Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Psychiatry ; 25(11): 2832-2843, 2020 11.
Article in English | MEDLINE | ID: mdl-30038231

ABSTRACT

Recent findings from in vivo-imaging and human post-mortem tissue studies in schizophrenic psychosis (SzP), have demonstrated functional and molecular changes in hippocampal subfields that can be associated with hippocampal hyperexcitability. In this study, we used a subfield-specific GluN1 knockout mouse with a disease-like molecular perturbation expressed only in hippocampal dentate gyrus (DG) and assessed its association with hippocampal physiology and psychosis-like behaviors. First, we used whole-cell patch-clamp recordings to measure the physiological changes in hippocampal subfields and cFos immunohistochemistry to examine cellular excitability. DG-GluN1 KO mice show CA3 cellular hyperactivity, detected using two approaches: (1) increased excitatory glutamate transmission at mossy fibers (MF)-CA3 synapses, and (2) an increased number of cFos-activated pyramidal neurons in CA3, an outcome that appears to project downstream to CA1 and basolateral amygdala (BLA). Furthermore, we examined psychosis-like behaviors and pathological memory processing; these show an increase in fear conditioning (FC), a reduction in prepulse inhibition (PPI) in the KO animal, along with a deterioration in memory accuracy with Morris Water Maze (MWM) and reduced social memory (SM). Moreover, with DREADD vectors, we demonstrate a remarkably similar behavioral profile when we induce CA3 hyperactivity. These hippocampal subfield changes could provide the basis for the observed increase in human hippocampal activity in SzP, based on the shared DG-specific GluN1 reduction. With further characterization, these animal model systems may serve as targets to test psychosis mechanisms related to hippocampus and assess potential hippocampus-directed treatments.


Subject(s)
CA3 Region, Hippocampal/physiopathology , Dentate Gyrus/metabolism , Nerve Tissue Proteins/deficiency , Psychotic Disorders/physiopathology , Receptors, N-Methyl-D-Aspartate/deficiency , Animals , CA3 Region, Hippocampal/cytology , Female , Male , Mice , Mice, Inbred C57BL , Pyramidal Cells
2.
Hippocampus ; 29(8): 726-735, 2019 08.
Article in English | MEDLINE | ID: mdl-30779299

ABSTRACT

Dentate gyrus adult neurogenesis is implicated in the formation of hippocampal-dependent contextual associations. However, the role of adult neurogenesis during reward-based context-dependent paradigms-such as conditioned place preference (CPP)-is understudied. Therefore, we used image-guided, hippocampal-targeted X-ray irradiation (IG-IR) and morphine CPP to explore whether dentate gyrus adult neurogenesis plays a role in reward memories created in adult C57BL/6J male mice. In addition, as adult neurogenesis appears to participate to a greater extent in retrieval and extinction of recent (<48 hr posttraining) versus remote (>1 week posttraining) memories, we specifically examined the role of adult neurogenesis in reward-associated contextual memories probed at recent and remote timepoints. Six weeks post-IG-IR or Sham treatment, mice underwent morphine CPP. Using separate groups, retrieval of recent and remote reward memories was found to be similar between IG-IR and Sham treatments. Interestingly, IG-IR mice showed impaired extinction-or increased persistence-of the morphine-associated reward memory when it was probed 24-hr (recent) but not 3-weeks (remote) postconditioning relative to Sham mice. Taken together, these data show that hippocampal-directed irradiation and the associated decrease in dentate gyrus adult neurogenesis affect the persistence of recently-but not remotely-probed reward memory. These data indicate a novel role for adult neurogenesis in reward-based memories and particularly the extinction rate of these memories. Consideration of this work may lead to better understanding of extinction-based behavioral interventions for psychiatric conditions characterized by dysregulated reward processing.


