Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Neural Transm (Vienna) ; 130(8): 1003-1012, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37014414

RESUMEN

Therapeutic approaches providing effective medication for Alzheimer's disease (AD) patients after disease onset are urgently needed. Previous studies in AD mouse models and in humans suggested that physical exercise or changed lifestyle can delay AD-related synaptic and memory dysfunctions when treatment started in juvenile animals or in elderly humans before onset of disease symptoms. However, a pharmacological treatment that can reverse memory deficits in AD patients was thus far not identified. Importantly, AD disease-related dysfunctions have increasingly been associated with neuro-inflammatory mechanisms and searching for anti-inflammatory medication to treat AD seems promising. Like for other diseases, repurposing of FDA-approved drugs for treatment of AD is an ideally suited strategy to reduce the time to bring such medication into clinical practice. Of note, the sphingosine-1-phosphate analogue fingolimod (FTY720) was FDA-approved in 2010 for treatment of multiple sclerosis patients. It binds to the five different isoforms of Sphingosine-1-phosphate receptors (S1PRs) that are widely distributed across human organs. Interestingly, recent studies in five different mouse models of AD suggest that FTY720 treatment, even when starting after onset of AD symptoms, can reverse synaptic deficits and memory dysfunction in these AD mouse models. Furthermore, a very recent multi-omics study identified mutations in the sphingosine/ceramide pathway as a risk factor for sporadic AD, suggesting S1PRs as promising drug target in AD patients. Therefore, progressing with FDA-approved S1PR modulators into human clinical trials might pave the way for these potential disease modifying anti-AD drugs.


Asunto(s)
Enfermedad de Alzheimer , Esclerosis Múltiple , Ratones , Animales , Humanos , Anciano , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Reposicionamiento de Medicamentos , Esclerosis , Esclerosis Múltiple/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo
2.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255764

RESUMEN

Therapeutic approaches providing effective medication for Alzheimer's disease (AD) patients after disease onset are urgently needed. Previous studies in AD mouse models suggested that physical exercise or changed lifestyle can delay AD-related synaptic and memory dysfunctions when treatment started in juvenile animals long before onset of disease symptoms, while a pharmacological treatment that can reverse synaptic and memory deficits in AD mice was thus far not identified. Repurposing food and drug administration (FDA)-approved drugs for treatment of AD is a promising way to reduce the time to bring such medication into clinical practice. The sphingosine-1 phosphate analog fingolimod (FTY720) was approved recently for treatment of multiple sclerosis patients. Here, we addressed whether fingolimod rescues AD-related synaptic deficits and memory dysfunction in an amyloid precursor protein/presenilin-1 (APP/PS1) AD mouse model when medication starts after onset of symptoms (at five months). Male mice received intraperitoneal injections of fingolimod for one to two months starting at five to six months. This treatment rescued spine density as well as long-term potentiation in hippocampal cornu ammonis-1 (CA1) pyramidal neurons, that were both impaired in untreated APP/PS1 animals at six to seven months of age. Immunohistochemical analysis with markers of microgliosis (ionized calcium-binding adapter molecule 1; Iba1) and astrogliosis (glial fibrillary acid protein; GFAP) revealed that our fingolimod treatment regime strongly down regulated neuroinflammation in the hippocampus and neocortex of this AD model. These effects were accompanied by a moderate reduction of Aß accumulation in hippocampus and neocortex. Our results suggest that fingolimod, when applied after onset of disease symptoms in an APP/PS1 mouse model, rescues synaptic pathology that is believed to underlie memory deficits in AD mice, and that this beneficial effect is mediated via anti-neuroinflammatory actions of the drug on microglia and astrocytes.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Precursor de Proteína beta-Amiloide/genética , Inflamación/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Presenilina-1/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Antiinflamatorios/farmacología , Astrocitos/metabolismo , Astrocitos/patología , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Sinapsis/genética , Sinapsis/patología
3.
Glia ; 67(10): 1893-1909, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31246351

