Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Alzheimers Res Ther ; 15(1): 16, 2023 01 14.
Article in English | MEDLINE | ID: mdl-36641439

ABSTRACT

BACKGROUND: Hyperphosphorylation and intraneuronal aggregation of the microtubule-associated protein tau is a major pathological hallmark of Alzheimer's disease (AD) brain. Of special interest is the effect of cerebral amyloid beta deposition, the second main hallmark of AD, on human tau pathology. Therefore, studying the influence of cerebral amyloidosis on human tau in a novel human tau knock-in (htau-KI) mouse model could help to reveal new details on their interplay. METHODS: We studied the effects of a novel human htau-KI under fast-progressing amyloidosis in 5xFAD mice in terms of correlation of gene expression data with human brain regions, development of Alzheimer's-like pathology, synaptic transmission, and behavior. RESULTS: The main findings are an interaction of human beta-amyloid and human tau in crossbred 5xFADxhtau-KI observed at transcriptional level and corroborated by electrophysiology and histopathology. The comparison of gene expression data of the 5xFADxhtau-KI mouse model to 5xFAD, control mice and to human AD patients revealed conspicuous changes in pathways related to mitochondria biology, extracellular matrix, and immune function. These changes were accompanied by plaque-associated MC1-positive pathological tau that required the htau-KI background. LTP deficits were noted in 5xFAD and htau-KI mice in contrast to signs of rescue in 5xFADxhtau-KI mice. Increased frequencies of miniature EPSCs and miniature IPSCs indicated an upregulated presynaptic function in 5xFADxhtau-KI. CONCLUSION: In summary, the multiple interactions observed between knocked-in human tau and the 5xFAD-driven progressing amyloidosis have important implications for future model development in AD.


Subject(s)
Alzheimer Disease , Amyloidosis , Mice , Humans , Animals , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Alzheimer Disease/pathology , tau Proteins/genetics , tau Proteins/metabolism , Brain/metabolism , Disease Models, Animal , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism
2.
Alzheimers Res Ther ; 12(1): 100, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32838792

ABSTRACT

BACKGROUND: Intensive basic and preclinical research into Alzheimer's disease (AD) has yielded important new findings, but they could not yet been translated into effective therapies. One of the reasons is the lack of animal models that sufficiently reproduce the complexity of human AD and the response of human brain circuits to novel treatment approaches. As a step in overcoming these limitations, new App knock-in models have been developed that avoid transgenic APP overexpression and its associated side effects. These mice are proposed to serve as valuable models to examine Aß-related pathology in "preclinical AD." METHODS: Since AD as the most common form of dementia progresses into synaptic failure as a major cause of cognitive deficits, the detailed characterization of synaptic dysfunction in these new models is essential. Here, we addressed this by extracellular and whole-cell patch-clamp recordings in AppNL-G-F mice compared to AppNL animals which served as controls. RESULTS: We found a beginning synaptic impairment (LTP deficit) at 3-4 months in the prefrontal cortex of AppNL-G-F mice that is further aggravated and extended to the hippocampus at 6-8 months. Measurements of miniature EPSCs and IPSCs point to a marked increase in excitatory and inhibitory presynaptic activity, the latter accompanied by a moderate increase in postsynaptic inhibitory function. CONCLUSIONS: Our data reveal a marked impairment of primarily postsynaptic processes at the level of synaptic plasticity but the dominance of a presumably compensatory presynaptic upregulation at the level of elementary miniature synaptic function.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/genetics , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Cereb Cortex ; 30(8): 4306-4324, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32147734

ABSTRACT

Schizophrenia is associated with cognitive and behavioral dysfunctions thought to reflect imbalances in neurotransmission systems. Recent screenings suggested that lack of (functional) syndapin I (PACSIN1) may be linked to schizophrenia. We therefore studied syndapin I KO mice to address the suggested causal relationship to schizophrenia and to analyze associated molecular, cellular, and neurophysiological defects. Syndapin I knockout (KO) mice developed schizophrenia-related behaviors, such as hyperactivity, reduced anxiety, reduced response to social novelty, and an exaggerated novel object response and exhibited defects in dendritic arborization in the cortex. Neuromorphogenic deficits were also observed for a schizophrenia-associated syndapin I mutant in cultured neurons and coincided with a lack of syndapin I-mediated membrane recruitment of cytoskeletal effectors. Syndapin I KO furthermore caused glutamatergic hypofunctions. Syndapin I regulated both AMPAR and NMDAR availabilities at synapses during basal synaptic activity and during synaptic plasticity-particularly striking were a complete lack of long-term potentiation and defects in long-term depression in syndapin I KO mice. These synaptic plasticity defects coincided with alterations of postsynaptic actin dynamics, synaptic GluA1 clustering, and GluA1 mobility. Both GluA1 and GluA2 were not appropriately internalized. Summarized, syndapin I KO led to schizophrenia-like behavior, and our analyses uncovered associated molecular and cellular mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain/metabolism , Neuronal Plasticity/physiology , Schizophrenia/metabolism , Animals , Behavior, Animal/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Brain Struct Funct ; 223(5): 2073-2095, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29372324

ABSTRACT

Unilateral vision loss through monocular enucleation (ME) results in partial reallocation of visual cortical territory to another sense in adult mice. The functional recovery of the visual cortex occurs through a combination of spared-eye potentiation and cross-modal reactivation driven by whisker-related, somatosensory inputs. Brain region-specific intracortical inhibition was recently recognized as a crucial regulator of the cross-modal component, yet the contribution of specific inhibitory neuron subpopulations remains poorly understood. Somatostatin (SST)-interneurons are ideally located within the cortical circuit to modulate sensory integration. Here we demonstrate that optogenetic stimulation of visual cortex SST-interneurons prior to eye removal decreases ME-induced cross-modal recovery at the stimulation site. Our results suggest that SST-interneurons act as local hubs, which are able to control the influx and extent of cortical cross-modal inputs into the deprived cortex. These insights critically expand our understanding of SST-interneuron-specific regulation of cortical plasticity induced by sensory loss.


Subject(s)
Blindness/pathology , Gene Expression Regulation/physiology , Interneurons/physiology , Neuronal Plasticity/physiology , Optogenetics/methods , Somatostatin/metabolism , Visual Cortex/pathology , Animals , Blindness/metabolism , Blindness/surgery , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Disease Models, Animal , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Eye Enucleation , Female , Functional Laterality , Male , Mice , Mice, Transgenic , Optic Nerve/physiology , Optic Nerve/transplantation , Recovery of Function/physiology , Sensory Deprivation/physiology , Somatostatin/genetics , Vibrissae/innervation
5.
Sci Rep ; 7(1): 5157, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698637

ABSTRACT

The hippocampus is important for spatial navigation, episodic memory and affective behaviour. Increasing evidence suggests that these multiple functions are accomplished by different segments along the dorsal-ventral (septal-temporal) axis. Long-term potentiation (LTP), the best-investigated cellular correlate of learning and memory, has distinct properties along this axis in the CA1 region, but so far, little is known about longitudinal differences in dentate gyrus (DG). Therefore, here we examined potential dorsoventral differences in DG-LTP using in vitro multi-electrode array recordings. In young mice, we found higher basal synaptic transmission in the dorsal DG, while the LTP magnitude markedly increased towards the ventral pole. Strikingly, these differences were greatly reduced in slices from middle-aged mice. Short-term plasticity, evaluated by paired-pulse ratios, was similar across groups. Recordings in the presence and absence of GABAA-receptor blocker picrotoxin suggested a higher inhibitory tone in the ventral DG of young mice, confirmed by an increased frequency of miniature inhibitory postsynaptic currents. Our findings support the view that the hippocampus contains discrete functional domains along its dorsoventral axis and demonstrate that these are subject to age-dependent changes. Since these characteristics are presumably conserved in the human hippocampus, our findings have important clinical implications for hippocampus- and age-related disorders.


Subject(s)
Aging/physiology , Dentate Gyrus/physiology , Long-Term Potentiation , Animals , Dentate Gyrus/drug effects , Female , Long-Term Potentiation/drug effects , Male , Memory, Short-Term/drug effects , Mice , Picrotoxin/pharmacology
6.
Neuropharmacology ; 116: 71-81, 2017 04.
Article in English | MEDLINE | ID: mdl-28012946

ABSTRACT

Many clinical and molecular features of the fragile X syndrome, a common form of intellectual disability and autism, can be modeled by deletion of the Fmr1 protein (Fmrp) in mice. Previous studies showed a decreased expression of several components of the GABAergic system in Fmr1 knockout mice. Here, we used this mouse model to investigate the functional consequences of Fmrp deletion on hippocampal GABAergic inhibition in the CA1-region of the hippocampus. Whole-cell patch-clamp recordings demonstrated a significantly reduced amplitude of evoked inhibitory postsynaptic currents (eIPSCs) and a decrease in the amplitude and frequency of spontaneous IPSCs. In addition, miniature IPSCs were reduced in amplitude and frequency and decayed significantly slower than mIPSCs in controls. Quantitative real-time PCR revealed a significantly lower expression of α2, ß1 and δ GABAA receptor subunits in the hippocampus of the juvenile mice (P22) compared to wild-type littermates. Correspondingly, we found also at the protein level reduced amounts of α2, ß1 and δ subunits in Fmr1 knockout mice. Overall, these results demonstrate that the reduction in several components of the GABAergic system is already present at young age and that this reduction results in measurable abnormalities on GABAA receptor-mediated phasic inhibition. These abnormalities might contribute to the behavioral and cognitive deficits of this fragile X mouse model.


Subject(s)
CA1 Region, Hippocampal/metabolism , Fragile X Mental Retardation Protein/metabolism , Inhibitory Postsynaptic Potentials/physiology , Pyramidal Cells/metabolism , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , CA1 Region, Hippocampal/drug effects , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , GABA-A Receptor Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , RNA, Messenger/metabolism , Tissue Culture Techniques
7.
Biol Psychiatry ; 81(2): 124-135, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27215477

ABSTRACT

BACKGROUND: Neuroplastin cell recognition molecules have been implicated in synaptic plasticity. Polymorphisms in the regulatory region of the human neuroplastin gene (NPTN) are correlated with cortical thickness and intellectual abilities in adolescents and in individuals with schizophrenia. METHODS: We characterized behavioral and functional changes in inducible conditional neuroplastin-deficient mice. RESULTS: We demonstrate that neuroplastins are required for associative learning in conditioning paradigms, e.g., two-way active avoidance and fear conditioning. Retrograde amnesia of learned associative memories is elicited by inducible neuron-specific ablation of Nptn gene expression in adult mice, which shows that neuroplastins are indispensable for the availability of previously acquired associative memories. Using single-photon emission computed tomography imaging in awake mice, we identified brain structures activated during memory recall. Constitutive neuroplastin deficiency or Nptn gene ablation in adult mice causes substantial electrophysiologic deficits such as reduced long-term potentiation. In addition, neuroplastin-deficient mice reveal profound physiologic and behavioral deficits, some of which are related to depression and schizophrenia, which illustrate neuroplastin's essential functions. CONCLUSIONS: Neuroplastins are essential for learning and memory. Retrograde amnesia after an associative learning task can be induced by ablation of the neuroplastin gene. The inducible neuroplastin-deficient mouse model provides a new and unique means to analyze the molecular and cellular mechanisms underlying retrograde amnesia and memory.


Subject(s)
Amnesia, Retrograde/physiopathology , Association Learning/physiology , Membrane Glycoproteins/physiology , Memory/physiology , Amnesia, Retrograde/genetics , Animals , Avoidance Learning/physiology , Behavior, Animal/physiology , Excitatory Postsynaptic Potentials , Fear/physiology , Hippocampus/physiology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Pflugers Arch ; 468(4): 593-607, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26631168