Subject(s)
Dentate Gyrus/physiology , Extinction, Psychological/physiology , Memory/physiology , Morphine/administration & dosage , Narcotics/administration & dosage , Neurogenesis/physiology , Reward , Animals , Cranial Irradiation/methods , Dentate Gyrus/drug effects , Dentate Gyrus/radiation effects , Extinction, Psychological/drug effects , Extinction, Psychological/radiation effects , Male , Memory/drug effects , Memory/radiation effects , Mice , Neurogenesis/drug effects , Neurogenesis/radiation effects , Neurons/drug effects , Neurons/pathology , Neurons/radiation effects
3.
J Neurosci ; 37(45): 10917-10931, 2017 11 08.
Article in English | MEDLINE | ID: mdl-28978667

ABSTRACT

Genetic perturbations of the transcription factor Forkhead Box P1 (FOXP1) are causative for severe forms of autism spectrum disorder that are often comorbid with intellectual disability. Recent work has begun to reveal an important role for FoxP1 in brain development, but the brain-region-specific contributions of Foxp1 to autism and intellectual disability phenotypes have yet to be determined fully. Here, we describe Foxp1 conditional knock-out (Foxp1cKO) male and female mice with loss of Foxp1 in the pyramidal neurons of the neocortex and the CA1/CA2 subfields of the hippocampus. Foxp1cKO mice exhibit behavioral phenotypes that are of potential relevance to autism spectrum disorder, including hyperactivity, increased anxiety, communication impairments, and decreased sociability. In addition, Foxp1cKO mice have gross deficits in learning and memory tasks of relevance to intellectual disability. Using a genome-wide approach, we identified differentially expressed genes in the hippocampus of Foxp1cKO mice associated with synaptic function and development. Furthermore, using magnetic resonance imaging, we uncovered a significant reduction in the volumes of both the entire hippocampus as well as individual hippocampal subfields of Foxp1cKO mice. Finally, we observed reduced maintenance of LTP in area CA1 of the hippocampus in these mutant mice. Together, these data suggest that proper expression of Foxp1 in the pyramidal neurons of the forebrain is important for regulating gene expression pathways that contribute to specific behaviors reminiscent of those seen in autism and intellectual disability. In particular, Foxp1 regulation of gene expression appears to be crucial for normal hippocampal development, CA1 plasticity, and spatial learning.SIGNIFICANCE STATEMENT Loss-of-function mutations in the transcription factor Forkhead Box P1 (FOXP1) lead to autism spectrum disorder and intellectual disability. Understanding the potential brain-region-specific contributions of FOXP1 to disease-relevant phenotypes could be a critical first step in the management of patients with these mutations. Here, we report that Foxp1 conditional knock-out (Foxp1cKO) mice with loss of Foxp1 in the neocortex and hippocampus display autism and intellectual-disability-relevant behaviors. We also show that these phenotypes correlate with changes in both the genomic and physiological profiles of the hippocampus in Foxp1cKO mice. Our work demonstrates that brain-region-specific FOXP1 expression may relate to distinct, clinically relevant phenotypes.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Gene Expression/genetics , Gene Expression/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Spatial Learning/physiology , Synapses/physiology , Animals , Autism Spectrum Disorder , Behavior, Animal/physiology , CA1 Region, Hippocampal/physiology , Female , Learning Disabilities/genetics , Learning Disabilities/psychology , Male , Memory Disorders/genetics , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neocortex/cytology , Neocortex/physiology , Pyramidal Cells/metabolism , Vocalization, Animal/physiology
4.
Dev Biol ; 431(2): 179-193, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28947178

ABSTRACT

While several studies indicate the importance of ephrin-B/EphB bidirectional signaling in excitatory neurons, potential roles for these molecules in inhibitory neurons are largely unknown. We identify here an autonomous receptor-like role for ephrin-B reverse signaling in the tangential migration of interneurons into the neocortex using ephrin-B (EfnB1/B2/B3) conditional triple mutant (TMlz) mice and a forebrain inhibitory neuron specific Cre driver. Inhibitory neuron deletion of the three EfnB genes leads to reduced interneuron migration, abnormal cortical excitability, and lethal audiogenic seizures. Truncated and intracellular point mutations confirm the importance of ephrin-B reverse signaling in interneuron migration and cortical excitability. A non-autonomous ligand-like role was also identified for ephrin-B2 that is expressed in neocortical radial glial cells and required for proper tangential migration of GAD65-positive interneurons. Our studies thus define both receptor-like and ligand-like roles for the ephrin-B molecules in controlling the migration of interneurons as they populate the neocortex and help establish excitatory/inhibitory (E/I) homeostasis.