RESUMEN

Human astrocytes differ dramatically in cell morphology and gene expression from murine astrocytes. The latter are well known to be of major importance in the formation of neuronal networks by promoting synapse maturation. However, whether human astrocyte lineage cells have a similar role in network formation has not been firmly established. Here, we investigated the impact of human astrocyte lineage cells on the functional maturation of neural networks that were derived from human induced pluripotent stem cells (hiPSCs). Initial in vitro differentiation of hiPSC-derived neural progenitor cells and immature neurons (glia+ cultures) resulted in spontaneously active neural networks as indicated by synchronous neuronal Ca2+ transients. Depleting proliferating neural progenitors from these cultures by short-term antimitotic treatment resulted in strongly astrocyte lineage cell-depleted neuronal networks (glia- cultures). Strikingly, in contrast to glia+ cultures, glia- cultures did not exhibit spontaneous network activity. Detailed analysis of the morphological and electrophysiological properties of neurons by patch clamp recordings revealed reduced dendritic arborization in glia- cultures. In addition, a reduced action potential frequency upon current injection in pyramidal-like neurons was observed, whereas the electrical excitability of multipolar neurons was unaltered. Furthermore, we found a reduced dendritic density of PSD95-positive excitatory synapses, and more immature properties of AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) miniature excitatory postsynaptic currents (mEPSCs) in glia- cultures, suggesting that the maturation of glutamatergic synapses depends on the presence of hiPSC-derived astrocyte lineage cells. Intriguingly, addition of the astrocyte-derived synapse maturation inducer cholesterol increased the dendritic density of PSD95-positive excitatory synapses in glia- cultures.


Asunto(s)
Astrocitos/fisiología , Linaje de la Célula , Células Madre Pluripotentes Inducidas/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Potenciales de Acción/fisiología , Células Cultivadas , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Humanos , Potenciales Postsinápticos Miniatura/fisiología , Vías Nerviosas/fisiología , Células-Madre Neurales/fisiología , Receptores AMPA/metabolismo
4.
Brain ; 139(Pt 2): 509-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26657517

RESUMEN

Despite amyloid plaques, consisting of insoluble, aggregated amyloid-ß peptides, being a defining feature of Alzheimer's disease, their significance has been challenged due to controversial findings regarding the correlation of cognitive impairment in Alzheimer's disease with plaque load. The amyloid cascade hypothesis defines soluble amyloid-ß oligomers, consisting of multiple amyloid-ß monomers, as precursors of insoluble amyloid-ß plaques. Dissecting the biological effects of single amyloid-ß oligomers, for example of amyloid-ß dimers, an abundant amyloid-ß oligomer associated with clinical progression of Alzheimer's disease, has been difficult due to the inability to control the kinetics of amyloid-ß multimerization. For investigating the biological effects of amyloid-ß dimers, we stabilized amyloid-ß dimers by an intermolecular disulphide bridge via a cysteine mutation in the amyloid-ß peptide (Aß-S8C) of the amyloid precursor protein. This construct was expressed as a recombinant protein in cells and in a novel transgenic mouse, termed tgDimer mouse. This mouse formed constant levels of highly synaptotoxic soluble amyloid-ß dimers, but not monomers, amyloid-ß plaques or insoluble amyloid-ß during its lifespan. Accordingly, neither signs of neuroinflammation, tau hyperphosphorylation or cell death were observed. Nevertheless, these tgDimer mice did exhibit deficits in hippocampal long-term potentiation and age-related impairments in learning and memory, similar to what was observed in classical Alzheimer's disease mouse models. Although the amyloid-ß dimers were unable to initiate the formation of insoluble amyloid-ß aggregates in tgDimer mice, after crossbreeding tgDimer mice with the CRND8 mouse, an amyloid-ß plaque generating mouse model, Aß-S8C dimers were sequestered into amyloid-ß plaques, suggesting that amyloid-ß plaques incorporate neurotoxic amyloid-ß dimers that by themselves are unable to self-assemble. Our results suggest that within the fine interplay between different amyloid-ß species, amyloid-ß dimer neurotoxic signalling, in the absence of amyloid-ß plaque pathology, may be involved in causing early deficits in synaptic plasticity, learning and memory that accompany Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Trastornos del Conocimiento/metabolismo , Plasticidad Neuronal/fisiología , Placa Amiloide/metabolismo , Multimerización de Proteína/fisiología , Péptidos beta-Amiloides/genética , Animales , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Placa Amiloide/genética , Placa Amiloide/patología
5.
J Cell Sci ; 126(Pt 21): 5062-73, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23986480

RESUMEN

Brain-derived neurotrophic factor (BDNF) is known to be a crucial regulator of neuronal survival and synaptic plasticity in the mammalian brain. Furthermore, BDNF positively influences differentiation of embryonic neural precursors, as well as that of neural stem cells from adult neurogenic niches. To study the impact of cell-released BDNF on neural differentiation of embryonic stem cells (ESCs), which represent an attractive source for cell transplantation studies, we have generated mouse ESC clones overexpressing BDNF-GFP by use of knock-in technology. After neural differentiation in vitro, we observed that ESC clones overexpressing BDNF-GFP gave rise to an increased number of neurons as compared to control ESCs. Neurons derived from BDNF-GFP-expressing ESCs harbored a more complex dendritic morphology and differentiated into the GABAergic lineage more than controls. Moreover, we show that ESC-derived neurons released BDNF-GFP in an activity-dependent manner and displayed similar electrophysiological properties as cortical neurons. Thus, our study describes the generation of ESCs stably overexpressing BDNF-GFP, which are ideally suited to investigate the ameliorating effects of BDNF in cell transplantation studies of various neuropathological conditions.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Proteínas Fluorescentes Verdes/metabolismo , Neuronas/citología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Células Madre Embrionarias/metabolismo , Proteínas Fluorescentes Verdes/genética , Ratones , Neuronas/metabolismo
6.
Brain ; 135(Pt 7): 2140-54, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22637581

RESUMEN

The aetiology of Alzheimer's disease is thought to include functional impairment of synapses and synapse loss as crucial pathological events leading to cognitive dysfunction and memory loss. Oligomeric amyloid-ß peptides are well known to induce functional damage, destabilization and loss of brain synapses. However, the complex molecular mechanisms of amyloid-ß action resulting ultimately in synapse elimination are incompletely understood, thus limiting knowledge of potential therapeutic targets. Under physiological conditions, long-term synapse stability is mediated by trans-synaptically interacting adhesion molecules such as the homophilically binding N-cadherin/catenin complexes. In this study, we addressed whether inhibition of N-cadherin function affects amyloid-ß-induced synapse impairment. We found that blocking N-cadherin function, both by specific peptides interfering with homophilic binding and by expression of a dominant-negative, ectodomain-deleted N-cadherin mutant, resulted in a strong acceleration of the effect of amyloid-ß on synapse function in cultured cortical neurons. The frequency of AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor-mediated miniature excitatory postsynaptic currents was reduced upon amyloid-ß application much earlier than observed in controls. We further hypothesized that ectodomain-shed, transmembrane C-terminal fragments that are generated during N-cadherin proteolytic processing might similarly enhance amyloid-ß-induced synapse damage. Indeed, expression of human N-cadherin C-terminal fragment 1 strongly accelerated amyloid-ß-triggered synapse impairment. Ectodomain-shed N-cadherin C-terminal fragment 1 is further proteolytically cleaved by γ-secretase. Therefore, both pharmacological inhibition of γ-secretase and expression of the dominant-negative presenilin 1 mutant L166P were used to increase the presence of endogeneous N-cadherin C-terminal fragment 1. Under these conditions, we again found a strong acceleration of amyloid-ß-induced synapse impairment, which could be compensated by over-expression of full-length N-cadherin. Intriguingly, western blot analysis of post-mortem brains from patients with Alzheimer's disease revealed an enhanced presence of N-cadherin C-terminal fragment 1. Thus, an inhibition of N-cadherin function by proteolytically generated N-cadherin C-terminal fragment 1 might play an important role in Alzheimer's disease progression by accelerating amyloid-ß-triggered synapse damage.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Antígenos CD/fisiología , Cadherinas/fisiología , Fragmentos de Péptidos/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Sinapsis/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/farmacología , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/antagonistas & inhibidores , Cadherinas/biosíntesis , Cadherinas/genética , Carbamatos/farmacología , Células Cultivadas , Dipéptidos/farmacología , Femenino , Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Presenilina-1/genética , Presenilina-1/fisiología , Proteolisis
7.
Cell Death Discov ; 9(1): 444, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38062019

RESUMEN

Synaptotoxic Aß oligomers are thought to play a major role in the early pathology of Alzheimer´s disease (AD). However, the molecular mechanisms involved in Aß-induced synaptic dysfunction and synapse damage remain largely unclear. Previously, Aß synaptotoxicity has been reported to be enhanced by increased levels of a C-terminal fragment of the synaptic adhesion molecule N-cadherin that is generated by proteolytic shedding of the extracellular domains [1]. To address the molecular mechanisms involved in this process, we have now studied the functional synaptic changes induced by C-terminal fragments (CTF1) of synaptic adhesion proteins. We used synaptophysin-pHluorin (SypHy) fluorescence imaging to monitor synaptic vesicle exo- and endocytosis in cultures of mouse cortical neurons. We increased the levels of C-terminal fragments of synaptic adhesion proteins by pharmacologically inhibiting γ-secretase, which further degrades CTF1 fragments. We found that this intervention caused a delay in synaptic vesicle endocytosis. A similar effect was induced by overexpression of N-cadherin CTF1, but not by overexpression of Neurexin3ß CTF1. Based on these observations, we further studied whether directly modulating synaptic vesicle endocytosis enhances Aß synaptotoxicity. We pharmacologically induced a delayed synaptic vesicle endocytosis by a low concentration of the endocytosis inhibitor dynasore. This treatment enhanced synaptoxicity of Aß oligomers as indicated by a reduced frequency of miniature postsynaptic currents. In conclusion, we propose that delayed endocytosis results in prolonged exposure of synaptic vesicle membranes to the extracellular space, thus enabling enhanced vesicle membrane binding of Aß oligomers. This might in turn promote the endocytic uptake of toxic Aß oligomers and might thus play an important role in intracellular Aß-mediated synaptotoxicity in AD.

