Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38719448

ABSTRACT

Physical exercise is a robust lifestyle intervention known for its enhancement of cognitive abilities. Nevertheless, the extent to which these benefits can be transmitted across generations (intergenerational inheritance to F1, and transgenerational to F2 and beyond) remains a topic of limited comprehension. We have already shown that cognitive improvements resulting from physical exercise can be inherited from parents to their offspring, proving intergenerational effects. So, we set out to explore whether these enhancements might extend transgenerationally, impacting the F2 generation. In this study, we initially examined the behavioral traits of second generation (F2) male mice, whose grandfathers (F0) had an exercise intervention. Our findings revealed that F2 mice with physically active grandpaternal F0 progenitors displayed significantly improved memory recall, encompassing both spatial and non-spatial information when compared to their counterparts from sedentary F0 progenitors, and proving for the first time the transgenerational inheritance of physical exercise induced cognitive enhancement. Surprisingly, while F2 memory improved (as was the case with F1), adult hippocampal neurogenesis remained unchanged between experimental and control groups (unlike in F1). Additionally, our analysis of small RNA sequences in the hippocampus identified 35 differentially expressed miRNAs linked to important brain function categories. Notably, two of these miRNAs, miRNA-144 and miRNA-298, displayed a robust negative correlation with cognitive performance. These findings highlight the enduring transgenerational transmission of cognitive benefits associated with exercise, even after two generations, suggesting that moderate exercise training can have lasting positive effects, possibly orchestrated by a specific set of miRNAs that exert their influence across multiple generations.


Subject(s)
Cognition , Hippocampus , Physical Conditioning, Animal , Animals , Male , Mice , Cognition/physiology , Physical Conditioning, Animal/physiology , Hippocampus/physiology , Hippocampus/metabolism , Female , Neurogenesis/physiology , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics
2.
Int J Mol Sci ; 25(8)2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38673915

ABSTRACT

Parkinson's disease (PD) is a chronic, age-related, progressive multisystem disease associated with neuroinflammation and immune dysfunction. This review discusses the methodological approaches used to study the changes in central and peripheral immunity in PD, the advantages and limitations of the techniques, and their applicability to humans. Although a single animal model cannot replicate all pathological features of the human disease, neuroinflammation is present in most animal models of PD and plays a critical role in understanding the involvement of the immune system (IS) in the pathogenesis of PD. The IS and its interactions with different cell types in the central nervous system (CNS) play an important role in the pathogenesis of PD. Even though culture models do not fully reflect the complexity of disease progression, they are limited in their ability to mimic long-term effects and need validation through in vivo studies. They are an indispensable tool for understanding the interplay between the IS and the pathogenesis of this disease. Understanding the immune-mediated mechanisms may lead to potential therapeutic targets for the treatment of PD. We believe that the development of methodological guidelines for experiments with animal models and PD patients is crucial to ensure the validity and consistency of the results.


Subject(s)
Disease Models, Animal , Parkinson Disease , Parkinson Disease/immunology , Parkinson Disease/pathology , Parkinson Disease/etiology , Animals , Humans , Immune System/immunology , Immune System/metabolism , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology
3.
Front Neurosci ; 17: 1335276, 2023.
Article in English | MEDLINE | ID: mdl-38089967
4.
Int J Mol Sci ; 23(22)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36430497

ABSTRACT

The relationship between Parkinson's disease (PD), the second-most common neurodegenerative disease after Alzheimer's disease, and palmitoylation, a post-translational lipid modification, is not well understood. In this study, to better understand the role of protein palmitoylation in PD and the pathways altered in this disease, we analyzed the differential palmitoyl proteome (palmitome) in the cerebral cortex of PD patients compared to controls (n = 4 per group). Data-mining of the cortical palmitome from PD patients and controls allowed us to: (i) detect a set of 150 proteins with altered palmitoylation in PD subjects in comparison with controls; (ii) describe the biological pathways and targets predicted to be altered by these palmitoylation changes; and (iii) depict the overlap between the differential palmitome identified in our study with protein interactomes of the PD-linked proteins α-synuclein, LRRK2, DJ-1, PINK1, GBA and UCHL1. In summary, we partially characterized the altered palmitome in the cortex of PD patients, which is predicted to impact cytoskeleton, mitochondrial and fibrinogen functions, as well as cell survival. Our study suggests that protein palmitoylation could have a role in the pathophysiology of PD, and that comprehensive palmitoyl-proteomics offers a powerful approach for elucidating novel cellular pathways modulated in this neurodegenerative disease.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Parkinson Disease/metabolism , Lipoylation , Neurodegenerative Diseases/metabolism , Cerebral Cortex/metabolism , Mitochondria/metabolism
5.
Int J Mol Sci ; 23(10)2022 May 11.
Article in English | MEDLINE | ID: mdl-35628158