Subject(s)
Cell Movement , Ephrins/metabolism , Interneurons/cytology , Interneurons/metabolism , Animals , Female , Gene Deletion , Ligands , Mice , Models, Biological , Mutation/genetics , Neocortex/cytology , Neocortex/metabolism , Neural Inhibition , Prosencephalon/cytology , Prosencephalon/metabolism , Pseudopodia/metabolism
5.
J Biol Chem ; 291(22): 11647-56, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27008863

ABSTRACT

The biological underpinnings and the pathological lesions of psychiatric disorders are centuries-old questions that have yet to be understood. Recent studies suggest that schizophrenia and related disorders likely have their origins in perturbed neurodevelopment and can result from a large number of common genetic variants or multiple, individually rare genetic alterations. It is thus conceivable that key neurodevelopmental pathways underline the various genetic changes and the still unknown pathological lesions in schizophrenia. Here, we report that mice defective of the nicastrin subunit of γ-secretase in oligodendrocytes have hypomyelination in the central nervous system. These mice have altered dopamine signaling and display profound abnormal phenotypes reminiscent of schizophrenia. In addition, we identify an association of the nicastrin gene with a human schizophrenia cohort. These observations implicate γ-secretase and its mediated neurodevelopmental pathways in schizophrenia and provide support for the "myelination hypothesis" of the disease. Moreover, by showing that schizophrenia and obsessive-compulsive symptoms could be modeled in animals wherein a single genetic factor is altered, our work provides a biological basis that schizophrenia with obsessive-compulsive disorder is a distinct subtype of schizophrenia.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Compulsive Behavior , Membrane Glycoproteins/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Schizophrenia/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Female , Humans , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Middle Aged , Schizophrenia/genetics
6.
J Neuroinflammation ; 13(1): 171, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27357503

ABSTRACT

BACKGROUND: A traumatic brain injury (TBI) event is a devastating injury to the brain that may result in heightened inflammation, neurodegeneration, and subsequent cognitive and mood deficits. TBI victims with co-morbidities such as heart disease, diabetes, or obesity may be more vulnerable to the secondary brain injury that follows the initial insult. Compared to lean individuals, obese subjects tend to have worse clinical outcomes and higher mortality rates after trauma. METHODS: To elucidate whether obesity predisposes individuals to worse outcomes after TBI, we subjected adult lean and obese male/female mice to a mild TBI. The injury was administered using a controlled skull impact (CSI) device. Lean or obese 6-month-old C57 BL/6 mice were subjected once to a mild TBI. Additionally, at day 30 after injury, both the lean and obese mice were tested for increased anxiety using the open field test. RESULTS: At day 30 after TBI, compared to the lean mice, we found heightened microglial (MG) activation in the cerebral cortex, corpus callosum, and hypothalamus. Another compelling finding was that, compared to the non-injured obese male control mice, the obese TBI mice had a decrease in the rate of weight gain and serum corticosterone levels at day 30 after injury. Additionally, the injured obese mice displayed higher levels of anxiety as determined by a significant decrease in time spent in the non-peripheral zones in the open field test. In contrast to the obese males, the obese female mice did not exhibit increases in the number of active MG in the brain, changes in weight gain/corticosterone levels, or increased anxiety at day 30 after TBI. CONCLUSIONS: The data presented here suggests that obese mice have worse outcomes compared to lean mice after mild TBI. Also, the obese males have worse outcomes than the injured female mice. This data may explain the sequela of chronic secondary brain injury in obese adults after a single mild TBI. Also, this report may help shape how the overweight/obese populations are monitored over the days and months following a TBI.


Subject(s)
Brain Concussion/metabolism , Brain Concussion/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Obesity/metabolism , Obesity/pathology , Animals , Brain Concussion/complications , Female , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Obesity/complications
7.
Hippocampus ; 25(11): 1374-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25786918

ABSTRACT

CA1 hippocampal N-methyl-d-aspartate-receptors (NMDARs) are necessary for contextually related learning and memory processes. Extinction, a form of learning, has been shown to require intact hippocampal NMDAR signalling. Renewal of fear expression can occur after fear extinction training, when the extinguished fear stimulus is presented in an environmental context different from the training context and thus, renewal is dependent on contextual memory. In this study, we show that a Grin1 knock-out (loss of the essential NR1 subunit for the NMDAR) restricted to the bilateral CA1 subfield of the dorsal hippocampus does not affect acquisition of learned fear, but does attenuate extinction of a cued fear response even when presented in the extinction-training context. We propose that failure to remember the (safe) extinction context is responsible for the abnormal fear response and suggest it is a dysfunctional renewal. The results highlight the difference in outcome of extinguished fear memory resulting from a partial rather than complete loss of function of the hippocampus and suggest a potential mechanism for abnormally increased fear expression in PTSD.


