 region. MF-mediated synaptic transmission plays an important role in hippocampal learning and memory. However, the molecular mechanisms underlying MF synaptic development and subsequent functional organization are not fully understood. We previously reported that calcium-dependent activator protein for secretion 2 (CADPS2, also known as CAPS2) regulates the secretion of dense-core vesicles (DCVs). Because CADPS2 is strongly expressed in MF terminals, we hypothesized that CADPS2 regulates the development and functional organization of MF synapses by controlling the secretion of DCVs and their contents. To test this, we compared the synaptic microstructures of hippocampal MF terminals in Cadps2 knockout (KO) mice and wild-type (WT) mice by electron microscopy (EM). On postnatal day 15 (P15), KO mice exhibited morphological abnormalities in MF boutons, including smaller bouton size, a larger number of DCVs and a smaller number of post-synaptic densities (PSDs), compared with WT mice. In adults (P56), MF boutons were larger in KO mice. Synaptic vesicles (SVs) were increased but with a lower density compared with the WT. Furthermore, the number of SVs was decreased near the active zone. Moreover, MF-innervated CA3 postsynapses in KO mice displayed aberrant structures at the postsynaptic density (PSD), with an increased number of PSDs (likely because of a larger number of perforated PSDs), compared with WT mice. Taken together, our findings suggest that CADPS2 plays a critical role in MF synaptic development and functional organization.


Subject(s)
Calcium-Binding Proteins/physiology , Mossy Fibers, Hippocampal/growth & development , Nerve Tissue Proteins/physiology , Synapses/physiology , Animals , Calcium-Binding Proteins/genetics , Male , Mice, Knockout , Mossy Fibers, Hippocampal/ultrastructure , Nerve Tissue Proteins/genetics , Synapses/ultrastructure
13.
Biol Pharm Bull ; 41(1): 138-141, 2018.
Article in English | MEDLINE | ID: mdl-29311476

ABSTRACT

Appropriate axonal pathfinding is a necessary step for the function of neuronal circuits. The mossy fibers (MFs) in the hippocampus of CaMKIIα heterozygous knockout (CaMKIIα-hKO) psychiatric model mice project onto not only the stratum lucidum but also the stratum oriens region in the CA3, which is a projection pattern distinct from that in normal mice. Thus, we examined the electrophysiological properties of the MF-CA3 connection in this mutant mouse on field recordings and found a lower synaptic connection. This study suggested that the phenotype of abnormal MF pathfindings could induce aberrant neuronal functions, which may link to cognition and memory.


Subject(s)
Axon Guidance , CA3 Region, Hippocampal/ultrastructure , Mental Disorders/pathology , Mossy Fibers, Hippocampal/ultrastructure , Neurons/ultrastructure , Animals , Axon Guidance/physiology , CA3 Region, Hippocampal/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Heterozygote , Male , Mental Disorders/physiopathology , Mice, Knockout , Mossy Fibers, Hippocampal/physiopathology , Neurons/metabolism
14.
Genes Cells ; 22(8): 742-755, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28695613

ABSTRACT

A hippocampal mossy fiber synapse, which is implicated in learning and memory, has a complex structure. We have previously shown using afadin-deficient mice that afadin plays multiple roles in the structural and functional differentiations of this synapse. We investigated here using a co-culture system with cultured hippocampal neurons and non-neuronal COS-7 cells expressing synaptogenic cell adhesion molecules (CAMs) whether afadin is involved in the presynaptic differentiation of hippocampal synapses. Postsynaptic CAMs NGL-3 (alias, a Lrrc4b gene product) and neuroligin induced presynaptic differentiation by trans-interacting with their respective presynaptic binding CAMs LAR (alias, a Ptprf gene product) and neurexin. This activity of NGL-3, but not neuroligin, was dependent on afadin, but not the afadin-binding presynaptic CAM nectin-1. The afadin-binding postsynaptic CAM nectin-3 did not induce presynaptic differentiation. Immunofluorescence and immunoelectron microscopy analyses showed that afadin was localized mainly at puncta adherentia junctions, but partly at synaptic junctions, of the mossy fiber synapse. ß-Catenin and γ-catenin known to bind to LAR were co-immunoprecipitated with afadin from the lysate of mouse brain. These results suggest that afadin is involved in the NGL-3-LAR system-induced presynaptic differentiation of hippocampal neurons cooperatively with ß-catenin and γ-catenin in a nectin-1-independent manner.