ABSTRACT

Neuroinflammation underlies neurodegenerative diseases. Herein, we test whether acute colon inflammation activates microglia and astrocytes, induces neuroinflammation, disturbs neuron intrinsic electrical properties in the primary motor cortex, and alters motor behaviors. We used a rat model of acute colon inflammation induced by dextran sulfate sodium. Inflammatory mediators and microglial activation were assessed in the primary motor cortex by PCR and immunofluorescence assays. Electrophysiological properties of the motor cortex neurons were determined by whole-cell patch-clamp recordings. Motor behaviors were examined using open-field and rotarod tests. We show that the primary motor cortex of rats with acute colon inflammation exhibited microglial and astrocyte activation and increased mRNA abundance of interleukin-6, tumor necrosis factor-alpha, and both inducible and neuronal nitric oxide synthases. These changes were accompanied by a reduction in resting membrane potential and rheobase and increased input resistance and action potential frequency, indicating motor neuron hyperexcitability. In addition, locomotion and motor coordination were impaired. In conclusion, acute colon inflammation induces motor cortex microglial and astrocyte activation and inflammation, which led to neurons' hyperexcitability and reduced motor coordination performance. The described disturbances resembled some of the early features found in amyotrophic lateral sclerosis patients and animal models, suggesting that colon inflammation might be a risk factor for developing this disease.


Subject(s)
Colitis , Motor Cortex , Animals , Colitis/chemically induced , Colitis/pathology , Humans , Inflammation/pathology , Motor Cortex/pathology , Motor Neurons/pathology , Neuroinflammatory Diseases , Rats
6.
J Neurosci ; 41(32): 6836-6849, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34210778

ABSTRACT

Adult neural plasticity is an important and intriguing phenomenon in the brain, and adult hippocampal neurogenesis is directly involved in modulating neural plasticity by mechanisms that are only partially understood. We have performed gain-of-function and loss-of-function experiments to study Smad2, a transcription factor selected from genes that are demethylated after exercise through the analysis of an array of physical activity-induced factors, and their corresponding gene expression, and an efficient inducer of plasticity. In these studies, changes in cell number and morphology were analyzed in the hippocampal dentate gyrus (cell proliferation and survival, including regional distribution, and structural maturation/differentiation, including arborization, dendritic spines, and neurotransmitter-specific vesicles) of sedentary male mice, after evaluation in a battery of behavioral tests. As a result, we reveal a role for Smad2 in the balance of proliferation versus maturation of differentiating immature cells (Smad2 silencing increases both the proliferation and survival of cycling cells in the dentate granule cell layer), and in the plasticity of both newborn and mature neurons in mice (by decreasing dendritic arborization and dendritic spine number). Moreover, Smad2 silencing specifically compromises spatial learning in mice (through impairments of spatial tasks acquisition both in long-term learning and working memory). These data suggest that Smad2 participates in adult neural plasticity by influencing the proliferation and maturation of dentate gyrus neurons.SIGNIFICANCE STATEMENT Smad2 is one of the main components of the transforming growth factor-ß (TGF-ß) pathway. The commitment of cell fate in the nervous system is tightly coordinated by SMAD2 signaling, as are further differentiation steps (e.g., dendrite and axon growth, myelination, and synapse formation). However, there are no studies that have directly evaluated the role of Smad2 gene in hippocampus of adult animals. Modulation of these parameters in the adult hippocampus can affect hippocampal-dependent behaviors, which may shed light on the mechanisms that regulate adult neurogenesis and behavior. We demonstrate here a role for Smad2 in the maturation of differentiating immature cells and in the plasticity of mature neurons. Moreover, Smad2 silencing specifically compromises the spatial learning abilities of adult male mice.