Subject(s)
Behavior, Animal/physiology , CA1 Region, Hippocampal/physiopathology , Extinction, Psychological/physiology , Fear/physiology , Nerve Tissue Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cues , Disease Models, Animal , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Stress Disorders, Post-Traumatic/physiopathology
8.
Hippocampus ; 25(4): 409-14, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25424867

ABSTRACT

Addiction has been proposed to emerge from associations between the drug and the reward-associated contexts. This associative learning has a cellular correlate, as there are more cFos+ neurons in the hippocampal dentate gyrus (DG) after psychostimulant conditioned place preference (CPP) versus saline controls. However, it is unknown whether morphine CPP leads to a similar DG activation, or whether DG activation is due to locomotion, handling, pharmacological effects, or-as data from contextual fear learning suggests-exposure to the drug-associated context. To explore this, we employed an unbiased, counterbalanced, and shortened CPP design that led to place preference and more DG cFos+ cells. Next, mice underwent morphine CPP but were then sequestered into the morphine-paired (conditioned stimulus+ [CS+]) or saline-paired (CS-) context on test day. Morphine-paired mice sequestered to CS+ had ∼30% more DG cFos+ cells than saline-paired mice. Furthermore, Bregma analysis revealed morphine-paired mice had more cFos+ cells in CS+ compared to CS- controls. Notably, there was no significant difference in DG cFos+ cell number after handling alone or after receiving morphine in home cage. Thus, retrieval of morphine-associated context is accompanied by activation of hippocampal DG granule cell neurons.


Subject(s)
Dentate Gyrus/cytology , Mental Recall/drug effects , Morphine/administration & dosage , Narcotics/administration & dosage , Neurons/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Mice , Time Factors
9.
Mol Genet Metab ; 116(1-2): 98-105, 2015.
Article in English | MEDLINE | ID: mdl-25982063

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs) are a group of related hereditary lysosomal storage disorders characterized by progressive loss of neurons in the central nervous system resulting in dementia, loss of motor skills, seizures and blindness. A characteristic intralysosomal accumulation of autofluorescent storage material occurs in the brain and other tissues. Three major forms and nearly a dozen minor forms of NCL are recognized. Infantile-onset NCL (CLN1 disease) is caused by severe deficiency in a soluble lysosomal enzyme, palmitoyl-protein thioesterase-1 (PPT1) and no therapy beyond supportive care is available. Homozygous Ppt1 knockout mice reproduce the known features of the disease, developing signs of motor dysfunction at 5 months of age and death around 8 months. Direct delivery of lysosomal enzymes to the cerebrospinal fluid is an approach that has gained traction in small and large animal models of several other neuropathic lysosomal storage diseases, and has advanced to clinical trials. In the current study, Ppt1 knockout mice were treated with purified recombinant human PPT1 enzyme delivered to the lumbar intrathecal space on each of three consecutive days at 6 weeks of age. Untreated PPT1 knockout mice and wild-type mice served as additional controls. Four enzyme concentration levels (0, 2.6, 5.3 and 10.6 mg/ml of specific activity 20 U/mg) were administered in a volume of 80 µl infused over 8 min. Each group consisted of 16-20 mice. The treatment was well tolerated. Disease-specific survival was 233, 267, 272, and 284days for each of the four treatment groups, respectively, and the effect of treatment was highly significant (p<0.0001). The timing of motor deterioration was also delayed. Neuropathology was improved as evidenced by decreased autofluorescent storage material in the spinal cord and a decrease in CD68 staining in the cortex and spinal cord. The improvements in motor function and survival are similar to results reported for preclinical studies involving other lysosomal storage disorders, such as CLN2/TPP1 deficiency, for which intraventricular ERT is being offered in clinical trials. If ERT delivery to the CSF proves to be efficacious in these disorders, PPT1 deficiency may also be amenable to this approach.


Subject(s)
Disease Models, Animal , Enzyme Replacement Therapy , Membrane Proteins/therapeutic use , Motion , Neuronal Ceroid-Lipofuscinoses/drug therapy , Animals , Brain/drug effects , Brain/immunology , Brain/pathology , Disease-Free Survival , Dose-Response Relationship, Drug , Humans , Injections, Spinal , Membrane Proteins/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thiolester Hydrolases/genetics , Tripeptidyl-Peptidase 1
10.
bioRxiv ; 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-37609208

ABSTRACT

Autism manifests differently in males and females and the brain mechanisms that mediate these sex-dependent differences are unknown. Here, we demonstrate that deletion of the ASD-risk gene, Pten, in neocortical pyramidal neurons (NSE Pten KO) results in robust hyperexcitability of local neocortical circuits in female, but not male, mice, observed as prolonged, spontaneous persistent activity states (UP states). Circuit hyperexcitability in NSE Pten KO mice is mediated by enhanced and/or altered signaling of metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) to ERK and protein synthesis selectively in Pten deleted female neurons. In support of this idea, Pten deleted Layer 5 cortical neurons have female-specific increases in mGluR5 and mGluR5-driven protein synthesis. In addition, mGluR5-ERα complexes are elevated in female cortex and genetic reduction of ERα in Pten KO cortical neurons rescues circuit excitability, protein synthesis and enlarged neurons selectively in females. Abnormal timing and hyperexcitability of neocortical circuits in female NSE Pten KO mice are associated with deficits in temporal processing of sensory stimuli and social behaviors as well as mGluR5-dependent seizures. Female-specific cortical hyperexcitability and mGluR5-dependent seizures are also observed in a human disease relevant mouse model, germline Pten +/- mice. Our results reveal molecular mechanisms by which sex and a high impact ASD-risk gene interact to affect brain function and behavior.

11.
bioRxiv ; 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38915673

ABSTRACT

Certain areas of the brain involved in episodic memory and behavior, such as the hippocampus, express high levels of insulin receptors and glucose transporter-4 (GLUT4) and are responsive to insulin. Insulin and neuronal glucose metabolism improve cognitive functions and regulate mood in humans. Insulin-dependent GLUT4 trafficking has been extensively studied in muscle and adipose tissue, but little work has demonstrated either how it is controlled in insulin-responsive brain regions or its mechanistic connection to cognitive functions. In this study, we demonstrate that inhibition of WNK (With-No-lysine (K)) kinases improves learning and memory in mice. Neuronal inhibition of WNK enhances in vivo hippocampal glucose uptake. Inhibition of WNK enhances insulin signaling output and insulin-dependent GLUT4 trafficking to the plasma membrane in mice primary neuronal cultures and hippocampal slices. Therefore, we propose that the extent of neuronal WNK kinase activity has an important influence on learning, memory and anxiety-related behaviors, in part, by modulation of neuronal insulin signaling.

12.
Cell Rep ; 43(4): 114056, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38581678

ABSTRACT

Little is known of the brain mechanisms that mediate sex-specific autism symptoms. Here, we demonstrate that deletion of the autism spectrum disorder (ASD)-risk gene, Pten, in neocortical pyramidal neurons (NSEPten knockout [KO]) results in robust cortical circuit hyperexcitability selectively in female mice observed as prolonged spontaneous persistent activity states. Circuit hyperexcitability in females is mediated by metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) signaling to mitogen-activated protein kinases (Erk1/2) and de novo protein synthesis. Pten KO layer 5 neurons have a female-specific increase in mGluR5 and mGluR5-dependent protein synthesis. Furthermore, mGluR5-ERα complexes are generally elevated in female cortices, and genetic reduction of ERα rescues enhanced circuit excitability, protein synthesis, and neuron size selectively in NSEPten KO females. Female NSEPten KO mice display deficits in sensory processing and social behaviors as well as mGluR5-dependent seizures. These results reveal mechanisms by which sex and a high-confidence ASD-risk gene interact to affect brain function and behavior.