8.
Front Mol Neurosci ; 15: 829506, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35386271

RESUMEN

One of the most fundamental organizing principles in the mammalian brain is that neurons do not establish synapses with the other major cell type, the astrocytes. However, induced synapse formation between neurons and astrocytes appears conceivable, because astrocytes are well known to express functional ionotropic glutamate receptors. Here, we attempted to trigger synapse formation between co-cultured neurons and astrocytes by overexpressing the strongly synaptogenic adhesion protein LRRTM2 in astrocytes physically contacted by cortical axons. Interestingly, control experiments with immature cortical astrocytes without any overexpression resulted in the induction of synaptic vesicle clustering in contacting axons (hemisynapse formation). This synaptogenic activity correlated with the endogenous expression of the synaptogenic protein Neuroligin1. Hemisynapse formation was further enhanced upon overexpression of LRRTM2 in cortical astrocytes. In contrast, cerebellar astrocytes required overexpression of LRRTM2 for induction of synaptic vesicle clustering in contacting axons. We further addressed, whether hemisynapse formation was accompanied by the appearance of fully functional glutamatergic synapses. We therefore attempted to record AMPA receptor-mediated miniature excitatory postsynaptic currents (mEPSCs) in innervated astrocytes using the whole-cell patch-clamp technique. Despite the endogenous expression of the AMPA receptor subunits GluA2 and to a lesser extent GluA1, we did not reliably observe spontaneous AMPA mEPSCs. In conclusion, overexpression of the synaptogenic protein LRRTM2 induced hemisynapse formation between co-cultured neurons and astrocytes. However, the formation of fully functional synapses appeared to require additional factors critical for nano-alignment of presynaptic vesicles and postsynaptic receptors.

9.
Acta Neuropathol Commun ; 10(1): 190, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36578035

RESUMEN

Semantic dementia (SD) is a clinical subtype of frontotemporal dementia consistent with the neuropathological diagnosis frontotemporal lobar degeneration (FTLD) TDP type C, with characteristic round TDP-43 protein inclusions in the dentate gyrus. Despite this striking clinicopathological concordance, the pathogenic mechanisms are largely unexplained forestalling the development of targeted therapeutics. To address this, we carried out laser capture microdissection of the dentate gyrus of 15 SD patients and 17 non-demented controls, and assessed relative protein abundance changes by label-free quantitative mass spectrometry. To identify SD specific proteins, we compared our results to eight other FTLD and Alzheimer's disease (AD) proteomic datasets of cortical brain tissue, parallel with functional enrichment analyses and protein-protein interactions (PPI). Of the total 5,354 quantified proteins, 151 showed differential abundance in SD patients (adjusted P-value < 0.01). Seventy-nine proteins were considered potentially SD specific as these were not detected, or demonstrated insignificant or opposite change in FTLD/AD. Functional enrichment indicated an overrepresentation of pathways related to the immune response, metabolic processes, and cell-junction assembly. PPI analysis highlighted a cluster of interacting proteins associated with adherens junction and cadherin binding, the cadherin-catenin complex. Multiple proteins in this complex showed significant upregulation in SD, including ß-catenin (CTNNB1), γ-catenin (JUP), and N-cadherin (CDH2), which were not observed in other neurodegenerative proteomic studies, and hence may resemble SD specific involvement. A trend of upregulation of all three proteins was observed by immunoblotting of whole hippocampus tissue, albeit only significant for N-cadherin. In summary, we discovered a specific increase of cell adhesion proteins in SD constituting the cadherin-catenin complex at the synaptic membrane, essential for synaptic signaling. Although further investigation and validation are warranted, we anticipate that these findings will help unravel the disease processes underlying SD.