Subject(s)
GPI-Linked Proteins/metabolism , Hippocampus/metabolism , Microfilament Proteins/metabolism , Mossy Fibers, Hippocampal/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , GPI-Linked Proteins/genetics , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microfilament Proteins/genetics , Mossy Fibers, Hippocampal/ultrastructure , Nectins/genetics , Nectins/metabolism , Nerve Tissue Proteins/genetics , Neurons/cytology , Protein Binding , beta Catenin/metabolism , gamma Catenin/metabolism
15.
eNeuro ; 4(3)2017.
Article in English | MEDLINE | ID: mdl-28670619

ABSTRACT

Neural circuits balance excitatory and inhibitory activity and disruptions in this balance are commonly found in neurodevelopmental disorders. Mice lacking the intellectual disability and autism-associated gene Kirrel3 have an excitation-inhibition imbalance in the hippocampus but the precise synaptic changes underlying this functional defect are unknown. Kirrel3 is a homophilic adhesion molecule expressed in dentate gyrus (DG) and GABA neurons. It was suggested that the excitation-inhibition imbalance of hippocampal neurons in Kirrel3 knockout mice is due to loss of mossy fiber (MF) filopodia, which are DG axon protrusions thought to excite GABA neurons and thereby provide feed-forward inhibition to CA3 pyramidal neurons. Fewer filopodial structures were observed in Kirrel3 knockout mice but neither filopodial synapses nor DG en passant synapses, which also excite GABA neurons, were examined. Here, we used serial block-face scanning electron microscopy (SBEM) with 3D reconstruction to define the precise connectivity of MF filopodia and elucidate synaptic changes induced by Kirrel3 loss. Surprisingly, we discovered wildtype MF filopodia do not synapse exclusively onto GABA neurons as previously thought, but instead synapse with similar frequency onto GABA neurons and CA3 neurons. Moreover, Kirrel3 loss selectively reduces MF filopodial synapses onto GABA neurons but not those made onto CA3 neurons or en passant synapses. In sum, the selective loss of MF filopodial synapses with GABA neurons likely underlies the hippocampal activity imbalance observed in Kirrel3 knockout mice and may impact neural function in patients with Kirrel3-dependent neurodevelopmental disorders.


Subject(s)
Hippocampus/cytology , Membrane Proteins/deficiency , Mossy Fibers, Hippocampal/ultrastructure , Pyramidal Cells/metabolism , Synapses/ultrastructure , Animals , Animals, Newborn , Dendrites/genetics , Dendrites/metabolism , Dendrites/ultrastructure , Female , Hippocampus/ultrastructure , Imaging, Three-Dimensional , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Electron , Pyramidal Cells/ultrastructure , Synapses/genetics , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure , gamma-Aminobutyric Acid/metabolism
16.
Genes Cells ; 22(8): 715-722, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28631873

ABSTRACT

A hippocampal mossy fiber synapse has a complex structure and is implicated in learning and memory. In this synapse, the mossy fiber boutons attach to the dendritic shaft by puncta adherentia junctions and wrap around a multiply-branched spine, forming synaptic junctions. We have recently shown using transmission electron microscopy, immunoelectron microscopy and serial block face-scanning electron microscopy that atypical puncta adherentia junctions are formed in the afadin-deficient mossy fiber synapse and that the complexity of postsynaptic spines and mossy fiber boutons, the number of spine heads, the area of postsynaptic densities and the density of synaptic vesicles docked to active zones are decreased in the afadin-deficient synapse. We investigated here the roles of afadin in the functional differentiations of the mossy fiber synapse using the afadin-deficient mice. The electrophysiological studies showed that both the release probability of glutamate and the postsynaptic responsiveness to glutamate were markedly reduced, but not completely lost, in the afadin-deficient mossy fiber synapse, whereas neither long-term potentiation nor long-term depression was affected. These results indicate that afadin plays roles in the functional differentiations of the presynapse and the postsynapse of the hippocampal mossy fiber synapse.


Subject(s)
Microfilament Proteins/metabolism , Mossy Fibers, Hippocampal/metabolism , Animals , Cells, Cultured , Glutamic Acid/metabolism , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Mossy Fibers, Hippocampal/physiology , Mossy Fibers, Hippocampal/ultrastructure , Post-Synaptic Density/metabolism , Post-Synaptic Density/physiology , Post-Synaptic Density/ultrastructure
17.
J Comp Neurol ; 525(12): 2719-2734, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28498492

ABSTRACT

A hippocampal mossy fiber synapse, which is implicated in learning and memory, has a complex structure in which mossy fiber boutons attach to the dendritic shaft by puncta adherentia junctions (PAJs) and wrap around a multiply-branched spine, forming synaptic junctions. Here, we electron microscopically analyzed the ultrastructure of this synapse in afadin-deficient mice. Transmission electron microscopy analysis revealed that typical PAJs with prominent symmetrical plasma membrane darkening undercoated with the thick filamentous cytoskeleton were observed in the control synapse, whereas in the afadin-deficient synapse, atypical PAJs with the symmetrical plasma membrane darkening, which was much less in thickness and darkness than those of the control typical PAJs, were observed. Immunoelectron microscopy analysis revealed that nectin-1, nectin-3, and N-cadherin were localized at the control typical PAJs, whereas nectin-1 and nectin-3 were localized at the afadin-deficient atypical PAJs to extents lower than those in the control synapse and N-cadherin was localized at their nonjunctional flanking regions. These results indicate that the atypical PAJs are formed by nectin-1 and nectin-3 independently of afadin and N-cadherin and that the typical PAJs are formed by afadin and N-cadherin cooperatively with nectin-1 and nectin-3. Serial block face-scanning electron microscopy analysis revealed that the complexity of postsynaptic spines and mossy fiber boutons, the number of spine heads, the area of postsynaptic densities, and the density of synaptic vesicles docked to active zones were decreased in the afadin-deficient synapse. These results indicate that afadin plays multiple roles in the complex ultrastructural morphogenesis of hippocampal mossy fiber synapses.


Subject(s)
Hippocampus/cytology , Microfilament Proteins/metabolism , Morphogenesis/physiology , Mossy Fibers, Hippocampal/ultrastructure , Neurons/ultrastructure , Synapses/metabolism , Animals , Cadherins/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , Gene Expression Regulation/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Models, Neurological , Mossy Fibers, Hippocampal/metabolism , Nectins/metabolism , Neurons/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels, Sodium-Activated , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Synapses/ultrastructure , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Neuroimage ; 128: 227-237, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26254115

ABSTRACT

Hippocampal mossy fibers (MFs) can show plasticity of their axon terminal arbor consequent to learning a spatial memory task. Such plasticity is seen as translaminar sprouting from the stratum lucidum (SL) of CA3 into the stratum pyramidale (SP) and the stratum oriens (SO). However, the functional role of this presynaptic remodeling is still obscure. In vivo imaging that allows longitudinal observation of such remodeling could provide a deeper understanding of this presynaptic growth phenomenon as it occurs over time. Here we used manganese-enhanced magnetic resonance imaging (MEMRI), which shows a high-contrast area that co-localizes with the MFs. This technique was applied in the detection of learning-induced MF plasticity in two strains of rats. Quantitative analysis of a series of sections in the rostral dorsal hippocampus showed increases in the CA3a' area in MEMRI of trained Wistar rats consistent with the increased SO+SP area seen in the Timm's staining. MF plasticity was not seen in the trained Lister-Hooded rats in either MEMRI or in Timm's staining. This indicates the potential of MEMRI for revealing neuro-architectures and plasticity of the hippocampal MF system in vivo in longitudinal studies.


Subject(s)
Brain Mapping/methods , CA3 Region, Hippocampal/cytology , Magnetic Resonance Imaging/methods , Mossy Fibers, Hippocampal/ultrastructure , Neuronal Plasticity/physiology , Spatial Memory/physiology , Animals , CA3 Region, Hippocampal/physiology , Image Processing, Computer-Assisted , Male , Manganese , Maze Learning/physiology , Mossy Fibers, Hippocampal/physiology , Rats , Rats, Wistar
19.
Bull Exp Biol Med ; 160(1): 91-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26597685

ABSTRACT

We studied the role of neurotransmitter signaling mediated by synaptic vesicles in the formation of aberrant functional connections between fascia dentata grafts and the somatosensory neocortex in adult rats. Quantitative analysis of the different populations of synaptic vesicles in the ectopic giant axonal endings of granular neurons was performed and the results were compared with the normal. Two pools of small clear vesicles (rapidly releasable pool and pool of reserve vesicles circulating in the active zone) and one pool of large dense-core vesicles were analyzed. Significant differences from the control suggest that synaptic integration of the transplants into the recipient brain is coordinated by transsynaptic signaling and mediated by different populations of synaptic vesicles.


Subject(s)
Brain Tissue Transplantation , Dentate Gyrus/transplantation , Fetal Tissue Transplantation , Mossy Fibers, Hippocampal/ultrastructure , Neurotransmitter Agents/physiology , Somatosensory Cortex/ultrastructure , Synaptic Vesicles/ultrastructure , Animals , Cell Communication , Dentate Gyrus/ultrastructure , Graft Survival , Male , Microscopy, Electron, Scanning , Mossy Fibers, Hippocampal/metabolism , Neurons/ultrastructure , Rats , Rats, Wistar , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Somatosensory Cortex/surgery , Synaptic Vesicles/metabolism
20.
Brain ; 137(Pt 8): 2312-28, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24941947

ABSTRACT

The study of neurogenesis during chronic neurodegeneration is crucial in order to understand the intrinsic repair mechanisms of the brain, and key to designing therapeutic strategies. In this study, using an experimental model of progressive chronic neurodegeneration, murine prion disease, we define the temporal dynamics of the generation, maturation and integration of new neurons in the hippocampal dentate gyrus, using dual pulse-chase, multicolour γ-retroviral tracing, transmission electron microscopy and patch-clamp. We found increased neurogenesis during the progression of prion disease, which partially counteracts the effects of chronic neurodegeneration, as evidenced by blocking neurogenesis with cytosine arabinoside, and helps to preserve the hippocampal function. Evidence obtained from human post-mortem samples, of both variant Creutzfeldt-Jakob disease and Alzheimer's disease patients, also suggests increased neurogenic activity. These results open a new avenue into the exploration of the effects and regulation of neurogenesis during chronic neurodegeneration, and offer a new model to reproduce the changes observed in human neurodegenerative diseases.


Subject(s)
Hippocampus/pathology , Neural Pathways/pathology , Neurodegenerative Diseases/pathology , Neurogenesis/physiology , Prion Diseases/pathology , Tissue Banks , Adult , Aged , Alzheimer Disease/pathology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation , Chronic Disease , Creutzfeldt-Jakob Syndrome/pathology , Cytarabine/administration & dosage , Cytarabine/pharmacology , Dentate Gyrus/cytology , Dentate Gyrus/pathology , Dentate Gyrus/ultrastructure , Disease Models, Animal , Disease Progression , Female , Genetic Vectors , Hippocampus/cytology , Hippocampus/ultrastructure , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mossy Fibers, Hippocampal/ultrastructure , Neural Pathways/cytology , Neural Pathways/ultrastructure , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neural Stem Cells/ultrastructure , Neuroanatomical Tract-Tracing Techniques , Patch-Clamp Techniques , Prions/pathogenicity , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...