Subject(s)
Autistic Disorder , Disease Models, Animal , Estrogen Receptor alpha , Mice, Knockout , Neocortex , PTEN Phosphohydrolase , Receptor, Metabotropic Glutamate 5 , Animals , Female , Male , Mice , Autistic Disorder/metabolism , Autistic Disorder/physiopathology , Autistic Disorder/genetics , Autistic Disorder/pathology , Estrogen Receptor alpha/metabolism , Mice, Inbred C57BL , Neocortex/metabolism , Neocortex/pathology , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Pyramidal Cells/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Social Behavior
13.
bioRxiv ; 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38895206

ABSTRACT

Most antipsychotic drugs (APDs) induce hyperphagia and weight gain. However, the neural mechanisms are poorly understood, partly due to challenges replicating their metabolic effects in rodents. Here, we report a new mouse model that recapitulates overeating induced by clozapine, a widely prescribed APD. Our study shows that clozapine boosts food intake by inhibiting melanocortin 4 receptor (MC4R) expressing neurons in the paraventricular nucleus of the hypothalamus. Interestingly, neither clozapine nor risperidone, another commonly used APD, affects receptor-ligand binding or the canonical Gαs signaling of MC4R. Instead, they inhibit neuronal activity by enhancing the coupling between MC4R and Kir7.1, leading to the open state of the inwardly rectifying potassium channel. Deletion of Kir7.1 in Mc4r-Cre neurons prevents clozapine-induced weight gain, while treatment with a selective Kir7.1 blocker mitigates overeating in clozapine-fed mice. Our findings unveil a molecular pathway underlying the effect of APDs on feeding behavior and suggest its potential as a therapeutic target.

14.
J Neurosci ; 32(17): 5880-90, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22539849

ABSTRACT

Adult neurogenesis persists throughout life in restricted brain regions in mammals and is affected by various physiological and pathological conditions. The tumor suppressor gene Pten is involved in adult neurogenesis and is mutated in a subset of autism patients with macrocephaly; however, the link between the role of PTEN in adult neurogenesis and the etiology of autism has not been studied before. Moreover, the role of hippocampus, one of the brain regions where adult neurogenesis occurs, in development of autism is not clear. Here, we show that ablating Pten in adult neural stem cells in the subgranular zone of hippocampal dentate gyrus results in higher proliferation rate and accelerated differentiation of the stem/progenitor cells, leading to depletion of the neural stem cell pool and increased differentiation toward the astrocytic lineage at later stages. Pten-deleted stem/progenitor cells develop into hypertrophied neurons with abnormal polarity. Additionally, Pten mutant mice have macrocephaly and exhibit impairment in social interactions and seizure activity. Our data reveal a novel function for PTEN in adult hippocampal neurogenesis and indicate a role in the pathogenesis of abnormal social behaviors.


Subject(s)
Adult Stem Cells/physiology , Dentate Gyrus/cytology , Gene Expression Regulation/genetics , Neurogenesis/genetics , Neurons/physiology , PTEN Phosphohydrolase/deficiency , Adaptation, Ocular/genetics , Adult Stem Cells/drug effects , Age Factors , Animals , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Electroencephalography , Electromyography , Exploratory Behavior/physiology , Intercellular Signaling Peptides and Proteins/pharmacology , Intermediate Filament Proteins/genetics , Interpersonal Relations , Lateral Ventricles/cytology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurogenesis/drug effects , Neurons/cytology , Propanolamines/metabolism , Proto-Oncogene Proteins c-akt/metabolism
15.
Eur J Neurosci ; 37(2): 242-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23039899

ABSTRACT

It has been suggested for some time that circadian rhythm abnormalities underlie the development of multiple psychiatric disorders. However, it is unclear how disruptions in individual circadian genes might regulate mood and anxiety. Here we found that mice lacking functional mPeriod 1 (mPer1) or mPeriod 2 (mPer2) individually did not have consistent behavioral abnormalities in measures of anxiety-related behavior. However, mice deficient in both mPer1 and mPer2 had an increase in levels of anxiety-like behavior in multiple measures. Moreover, we found that mPer1 and mPer2 expression was reduced in the nucleus accumbens (NAc) after exposure to chronic social defeat stress, a paradigm that led to increased anxiety-related behavior. Following social defeat, chronic treatment with fluoxetine normalized Per gene expression towards wild-type levels. Knockdown of both mPer1 and mPer2 expression via RNA interference specifically in the NAc led to a similar increase in anxiety-like behavior as seen in the mutant animals. Taken together, these results implicate the Per genes in the NAc in response to stress and the development of anxiety.


Subject(s)
Anxiety/genetics , Nucleus Accumbens/metabolism , Period Circadian Proteins/metabolism , Animals , Antidepressive Agents, Second-Generation/pharmacology , Anxiety/metabolism , Avoidance Learning , Fluoxetine/pharmacology , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Nucleus Accumbens/physiology , Period Circadian Proteins/genetics , RNA, Small Interfering , Stress, Psychological , Transcription, Genetic/drug effects
16.
FASEB J ; 26(8): 3148-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22542682

ABSTRACT

Adult neurogenesis occurs throughout life in the mammalian hippocampus and is essential for memory and mood control. There is significant interest in identifying ways to promote neurogenesis and ensure maintenance of these hippocampal functions. Previous work with a synthetic small molecule, isoxazole 9 (Isx-9), highlighted its neuronal-differentiating properties in vitro. However, the ability of Isx-9 to drive neurogenesis in vivo or improve hippocampal function was unknown. Here we show that Isx-9 promotes neurogenesis in vivo, enhancing the proliferation and differentiation of hippocampal subgranular zone (SGZ) neuroblasts, and the dendritic arborization of adult-generated dentate gyrus neurons. Isx-9 also improves hippocampal function, enhancing memory in the Morris water maze. Notably, Isx-9 enhances neurogenesis and memory without detectable increases in cellular or animal activity or vascularization. Molecular exploration of Isx-9-induced regulation of neurogenesis (via FACS and microarray of SGZ stem and progenitor cells) suggested the involvement of the myocyte-enhancer family of proteins (Mef2). Indeed, transgenic-mediated inducible knockout of all brain-enriched Mef2 isoforms (Mef2a/c/d) specifically from neural stem cells and their progeny confirmed Mef2's requirement for Isx-9-induced increase in hippocampal neurogenesis. Thus, Isx-9 enhances hippocampal neurogenesis and memory in vivo, and its effects are reliant on Mef2, revealing a novel cell-intrinsic molecular pathway regulating adult neurogenesis.


Subject(s)
Hippocampus/physiology , Isoxazoles/pharmacology , Neurogenesis/drug effects , Thiophenes/pharmacology , Animals , Blood-Brain Barrier/metabolism , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dentate Gyrus/physiology , Hippocampus/drug effects , Isoxazoles/metabolism , MEF2 Transcription Factors , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic , Myogenic Regulatory Factors/physiology , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Thiophenes/metabolism
17.
Proc Natl Acad Sci U S A ; 107(33): 14845-50, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20682746

ABSTRACT

The direct and indirect pathways of the basal ganglia have been proposed to oppositely regulate locomotion and differentially contribute to pathological behaviors. Analysis of the distinct contributions of each pathway to behavior has been a challenge, however, due to the difficulty of selectively investigating the neurons comprising the two pathways using conventional techniques. Here we present two mouse models in which the function of striatonigral or striatopallidal neurons is selectively disrupted due to cell type-specific deletion of the striatal signaling protein dopamine- and cAMP-regulated phosphoprotein Mr 32kDa (DARPP-32). Using these mice, we found that the loss of DARPP-32 in striatonigral neurons decreased basal and cocaine-induced locomotion and abolished dyskinetic behaviors in response to the Parkinson's disease drug L-DOPA. Conversely, the loss of DARPP-32 in striatopallidal neurons produced a robust increase in locomotor activity and a strongly reduced cataleptic response to the antipsychotic drug haloperidol. These findings provide insight into the selective contributions of the direct and indirect pathways to striatal motor behaviors.


Subject(s)
Corpus Striatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/physiology , Motor Activity/physiology , Neurons/metabolism , Animals , Catalepsy/chemically induced , Catalepsy/physiopathology , Cocaine/pharmacology , Corpus Striatum/cytology , Dopamine Agents/toxicity , Dopamine Uptake Inhibitors/pharmacology , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Female , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Haloperidol/toxicity , Immunohistochemistry , Levodopa/toxicity , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Activity/drug effects , Neuronal Plasticity/physiology , Neurons/classification , Neurons/cytology , Synaptic Potentials/physiology
18.
Mol Genet Metab ; 107(1-2): 213-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22704978

ABSTRACT

PPT1-related neuronal ceroid lipofuscinosis (NCL) is a lysosomal storage disorder caused by deficiency in a soluble lysosomal enzyme, palmitoyl-protein thioesterase-1 (PPT1). Enzyme replacement therapy (ERT) has not been previously examined in a preclinical animal model. Homozygous PPT1 knockout mice reproduce the known features of the disease, developing signs of motor dysfunction at 5 months of age and death by around 8 months. In the current study, PPT1 knockout mice were treated with purified recombinant PPT1 (0.3 mg, corresponding to 12 mg/kg or 180 U/kg for a 25 g mouse) administered intravenously weekly either 1) from birth; or 2) beginning at 8 weeks of age. The treatment was surprisingly well tolerated and neither anaphylaxis nor antibody formation was observed. In mice treated from birth, survival increased from 236 to 271 days (p<0.001) and the onset of motor deterioration was similarly delayed. In mice treated beginning at 8 weeks, no increases in survival or motor performance were seen. An improvement in neuropathology in the thalamus was seen at 3 months in mice treated from birth, and although this improvement persisted it was attenuated by 7 months. Outside the central nervous system, substantial clearance of autofluorescent storage material in many tissues was observed. Macrophages in spleen, liver and intestine were especially markedly improved, as were acinar cells of the pancreas and tubular cells of the kidney. These findings suggest that ERT may be an option for addressing visceral storage as part of a comprehensive approach to PPT1-related NCL, but more effective delivery methods to target the brain are needed.


Subject(s)
Enzyme Replacement Therapy , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neuronal Ceroid-Lipofuscinoses/mortality , Recombinant Proteins/administration & dosage , Thiolester Hydrolases/administration & dosage , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Rotarod Performance Test , Thiolester Hydrolases/adverse effects , Viscera/drug effects , Viscera/metabolism , Viscera/pathology
19.
Nat Neurosci ; 11(7): 752-3, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18552842

ABSTRACT

We found that increasing ghrelin levels, through subcutaneous injections or calorie restriction, produced anxiolytic- and antidepressant-like responses in the elevated plus maze and forced swim test. Moreover, chronic social defeat stress, a rodent model of depression, persistently increased ghrelin levels, whereas growth hormone secretagogue receptor (Ghsr) null mice showed increased deleterious effects of chronic defeat. Together, these findings demonstrate a previously unknown function for ghrelin in defending against depressive-like symptoms of chronic stress.


Subject(s)
Depression/prevention & control , Ghrelin/administration & dosage , Stress, Psychological/complications , Analysis of Variance , Animals , Behavior, Animal , Caloric Restriction/methods , Depression/etiology , Disease Models, Animal , Eating/drug effects , Eating/genetics , Enzyme-Linked Immunosorbent Assay , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Ghrelin/metabolism , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Orexin Receptors , Reaction Time/physiology , Receptors, G-Protein-Coupled/deficiency , Receptors, Ghrelin/deficiency , Receptors, Neuropeptide/deficiency , Stress, Psychological/genetics , Swimming , Time Factors
20.
Neuron ; 55(2): 289-300, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17640529

ABSTRACT

We analyzed the influence of the transcription factor DeltaFosB on learned helplessness, an animal model of affective disorder wherein a subset of mice exposed to inescapable stress (IS) develop a deficit in escape behavior. Repeated IS induces DeltaFosB in the ventrolateral periaqueductal gray (vlPAG), and levels of the protein are highly predictive of an individual's subsequent behavorial deficit-with the strongest DeltaFosB induction observed in the most resilient animals. Induction of DeltaFosB by IS predominates in substance P-positive neurons in the vlPAG, and the substance P gene, a direct target for DeltaFosB, is downregulated upon DeltaFosB induction. Local overexpression of DeltaFosB in the vlPAG using viral-mediated gene transfer dramatically reduces depression-like behaviors and inhibits stress-induced release of substance P. These results indicate that IS-induced accumulation of DeltaFosB in the vlPAG desensitizes substance P neurons enriched in this area and opposes behavioral despair by promoting active defense responses.


Subject(s)
Escape Reaction/physiology , Helplessness, Learned , Periaqueductal Gray/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Stress, Psychological/metabolism , Adaptation, Physiological , Adaptation, Psychological/physiology , Analysis of Variance , Animals , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Nucleus Accumbens/metabolism , Periaqueductal Gray/cytology , Rats , Rats, Sprague-Dawley , Substance P/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL