Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609585

ABSTRACT

The hippocampus is crucial for acquiring and retrieving episodic and contextual memories. In previous studies, the inactivation of dentate gyrus (DG) neurons by chemogenetic- and optogenetic-mediated hyperpolarization led to opposing conclusions about DG's role in memory retrieval. One study used Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-mediated clozapine N-oxide (CNO)-induced hyperpolarization and reported that the previously formed memory was erased, thus concluding that denate gyrus is needed for memory maintenance. The other study used optogenetic with halorhodopsin induced hyperpolarization and reported and dentate gyrus is needed for memory retrieval. We hypothesized that this apparent discrepancy could be due to the length of hyperpolarization in previous studies; minutes by optogenetics and several hours by DREADD/CNO. Since hyperpolarization interferes with anterograde and retrograde neuronal signaling, it is possible that the memory engram in the dentate gyrus and the entorhinal to hippocampus trisynaptic circuit was erased by long-term, but not with short-term hyperpolarization. We developed and applied an advanced chemogenetic technology to selectively silence synaptic output by blocking neurotransmitter release without hyperpolarizing DG neurons to explore this apparent discrepancy. We performed in vivo electrophysiology during trace eyeblink in a rabbit model of associative learning. Our work shows that the DG output is required for memory retrieval. Based on previous and recent findings, we propose that the actively functional anterograde and retrograde neuronal signaling is necessary to preserve synaptic memory engrams along the entorhinal cortex to the hippocampal trisynaptic circuit.

2.
iScience ; 26(11): 108050, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37876798

ABSTRACT

The organization of fear memory involves the participation of multiple brain regions. However, it is largely unknown how fear memory is formed, which circuit pathways are used for "printing" memory engrams across brain regions, and the role of identified brain circuits in memory retrieval. With advanced genetic methods, we combinatorially blocked presynaptic output and manipulated N-methyl-D-aspartate receptor (NMDAR) in the basolateral amygdala (BLA) and medial prefrontal cortex (mPFC) before and after cued fear conditioning. Further, we tagged fear-activated neurons during associative learning for optogenetic memory recall. We found that presynaptic mPFC and postsynaptic BLA NMDARs are required for fear memory formation, but not expression. Our results provide strong evidence that NMDAR-dependent synaptic plasticity drives multi-trace systems consolidation for the sequential printing of fear memory engrams from BLA to mPFC and, subsequently, to the other regions, for flexible memory retrieval.

3.
J Cereb Blood Flow Metab ; 42(1): 104-120, 2022 01.
Article in English | MEDLINE | ID: mdl-34427142

ABSTRACT

Gene vectors targeting CNS endothelial cells allow to manipulate the blood-brain barrier and to correct genetic defects in the CNS. Because vectors based on the adeno-associated virus (AAV) have a limited capacity, it is essential that the DNA sequence controlling gene expression is short. In addition, it must be specific for endothelial cells to avoid off-target effects. To develop improved regulatory sequences with selectivity for brain endothelial cells, we tested the transcriptional activity of truncated promoters of eleven (brain) endothelial-specific genes in combination with short regulatory elements, i.e., the woodchuck post-transcriptional regulatory element (W), the CMV enhancer element (C), and a fragment of the first intron of the Tie2 gene (S), by transfecting brain endothelial cells of three species. Four combinations of regulatory elements and short promoters (Cdh5, Ocln, Slc2a1, and Slco1c1) progressed through this in-vitro pipeline displaying suitable activity. When tested in mice, the regulatory sequences C-Ocln-W and C-Slc2a1-S-W enabled a stronger and more specific gene expression in brain endothelial cells than the frequently used CAG promoter. In summary, the new regulatory elements efficiently control gene expression in brain endothelial cells and may help to specifically target the blood-brain barrier with gene therapy vectors.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Gene Expression , Gene Targeting , Genetic Therapy , Response Elements , Transfection , Animals , Mice
4.
Cereb Cortex ; 31(1): 281-300, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32885230

ABSTRACT

It is assumed that the claustrum (CL) is involved in sensorimotor integration and cognitive processes. We recorded the firing activity of identified CL neurons during classical eyeblink conditioning in rabbits, using a delay paradigm in which a tone was presented as conditioned stimulus (CS), followed by a corneal air puff as unconditioned stimulus (US). Neurons were identified by their activation from motor (MC), cingulate (CC), and medial prefrontal (mPFC) cortices. CL neurons were rarely activated by single stimuli of any modality. In contrast, their firing was significantly modulated during the first sessions of paired CS/US presentations, but not in well-trained animals. Neuron firing rates did not correlate with the kinematics of conditioned responses (CRs). CL local field potentials (LFPs) changed their spectral power across learning and presented well-differentiated CL-mPFC/CL-MC network dynamics, as shown by crossfrequency spectral measurements. CL electrical stimulation did not evoke eyelid responses, even in trained animals. Silencing of synaptic transmission of CL neurons by the vINSIST method delayed the acquisition of CRs but did not affect their presentation rate. The CL plays an important role in the acquisition of associative learning, mostly in relation to the novelty of CS/US association, but not in the expression of CRs.


Subject(s)
Action Potentials/physiology , Cognition/physiology , Conditioning, Classical/physiology , Eyelids/physiology , Animals , Blinking/physiology , Conditioning, Eyelid/physiology , Electric Stimulation/methods , Neurons/physiology , Prefrontal Cortex/physiology , Rabbits
5.
J Cereb Blood Flow Metab ; 40(6): 1338-1350, 2020 06.
Article in English | MEDLINE | ID: mdl-31357902

ABSTRACT

In Tay-Sachs and Sandhoff disease, a deficiency of the lysosomal enzyme ß-hexosaminidase causes GM2 and other gangliosides to accumulate in neurons and triggers neurodegeneration. Although the pathology centers on neurons, ß-hexosaminidase is mainly expressed outside of neurons, suggesting that gene therapy of these diseases should target non-neuronal cells to reconstitute physiological conditions. Here, we tested in Hexb-/- mice, a model of Sandhoff disease, to determine whether endothelial expression of the genes for human ß-hexosaminidase subunit A and B (HEXA, HEXB) is able to reduce disease symptoms and prolong survival of the affected mice. The brain endothelial selective vectors AAV-BR1-CAG-HEXA and AAV-BR1-CAG-HEXB transduced brain endothelial cells, which subsequently released ß-hexosaminidase enzyme. In vivo intravenous administration of the gene vectors to adult and neonatal mice prolonged survival. They improved neurological function and reduced accumulation of the ganglioside GM2 and the glycolipid GA2 as well as astrocytic activation. Overall, the data demonstrate that endothelial cells are a suitable target for intravenous gene therapy of GM2 gangliosidoses and possibly other lysosomal storage disorders.


Subject(s)
Endothelial Cells , Genetic Therapy/methods , Sandhoff Disease , beta-Hexosaminidase alpha Chain/administration & dosage , beta-Hexosaminidase beta Chain/administration & dosage , Animals , Brain , Dependovirus , Disease Models, Animal , Genetic Vectors , Humans , Mice , Mice, Knockout , Transduction, Genetic , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
6.
Neuron ; 103(1): 133-146.e8, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31104950

ABSTRACT

Oxytocin (OT) release by axonal terminals onto the central nucleus of the amygdala exerts anxiolysis. To investigate which subpopulation of OT neurons contributes to this effect, we developed a novel method: virus-delivered genetic activity-induced tagging of cell ensembles (vGATE). With the vGATE method, we identified and permanently tagged a small subpopulation of OT cells, which, by optogenetic stimulation, strongly attenuated contextual fear-induced freezing, and pharmacogenetic silencing of tagged OT neurons impaired context-specific fear extinction, demonstrating that the tagged OT neurons are sufficient and necessary, respectively, to control contextual fear. Intriguingly, OT cell terminals of fear-experienced rats displayed enhanced glutamate release in the amygdala. Furthermore, rats exposed to another round of fear conditioning displayed 5-fold more activated magnocellular OT neurons in a novel environment than a familiar one, possibly for a generalized fear response. Thus, our results provide first evidence that hypothalamic OT neurons represent a fear memory engram.


Subject(s)
Fear/physiology , Hypothalamus/physiology , Memory/physiology , Oxytocin/physiology , Amygdala/metabolism , Amygdala/physiology , Animals , Environment , Extinction, Psychological/physiology , Fear/psychology , Female , Freezing Reaction, Cataleptic , Gene Silencing , Glutamic Acid/metabolism , Hypothalamus/cytology , Neuronal Plasticity/physiology , Neurons/physiology , Optogenetics , Oxytocin/genetics , Rats , Rats, Wistar
7.
Ann Neurol ; 82(1): 93-104, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28628231

ABSTRACT

OBJECTIVE: Incontinentia pigmenti (IP) is a genetic disease leading to severe neurological symptoms, such as epileptic seizures, but no specific treatment is available. IP is caused by pathogenic variants that inactivate the Nemo gene. Replacing Nemo through gene therapy might provide therapeutic benefits. METHODS: In a mouse model of IP, we administered a single intravenous dose of the adeno-associated virus (AAV) vector, AAV-BR1-CAG-NEMO, delivering the Nemo gene to the brain endothelium. Spontaneous epileptic seizures and the integrity of the blood-brain barrier (BBB) were monitored. RESULTS: The endothelium-targeted gene therapy improved the integrity of the BBB. In parallel, it reduced the incidence of seizures and delayed their occurrence. Neonate mice intravenously injected with the AAV-BR1-CAG-NEMO vector developed no hepatocellular carcinoma or other major adverse effects 11 months after vector injection, demonstrating that the vector has a favorable safety profile. INTERPRETATION: The data show that the BBB is a target of antiepileptic treatment and, more specifically, provide evidence for the therapeutic benefit of a brain endothelial-targeted gene therapy in IP. Ann Neurol 2017;82:93-104.


Subject(s)
Genetic Therapy , Incontinentia Pigmenti/therapy , Intracellular Signaling Peptides and Proteins/genetics , Seizures/therapy , Animals , Blood-Brain Barrier/metabolism , Cells, Cultured , Dependovirus , Female , Genetic Vectors/adverse effects , Humans , Incontinentia Pigmenti/complications , Male , Mice , Mice, Knockout , Permeability , Seizures/complications
8.
EMBO Mol Med ; 8(6): 609-25, 2016 06.
Article in English | MEDLINE | ID: mdl-27137490

ABSTRACT

Gene therapy critically relies on vectors that combine high transduction efficiency with a high degree of target specificity and that can be administered through a safe intravenous route. The lack of suitable vectors, especially for gene therapy of brain disorders, represents a major obstacle. Therefore, we applied an in vivo screening system of random ligand libraries displayed on adeno-associated viral capsids to select brain-targeted vectors for the treatment of neurovascular diseases. We identified a capsid variant showing an unprecedented degree of specificity and long-lasting transduction efficiency for brain microvasculature endothelial cells as the primary target of selection. A therapeutic vector based on this selected viral capsid was used to markedly attenuate the severe cerebrovascular pathology of mice with incontinentia pigmenti after a single intravenous injection. Furthermore, the versatility of this selection system will make it possible to select ligands for additional in vivo targets without requiring previous identification of potential target-specific receptors.


Subject(s)
Brain/pathology , Dependovirus/genetics , Endothelial Cells/pathology , Genetic Therapy/methods , Genetic Vectors , Incontinentia Pigmenti/therapy , Microvessels/pathology , Animals , Disease Models, Animal , Injections, Intravenous , Mice , Transduction, Genetic , Treatment Outcome
9.
Mol Ther Nucleic Acids ; 5: e309, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27070301

ABSTRACT

Controlling gene expression in mammalian brain is of utmost importance to causally link the role of gene function to cell circuit dynamics under normal conditions and disease states. We have developed recombinant adeno-associated viruses equipped with tetracycline-controlled genetic switches for inducible and reversible control of gene expression in a cell type specific and brain subregion selective manner. Here, we characterize a two-virus approach to efficiently and reliably switch gene expression on and off, repetitively, both in vitro and in vivo. Our recombinant adeno-associated virus (rAAV)-Tet approach is highly flexible and it has great potential for application in basic and biomedical neuroscience research and gene therapy.

10.
Front Cell Neurosci ; 9: 142, 2015.
Article in English | MEDLINE | ID: mdl-25954155

ABSTRACT

We have deployed recombinant adeno-associated viruses equipped with tetracycline-controlled genetic switches to manipulate gene expression in mouse brain. Here, we show a combinatorial genetic approach for inducible, cell type-specific gene expression and Cre/loxP mediated gene recombination in different brain regions. Our chemical-genetic approach will help to investigate 'when', 'where', and 'how' gene(s) control neuronal circuit dynamics, and organize, for example, sensory signal processing, learning and memory, and behavior.

11.
BMC Neurosci ; 15: 38, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24618040

ABSTRACT

BACKGROUND: Bone morphogenic proteins (BMPs) promote the survival of neurons, suggesting a therapeutic application of BMPs in the treatment of acute and chronic neurodegenerative disorders. However, the application of recombinant BMPs in vivo is limited by their short half-life. To provide a continuous supply for functionally active BMPs, we expressed BMP7, BMP2 and the BMP inhibitor Noggin under the control of rAAV vectors in vivo. For visual control of rAAV-mediated BMP (v-BMP) expression we fused the secreted morphogenic polypeptides and the fluorescent reporter protein Venus via the 'ribosomal skip' promoting 2A peptide-bridge. RESULTS: In primary cortical neurons, the rAAV-expressed morphogenic polypeptides were efficiently released from the 2A-Venus fusion precursors, were secreted, correctly processed and functionally active as shown by their effects on Smad phosphorylation in HeLa cells and in primary neurons, by the protection of v-BMP7-transduced primary cortical neurons against oxidative stress, and by the activation of BMP responsive GFP in v-BMP2 transduced reporter mice. In the stroke model of middle cerebral artery occlusion rAAV-transduced v-BMP7 reduced the infarct size in mice. CONCLUSION: Polycistronic rAAV vectors encoding secreted polypeptides and 2A-linked reporter proteins are potential novel therapeutic tools for the treatment of neurological and neurodegenerative diseases. Using this technique we documented that rAAV delivery of BMP7 reduced ischemic cell death in mice.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/therapeutic use , Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Dependovirus/genetics , Neurons/metabolism , Transduction, Genetic/methods , Animals , Cerebral Cortex/metabolism , Genetic Vectors/genetics , HeLa Cells , Humans , Mice , Neuroprotective Agents/therapeutic use , Treatment Outcome
12.
Nat Commun ; 4: 2258, 2013.
Article in English | MEDLINE | ID: mdl-23978820

ABSTRACT

The primary motor cortex has an important role in the precise execution of learned motor responses. During motor learning, synaptic efficacy between sensory and primary motor cortical neurons is enhanced, possibly involving long-term potentiation and N-methyl-D-aspartate (NMDA)-specific glutamate receptor function. To investigate whether NMDA receptor in the primary motor cortex can act as a coincidence detector for activity-dependent changes in synaptic strength and associative learning, here we generate mice with deletion of the Grin1 gene, encoding the essential NMDA receptor subunit 1 (GluN1), specifically in the primary motor cortex. The loss of NMDA receptor function impairs primary motor cortex long-term potentiation in vivo. Importantly, it impairs the synaptic efficacy between the primary somatosensory and primary motor cortices and significantly reduces classically conditioned eyeblink responses. Furthermore, compared with wild-type littermates, mice lacking NMDA receptors in the [corrected] primary motor cortex show slower learning in Skinner-box tasks. Thus, primary motor cortex NMDA receptors are necessary for activity-dependent synaptic strengthening and associative learning.


Subject(s)
Behavior, Animal/physiology , Learning , Motor Cortex/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Conditioning, Operant/physiology , Electrophysiological Phenomena , Gene Deletion , Gene Knockout Techniques , Integrases/metabolism , Long-Term Potentiation/physiology , Mice , Nerve Tissue Proteins/genetics , Organ Specificity , Receptors, N-Methyl-D-Aspartate/genetics , Task Performance and Analysis
13.
Mol Ther ; 21(8): 1497-506, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23774792

ABSTRACT

Both genetic inactivation and pharmacological inhibition of the cholesteryl ester synthetic enzyme acyl-CoA:cholesterol acyltransferase 1 (ACAT1) have shown benefit in mouse models of Alzheimer's disease (AD). In this study, we aimed to test the potential therapeutic applications of adeno-associated virus (AAV)-mediated Acat1 gene knockdown in AD mice. We constructed recombinant AAVs expressing artificial microRNA (miRNA) sequences, which targeted Acat1 for knockdown. We demonstrated that our AAVs could infect cultured mouse neurons and glia and effectively knockdown ACAT activity in vitro. We next delivered the AAVs to mouse brains neurosurgically, and demonstrated that Acat1-targeting AAVs could express viral proteins and effectively diminish ACAT activity in vivo, without inducing appreciable inflammation. We delivered the AAVs to the brains of 10-month-old AD mice and analyzed the effects on the AD phenotype at 12 months of age. Acat1-targeting AAV delivered to the brains of AD mice decreased the levels of brain amyloid-ß and full-length human amyloid precursor protein (hAPP), to levels similar to complete genetic ablation of Acat1. This study provides support for the potential therapeutic use of Acat1 knockdown gene therapy in AD.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , MicroRNAs/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Dependovirus/genetics , Disease Models, Animal , Female , Gene Expression , Gene Knockdown Techniques , Gene Order , Genetic Therapy , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Knockout , MicroRNAs/metabolism , Neurons/metabolism , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...