ABSTRACT

TRPM4 is a calcium-activated but calcium-impermeable non-selective cation (CAN) channel. Previous studies have shown that TRPM4 is an important regulator of Ca(2+)-dependent changes in membrane potential in excitable and non-excitable cell types. However, its physiological significance in neurons of the central nervous system remained unclear. Here, we report that TRPM4 proteins form a CAN channel in CA1 neurons of the hippocampus and we show that TRPM4 is an essential co-activator of N-methyl-D-aspartate (NMDA) receptors (NMDAR) during the induction of long-term potentiation (LTP). Disrupting the Trpm4 gene in mice specifically eliminates NMDAR-dependent LTP, while basal synaptic transmission, short-term plasticity, and NMDAR-dependent long-term depression are unchanged. The induction of LTP in Trpm4 (-/-) neurons was rescued by facilitating NMDA receptor activation or post-synaptic membrane depolarization. Accordingly, we obtained normal LTP in Trpm4 (-/-) neurons in a pairing protocol, where post-synaptic depolarization was applied in parallel to pre-synaptic stimulation. Taken together, our data are consistent with a novel model of LTP induction in CA1 hippocampal neurons, in which TRPM4 is an essential player in a feed-forward loop that generates the post-synaptic membrane depolarization which is necessary to fully activate NMDA receptors during the induction of LTP but which is dispensable for the induction of long-term depression (LTD). These results have important implications for the understanding of the induction process of LTP and the development of nootropic medication.


Subject(s)
CA1 Region, Hippocampal/metabolism , Long-Term Potentiation , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Potentials , TRPM Cation Channels/metabolism , Animals , CA1 Region, Hippocampal/cytology , Cells, Cultured , Feedback, Physiological , Mice , Neurons/physiology , TRPM Cation Channels/genetics
9.
Brain Struct Funct ; 220(3): 1273-90, 2015.
Article in English | MEDLINE | ID: mdl-24562414

ABSTRACT

Although the Morris water maze (MWM) is the most frequently used protocol to examine hippocampus-dependent learning in mice, not much is known about the spatio-temporal dynamics of underlying plasticity processes. Here, we studied molecular and cellular hippocampal plasticity mechanisms during early and late phases of spatial learning in the MWM. Quantitative in situ hybridization for the immediate early genes zif268 and Homer1a (H1a) revealed phase-dependent differences in their expression between areas CA1 and CA3. During the initial learning phase, CA1 expression levels of the molecular plasticity marker H1a, but not of the activity reporter gene zif268, were related to task proficiency; whereas no learning-specific changes could be detected in CA3. Simultaneously, the ratio of surface-expressed NMDAR subunits NR2A and NR2B was downregulated as measured by acute slice biotinylation assay, while the total number of surface NMDARs was unaltered. When intrinsic 'somatic' and synaptic plasticity in the CA1-region of hippocampal slices were examined, we found that early learning promotes intrinsic neuronal plasticity as manifested by a reduction of spike frequency adaptation and postburst afterhyperpolarization. At the synaptic level, however, maintenance of long-term potentiation (LTP) in all learning groups was impaired which is most likely due to 'intrinsic' learning-induced LTP which occluded any further electrically induced LTP. Late learning, in contrast, was characterized by re-normalized H1a, NR2A and NR2B expression and neuronal firing, yet a further strengthening of learning-induced LTP. Together, our data support a precisely timed cascade of complex molecular and subcellular transformations occurring from early to late MWM learning.


Subject(s)
Hippocampus/metabolism , Long-Term Potentiation/physiology , Maze Learning/physiology , Neuronal Plasticity/physiology , Animals , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Down-Regulation , Early Growth Response Protein 1/genetics , Female , Hippocampus/physiology , Mice , Mice, Inbred C57BL , Receptors, N-Methyl-D-Aspartate/genetics , Time Factors
10.
Elife ; 32014 Jun 02.
Article in English | MEDLINE | ID: mdl-24891237

ABSTRACT

Neuregulin 1 (NRG1) and the γ-secretase subunit APH1B have been previously implicated as genetic risk factors for schizophrenia and schizophrenia relevant deficits have been observed in rodent models with loss of function mutations in either gene. Here we show that the Aph1b-γ-secretase is selectively involved in Nrg1 intracellular signalling. We found that Aph1b-deficient mice display a decrease in excitatory synaptic markers. Electrophysiological recordings show that Aph1b is required for excitatory synaptic transmission and plasticity. Furthermore, gain and loss of function and genetic rescue experiments indicate that Nrg1 intracellular signalling promotes dendritic spine formation downstream of Aph1b-γ-secretase in vitro and in vivo. In conclusion, our study sheds light on the physiological role of Aph1b-γ-secretase in brain and provides a new mechanistic perspective on the relevance of NRG1 processing in schizophrenia.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Endopeptidases/metabolism , Gene Expression Regulation , Hippocampus/embryology , Neuregulin-1/metabolism , Alzheimer Disease/genetics , Animals , Brain/metabolism , Disease Models, Animal , Electrophysiology , Gene Deletion , Hippocampus/metabolism , Membrane Proteins , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , Patch-Clamp Techniques , Schizophrenia/metabolism , Signal Transduction , Synapses/metabolism
11.
EMBO J ; 30(24): 4955-69, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21926968

ABSTRACT

Synaptic transmission relies on effective and accurate compensatory endocytosis. F-BAR proteins may serve as membrane curvature sensors and/or inducers and thereby support membrane remodelling processes; yet, their in vivo functions urgently await disclosure. We demonstrate that the F-BAR protein syndapin I is crucial for proper brain function. Syndapin I knockout (KO) mice suffer from seizures, a phenotype consistent with excessive hippocampal network activity. Loss of syndapin I causes defects in presynaptic membrane trafficking processes, which are especially evident under high-capacity retrieval conditions, accumulation of endocytic intermediates, loss of synaptic vesicle (SV) size control, impaired activity-dependent SV retrieval and defective synaptic activity. Detailed molecular analyses demonstrate that syndapin I plays an important role in the recruitment of all dynamin isoforms, central players in vesicle fission reactions, to the membrane. Consistently, syndapin I KO mice share phenotypes with dynamin I KO mice, whereas their seizure phenotype is very reminiscent of fitful mice expressing a mutant dynamin. Thus, syndapin I acts as pivotal membrane anchoring factor for dynamins during regeneration of SVs.


Subject(s)
Neurons/physiology , Neuropeptides/physiology , Phosphoproteins/physiology , Synaptic Vesicles/physiology , Adaptor Proteins, Signal Transducing , Animals , Dynamins/metabolism , Endocytosis , Hippocampus/physiopathology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Neurons/ultrastructure , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Retina/physiology , Retina/ultrastructure , Retinal Rod Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/ultrastructure , Seizures/genetics , Synaptic Transmission , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure
12.
Biochim Biophys Acta ; 1768(11): 2714-25, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17716619

ABSTRACT

The Cl(-) channels of brown adipocytes electrophysiologically resemble outwardly rectifying Cl(-) channels (ORCC). To study tentative Ca(2+) regulation of these channels, we attempted to control Ca(2+) levels at the cytoplasmic side of the inside-out membrane patches with Ca(2+)-chelating agents. However, we found that the commonly used Ca(2+)-chelators EGTA and BAPTA by themselves influenced the Cl(-) channel currents, unrelated to their calcium chelating effects. Consequently, in this report we delineate effects of Ca(2+)-chelators (acting from the cytoplasmic side) on the single Cl(-) channel currents in patch-clamp experiments. Using fixed (1-2 mM) concentrations of chelators, two types of Cl(-) channels were identified, as discriminated by their reaction to the Ca(2+)-chelators and by their conductance: true-blockage channels (31 pS) and quasi-blockage channels (52 pS). In true-blockage channels, EGTA and BAPTA inhibited channel activity in a classical flickery type manner. In quasi-blockage channels, chelators significantly shortened the duration of individual openings, as in a flickering block, but the overall channel activity tended to increase. This dual effect of mean open time decrease accompanied by a tendency of open probability to increase we termed a quasi-blockage. Despite the complications due to the chelators as such, we could detect a moderate inhibitory effect of Ca(2+). The anionic classical Cl(-) channel blockers DIDS and SITS could mimic the true/quasi blockage of EGTA and BAPTA. It was concluded that at least in this experimental system, standard techniques for Ca(2+) level control in themselves could fundamentally affect the behaviour of Cl(-) channels.


Subject(s)
Adipocytes, Brown/metabolism , Calcium/physiology , Chelating Agents/pharmacology , Chloride Channels/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Animals , Cells, Cultured , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...