Subject(s)
Dentate Gyrus/physiology , Neuronal Plasticity/physiology , Smad2 Protein/metabolism , Spatial Learning/physiology , Spatial Memory/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Neurogenesis/physiology
7.
Front Oncol ; 10: 602217, 2020.
Article in English | MEDLINE | ID: mdl-33330101

ABSTRACT

Neural stem cells (NSCs) persist in the adult mammalian brain in two neurogenic regions: the subventricular zone lining the lateral ventricles and the dentate gyrus of the hippocampus. Compelling evidence suggests that NSCs of the subventricular zone could be the cell type of origin of glioblastoma, the most devastating brain tumor. Studies in glioblastoma patients revealed that NSCs of the tumor-free subventricular zone, harbor cancer-driver mutations that were found in the tumor cells but were not present in normal cortical tissue. Endogenous mutagenesis can also take place in hippocampal NSCs. However, to date, no conclusive studies have linked hippocampal mutations with glioblastoma development. In addition, glioblastoma cells often invade or are closely located to the subventricular zone, whereas they do not tend to infiltrate into the hippocampus. In this review we will analyze possible causes by which subventricular zone NSCs might be more susceptible to malignant transformation than their hippocampal counterparts. Cellular and molecular differences between the two neurogenic niches, as well as genotypic and phenotypic characteristics of their respective NSCs will be discussed regarding why the cell type originating glioblastoma brain tumors has been linked mainly to subventricular zone, but not to hippocampal NSCs.

8.
Proc Natl Acad Sci U S A ; 116(20): 10103-10112, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31010925

ABSTRACT

Physical exercise has positive effects on cognition, but very little is known about the inheritance of these effects to sedentary offspring and the mechanisms involved. Here, we use a patrilineal design in mice to test the transmission of effects from the same father (before or after training) and from different fathers to compare sedentary- and runner-father progenies. Behavioral, stereological, and whole-genome sequence analyses reveal that paternal cognition improvement is inherited by the offspring, along with increased adult neurogenesis, greater mitochondrial citrate synthase activity, and modulation of the adult hippocampal gene expression profile. These results demonstrate the inheritance of exercise-induced cognition enhancement through the germline, pointing to paternal physical activity as a direct factor driving offspring's brain physiology and cognitive behavior.


Subject(s)
Brain/physiology , Cognition/physiology , Fathers/psychology , Paternal Inheritance , Running/physiology , Animals , Female , Gene Expression , Male , Mice , Pregnancy
9.
Proc Natl Acad Sci U S A ; 115(45): 11625-11630, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30352848

ABSTRACT

Increasing age is the greatest known risk factor for the sporadic late-onset forms of neurodegenerative disorders such as Alzheimer's disease (AD). One of the brain regions most severely affected in AD is the hippocampus, a privileged structure that contains adult neural stem cells (NSCs) with neurogenic capacity. Hippocampal neurogenesis decreases during aging and the decrease is exacerbated in AD, but the mechanistic causes underlying this progressive decline remain largely unexplored. We here investigated the effect of age on NSCs and neurogenesis by analyzing the senescence accelerated mouse prone 8 (SAMP8) strain, a nontransgenic short-lived strain that spontaneously develops a pathological profile similar to that of AD and that has been employed as a model system to study the transition from healthy aging to neurodegeneration. We show that SAMP8 mice display an accelerated loss of the NSC pool that coincides with an aberrant rise in BMP6 protein, enhanced canonical BMP signaling, and increased astroglial differentiation. In vitro assays demonstrate that BMP6 severely impairs NSC expansion and promotes NSC differentiation into postmitotic astrocytes. Blocking the dysregulation of the BMP pathway and its progliogenic effect in vivo by intracranial delivery of the antagonist Noggin restores hippocampal NSC numbers, neurogenesis, and behavior in SAMP8 mice. Thus, manipulating the local microenvironment of the NSC pool counteracts hippocampal dysfunction in pathological aging. Our results shed light on interventions that may allow taking advantage of the brain's natural plastic capacity to enhance cognitive function in late adulthood and in chronic neurodegenerative diseases such as AD.


Subject(s)
Aging/genetics , Alzheimer Disease/drug therapy , Bone Morphogenetic Protein 6/genetics , Carrier Proteins/pharmacology , Neural Stem Cells/drug effects , Neuroprotective Agents/pharmacology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/metabolism , Cell Differentiation , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Injections, Intraventricular , Male , Mice , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Signal Transduction
10.
Neurobiol Aging ; 56: 50-66, 2017 08.
Article in English | MEDLINE | ID: mdl-28494436

ABSTRACT

A controversy exists as to whether de novo-generated neuronal tetraploidy (dnNT) occurs in Alzheimer's disease. In addition, the presence of age-associated dnNT in the normal brain remains unexplored. Here we show that age-associated dnNT occurs in both superficial and deep layers of the cerebral cortex of adult mice, a process that is blocked in the absence of E2F1, a known regulator of cell cycle progression. This blockage correlates with improved cognition despite compromised neurogenesis in the adult hippocampus was confirmed in mice lacking the E2f1 gene. We also show that the human cerebral cortex contains tetraploid neurons. In normal humans, age-associated dnNT specifically occurs in the entorhinal cortex whereas, in Alzheimer, dnNT also affects association cortices prior to neurofibrillary tangle formation. Alzheimer-associated dnNT is likely potentiated by altered amyloid precursor protein (APP) processing as it is enhanced in the cerebral cortex of young APPswe/PS1deltaE9 mice, long before the first amyloid plaques are visible in their brains. In contrast to age-associated dnNT, enhanced dnNT in APPswe/PS1deltaE9 mice mostly affects the superficial cortical layers. The correlation of dnNT with reduced cognition in mice and its spatiotemporal course, preceding and recapitulating Alzheimer-associated neuropathology, makes this process a potential target for intervention in Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cerebral Cortex/pathology , Cognition/physiology , Neurons/pathology , Tetraploidy , Aged , Aged, 80 and over , Aging/genetics , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Cycle/genetics , Cerebral Cortex/cytology , E2F1 Transcription Factor/physiology , Female , Hippocampus , Humans , Male , Mice, Transgenic , Middle Aged , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Neurogenesis/genetics
11.
Front Neurosci ; 10: 93, 2016.
Article in English | MEDLINE | ID: mdl-27013955

ABSTRACT

Exercise can make you smarter, happier and have more neurons depending on the dose (intensity) of the training program. It is well recognized that exercise protocols induce both positive and negative effects depending on the intensity of the exercise, among other key factors, a process described as a hormetic-like biphasic dose-response. However, no evidences have been reported till very recently about the biphasic response of some of the potential mediators of the exercise-induced actions. This hypothesis and theory will focus on the adult hippocampal neurogenesis (AHN) as a putative physical substrate for hormesis responses to exercise in the context of exercise-induced actions on cognition and mood, and on the molecular pathways which might potentially be mediating these actions.

12.
PLoS One ; 6(12): e28927, 2011.
Article in English | MEDLINE | ID: mdl-22174927

ABSTRACT

Defects in the ubiquitin-proteasome system have been related to aging and the development of neurodegenerative disease, although the effects of deficient proteasome activity during early postnatal development are poorly understood. Accordingly, we have assessed how proteasome dysfunction during early postnatal development, induced by administering proteasome inhibitors daily during the first 10 days of life, affects the behaviour of adult mice. We found that this regime of exposure to the proteasome inhibitors MG132 or lactacystin did not produce significant behavioural or morphological changes in the first 15 days of life. However, towards the end of the treatment with proteasome inhibitors, there was a loss of mitochondrial markers and activity, and an increase in DNA oxidation. On reaching adulthood, the memory of mice that were injected with proteasome inhibitors postnatally was impaired in hippocampal and amygdala-dependent tasks, and they suffered motor dysfunction and imbalance. These behavioural deficiencies were correlated with neuronal loss in the hippocampus, amygdala and brainstem, and with diminished adult neurogenesis. Accordingly, impairing proteasome activity at early postnatal ages appears to cause morphological and behavioural alterations in adult mice that resemble those associated with certain neurodegenerative diseases and/or syndromes of mental retardation.


Subject(s)
Cognition Disorders/complications , Nerve Degeneration/complications , Nervous System/growth & development , Nervous System/pathology , Proteasome Inhibitors , Amygdala/drug effects , Amygdala/pathology , Amygdala/physiopathology , Animals , Animals, Newborn , Ataxia/complications , Ataxia/physiopathology , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/physiopathology , DNA/metabolism , Depression/complications , Depression/physiopathology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Leupeptins/administration & dosage , Leupeptins/pharmacology , Memory/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Motor Activity/drug effects , Nerve Degeneration/physiopathology , Nervous System/drug effects , Oxidation-Reduction/drug effects , Proteasome Endopeptidase Complex/metabolism , Ubiquitinated Proteins/metabolism
13.
PLoS One ; 6(9): e24915, 2011.
Article in English | MEDLINE | ID: mdl-21966384

ABSTRACT

Synaptic plasticity involves short- and long-term events, although the molecular mechanisms that underlie these processes are not fully understood. The transient A-type K(+) current (I(A)) controls the excitability of the dendrites from CA1 pyramidal neurons by regulating the back-propagation of action potentials and shaping synaptic input. Here, we have studied how decreases in I(A) affect cognitive processes and synaptic plasticity. Using wild-type mice treated with 4-AP, an I(A) inhibitor, and mice lacking the DREAM protein, a transcriptional repressor and modulator of the I(A), we demonstrate that impairment of I(A) decreases the stimulation threshold for learning and the induction of early-LTP. Hippocampal electrical recordings in both models revealed alterations in basal electrical oscillatory properties toward low-theta frequencies. In addition, we demonstrated that the facilitated learning induced by decreased I(A) requires the activation of NMDA receptors containing the NR2B subunit. Together, these findings point to a balance between the I(A) and the activity of NR2B-containing NMDA receptors in the regulation of learning.


Subject(s)
CA1 Region, Hippocampal/metabolism , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/genetics , Action Potentials , Animals , Behavior, Animal , Electrophysiology/methods , Memory , Mice , Mice, Transgenic , Models, Biological , Models, Genetic , Oscillometry/methods , Potassium/metabolism , Protein Structure, Tertiary , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission
14.
Hippocampus ; 21(1): 22-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-19921704

ABSTRACT

Aging, mental retardation, number of psychiatric and neurological disorders are all associated with learning and memory impairments. As the underlying causes of such conditions are very heterogeneous, manipulations that can enhance learning and memory in mice under different circumstances might be able to overcome the cognitive deficits in patients. The M-current regulates neuronal excitability and action potential firing, suggesting that its inhibition may increase cognitive capacities. We demonstrate that XE991, a specific M-current blocker, enhances learning and memory in healthy mice. This effect may be achieved by altering basal hippocampal synaptic activity and by diminishing the stimulation threshold for long-term changes in synaptic efficacy and learning-related gene expression. We also show that training sessions regulate the M-current by transiently decreasing the levels of KCNQ/Kv7.3 protein, a pivotal subunit for the M-current. Furthermore, we found that XE991 can revert the cognitive impairment associated with acetylcholine depletion and the neurodegeneration induced by kainic acid. Together, these results show that inhibition of the M-current as a general strategy may be useful to enhance cognitive capacities in healthy and aging individuals, as well as in those with neurodegenerative diseases.


Subject(s)
Anthracenes/pharmacology , Brain/physiology , Cognition Disorders/physiopathology , KCNQ3 Potassium Channel/drug effects , Neuronal Plasticity/drug effects , Potassium Channel Blockers/pharmacology , Animals , Brain/drug effects , Disease Models, Animal , Electrophysiology , Gene Expression Profiling , Immunohistochemistry , KCNQ3 Potassium Channel/biosynthesis , Learning/drug effects , Learning/physiology , Male , Memory/drug effects , Memory/physiology , Mice , Neuronal Plasticity/physiology , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
15.
PLoS One ; 5(11): e13900, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21085477

ABSTRACT

Every other day feeding (EOD) and exercise induce changes in cell metabolism. The aim of the present work was to know if both EOD and exercise produce similar effects on physical capacity, studying their physiological, biochemical and metabolic effects on muscle. Male OF-1 mice were fed either ad libitum (AL) or under EOD. After 18 weeks under EOD, animals were also trained by using a treadmill for another 6 weeks and then analyzed for physical activity. Both, EOD and endurance exercise increased the resistance of animals to extenuating activity and improved motor coordination. Among the groups that showed the highest performance, AL and EOD trained animals, ALT and EODT respectively, only the EODT group was able to increase glucose and triglycerides levels in plasma after extenuating exercise. No high effects on mitochondrial respiratory chain activities or protein levels neither on coenzyme Q levels were found in gastrocnemius muscle. However, exercise and EOD did increase ß-oxidation activity in this muscle accompanied by increased CD36 levels in animals fed under EOD and by changes in shape and localization of mitochondria in muscle fibers. Furthermore, EOD and training decreased muscle damage after strenuous exercise. EOD also reduced the levels of lipid peroxidation in muscle. Our results indicate that EOD improves muscle performance and resistance by increasing lipid catabolism in muscle mitochondria at the same time that prevents lipid peroxidation and muscle damage.


Subject(s)
Feeding Behavior/physiology , Motor Activity/physiology , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Animals , Blood Glucose/metabolism , CD36 Antigens/metabolism , Cholesterol/blood , Exercise Test , Lactates/blood , Lipid Metabolism/physiology , Lipid Peroxidation/physiology , Male , Malondialdehyde/metabolism , Mice , Microscopy, Electron , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Oxidation-Reduction , Triglycerides/blood , Ubiquinone/metabolism
16.
J Neurosci ; 30(40): 13305-13, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926656

ABSTRACT

Memory formation requires changes in gene expression, which are regulated by the activation of transcription factors and by changes in epigenetic factors. Poly[ADP]-ribosylation of nuclear proteins has been postulated as a chromatin modification involved in memory consolidation, although the mechanisms involved are not well characterized. Here we demonstrate that poly[ADP]-ribose polymerase 1 (PARP-1) activity and the poly[ADP]-ribosylation of proteins over a specific time course is required for the changes in synaptic plasticity related to memory stabilization in mice. At the molecular level, histone H1 poly[ADP]-ribosylation was evident in the hippocampus after the acquisition period, and it was selectively released in a PARP-1-dependent manner at the promoters of cAMP response element-binding protein and nuclear factor-κB dependent genes associated with learning and memory. These findings suggest that histone H1 poly[ADP]-ribosylation, and its loss at specific loci, is an epigenetic mechanism involved in the reprogramming of neuronal gene expression required for memory consolidation.


Subject(s)
Chromatin/metabolism , Histones/metabolism , Learning/physiology , Poly Adenosine Diphosphate Ribose/metabolism , Proteins/metabolism , Animals , Chromatin/genetics , Epigenesis, Genetic/genetics , Exploratory Behavior/physiology , Gene Expression Regulation/physiology , Genetic Loci/genetics , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Histones/physiology , Male , Memory/physiology , Mice , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/physiology , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/physiology , Promoter Regions, Genetic/genetics , Proteins/physiology , Synaptic Transmission/genetics
17.
Hippocampus ; 20(5): 584-95, 2010 May.
Article in English | MEDLINE | ID: mdl-19603520

ABSTRACT

Neuropsychological analyses of amnesic patients, as well as lesion experiments, indicate that the temporal lobe is essential for the encoding, storage, and expression of object recognition memory (ORM). However, temporal lobe structures directly involved in the consolidation and reconsolidation of these memories are not yet well-defined. We report here that systemic administration of a protein synthesis inhibitor before or up to 4 h after training or reactivation sessions impairs consolidation and reconsolidation of ORM, without affecting short-term memory. We have also observed that ORM reconsolidation is sensitive to protein synthesis inhibition, independently of the ORM trace age. Using bdnf and egr-1 gene expression analysis, we defined temporal lobe areas related to consolidation and reconsolidation of ORM. Training and reactivation 21 days after ORM acquisition sessions provoked changes in bdnf mRNA in somatosensory, perirhinal, and hippocampal cortices. Reactivation 2 days after the training session elicited changes in bdnf and egr-1 mRNA in entorhinal and prefrontal cortices, while reactivation 9 days post-training provoked an increase in egr-1 transcription in somatosensory and entorhinal cortices. The differences in activated circuits and in the capacity to recall the memory trace after 9 or 21 days post-training suggest that memory trace suffers functional changes in this period of time. All these results indicate that the functional state of the recognition memory trace, from acquisition to forgetting, can be specifically defined by behavioral, circuitry, and molecular properties.


Subject(s)
Brain/metabolism , Discrimination Learning/physiology , Exploratory Behavior/physiology , Gene Expression Regulation/physiology , Memory Disorders/physiopathology , Recognition, Psychology/physiology , 4-Aminopyridine/pharmacology , Age Factors , Animals , Anisomycin/pharmacology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/anatomy & histology , Brain/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Discrimination Learning/drug effects , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Exploratory Behavior/drug effects , Gene Expression Regulation/drug effects , Imidazoles , Male , Memory Disorders/chemically induced , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Neural Pathways/drug effects , Neural Pathways/metabolism , Potassium Channel Blockers/pharmacology , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Pyridines , RNA, Messenger/metabolism , Recognition, Psychology/drug effects , Time Factors
18.
Curr Biol ; 19(1): 54-60, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19110430

ABSTRACT

Memory deficits in aging affect millions of people and are often disturbing to those concerned. Dissection of the molecular control of learning and memory is paramount to understand and possibly enhance cognitive functions. Old-age memory loss also has been recently linked to altered Ca(2+) homeostasis. We have previously identified DREAM (downstream regulatory element antagonistic modulator), a member of the neuronal Ca(2+) sensor superfamily of EF-hand proteins, with specific roles in different cell compartments. In the nucleus, DREAM is a Ca(2+)-dependent transcriptional repressor, binding to specific DNA signatures, or interacting with nucleoproteins regulating their transcriptional properties. Also, we and others have shown that dream mutant (dream(-/-)) mice exhibit marked analgesia. Here we report that dream(-/-) mice exhibit markedly enhanced learning and synaptic plasticity related to improved cognition. Mechanistically, DREAM functions as a negative regulator of the key memory factor CREB in a Ca(2+)-dependent manner, and loss of DREAM facilitates CREB-dependent transcription during learning. Intriguingly, 18-month-old dream(-/-) mice display learning and memory capacities similar to young mice. Moreover, loss of DREAM protects from brain degeneration in aging. These data identify the Ca(2+)-regulated "pain gene" DREAM as a novel key regulator of memory and brain aging.


Subject(s)
Aging/physiology , Kv Channel-Interacting Proteins/deficiency , Learning/physiology , Memory/physiology , Aging/genetics , Analysis of Variance , Animals , Blotting, Western , Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/metabolism , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Electrophysiology , Hippocampus/physiology , Immunohistochemistry , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Mice , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Mol Neurobiol ; 38(2): 167-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18759009

ABSTRACT

Aging is associated with the decline of cognitive properties. This situation is magnified when neurodegenerative processes associated with aging appear in human patients. Neuronal synaptic plasticity events underlie cognitive properties in the central nervous system. Caloric restriction (CR; either a decrease in food intake or an intermittent fasting diet) can extend life span and increase disease resistance. Recent studies have shown that CR can have profound effects on brain function and vulnerability to injury and disease. Moreover, CR can stimulate the production of new neurons from stem cells (neurogenesis) and can enhance synaptic plasticity, which modulate pain sensation, enhance cognitive function, and may increase the ability of the brain to resist aging. The beneficial effects of CR appear to be the result of a cellular stress response stimulating the production of proteins that enhance neuronal plasticity and resistance to oxidative and metabolic insults; they include neurotrophic factors, neurotransmitter receptors, protein chaperones, and mitochondrial biosynthesis regulators. In this review, we will present and discuss the effect of CR in synaptic processes underlying analgesia and cognitive improvement in healthy, sick, and aging animals. We will also discuss the possible role of mitochondrial biogenesis induced by CR in regulation of neuronal synaptic plasticity.


Subject(s)
Caloric Restriction , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/metabolism , Aging/physiology , Analgesia , Animals , Humans , Learning/physiology , Mitochondria/metabolism , Neurodegenerative Diseases/diet therapy , Neurodegenerative Diseases/physiopathology
20.
Mol Cell Neurosci ; 39(2): 193-201, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18638560

ABSTRACT

Histone deacetylases (HDAC) are enzymes that maintain chromatin in a condensate state, related with absence of transcription. We have studied the role of HDAC on learning and memory processes. Both eyeblink classical conditioning (EBCC) and object recognition memory (ORM) induced an increase in histone H3 acetylation (Ac-H3). Systemic treatment with HDAC inhibitors improved cognitive processes in EBCC and in ORM tests. Immunohistochemistry and gene expression analyses indicated that administration of HDAC inhibitors decreased the stimulation threshold for Ac-H3, and gene expression to reach the levels required for learning and memory. Finally, we evaluated the effect of systemic administration of HDAC inhibitors to mice models of neurodegeneration and aging. HDAC inhibitors reversed learning and consolidation deficits in ORM in these models. These results point out HDAC inhibitors as candidate agents for the palliative treatment of learning and memory impairments in aging and in neurodegenerative disorders.


Subject(s)
Aging/genetics , Conditioning, Classical/drug effects , Histone Deacetylase Inhibitors , Memory/physiology , Neurodegenerative Diseases/physiopathology , Acetylation/drug effects , Analysis of Variance , Animals , Association Learning/drug effects , Blinking/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Histones/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Kainic Acid , Male , Memory/drug effects , Mice , Mice, Mutant Strains , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/drug therapy , Pattern Recognition, Visual/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...