Asunto(s)
Enfermedad de Alzheimer , Demencia Frontotemporal , Degeneración Lobar Frontotemporal , Humanos , Demencia Frontotemporal/patología , Patología Molecular , Proteómica , Degeneración Lobar Frontotemporal/patología , Enfermedad de Alzheimer/patología , Giro Dentado/metabolismo , Cadherinas/metabolismo , Cateninas/metabolismo
10.
Front Cell Neurosci ; 15: 713693, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34759800

RESUMEN

At mammalian glutamatergic synapses, most basic elements of synaptic transmission have been shown to be modulated by specific transsynaptic adhesion complexes. However, although crucial for synapse homeostasis, a physiological regulation of synaptic vesicle endocytosis by adhesion molecules has not been firmly established. The homophilic adhesion protein N-cadherin is localized at the peri-active zone, where the highly temperature-dependent endocytosis of vesicles occurs. Here, we demonstrate an important modulatory role of N-cadherin in endocytosis at near physiological temperature by synaptophysin-pHluorin imaging. Different modes of endocytosis including bulk endocytosis were dependent on N-cadherin expression and function. N-cadherin modulation might be mediated by actin filaments because actin polymerization ameliorated the knockout-induced endocytosis defect. Using super-resolution imaging, we found strong recruitment of N-cadherin to glutamatergic synapses upon massive vesicle release, which might in turn enhance vesicle endocytosis. This provides a novel, adhesion protein-mediated mechanism for efficient coupling of exo- and endocytosis.

11.
Neuron ; 51(6): 741-54, 2006 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-16982420

RESUMEN

Synaptogenesis, the generation and maturation of functional synapses between nerve cells, is an essential step in the development of neuronal networks in the brain. It is thought to be triggered by members of the neuroligin family of postsynaptic cell adhesion proteins, which may form transsynaptic contacts with presynaptic alpha- and beta-neurexins and have been implicated in the etiology of autism. We show that deletion mutant mice lacking neuroligin expression die shortly after birth due to respiratory failure. This respiratory failure is a consequence of reduced GABAergic/glycinergic and glutamatergic synaptic transmission and network activity in brainstem centers that control respiration. However, the density of synaptic contacts is not altered in neuroligin-deficient brains and cultured neurons. Our data show that neuroligins are required for proper synapse maturation and brain function, but not for the initial formation of synaptic contacts.


Asunto(s)
Encéfalo/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Sinapsis/fisiología , Proteínas Adaptadoras del Transporte Vesicular , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/citología , Encéfalo/metabolismo , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiología , Moléculas de Adhesión Celular Neuronal , Células Cultivadas , Expresión Génica/genética , Hibridación in Situ , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neuronas/ultraestructura , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/fisiopatología , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/metabolismo , Sinapsis/genética , Sinapsis/ultraestructura
12.
Exp Brain Res ; 200(2): 169-82, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19862508

RESUMEN

Following initial contact formation, glutamatergic synapses in cortical neurons undergo pronounced functional maturation. These maturational events, occurring both pre- and postsynaptically, have been well described in the developing hippocampus. In this paper, we characterized glutamatergic synapses in immature layer Vb pyramidal neurons of the mouse somatosensory cortex during early postnatal development. At postnatal day 7, a significant subpopulation of glutamatergic synapses exhibited a low release probability that was accompanied by strong paired-pulse facilitation of AMPA EPSCs (paired-pulse ratio C > or = 2). Increasing extracellular Ca(2+) concentration increased release probability and led to paired-pulse depression. During further postnatal development, these functionally immature synapses disappeared. As shown pharmacologically,these synapses expressed postsynaptic NMDA receptors containing NR2B subunits, while NMDA receptors with NR2A subunits were lacking. Taken together, a low release probability presynaptically was coupled to postsynaptic NR2B signaling. This subpopulation of neocortical synapses thus differed from the majority of synapses in the developing hippocampus, where high release probability is coupled to NR2B signaling. The novel type of functionally immature glutamatergic synapse described here might play an important role in early developmental synapse elimination and in the activity-dependent refinement of the neocortical synaptic microcircuitry.


Asunto(s)
Ácido Glutámico/metabolismo , Células Piramidales/fisiología , Corteza Somatosensorial/crecimiento & desarrollo , Corteza Somatosensorial/fisiología , Sinapsis/fisiología , Envejecimiento , Animales , Calcio/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Técnicas de Placa-Clamp , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/fisiología , Probabilidad , Células Piramidales/efectos de los fármacos , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Corteza Somatosensorial/efectos de los fármacos , Sinapsis/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
13.
Nature ; 423(6943): 939-48, 2003 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-12827191

RESUMEN

Synapses are specialized intercellular junctions in which cell adhesion molecules connect the presynaptic machinery for neurotransmitter release to the postsynaptic machinery for receptor signalling. Neurotransmitter release requires the presynaptic co-assembly of Ca2+ channels with the secretory apparatus, but little is known about how synaptic components are organized. Alpha-neurexins, a family of >1,000 presynaptic cell-surface proteins encoded by three genes, link the pre- and postsynaptic compartments of synapses by binding extracellularly to postsynaptic cell adhesion molecules and intracellularly to presynaptic PDZ domain proteins. Using triple-knockout mice, we show that alpha-neurexins are not required for synapse formation, but are essential for Ca2+-triggered neurotransmitter release. Neurotransmitter release is impaired because synaptic Ca2+ channel function is markedly reduced, although the number of cell-surface Ca2+ channels appears normal. These data suggest that alpha-neurexins organize presynaptic terminals by functionally coupling Ca2+ channels to the presynaptic machinery.


Asunto(s)
Canales de Calcio/fisiología , Exocitosis/fisiología , Proteínas del Tejido Nervioso/fisiología , Sinapsis/fisiología , Vesículas Sinápticas/fisiología , Animales , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Calcio/metabolismo , Señalización del Calcio , Adhesión Celular , Moléculas de Adhesión Celular Neuronal/fisiología , Células Cultivadas , Potenciales Evocados , Glicoproteínas , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neocórtex/citología , Neocórtex/fisiología , Proteínas del Tejido Nervioso/genética , Neuropéptidos , Neurotransmisores/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transmisión Sináptica
14.
J Neurosci Methods ; 341: 108797, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32479974

RESUMEN

BACKGROUND: Spine loss is a hallmark of Alzheimer´s and other neurodegenerative diseases, and testing candidate therapeutic drugs needs quantitative analysis of dendritic spine densities. Golgi-Cox impregnation of neurons is a classical method to visualize dendritic spines in diseased brains. Importantly, at early disease stages spine loss occurs locally in the vicinity of amyloid plaques, and concomitant fluorescence labeling of amyloid plaques is required to detect local spine damage. NEW METHOD: Because Golgi-Cox impregnation is done on unsectioned brains, whereas fluorescence staining is performed on sectioned material, the combination is technically challenging. We have now developed a novel combination of Golgi-Cox impregnation with methoxy-X04 fluorescence labeling of plaques that is performed on unsectioned brains. RESULTS: We used this new combination method to quantify dendritic spine densities in mouse hippocampal CA1 pyramidal neurons. Comparison of neurons from wildtype and APP/PS1 mice revealed local spine loss in the vicinity of amyloid plaques in both male and female APP/PS1 mice. COMPARISON WITH EXISTING METHOD: Golgi-Cox impregnation of neurons combined with methoxy-X04 staining of amyloid plaques is a highly reliable, easy-to-use method for permanent visualization of spines as compared to the technically more sophisticated and less stable fluorescence imaging of spines. CONCLUSION: Our novel combination method will be highly useful for testing potential therapeutic drugs in Alzheimer mouse models.


Asunto(s)
Enfermedad de Alzheimer , Placa Amiloide , Enfermedad de Alzheimer/diagnóstico por imagen , Animales , Espinas Dendríticas , Femenino , Masculino , Ratones , Ratones Transgénicos , Coloración y Etiquetado
15.
Exp Brain Res ; 199(3-4): 203-34, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19777221

RESUMEN

In the past 15 years numerous reports provided strong evidence that brain-derived neurotrophic factor (BDNF) is one of the most important modulators of glutamatergic and GABAergic synapses. Remarkable progress regarding localization, kinetics, and molecular mechanisms of BDNF secretion has been achieved, and a large number of studies provided evidence that continuous extracellular supply of BDNF is important for the proper formation and functional maturation of glutamatergic and GABAergic synapses. BDNF can play a permissive role in shaping synaptic networks, making them more susceptible for the occurrence of plastic changes. In addition, BDNF appears to be also an instructive factor for activity-dependent long-term synaptic plasticity. BDNF release just in response to synaptic stimulation might be a molecular trigger to convert high-frequency synaptic activity into long-term synaptic memories. This review attempts to summarize the current knowledge in synaptic secretion and synaptic action of BDNF, including both permissive and instructive effects of BDNF in synaptic plasticity.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Ácido Glutámico/fisiología , Plasticidad Neuronal/fisiología , Transducción de Señal/fisiología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Humanos , Sinapsis/metabolismo , Potenciales Sinápticos/fisiología , Ácido gamma-Aminobutírico/metabolismo
16.
Front Mol Neurosci ; 12: 269, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31780894

RESUMEN

Synaptic cell adhesion molecules are well established to exhibit synaptogenic activity when overexpressed in target cells, indicating that they are involved in formation and functional maturation of synapses. The postsynaptic adhesion proteins Neuroligin1 and LRRTM2 both induce synaptic vesicle clusters in presynaptic axons in vitro by transsynaptically interacting with neurexins. In neurons, this is accompanied by the induction of glutamatergic, but not GABAergic synapses. Although the synaptogenic activity of Neuroligin1 has been well characterized, the properties of the synaptogenic activities of other synaptic adhesion molecules are largely unknown. In this paper, we now compared characteristics of the synaptogenic activities of Neuroligin1 and LRRTM2 upon overexpression in cultured mouse cortical neurons. Individual cortical neurons were transfected with Neuroligin1 and LRRTM2 expression plasmids, respectively, and synaptic vesicle clustering in contacting axons was examined by immunostaining for the vesicle membrane protein VAMP2. In immature neurons at 6-7 days in vitro (DIV) both Neuroligin1 and LRRTM2 exhibited strong synaptogenic activity. However, upon further neuronal differentiation only LRRTM2 retained significant synaptogenic activity at 12-13 DIV. A similar differential developmental maturation of the synaptogenic activities of Neuroligin1 and LRRTM2 was observed for the induction of glutamatergic synapses, which were detected by co-immunostaining for VGLUT1 and Homer1. Most interestingly, the synaptogenic activity of Neuroligin1 was strongly dependent on the expression and function of the synaptic adhesion molecule N-cadherin in immature neurons. In contrast, the synaptogenic activity of LRRTM2 was independent of N-cadherin expression and function in both immature (6-7 DIV) and more mature neurons (14-15 DIV). Taken together, our results with overexpression in cultured cortical neurons revealed striking differences in the properties of the synaptogenic activities of Neuroligin1 and LRRTM2, although both transsynaptically interact with presynaptic neurexins.

17.
J Neurosci Res ; 86(2): 223-32, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17787017

RESUMEN

Delicate control of the synaptic vesicle cycle is required to meet the demands imposed on synaptic transmission by the brain's complex information processing. In addition to intensively analyzed intrinsic regulation, extrinsic modulation of the vesicle cycle by the postsynaptic target neuron has become evident. Recent studies have demonstrated that several families of synaptic cell-adhesion molecules play a significant role in transsynaptic retrograde signaling. Different adhesion systems appear to specifically target distinct steps of the synaptic vesicle cycle. Signaling via classical cadherins regulates the recruitment of synaptic vesicles to the active zone. The neurexin/neuroligin system has been shown to modulate presynaptic release probability. In addition, reverse signaling via the EphB/ephrinB system plays an important role in the activity-dependent induction of long-term potentiation of presynaptic transmitter release. Moreover, the first hints of involvement of cell-adhesion molecules in vesicle endocytosis have been published. A general hypothesis is that specific adhesion systems might use different but parallel transsynaptic signaling pathways able to selectively modulate each step of the synaptic vesicle cycle in a tightly coordinated manner.


Asunto(s)
Encéfalo/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Animales , Humanos , Plasticidad Neuronal/fisiología
18.
J Neurosci ; 26(26): 6968-78, 2006 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-16807326

RESUMEN

The cell adhesion molecule N-cadherin has been proposed to regulate synapse formation in mammalian central neurons. This is based on its synaptic localization enabling alignment of presynaptic and postsynaptic specializations by an adhesion mechanism. However, a potential role of N-cadherin in regulating synaptic transmission has remained elusive. In this paper, a functional analysis of N-cadherin knock-out synapses was enabled by in vitro neuronal differentiation of mouse embryonic stem cells circumventing the early embryonic lethality of mice genetically null for N-cadherin. In our in vitro system, initial synapse formation was not altered in the absence of N-cadherin, which might be attributable to compensatory mechanisms. Here, we demonstrate that N-cadherin is required for regulating presynaptic function at glutamatergic synapses. An impairment in the availability of vesicles for exocytosis became apparent selectively during high activity. Short-term plasticity was strongly altered with synaptic depression enhanced in the absence of N-cadherin. Most intriguingly, facilitation was converted to depression under specific stimulation conditions. This indicates an important role of N-cadherin in the control of short-term plasticity. To analyze, whether N-cadherin regulates presynaptic function by a transsynaptic mechanism, we studied chimeric cultures consisting of wild-type neocortical neurons and ES cell-derived neurons. With N-cadherin absent only postsynaptically, we observed a similar increase in short-term synaptic depression as found in its complete absence. This indicates a retrograde control of short-term plasticity by N-cadherin. In summary, our results revealed an unexpected involvement of a synaptic adhesion molecule in the regulation of short-term plasticity at glutamatergic synapses.


Asunto(s)
Cadherinas/fisiología , Embrión de Mamíferos/citología , Ácido Glutámico/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Células Madre/citología , Sinapsis/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Conductividad Eléctrica , Exocitosis/fisiología , Colorantes Fluorescentes , Ratones , Ratones Noqueados , Neuronas/citología , Compuestos de Piridinio , Compuestos de Amonio Cuaternario , Coloración y Etiquetado , Sacarosa/metabolismo , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/fisiología , Factores de Tiempo
19.
Sci Rep ; 7: 40865, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-28106089

RESUMEN

At synapses in the mammalian brain, continuous information transfer requires the long-term maintenance of homeostatic coupling between exo- and endocytosis of synaptic vesicles. Because classical endocytosis is orders of magnitude slower than the millisecond-range exocytosis of vesicles, high frequency vesicle fusion could potentially compromise structural stability of synapses. However, the molecular mechanisms mediating the tight coupling of exo- and endocytosis are largely unknown. Here, we investigated the role of the transsynaptic adhesion molecules N-cadherin and Neuroligin1 in regulating vesicle exo- and endocytosis by using activity-induced FM4-64 staining and by using synaptophysin-pHluorin fluorescence imaging. The synaptic adhesion molecules N-cadherin and Neuroligin1 had distinct impacts on exo- and endocytosis at mature cortical synapses. Expression of Neuroligin1 enhanced vesicle release in a N-cadherin-dependent way. Most intriguingly, expression of N-cadherin enhanced both vesicle exo- and endocytosis. Further detailed analysis of N-cadherin knockout neurons revealed that the boosting of endocytosis by N-cadherin was largely dependent on preceding high levels of vesicle release activity. In summary, regulation of vesicle endocytosis was mediated at the molecular level by N-cadherin in a release activity-dependent manner. Because of its endocytosis enhancing function, N-cadherin might play an important role in the coupling of vesicle exo- and endocytosis.


Asunto(s)
Cadherinas/metabolismo , Endocitosis , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Transporte Biológico , Cadherinas/deficiencia , Cadherinas/genética , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Cultivadas , Exocitosis , Técnica del Anticuerpo Fluorescente , Expresión Génica , Técnicas de Inactivación de Genes , Genes Reporteros , Ratones , Neuronas/metabolismo
20.
Prog Neurobiol ; 69(5): 341-74, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12787574

RESUMEN

The proteins of the mammalian neurotrophin family (nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and neurotrophin-4/5 (NT-4/5)) were originally identified as neuronal survival factors. During the last decade, evidence has accumulated implicating them (especially BDNF) in addition in the regulation of synaptic transmission and synaptogenesis in the CNS. However, a detailed understanding of the secretion of neurotrophins from neurons is required to delineate their role in regulating synaptic function. Some crucial questions that need to be addressed include the sites of neurotrophin secretion (i.e. axonal versus dendritic; synaptic versus extrasynaptic) and the neuronal and synaptic activity patterns that trigger the release of neurotrophins. In this article, we review the current knowledge in the field of neurotrophin secretion, focussing on activity-dependent synaptic release of BDNF. The modality and the site of neurotrophin secretion are dependent on the processing and subsequent targeting of the neurotrophin precursor molecules. Therefore, the available data regarding formation and trafficking of neurotrophins in the secreting neurons are critically reviewed. In addition, we discuss existing evidence that the characteristics of neurotrophin secretion are similar (but not identical) to those of other neuropeptides. Finally, since BDNF has been proposed to play a critical role as an intercellular synaptic messenger in long-term potentiation (LTP) in the hippocampus, we try to reconcile this possible role of BDNF in LTP with the recently described features of synaptic BDNF secretion.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Animales , Línea Celular/metabolismo , Supervivencia Celular/fisiología , Sistema Nervioso Central/metabolismo , Predicción , Humanos , Factores de Crecimiento Nervioso/clasificación , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Péptidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA