Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biomolecules ; 12(11)2022 11 15.
Article in English | MEDLINE | ID: mdl-36421705

ABSTRACT

Autism Spectrum Disorder (ASD) is a progressive neurodevelopmental disorder mainly characterized by deficits in social communication and stereotyped behaviors and interests. Here, we aimed to investigate the state of several key players in the dopamine and glutamate neurotransmission systems in the valproic acid (VPA) animal model that was administered to E12.5 pregnant females as a single dose (450 mg/kg). We report no alterations in the number of mesencephalic dopamine neurons or in protein levels of tyrosine hydroxylase in either the striatum or the nucleus accumbens. In females prenatally exposed to VPA, levels of dopamine were slightly decreased while the ratio of DOPAC/dopamine was increased in the dorsal striatum, suggesting increased turn-over of dopamine tone. In turn, levels of D1 and D2 dopamine receptor mRNAs were increased in the nucleus accumbens of VPA mice suggesting upregulation of the corresponding receptors. We also report decreased protein levels of striatal parvalbumin and increased levels of p-mTOR in the cerebellum and the motor cortex of VPA mice. mRNA levels of mGluR1, mGluR4, and mGluR5 and the glutamate receptor subunits NR1, NR2A, and NR2B were not altered by VPA, nor were protein levels of NR1, NR2A, and NR2B and those of BDNF and TrkB. These findings are of interest as clinical trials aiming at the dopamine and glutamate systems are being considered.


Subject(s)
Autism Spectrum Disorder , Valproic Acid , Pregnancy , Female , Mice , Animals , Valproic Acid/pharmacology , Dopamine/metabolism , Disease Models, Animal , Glutamates
2.
Neurosci Lett ; 745: 135629, 2021 02 06.
Article in English | MEDLINE | ID: mdl-33440236

ABSTRACT

There is converging evidence of dendritic spine dysfunction in schizophrenia. In the present study we hypothesized that the expression of key proteins involved in dendritic spine development and stability may be affected in schizophrenia. Postmortem frontal cortex (BA6) from patients with schizophrenia, major depressive disorder, bipolar disorder and healthy controls was processed for glutamate post-synaptic fraction extraction and post-synaptic density purification. Protein expression of the post-synaptic fraction and the post-synaptic density was assessed using immunoprecipitation and Western blotting respectively. The expression of the N-methyl-d-aspartate glutamate receptor (NMDAR) subunit NR2A, post-synaptic density 95 (PSD-95), Ca2+/calmodulin-dependent protein kinase II subunits α and ß (CaMKIIα and ß) were significantly reduced in schizophrenia. A significant decrease in the expression of NR2A was also observed in patients with major depressive disorder relative to controls, but not in patients with bipolar disorder. These results add to existing evidence for disturbed post-synaptic glutamate function and synaptic plasticity in schizophrenia. There may also be subtle disturbances in the post-synaptic glutamatergic function in major depressive disorder.


Subject(s)
Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neuronal Plasticity/physiology , Prefrontal Cortex/metabolism , Schizophrenia/metabolism , Adult , Disks Large Homolog 4 Protein/biosynthesis , Disks Large Homolog 4 Protein/genetics , Female , Gene Expression , Humans , Male , Membrane Proteins/genetics , Middle Aged , Nerve Tissue Proteins/genetics , Prefrontal Cortex/pathology , Proteins/genetics , Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Schizophrenia/pathology
3.
Mol Autism ; 12(1): 2, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33468258

ABSTRACT

BACKGROUND: Contrasting findings were reported in several animal models with a Shank3 mutation used to induce various autism spectrum disorder (ASD) symptoms. Here, we aimed at investigating behavioral, cellular, and molecular consequences of a C-terminal (frameshift in exon 21) deletion in Shank3 protein in mice, a mutation that is also found in clinical conditions and which results in loss of major isoforms of Shank3. A special focus was made on cerebellar related parameters. METHODS: All three genotypes were analyzed [wild type (WT), heterozygote (Shank3+/ΔC) and homozygote (Shank3 ΔC/ΔC)] and males and females were separated into two distinct groups. Motor and social behavior, gait, Purkinje cells (PC) and glutamatergic protein levels were determined. Behavioral and cellular procedures used here were previously validated using two environmental animal models of ASD. ANOVA and post-hoc analysis were used for statistical analysis. RESULTS: Shank3 ΔC/ΔC mice showed significant impairments in social novelty preference, stereotyped behavior and gait. These were accompanied by a decreased number of PC in restricted cerebellar sub-regions and decreased cerebellar expression of mGluR5. Females Shank3 ΔC/ΔC were less affected by the mutation than males. Shank3+/ΔC mice showed impairments only in social novelty preference, grooming, and decreased mGluR5 expression and that were to a much lesser extent than in Shank3 ΔC/ΔC mice. LIMITATIONS: As Shank3 mutation is a haploinsufficiency, it is of interest to emphasize that Shank3+/ΔC mice showed only mild to no deficiencies compared to Shank3 ΔC/ΔC. CONCLUSIONS: Our findings indicate that several behavioral, cellular, and molecular parameters are affected in this animal model. The reported deficits are more pronounced in males than in females. Additionally, male Shank3 ΔC/ΔC mice show more pronounced alterations than Shank3+/ΔC. Together with our previous findings in two environmental animal models of ASD, our studies indicate that gait dysfunction constitutes a robust set of motor ASD symptoms that may be considered for implementation in clinical settings as an early and quantitative diagnosis criteria.


Subject(s)
Gait , Genetic Predisposition to Disease , Microfilament Proteins , Motor Activity , Mutation , Nerve Tissue Proteins , Psychomotor Disorders/genetics , Psychomotor Disorders/physiopathology , Animals , Behavior, Animal , Biomarkers , Disease Models, Animal , Female , Genetic Association Studies , Immunohistochemistry , Male , Mice , Mice, Knockout , Phenotype , Psychomotor Disorders/diagnosis , Sex Factors , Social Behavior
4.
Mol Psychiatry ; 25(10): 2641, 2020 Oct.
Article in English | MEDLINE | ID: mdl-31520066

ABSTRACT

A correction to this paper has been published and can be accessed via a link at the top of the paper.

5.
Mol Psychiatry ; 25(10): 2517-2533, 2020 10.
Article in English | MEDLINE | ID: mdl-30659288

ABSTRACT

Many of the genes disrupted in autism are identified as histone-modifying enzymes and chromatin remodelers, most prominently those that mediate histone methylation/demethylation. However, the role of histone methylation enzymes in the pathophysiology and treatment of autism remains unknown. To address this, we used mouse models of haploinsufficiency of the Shank3 gene (a highly penetrant monogenic autism risk factor), which exhibits prominent autism-like social deficits. We found that histone methyltransferases EHMT1 and EHMT2, as well as histone lysine 9 dimethylation (specifically catalyzed by EHMT1/2), were selectively increased in the prefrontal cortex (PFC) of Shank3-deficient mice and autistic human postmortem brains. Treatment with the EHMT1/2 inhibitor UNC0642 or knockdown of EHMT1/2 in PFC induced a robust rescue of autism-like social deficits in Shank3-deficient mice, and restored NMDAR-mediated synaptic function. Activity-regulated cytoskeleton-associated protein (Arc) was identified as one of the causal factors underlying the rescuing effects of UNC0642 on NMDAR function and social behaviors in Shank3-deficient mice. UNC0642 treatment also restored a large set of genes involved in neural signaling in PFC of Shank3-deficient mice. These results suggest that targeting histone methylation enzymes to adjust gene expression and ameliorate synaptic defects could be a potential therapeutic strategy for autism.


Subject(s)
Autistic Disorder/drug therapy , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Microfilament Proteins/deficiency , Nerve Tissue Proteins/deficiency , Animals , Autistic Disorder/genetics , Disease Models, Animal , Female , Haploinsufficiency , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Male , Methylation/drug effects , Mice , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Quinazolines/pharmacology
6.
Neurosci Biobehav Rev ; 117: 281-296, 2020 10.
Article in English | MEDLINE | ID: mdl-28571876

ABSTRACT

This review focuses on the inter- and transgenerational effects of stress experience prior to and during gestation. We provide an overview of findings from studies in humans as well as in animal models on brain structural and physiological functions and on the development of cognitive and executive functions. We also discuss the concept of stress-induced (re-)programming in more detail by highlighting epigenetic mechanisms and particularly those affecting the development of monoaminergic transmitter systems, which constitute the brains reward system. As the majority of studies have focused on male individuals we will emphasize sex-specific differences in stress vulnerability and resilience. Finally, we offer some perspectives on the development of protective and therapeutic interventions in cognitive and emotional disturbances resulting from pre-conception and prenatal stress.


Subject(s)
Prenatal Exposure Delayed Effects , Animals , Brain , Epigenesis, Genetic , Female , Humans , Male , Pregnancy , Stress, Psychological
7.
Transl Psychiatry ; 9(1): 124, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30923308

ABSTRACT

Infections during gestation and the consequent maternal immune activation (MIA) increase the risk of developing neuropsychiatric disorders in infants and throughout life, including autism spectrum disorders (ASD). ASD is a neurodevelopmental disorder that affects three times more males than females and is mainly characterized by deficits in social communication and restricted interests. Consistent findings also indicate that ASD patients suffer from movement disorders, although these symptoms are not yet considered as diagnosis criteria. Here we used the double-stranded RNA analog polyinosinic:polycytidylic acid (poly I:C) MIA animal model of ASD in mice and explored its effects in males and females on social and motor behavior. We then investigated brain areas implicated in controlling and coordinating movements, namely the nigro-striatal pathway, motor cortex and cerebellum. We show that male mice are more affected by this treatment than females as they show reduced social interactions as well as motor development and coordination deficits. Reduced numbers of Purkinje cells in the cerebellum was found more widespread and within distinct lobules in males than in females. Moreover, a reduced number of neurons was found in the motor cortex of males only. These results suggest that females are better protected against developmental insults leading to ASD symptoms in mice. They also point to brain areas that may be targeted to better manage social and motor consequences of ASD.


Subject(s)
Autism Spectrum Disorder/immunology , Behavior, Animal , Brain/pathology , Neurons/pathology , Sex Factors , Animals , Autism Spectrum Disorder/chemically induced , Disease Models, Animal , Female , Male , Mice , Poly I-C/pharmacology , Pregnancy
8.
Brain ; 142(3): 787-807, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30668640

ABSTRACT

Epigenetic dysregulation, which leads to the alteration of gene expression in the brain, is suggested as one of the key pathophysiological bases of ageing and neurodegeneration. Here we found that, in the late-stage familial Alzheimer's disease (FAD) mouse model, repressive histone H3 dimethylation at lysine 9 (H3K9me2) and euchromatic histone methyltransferases EHMT1 and EHMT2 were significantly elevated in the prefrontal cortex, a key cognitive region affected in Alzheimer's disease. Elevated levels of H3K9me2 were also detected in the prefrontal cortex region of post-mortem tissues from human patients with Alzheimer's disease. Concomitantly, H3K9me2 at glutamate receptors was increased in prefrontal cortex of aged FAD mice, which was linked to the diminished transcription, expression and function of AMPA and NMDA receptors. Treatment of FAD mice with specific EHMT1/2 inhibitors reversed histone hyper-methylation and led to the recovery of glutamate receptor expression and excitatory synaptic function in prefrontal cortex and hippocampus. Chromatin immunoprecipitation-sequencing (ChIP-seq) data indicated that FAD mice exhibited genome-wide increase of H3K9me2 enrichment at genes involved in neuronal signalling (including glutamate receptors), which was reversed by EHMT1/2 inhibition. Moreover, the impaired recognition memory, working memory, and spatial memory in aged FAD mice were rescued by the treatment with EHMT1/2 inhibitors. These results suggest that disrupted epigenetic regulation of glutamate receptor transcription underlies the synaptic and cognitive deficits in Alzheimer's disease, and targeting histone methylation enzymes may represent a novel therapeutic strategy for this prevalent neurodegenerative disorder.


Subject(s)
Alzheimer Disease/metabolism , Histocompatibility Antigens/physiology , Histone-Lysine N-Methyltransferase/physiology , Animals , Chromosome Deletion , Cognition/physiology , Cognition Disorders/genetics , Cognitive Dysfunction/metabolism , DNA Methylation/genetics , Disease Models, Animal , Epigenesis, Genetic/genetics , Hippocampus/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Lysine/genetics , Memory Disorders/genetics , Methylation , Mice , Mice, Transgenic , Prefrontal Cortex/metabolism , Synapses/metabolism
9.
Nat Neurosci ; 21(8): 1139, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29872123

ABSTRACT

In the version of this article initially published, the blue diamonds in Fig. 2a-d were defined as Shank3+/Δc + saline; the correct definition is Shank3+/Δc + RMD. The error has been corrected in the HTML and PDF versions of the article.

10.
Neuropsychopharmacology ; 43(8): 1779-1788, 2018 07.
Article in English | MEDLINE | ID: mdl-29760409

ABSTRACT

Autism is a neurodevelopmental disorder characterized by social deficits and repetitive behaviors. Genetic screening has identified synaptic, transcriptional, and chromatin genes disrupted in autistic patients. Haploinsufficiency of Shank3, which encodes a scaffold protein at glutamatergic synapses, is causally linked to autism. Using a Shank3-deficient mouse model that exhibits prominent autism-like phenotypes, we have found that histone acetylation in the prefrontal cortex (PFC) is abnormally low, which can be reversed by MS-275 (also known as Entinostat, SNDX-275), a class I histone deacetylase (HDAC) inhibitor that is selectively potent in PFC. A brief (3-day) treatment with MS-275 (i.p.) led to the sustained (11 days) rescue of autistic social preference deficits in Shank3-deficient mice, without altering locomotion, motor coordination, anxiety, or the increased grooming. MS-275 treatment also rescued the diminished NMDAR surface expression and NMDAR function induced by Shank3 deficiency. Moreover, F-actin at synapses was restored and the transcription of actin regulators was elevated by MS-275 treatment of Shank3-deficient mice, which may contribute to the recovery of actin-based NMDAR synaptic delivery. Taken together, these results suggest that MS-275 treatment could normalize the aberrant epigenetic regulation of genes, leading to the amelioration of synaptic and social deficits associated with autism.


Subject(s)
Autistic Disorder/drug therapy , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Pyridines/pharmacology , Social Behavior , Synapses/drug effects , Actins/metabolism , Animals , Autistic Disorder/physiopathology , Brain/drug effects , Brain/physiopathology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Disease Models, Animal , Epigenesis, Genetic/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Rats , Synapses/physiology
11.
Int J Neuropsychopharmacol ; 21(9): 871-882, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29762671

ABSTRACT

Background: Motor impairments are amongst the earliest and most consistent signs of autism spectrum disorders but are not used as diagnostic criteria. In addition, the relationship between motor and cognitive impairments and their respective neural substrates remain unknown. Methods: Here, we aimed at determining whether a well-acknowledged animal model of autism spectrum disorders, the valproic acid model, displays motor impairments and whether they may correlate with social deficits and neuronal loss within motor brain areas. For this, pregnant female mice (C57BL/6J) received valproic acid (450 mg/kg) at embryonic day 12.5 and offspring underwent a battery of behavioral analyses before being killed for histological correlates in motor cortex, nigrostriatal pathway, and cerebellum. Results: We show that while valproic acid male mice show both social and motor impairments, female mice only show motor impairments. Prenatal valproic acid exposure induces specific cell loss within the motor cortex and cerebellum and that is of higher magnitude in males than in females. Finally, we demonstrate that motor dysfunction correlates with reduced social behavior and that motor and social deficits both correlate with a loss of Purkinje cells within the Crus I cerebellar area. Conclusions: Our results suggest that motor dysfunction could contribute to social and communication deficits in autism spectrum disorders and that motor and social deficits may share common neuronal substrates in the cerebellum. A systematic assessment of motor function in autism spectrum disorders may potentially help the quantitative diagnosis of autism spectrum disorders and strategies aimed at improving motor behavior may provide a global therapeutic benefit.


Subject(s)
Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/psychology , Brain/pathology , Neurons/pathology , Social Behavior , Animals , Disease Models, Animal , Female , Gait , Male , Mice, Inbred C57BL , Motor Skills , Movement Disorders/pathology , Movement Disorders/psychology , Pregnancy , Prenatal Exposure Delayed Effects , Random Allocation , Sex Factors , Valproic Acid
12.
Nat Neurosci ; 21(4): 564-575, 2018 04.
Article in English | MEDLINE | ID: mdl-29531362

ABSTRACT

Haploinsufficiency of the SHANK3 gene is causally linked to autism spectrum disorder (ASD), and ASD-associated genes are also enriched for chromatin remodelers. Here we found that brief treatment with romidepsin, a highly potent class I histone deacetylase (HDAC) inhibitor, alleviated social deficits in Shank3-deficient mice, which persisted for ~3 weeks. HDAC2 transcription was upregulated in these mice, and knockdown of HDAC2 in prefrontal cortex also rescued their social deficits. Nuclear localization of ß-catenin, a Shank3-binding protein that regulates cell adhesion and transcription, was increased in Shank3-deficient mice, which induced HDAC2 upregulation and social deficits. At the downstream molecular level, romidepsin treatment elevated the expression and histone acetylation of Grin2a and actin-regulatory genes and restored NMDA-receptor function and actin filaments in Shank3-deficient mice. Taken together, these findings highlight an epigenetic mechanism underlying social deficits linked to Shank3 deficiency, which may suggest potential therapeutic strategies for ASD patients bearing SHANK3 mutations.


Subject(s)
Autistic Disorder/complications , Gene Expression Regulation/genetics , Histone Deacetylases/metabolism , Nerve Tissue Proteins/deficiency , Social Behavior Disorders , Animals , Autistic Disorder/genetics , Depsipeptides/therapeutic use , Disease Models, Animal , Exploratory Behavior/drug effects , Gene Expression Regulation/drug effects , Grooming/drug effects , Grooming/physiology , Histone Deacetylase Inhibitors/therapeutic use , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/pathology , Psychomotor Performance/drug effects , Social Behavior Disorders/enzymology , Social Behavior Disorders/etiology , Social Behavior Disorders/therapy , Synaptic Potentials/drug effects , Synaptic Potentials/genetics
13.
Psychopathology ; 49(4): 201-210, 2016.
Article in English | MEDLINE | ID: mdl-27668788

ABSTRACT

The development of the brain depends on an individual's nature (genes) and nurture (environments). This interaction between genetic predispositions and environmental events during brain development drives the maturation of functional brain circuits such as sensory, motor, emotional, and complex cognitive pathways. Adverse environmental conditions such as early life stress can interfere with the functional development of emotional and cognitive brain systems and thereby increase the risk of developing psychiatric disorders later in life. In order to develop more efficient and individualized protective and therapeutic interventions, it is essential to understand how environmental stressors during infancy affect cellular and molecular mechanisms involved in brain maturation. Animal models of early life stress have been able to reveal brain structural and metabolic changes in prefrontolimbic circuits, which are time, brain region, neuron, and sex specific. By focusing on animal models of separation stress during infancy, this review highlights epigenetic and cytoarchitectural modifications which are assumed to mediate lasting changes of brain function and behavior.


Subject(s)
Adaptation, Physiological/physiology , Brain/growth & development , Epigenesis, Genetic/physiology , Neuronal Plasticity/physiology , Parent-Child Relations , Synapses/physiology , Animals , Emotions/physiology , Female , Humans , Infant , Male , Stress, Psychological/psychology
14.
J Neural Transm (Vienna) ; 123(9): 1037-52, 2016 09.
Article in English | MEDLINE | ID: mdl-27169537

ABSTRACT

The view that the functional maturation of the brain is the result of an environmentally driven adaptation of genetically preprogrammed neuronal networks is an important current concept in developmental neuroscience and psychology. This hypothesis proposes that early traumatic experiences or early life stress (ELS) as a negative environmental experience provide a major risk factor for the development of dysfunctional brain circuits and as a consequence for the emergence of behavioral dysfunctions and mental disorders in later life periods. This view is supported by an increasing number of clinical as well as experimental animal studies revealing that early life traumas can induce functional 'scars' in the brain, especially in brain circuits, which are essential for emotional control, learning, and memory functions. Such gene × environment interactions are modulated by specific epigenetic mechanisms, which are suggested to be the key factors of transgenerational epigenetic inheritance. Indeed, there is increasing evidence for inter- and transgenerational cycles of environmentally driven neuronal and behavioral adaptations mediated by epigenetic mechanisms. Finally, recent concepts postulate that, dependent on type, time point, and duration of ELS exposure, also positive functional adaptations may occur in the relevant brain pathways, leading to better stress coping and resilience against adversities later in life.


Subject(s)
Brain/pathology , Epigenesis, Genetic/physiology , Inheritance Patterns , Prenatal Exposure Delayed Effects , Stress, Psychological , Animals , Female , Humans , Pregnancy , Stress, Psychological/genetics , Stress, Psychological/pathology , Stress, Psychological/physiopathology
15.
J Neurosci ; 36(7): 2119-30, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888924

ABSTRACT

Stress and the major stress hormone corticosterone induce profound influences in the brain. Altered histone modification and transcriptional dysfunction have been implicated in stress-related mental disorders. We previously found that repeated stress caused an impairment of prefrontal cortex (PFC)-mediated cognitive functions by increasing the ubiquitination and degradation of AMPA-type glutamate receptors via a mechanism depending on the E3 ubiquitin ligase Nedd4. Here, we demonstrated that in PFC of repeatedly stressed rats, active glucocorticoid receptor had the increased binding to the glucocorticoid response element of histone deacetylase 2 (HDAC2) promoter, resulting in the upregulation of HDAC2. Inhibition or knock-down of HDAC2 blocked the stress-induced impairment of synaptic transmission, AMPAR expression, and recognition memory. Furthermore, we found that, in stressed animals, the HDAC2-dependent downregulation of histone methyltransferase Ehmt2 (G9a) led to the loss of repressive histone methylation at the Nedd4-1 promoter and the transcriptional activation of Nedd4. These results have provided an epigenetic mechanism and a potential treatment strategy for the detrimental effects of chronic stress. SIGNIFICANCE STATEMENT: Prolonged stress exposure can induce altered histone modification and transcriptional dysfunction, which may underlie the profound influence of stress in regulating brain functions. We report an important finding about the epigenetic mechanism controlling the detrimental effects of repeated stress on synaptic transmission and cognitive function. First, it has revealed the stress-induced alteration of key epigenetic regulators HDAC2 and Ehmt2, which determines the synaptic and behavioral effects of repeated stress. Second, it has uncovered the stress-induced histone modification of the target gene Nedd4, an E3 ligase that is critically involved in the ubiquitination and degradation of AMPA receptors and cognition. Third, it has provided the epigenetic approach, HDAC2 inhibition or knock-down, to rescue synaptic and cognitive functions in stressed animals.


Subject(s)
Cognition Disorders/etiology , Endosomal Sorting Complexes Required for Transport , Histones/metabolism , Receptors, AMPA , Stress, Psychological/complications , Ubiquitin-Protein Ligases , Animals , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Gene Knockdown Techniques , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/chemistry , Male , Nedd4 Ubiquitin Protein Ligases , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Recognition, Psychology/drug effects , Stress, Psychological/metabolism , Synaptic Transmission/drug effects , Ubiquitination/genetics
16.
Cell Rep ; 11(9): 1400-1413, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26027926

ABSTRACT

Haploinsufficiency of the Shank3 gene, which encodes a scaffolding protein at glutamatergic synapses, is a highly prevalent and penetrant risk factor for autism. Using combined behavioral, electrophysiological, biochemical, imaging, and molecular approaches, we find that Shank3-deficient mice exhibit autism-like social deficits and repetitive behaviors, as well as the significantly diminished NMDA receptor (NMDAR) synaptic function and synaptic distribution in prefrontal cortex. Concomitantly, Shank3-deficient mice have a marked loss of cortical actin filaments, which is associated with the reduced Rac1/PAK activity and increased activity of cofilin, the major actin depolymerizing factor. The social deficits and NMDAR hypofunction are rescued by inhibiting cofilin or activating Rac1 in Shank3-deficient mice and are induced by inhibiting PAK or Rac1 in wild-type mice. These results indicate that the aberrant regulation of synaptic actin filaments and loss of synaptic NMDARs contribute to the manifestation of autism-like phenotypes. Thus, targeting actin regulators provides a strategy for autism treatment.


Subject(s)
Actin Depolymerizing Factors/metabolism , Autistic Disorder/metabolism , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/physiopathology , Animals , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
17.
Curr Top Med Chem ; 13(20): 2541-50, 2013.
Article in English | MEDLINE | ID: mdl-24066892

ABSTRACT

Vaccines are powerful public health tools that have been of tremendous benefit in protecting vulnerable populations worldwide from many pathogens. However, vaccine- preventable diseases still remain a considerable burden and this is particularly true among aging and aged populations in industrialized countries. The predicted demographic shift in the population landscape towards an ever-increasing aging population and the evidence suggesting that older individuals mount less-than optimal immune response to vaccination have raised the question of improving vaccine responses in older individuals. This review presents recent progress in the understanding at the cellular and molecular levels of age related immune decline and strategies to translate current knowledge into the development of immunization strategies to promote healthy aging, keeping older members of our society autonomous and independent.


Subject(s)
Aging/immunology , Vaccination , Vaccines/immunology , Aged , Humans , Public Health
18.
Rejuvenation Res ; 15(4): 414-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22663183

ABSTRACT

Age-associated atrophy of the thymus with coincident reduction in thymopoeisis, decline in thymic output, and subsequent immune dysfunction has been reversed by the use of interleukin-7 (IL-7). In the earlier studies and in clinical trials, delivery of IL-7 has been by multiple injections over several days to maintain effective activity levels in the tissues. This is unlikely to meet with high compliance rates in future clinical use, and so we tested alternate routes of delivery using a technique involving tagging IL-7 with fluorescent dye that emits in the near-infrared region and whose fluorescence can be visualized within the tissues of live animals. We have shown that intratracheal instillation, enabling transfer through the lungs, provides an effective route for delivering IL-7 into the bloodstream and from there into the tissues in older animals. Delivery is rapid and widespread tissue distribution is seen. Comparison of administration either subcutaneously or by instillation reveals that IL-7 delivery by the pulmonary route provides significantly greater transmission to lymphoid tissues when compared with injection. In functional assessment studies, pulmonary administration led to significantly improved intrathymic T cell development in older animals when compared with IL-7 delivered by injection. Furthermore, in these older animals, delivery of IL-7 by intratracheal instillation was not accompanied by any apparent adverse events when compared with controls receiving saline vehicle by instillation or animals receiving IL-7 by subcutaneous injection.


Subject(s)
Aging/immunology , Aging/metabolism , Interleukin-7/administration & dosage , Administration, Inhalation , Animals , Female , Immune System , Instillation, Drug , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Tissue Distribution , Trachea
SELECTION OF CITATIONS
SEARCH DETAIL
...