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










Publication year range
1.
JCI Insight ; 7(15)2022 08 08.
Article in English | MEDLINE | ID: mdl-35737457

ABSTRACT

Aging is known to be associated with hippocampus-dependent memory decline, but the underlying causes of this age-related memory impairment remain highly debated. Here, we show that fecal microbiota transplantation (FMT) from aged, but not young, animal donors into young mice is sufficient to trigger profound hippocampal alterations, including astrogliosis, decreased adult neurogenesis, decreased novelty-induced neuronal activation, and impairment in hippocampus-dependent memory. Furthermore, similar alterations were reported when mice were subjected to an FMT from aged human donors. To decipher the mechanisms involved in mediating these microbiota-induced effects on brain function, we mapped the vagus nerve-related (VN-related) neuronal activity patterns and report that aged FMT animals showed a reduction in neuronal activity in the ascending-VN output brain structure, whether under basal condition or after VN stimulation. Targeted pharmacogenetic manipulation of VN-ascending neurons demonstrated that the decrease in vagal activity is detrimental to hippocampal functions. In contrast, increasing vagal ascending activity alleviated the adverse effects of aged mouse FMT on hippocampal functions and had a promnesic effect in aged mice. Thus, pharmacogenetic VN stimulation is a potential therapeutic strategy to lessen microbiota-dependent age-associated impairments in hippocampal functions.


Subject(s)
Gastrointestinal Microbiome , Adult , Aged , Animals , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Hippocampus/physiology , Humans , Mice , Neurogenesis , Vagus Nerve
2.
Brain ; 145(4): 1391-1409, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35441215

ABSTRACT

Sepsis is a life-threatening condition induced by a deregulated host response to severe infection. Post-sepsis syndrome includes long-term psychiatric disorders, such as persistent anxiety and post-traumatic stress disorder, whose neurobiological mechanisms remain unknown. Using a reference mouse model of sepsis, we showed that mice that recovered from sepsis further developed anxiety-related behaviours associated with an exaggerated fear memory. In the brain, sepsis induced an acute pathological activation of a specific neuronal population of the central nucleus of the amygdala, which projects to the ventral bed nucleus of the stria terminalis. Using viral-genetic circuit tracing and in vivo calcium imaging, we observed that sepsis induced persistent changes in the connectivity matrix and in the responsiveness of these central amygdala neurons projecting to the ventral bed nucleus of the stria terminalis. The transient and targeted silencing of this subpopulation only during the acute phase of sepsis with a viral pharmacogenetic approach, or with the anti-epileptic and neuroprotective drug levetiracetam, prevented the subsequent development of anxiety-related behaviours. Specific inhibition of brain anxiety and fear circuits during the sepsis acute phase constitutes a preventive approach to preclude the post-infection psychiatric outcomes.


Subject(s)
Central Amygdaloid Nucleus , Sepsis , Animals , Anxiety , Anxiety Disorders , Fear/physiology , Humans , Mice , Sepsis/complications
3.
Cell Res ; 26(9): 1033-47, 2016 09.
Article in English | MEDLINE | ID: mdl-27325298

ABSTRACT

Once generated, neurons are thought to permanently exit the cell cycle and become irreversibly differentiated. However, neither the precise point at which this post-mitotic state is attained nor the extent of its irreversibility is clearly defined. Here we report that newly born neurons from the upper layers of the mouse cortex, despite initiating axon and dendrite elongation, continue to drive gene expression from the neural progenitor tubulin α1 promoter (Tα1p). These observations suggest an ambiguous post-mitotic neuronal state. Whole transcriptome analysis of sorted upper cortical neurons further revealed that neurons continue to express genes related to cell cycle progression long after mitotic exit until at least post-natal day 3 (P3). These genes are however down-regulated thereafter, associated with a concomitant up-regulation of tumor suppressors at P5. Interestingly, newly born neurons located in the cortical plate (CP) at embryonic day 18-19 (E18-E19) and P3 challenged with calcium influx are found in S/G2/M phases of the cell cycle, and still able to undergo division at E18-E19 but not at P3. At P5 however, calcium influx becomes neurotoxic and leads instead to neuronal loss. Our data delineate an unexpected flexibility of cell cycle control in early born neurons, and describe how neurons transit to a post-mitotic state.


Subject(s)
Cerebral Cortex/cytology , Mitosis , Neurons/cytology , Animals , Axons/drug effects , Axons/metabolism , Calcium/pharmacology , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Shape/drug effects , Dendrites/drug effects , Dendrites/metabolism , Mice , Mitosis/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Transcription, Genetic/drug effects
4.
Proc Natl Acad Sci U S A ; 112(23): 7291-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25995364

ABSTRACT

Repeated stress has been suggested to underlie learning and memory deficits via the basolateral amygdala (BLA) and the hippocampus; however, the functional contribution of BLA inputs to the hippocampus and their molecular repercussions are not well understood. Here we show that repeated stress is accompanied by generation of the Cdk5 (cyclin-dependent kinase 5)-activator p25, up-regulation and phosphorylation of glucocorticoid receptors, increased HDAC2 expression, and reduced expression of memory-related genes in the hippocampus. A combination of optogenetic and pharmacosynthetic approaches shows that BLA activation is both necessary and sufficient for stress-associated molecular changes and memory impairments. Furthermore, we show that this effect relies on direct glutamatergic projections from the BLA to the dorsal hippocampus. Finally, we show that p25 generation is necessary for the stress-induced memory dysfunction. Taken together, our data provide a neural circuit model for stress-induced hippocampal memory deficits through BLA activity-dependent p25 generation.


Subject(s)
Basolateral Nuclear Complex/physiopathology , Cyclin-Dependent Kinase 5/metabolism , Hippocampus/physiopathology , Learning Disabilities/physiopathology , Memory Disorders/physiopathology , Animals , Basolateral Nuclear Complex/radiation effects , Hippocampus/radiation effects , Light , Mice , Stress, Physiological
5.
Cell ; 156(1-2): 261-76, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439381

ABSTRACT

Traumatic events generate some of the most enduring forms of memories. Despite the elevated lifetime prevalence of anxiety disorders, effective strategies to attenuate long-term traumatic memories are scarce. The most efficacious treatments to diminish recent (i.e., day-old) traumata capitalize on memory updating mechanisms during reconsolidation that are initiated upon memory recall. Here, we show that, in mice, successful reconsolidation-updating paradigms for recent memories fail to attenuate remote (i.e., month-old) ones. We find that, whereas recent memory recall induces a limited period of hippocampal neuroplasticity mediated, in part, by S-nitrosylation of HDAC2 and histone acetylation, such plasticity is absent for remote memories. However, by using an HDAC2-targeting inhibitor (HDACi) during reconsolidation, even remote memories can be persistently attenuated. This intervention epigenetically primes the expression of neuroplasticity-related genes, which is accompanied by higher metabolic, synaptic, and structural plasticity. Thus, applying HDACis during memory reconsolidation might constitute a treatment option for remote traumata.


Subject(s)
Fear , Memory, Long-Term , Neuronal Plasticity , Animals , Epigenesis, Genetic , Hippocampus/metabolism , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Male , Memory, Long-Term/drug effects , Mice , Mice, Inbred C57BL , Transcriptome
6.
J Biol Chem ; 288(30): 22042-56, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23737518

ABSTRACT

Although Tau accumulation is a feature of several neurodegenerative conditions, treatment options for these conditions are nonexistent. Targeting Tau kinases represents a potential therapeutic approach. Small molecules in the diaminothiazole class are potent Tau kinase inhibitors that target CDK5 and GSK3ß. Lead compounds from the series have IC50 values toward CDK5/p25 and GSK3ß in the low nanomolar range and no observed toxicity in the therapeutic dose range. Neuronal protective effects and decreased PHF-1 immunoreactivity were observed in two animal models, 3×Tg-AD and CK-p25. Treatment nearly eliminated Sarkosyl-insoluble Tau with the most prominent effect on the phosphorylation at Ser-404. Treatment also induced the recovery of memory in a fear conditioning assay. Given the contribution of both CDK5/p25 and GSK3ß to Tau phosphorylation, effective treatment of tauopathies may require dual kinase targeting.


Subject(s)
Disease Models, Animal , Phosphorylation/drug effects , Tauopathies/prevention & control , Thiazoles/pharmacology , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Diamines/chemistry , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Injections, Intraperitoneal , Injections, Intraventricular , Injections, Subcutaneous , Learning/drug effects , Male , Memory/drug effects , Mice , Mice, Transgenic , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Tauopathies/genetics , Tauopathies/metabolism , Thiazoles/administration & dosage , Thiazoles/chemistry , Thiazoles/pharmacokinetics , Treatment Outcome , tau Proteins/genetics
7.
J Neurosci ; 33(21): 8951-60, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23699506

ABSTRACT

Caloric restriction (CR) is a dietary regimen known to promote lifespan by slowing down the occurrence of age-dependent diseases. The greatest risk factor for neurodegeneration in the brain is age, from which follows that CR might also attenuate the progressive loss of neurons that is often associated with impaired cognitive capacities. In this study, we used a transgenic mouse model that allows for a temporally and spatially controlled onset of neurodegeneration to test the potentially beneficial effects of CR. We found that in this model, CR significantly delayed the onset of neurodegeneration and synaptic loss and dysfunction, and thereby preserved cognitive capacities. Mechanistically, CR induced the expression of the known lifespan-regulating protein SIRT1, prompting us to test whether a pharmacological activation of SIRT1 might recapitulate CR. We found that oral administration of a SIRT1-activating compound essentially replicated the beneficial effects of CR. Thus, SIRT1-activating compounds might provide a pharmacological alternative to the regimen of CR against neurodegeneration and its associated ailments.


Subject(s)
Caloric Restriction/methods , Cognition Disorders/therapy , Neurodegenerative Diseases/complications , Sirtuin 1/metabolism , Analysis of Variance , Animals , Atrophy/etiology , Atrophy/prevention & control , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Case-Control Studies , Cognition Disorders/etiology , Cyclin-Dependent Kinase 5/genetics , Disease Models, Animal , Double-Blind Method , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Green Fluorescent Proteins/genetics , Immunoprecipitation , In Vitro Techniques , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Male , Memory Disorders/etiology , Memory Disorders/prevention & control , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/pathology , Phosphopyruvate Hydratase/metabolism , Phosphotransferases , Piperidines/therapeutic use , Silver Staining , Sirtuin 1/genetics , Synapses/pathology , Thiazoles/therapeutic use , Tumor Suppressor Protein p53/metabolism , Vitamin E/administration & dosage
8.
Nat Neurosci ; 15(7): 1022-31, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22683681

ABSTRACT

How neurons develop their morphology is an important question in neurobiology. Here we describe a new pathway that specifically affects the formation of basal dendrites and axonal projections in cortical pyramidal neurons. We report that thousand-and-one-amino acid 2 kinase (TAOK2), also known as TAO2, is essential for dendrite morphogenesis. TAOK2 downregulation impairs basal dendrite formation in vivo without affecting apical dendrites. Moreover, TAOK2 interacts with Neuropilin 1 (Nrp1), a receptor protein that binds the secreted guidance cue Semaphorin 3A (Sema3A). TAOK2 overexpression restores dendrite formation in cultured cortical neurons from Nrp1(Sema-) mice, which express Nrp1 receptors incapable of binding Sema3A. TAOK2 overexpression also ameliorates the basal dendrite impairment resulting from Nrp1 downregulation in vivo. Finally, Sema3A and TAOK2 modulate the formation of basal dendrites through the activation of the c-Jun N-terminal kinase (JNK). These results delineate a pathway whereby Sema3A and Nrp1 transduce signals through TAOK2 and JNK to regulate basal dendrite development in cortical neurons.


Subject(s)
Child Development Disorders, Pervasive/genetics , Dendrites/physiology , Genetic Predisposition to Disease/genetics , MAP Kinase Kinase Kinases/genetics , Neocortex/cytology , Neocortex/growth & development , Animals , Animals, Newborn , Cells, Cultured , Child , Child Development Disorders, Pervasive/pathology , Down-Regulation/genetics , Female , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/biosynthesis , Mice , Neocortex/pathology , Pregnancy , Protein Serine-Threonine Kinases , Rats
9.
Nature ; 483(7388): 222-6, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22388814

ABSTRACT

Cognitive decline is a debilitating feature of most neurodegenerative diseases of the central nervous system, including Alzheimer's disease. The causes leading to such impairment are only poorly understood and effective treatments are slow to emerge. Here we show that cognitive capacities in the neurodegenerating brain are constrained by an epigenetic blockade of gene transcription that is potentially reversible. This blockade is mediated by histone deacetylase 2, which is increased by Alzheimer's-disease-related neurotoxic insults in vitro, in two mouse models of neurodegeneration and in patients with Alzheimer's disease. Histone deacetylase 2 associates with and reduces the histone acetylation of genes important for learning and memory, which show a concomitant decrease in expression. Importantly, reversing the build-up of histone deacetylase 2 by short-hairpin-RNA-mediated knockdown unlocks the repression of these genes, reinstates structural and synaptic plasticity, and abolishes neurodegeneration-associated memory impairments. These findings advocate for the development of selective inhibitors of histone deacetylase 2 and suggest that cognitive capacities following neurodegeneration are not entirely lost, but merely impaired by this epigenetic blockade.


Subject(s)
Brain/physiopathology , Epigenesis, Genetic , Histone Deacetylase 2/genetics , Memory Disorders/genetics , Memory Disorders/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Acetylation/drug effects , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/toxicity , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Hippocampus/drug effects , Hippocampus/metabolism , Histone Deacetylase 2/deficiency , Histone Deacetylase 2/metabolism , Histones/metabolism , Humans , Hydrogen Peroxide/toxicity , Memory Disorders/complications , Mice , Neurodegenerative Diseases/complications , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Peptide Fragments/toxicity , Phosphorylation/drug effects , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , Receptors, Glucocorticoid/metabolism
11.
J Neurosci ; 31(44): 15751-6, 2011 Nov 02.
Article in English | MEDLINE | ID: mdl-22049418

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia, and is characterized by memory loss and cognitive decline, as well as amyloid ß (Aß) accumulation, and progressive neurodegeneration. Cdk5 is a proline-directed serine/threonine kinase whose activation by the p25 protein has been implicated in a number of neurodegenerative disorders. The CK-p25 inducible mouse model exhibits progressive neuronal death, elevated Aß, reduced synaptic plasticity, and impaired learning following p25 overexpression in forebrain neurons. Levels of Aß, as well as the APP processing enzyme, ß-secretase (BACE1), are also increased in CK-p25 mice. It is unknown what role increased Aß plays in the cognitive and neurodegenerative phenotype of the CK-p25 mouse. In the current work, we restored Aß levels in the CK-p25 mouse to those of wild-type mice via the partial genetic deletion of BACE1, allowing us to examine the Aß-independent phenotype of this mouse model. We show that, in the CK-p25 mouse, normalization of Aß levels led to a rescue of synaptic and cognitive deficits. Conversely, neuronal loss was not ameliorated. Our findings indicate that increases in p25/Cdk5 activity may mediate cognitive and synaptic impairment via an Aß-dependent pathway in the CK-p25 mouse. These findings explore the impact of targeting Aß production in a mouse model of neurodegeneration and cognitive impairment, and how this may translate into therapeutic approaches for sporadic AD.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Cyclin-Dependent Kinase 5/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/pathology , Synapses/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Avoidance Learning/physiology , Biophysics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Death/genetics , Disease Models, Animal , Electric Stimulation/methods , Enzyme-Linked Immunosorbent Assay/methods , Freezing Reaction, Cataleptic/physiology , Functional Laterality , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hippocampus/pathology , In Vitro Techniques , Long-Term Potentiation/genetics , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/therapy , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/complications , Patch-Clamp Techniques , Peptide Fragments/metabolism , Phosphorylation/genetics , Phosphotransferases , Prosencephalon/metabolism , Prosencephalon/pathology , Synapses/genetics , Synaptophysin/metabolism , tau Proteins/metabolism
12.
J Neurosci ; 30(31): 10391-406, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20685982

ABSTRACT

The mechanisms underlying the normal development of neuronal morphology remain a fundamental question in neurobiology. Studies in cultured neurons have suggested that the position of the centrosome and the Golgi may predict the site of axon outgrowth. During neuronal migration in the developing cortex, however, the centrosome and Golgi are oriented toward the cortical plate at a time when axons grow toward the ventricular zone. In the current work, we use in situ live imaging to demonstrate that the centrosome and the accompanying polarized cytoplasm exhibit apical translocation in newborn cortical neurons preceding initial axon outgrowth. Disruption of centrosomal activity or downregulation of the centriolar satellite protein PCM-1 affects axon formation. We further show that downregulation of the centrosomal protein Cep120 impairs microtubule organization, resulting in increased centrosome motility. Decreased centrosome motility resulting from microtubule stabilization causes an aberrant centrosomal localization, leading to misplaced axonal outgrowth. Our results reveal the dynamic nature of the centrosome in developing cortical neurons, and implicate centrosome translocation and microtubule organization during the multipolar stage as important determinants of axon formation.


Subject(s)
Axons/metabolism , Cell Movement/physiology , Centrosome/metabolism , Neocortex/metabolism , Neurons/metabolism , Analysis of Variance , Animals , Autoantigens/metabolism , Cell Cycle Proteins/metabolism , Cell Death , Cell Line , Cell Polarity , Cells, Cultured , Down-Regulation , Electroporation , Embryo Culture Techniques , Fluorescent Antibody Technique , Golgi Apparatus/metabolism , Humans , Mice , Microscopy, Confocal , Microtubules/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...