Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 384: 112553, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32057826

ABSTRACT

Chronic social defeat stress (CSDS) has been found to produce different impacts on anxiety-like behaviors, spatial cognitive function and memory in rodents with different susceptibilities. However, the impacts of chronic social defeat on social behaviors in adult male mice with different susceptibilities to social defeat and the underlying mechanisms in the brain remain unclear. In the present study, we found that ten days of social defeat reduced the tendency of susceptible adult male C57 mice to approach an unfamiliar individual and increased their avoidance of an unfamiliar CD-1 mouse but had no effects on resilient individuals. In addition, CSDS enhanced anxiety-like behavior in susceptible animals, but produced no effects in the resilient group. Meanwhile, CSDS increased the number of corticotropin-releasing factor (CRF)-positive neurons in the paraventricular nucleus of the hypothalamus and CRF-R2-positive neurons in the accumbens nucleus shell in both resilient and susceptible animals. CSDS increased the number of CRF-R1-positive neurons and CRF-R1 mRNA expression in the prelimbic cortex (PrL) and the number of CRF-R2-positive neurons in the basolateral amygdala, but reduced the number of CRF-R2-positive neurons and mRNA expression in the PrL in susceptible animals. Therefore, the different effects of CSDS on sociability and anxiety-like behavior in mice with different susceptibilities may be associated with region- and type-specific alterations in CRF receptor levels. These findings help us understand the underlying mechanism by which social stress affects emotion and social behavior and provides an important basis for the treatment of disorders of social and emotional behavior caused by social stress.


Subject(s)
Brain/metabolism , Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Social Behavior , Social Defeat , Stress, Psychological/genetics , Animals , Anxiety/genetics , Anxiety/metabolism , Anxiety/physiopathology , Avoidance Learning , Basolateral Nuclear Complex/metabolism , Limbic Lobe/metabolism , Male , Mice , Neurons/metabolism , Nucleus Accumbens/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
2.
Proc Natl Acad Sci U S A ; 116(51): 25958-25967, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31796600

ABSTRACT

Psychostimulant use is an ever-increasing socioeconomic burden, including a dramatic rise during pregnancy. Nevertheless, brain-wide effects of psychostimulant exposure are incompletely understood. Here, we performed Fos-CreERT2-based activity mapping, correlated for pregnant mouse dams and their fetuses with amphetamine, nicotine, and caffeine applied acutely during midgestation. While light-sheet microscopy-assisted intact tissue imaging revealed drug- and age-specific neuronal activation, the indusium griseum (IG) appeared indiscriminately affected. By using GAD67gfp/+ mice we subdivided the IG into a dorsolateral domain populated by γ-aminobutyric acidergic interneurons and a ventromedial segment containing glutamatergic neurons, many showing drug-induced activation and sequentially expressing Pou3f3/Brn1 and secretagogin (Scgn) during differentiation. We then combined Patch-seq and circuit mapping to show that the ventromedial IG is a quasi-continuum of glutamatergic neurons (IG-Vglut1+) reminiscent of dentate granule cells in both rodents and humans, whose dendrites emanate perpendicularly toward while their axons course parallel with the superior longitudinal fissure. IG-Vglut1+ neurons receive VGLUT1+ and VGLUT2+ excitatory afferents that topologically segregate along their somatodendritic axis. In turn, their efferents terminate in the olfactory bulb, thus being integral to a multisynaptic circuit that could feed information antiparallel to the olfactory-cortical pathway. In IG-Vglut1+ neurons, prenatal psychostimulant exposure delayed the onset of Scgn expression. Genetic ablation of Scgn was then found to sensitize adult mice toward methamphetamine-induced epilepsy. Overall, our study identifies brain-wide targets of the most common psychostimulants, among which Scgn+/Vglut1+ neurons of the IG link limbic and olfactory circuits.


Subject(s)
Brain Mapping , Brain/metabolism , Gene Expression Regulation , Limbic Lobe/metabolism , Animals , Axons/metabolism , Brain/diagnostic imaging , Dendrites/metabolism , Female , Glutamate Decarboxylase/genetics , Humans , Interneurons/metabolism , Limbic Lobe/anatomy & histology , Limbic Lobe/drug effects , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Olfactory Bulb/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Secretagogins/genetics , Secretagogins/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , gamma-Aminobutyric Acid/metabolism
3.
J Neuroimmune Pharmacol ; 14(4): 661-678, 2019 12.
Article in English | MEDLINE | ID: mdl-31372820

ABSTRACT

In the era of combined antiretroviral therapy, HIV-1 infected individuals are living longer lives; however, longevity is met with an increasing number of HIV-1 associated neurocognitive disorders (HAND) diagnoses. The transactivator of transcription (Tat) is known to mediate the neurotoxic effects in HAND by acting directly on neurons and also indirectly via its actions on glia. The Go/No-Go (GNG) task was used to examine HAND in the Tat transgenic mouse model. The GNG task involves subjects discriminating between two stimuli sets in order to determine whether or not to inhibit a previously trained response. Data reveal inhibitory control deficits in female Tat(+) mice (p = .048) and an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group (p < .05). A significant negative correlation was noted between inhibitory control and IL CB1R expression (r = -.543, p = .045), with CB1R expression predicting 30% of the variance of inhibitory control (R2 = .295, p = .045). Furthermore, there was a significant increase in spontaneous excitatory postsynaptic current (sEPSC) frequencies in Tat(+) compared to Tat(-) mice (p = .008, across sexes). The increase in sEPSC frequency was significantly attenuated by bath application of PF3845, a fatty acid amide hydrolase (FAAH) enzyme inhibitor (p < .001). Overall, the GNG task is a viable measure to assess inhibitory control deficits in Tat transgenic mice and results suggest a potential therapeutic treatment for the observed deficits with drugs which modulate endocannabinoid enzyme activity. Graphical Abstract Results of the Go/No-Go operant conditioning task reveal inhibitory control deficits in female transgenic Tat(+) mice without significantly affecting males. The demonstrated inhibitory control deficits appear to be associated with an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group.


Subject(s)
AIDS Dementia Complex/metabolism , Disease Models, Animal , HIV-1 , Inhibition, Psychological , Receptor, Cannabinoid, CB1/biosynthesis , tat Gene Products, Human Immunodeficiency Virus/biosynthesis , AIDS Dementia Complex/genetics , AIDS Dementia Complex/psychology , Animals , Female , Limbic Lobe/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurocognitive Disorders/genetics , Neurocognitive Disorders/metabolism , Psychomotor Performance/physiology , Receptor, Cannabinoid, CB1/genetics , Up-Regulation/physiology , tat Gene Products, Human Immunodeficiency Virus/genetics
4.
Neurotox Res ; 36(1): 132-143, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30879275

ABSTRACT

Designer drug mixtures popularized as "bath salts" often contain the synthetic cathinones 3,4 methylenedioxypyrovalerone (MDPV), mephedrone, and methylone in various combinations. However, most preclinical investigations have only assessed the effects of individual bath salt constituents, and little is known about whether co-exposure to MDPV, mephedrone, and methylone produces significant neuropharmacological interactions. This study evaluated and compared how MDPV, mephedrone, and methylone influence discrete brain tissue dopamine (DA) levels and motor stimulant responses in mice when administered alone and as a ternary mixture. Male adolescent Swiss-Webster mice received intraperitoneal injections of saline or 1 or 10 mg/kg doses of MDPV, mephedrone, or methylone, or a cocktail of all three cathinones at doses of 1, 3.3, or 10 mg/kg each. The effect of each treatment on DA and DA metabolite levels in mesolimbic and nigrostriatal brain tissue was quantified 15 min after a single exposure using HPLC-ECD. Additionally, locomotor activity was recorded in mice after acute (day 1) and chronic intermittent (day 7) dosing. MDPV, mephedrone, and methylone produced dose-related increases in mesolimbic and nigrostriatal DA levels that were significantly enhanced following their co-administration. In addition, mice treated with the cathinone cocktail displayed decreased locomotor activity on day 1 that was exacerbated by day 7 and not observed with any of the drugs alone. Our findings demonstrate a significant enhanced effect of MDPV, mephedrone, and methylone on both DA, and these effects on DA result in significant alterations in locomotor activity.


Subject(s)
Benzodioxoles/pharmacology , Brain/drug effects , Dopamine Agents/pharmacology , Dopamine/metabolism , Methamphetamine/analogs & derivatives , Pyrrolidines/pharmacology , Animals , Brain/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Designer Drugs/pharmacology , Dopamine/analysis , Dose-Response Relationship, Drug , Limbic Lobe/drug effects , Limbic Lobe/metabolism , Locomotion/drug effects , Male , Methamphetamine/pharmacology , Mice , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Synthetic Cathinone
5.
Schizophr Bull ; 45(5): 1092-1100, 2019 09 11.
Article in English | MEDLINE | ID: mdl-30388260

ABSTRACT

Psychotic experiences may be understood as altered information processing due to aberrant neural computations. A prominent example of such neural computations is the computation of prediction errors (PEs), which signal the difference between expected and experienced events. Among other areas showing PE coding, hippocampal-prefrontal-striatal neurocircuits play a prominent role in information processing. Dysregulation of dopaminergic signaling, often secondary to psychosocial stress, is thought to interfere with the processing of biologically important events (such as reward prediction errors) and result in the aberrant attribution of salience to irrelevant sensory stimuli and internal representations. Bayesian hierarchical predictive coding offers a promising framework for the identification of dysfunctional neurocomputational processes and the development of a mechanistic understanding of psychotic experience. According to this framework, mismatches between prior beliefs encoded at higher levels of the cortical hierarchy and lower-level (sensory) information can also be thought of as PEs, with important consequences for belief updating. Low levels of precision in the representation of prior beliefs relative to sensory data, as well as dysfunctional interactions between prior beliefs and sensory data in an ever-changing environment, have been suggested as a general mechanism underlying psychotic experiences. Translating the promise of the Bayesian hierarchical predictive coding into patient benefit will come from integrating this framework with existing knowledge of the etiology and pathophysiology of psychosis, especially regarding hippocampal-prefrontal-striatal network function and neural mechanisms of information processing and belief updating.


Subject(s)
Brain/physiopathology , Cognition/physiology , Learning/physiology , Psychotic Disorders/physiopathology , Schizophrenia/physiopathology , Schizophrenic Psychology , Bayes Theorem , Brain/metabolism , Cognitive Neuroscience , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Delusions/metabolism , Delusions/physiopathology , Delusions/psychology , Dopamine/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Limbic Lobe/metabolism , Limbic Lobe/physiopathology , Models, Neurological , Models, Psychological , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neurophysiology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Psychotic Disorders/metabolism , Psychotic Disorders/psychology , Reinforcement, Psychology , Reward , Schizophrenia/metabolism , Synaptic Transmission , Temporal Lobe/metabolism , Temporal Lobe/physiopathology
6.
Pharmacol Biochem Behav ; 171: 54-65, 2018 08.
Article in English | MEDLINE | ID: mdl-29908200

ABSTRACT

BACKGROUND: Chronic nicotine exposure produces neuroadaptations in brain reward systems and α4ß2 nicotinic acetylcholine receptors (nAChRs) in the corticolimbic brain areas. We previously demonstrated opposite effects of nicotine exposure delivered by self-administration or pumps on brain reward thresholds that can be attributed to the different temporal pattern and contingency of nicotine exposure. We investigated the effects of these two factors on reward thresholds and somatic signs during nicotine withdrawal, and on nAChRs binding in corticolimbic brain areas. METHODS: The intracranial self-stimulation procedure was used to assess reward thresholds in rats prepared with pumps delivering various doses of nicotine continuously or intermittently. Separate group of rats were randomly exposed to nicotine via pumps (non-contingent) or nicotine self-administration (contingent) to determine [125I]-epibatidine binding at α4ß2* nAChRs. RESULTS: Withdrawal from continuous non-contingent nicotine exposure led to significant elevations in thresholds and increases in somatic signs in rats, while there was no significant effect of withdrawal from intermittent non-contingent nicotine exposure at the same doses. nAChRs were upregulated during withdrawal from continuous non-contingent nicotine exposure. α4ß2* nAChRs were upregulated in the ventral tegmental area and prelimbic cortex during withdrawal from non-contingent intermittent exposure and in the nucleus accumbens during withdrawal from contingent intermittent nicotine exposure to the same dose. CONCLUSIONS: During non-contingent nicotine exposure, the temporal pattern of nicotine delivery differentially affected thresholds and somatic signs of withdrawal. Upregulation of α4ß2* nAChRs was brain site-specific and depended on both temporal pattern and contingency of nicotine exposure.


Subject(s)
Nicotine/adverse effects , Receptors, Nicotinic/metabolism , Reward , Substance Withdrawal Syndrome/diagnosis , Substance Withdrawal Syndrome/metabolism , Animals , Autoantigens , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Drug Administration Schedule , Infusion Pumps, Implantable , Iodine Radioisotopes/metabolism , Limbic Lobe/metabolism , Male , Nicotine/administration & dosage , Nucleus Accumbens/metabolism , Pyridines/metabolism , Radioligand Assay , Rats , Rats, Wistar , Up-Regulation , Ventral Tegmental Area/metabolism
7.
Cell Mol Neurobiol ; 38(1): 305-316, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28695320

ABSTRACT

The prelimbic cortex (PL) is an important structure in the neural pathway integrating stress responses. Brain angiotensin is involved in cardiovascular control and modulation of stress responses. Blockade of angiotensin receptors has been reported to reduce stress responses. Acute restraint stress (ARS) is a stress model, which evokes sustained blood pressure increase, tachycardia, and reduction in tail temperature. We therefore hypothesized that PL locally generated angiotensin and angiotensin receptors modulate stress autonomic responses. To test this hypothesis, we microinjected an angiotensin-converting enzyme (ACE) inhibitor or angiotensin antagonists into the PL, prior to ARS. Male Wistar rats were used; guide cannulas were bilaterally implanted in the PL for microinjection of vehicle or drugs. A polyethylene catheter was introduced into the femoral artery to record cardiovascular parameters. Tail temperature was measured using a thermal camera. ARS was started 10 min after PL treatment with drugs. Pretreatment with ACE inhibitor lisinopril (0.5 nmol/100 nL) reduced the pressor response, but did not affect ARS-evoked tachycardia. At a dose of 1 nmol/100 nL, it reduced both ARS pressor and tachycardic responses. Pretreatment with candesartan, AT1 receptor antagonist reduced ARS-evoked pressor response, but not tachycardia. Pretreatment with PD123177, AT2 receptor antagonist, reduced tachycardia, but did not affect ARS pressor response. No treatment affected ARS fall in tail temperature. Results suggest involvement of PL angiotensin in the mediation of ARS cardiovascular responses, with participation of both AT1 and AT2 receptors. In conclusion, results indicate that PL AT1-receptors modulate the ARS-evoked pressor response, while AT2-receptors modulate the tachycardic component of the autonomic response.


Subject(s)
Blood Pressure/physiology , Cerebral Cortex/metabolism , Heart Rate/physiology , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology , Stress, Psychological/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Heart Rate/drug effects , Limbic Lobe/drug effects , Limbic Lobe/metabolism , Male , Rats , Rats, Wistar , Restraint, Physical/physiology , Restraint, Physical/psychology , Stress, Psychological/psychology
8.
Neurosci Res ; 130: 23-38, 2018 May.
Article in English | MEDLINE | ID: mdl-28842244

ABSTRACT

The presence of substance P (SP) receptor (Neurokinin-1 receptor, NK1R) in the indusium griseum (IG) and anterior hippocampal continuation (AHC) during postnatal development was studied by immunocytochemistry (ICC). NK1R-immunopositive neurons (NK1RIP-n) first appeared in both areas on postnatal day (P) 5. From P5 onward, their distribution pattern was adult-like. In sagittal sections NK1RIP-n formed a narrow strip of neurons and dendrites that were located over the corpus callosum (cc); in coronal sections they were found in a roughly triangular area at the base of the cingulate cortex (Cg) on the dorsal surface of the cc. NK1RIP-n were also found in the AHC, which is considered as a subcallosal extension of the IG, located ventral to the genu of the cc. At all ages studied, IG NK1RIP-n sent dendrites to the contralateral IG, the underlying cc, and the Cg. Moreover, NK1RIP-n located in the Cg and the cc sent dendrites to the IG. The present findings are in line with previous ICC studies describing dopaminergic and serotoninergic afferents to the IG. Together these data suggest that, through NK1R, SP could play an important role in regulating the release mechanisms of these afferents and that it could be an important developmental factor. Notably, IG neurons could be activated by cortical and intracallosal afferents.


Subject(s)
Limbic Lobe/growth & development , Limbic Lobe/metabolism , Receptors, Neurokinin-1/metabolism , Animals , Axons/metabolism , Dendrites/metabolism , Immunohistochemistry , Limbic Lobe/cytology , Rats, Sprague-Dawley
9.
Addict Biol ; 23(5): 1032-1045, 2018 09.
Article in English | MEDLINE | ID: mdl-28971565

ABSTRACT

The hypocretin receptor 1 (HCRTr1) is a critical participant in the regulation of motivated behavior. Previous observations demonstrate that acute pharmacological blockade of HCRTr1 disrupts dopamine (DA) signaling and the motivation for cocaine when delivered systemically or directly into the ventral tegmental area (VTA). To further examine the involvement of HCRTr1 in regulating reward and reinforcement processing, we employed an adeno-associated virus to express a short hairpin RNA designed to knock down HCRTr1. We injected virus into the VTA and examined the effects of HCRTr1 knockdown on cocaine self-administration and DA signaling in the nucleus accumbens (NAc) core. We determined that the viral approach was effective at reducing HCRTr1 expression without affecting the expression of hypocretin receptor 2 or DA-related mRNAs. We next examined the effects of HCRTr1 knockdown on cocaine self-administration, observing delayed acquisition under a fixed-ratio schedule and reduced motivation for cocaine under a progressive ratio schedule. These effects did not appear to be associated with alterations in sleep/wake activity. Using fast-scan cyclic voltammetry, we then examined whether HCRTr1 knockdown alters DA signaling dynamics in the NAc core. We observed reduced DA release and slower uptake rate as well as attenuated cocaine-induced DA uptake inhibition in rats with knockdown of HCRTr1. These observations indicate that HCRTr1 within the VTA influence the motivation for cocaine, likely via alterations in DA signaling in the NAc.


Subject(s)
Cocaine-Related Disorders/genetics , Cocaine/administration & dosage , Dopamine/metabolism , Motivation/genetics , Orexin Receptors/genetics , Signal Transduction/drug effects , Ventral Tegmental Area/metabolism , Animals , Cocaine/genetics , Cocaine-Related Disorders/metabolism , Disease Models, Animal , Limbic Lobe/drug effects , Limbic Lobe/metabolism , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Motivation/drug effects , Motivation/physiology , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Reward , Self Administration , Signal Transduction/physiology , Ventral Tegmental Area/drug effects
10.
Psychopharmacology (Berl) ; 235(2): 447-458, 2018 02.
Article in English | MEDLINE | ID: mdl-29063964

ABSTRACT

Growing clinical and preclinical evidence suggests a potential role for the phytocannabinoid cannabidiol (CBD) as a pharmacotherapy for various neuropsychiatric disorders. In contrast, delta-9-tetrahydrocannabinol (THC), the primary psychoactive component in cannabis, is associated with acute and neurodevelopmental propsychotic side effects through its interaction with central cannabinoid type 1 receptors (CB1Rs). CB1R stimulation in the ventral hippocampus (VHipp) potentiates affective memory formation through inputs to the mesolimbic dopamine (DA) system, thereby altering emotional salience attribution. These changes in DA activity and salience attribution, evoked by dysfunctional VHipp regulatory actions and THC exposure, could predispose susceptible individuals to psychotic symptoms. Although THC can accelerate the onset of schizophrenia, CBD displays antipsychotic properties, can prevent the acquisition of emotionally irrelevant memories, and reverses amphetamine-induced neuronal sensitization through selective phosphorylation of the mechanistic target of rapamycin (mTOR) molecular signaling pathway. This review summarizes clinical and preclinical evidence demonstrating that distinct phytocannabinoids act within the VHipp and associated corticolimbic structures to modulate emotional memory processing through changes in mesolimbic DA activity states, salience attribution, and signal transduction pathways associated with schizophrenia-related pathology.


Subject(s)
Cannabinoids/therapeutic use , Dopamine/metabolism , Emotions/physiology , Hippocampus/metabolism , Memory/physiology , Mental Disorders/metabolism , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Cannabinoids/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Emotions/drug effects , Hippocampus/drug effects , Humans , Limbic Lobe/drug effects , Limbic Lobe/metabolism , Memory/drug effects , Mental Disorders/drug therapy , Mental Disorders/psychology , Phytochemicals/pharmacology , Phytochemicals/therapeutic use
11.
PLoS One ; 12(11): e0188542, 2017.
Article in English | MEDLINE | ID: mdl-29155879

ABSTRACT

The aim of our study was to test the hypothesis that the spatial distribution of breast cancer brain metastases (BM) differ according to their biological subtypes. MR images of 100 patients with BM from primary breast cancer were retrospectively reviewed. Patients were divided according to the biological subtype of the primary tumor, (triple-negative: 24, HER2 positive: 48, luminal: 28). All images marked with BMs were standardized to the human brain MRI atlas provided by the Montreal Neurological Institute 152 database. Distribution pattern of BM was evaluated with intra-group and intergroup analysis. In intra-group analysis, hot spots of metastases from triple-negative are evenly distributed in the brain, meanwhile BMs from HER2 positive and luminal type occur dominantly in occipital lobe and cerebellum. In intergroup analysis, BMs from triple-negative type occurred more often in frontal lobe, limbic region, and parietal lobe, compared with other types (P < .05). Breast cancer subtypes tend to demonstrate different spatial distributions of their BMs. These findings may have direct implications for dose modulation in prophylactic irradiation as well as for differential diagnoses. Thus, this result should be validated in future study with a larger population.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/classification , Breast Neoplasms/classification , Receptor, ErbB-2/genetics , Adult , Atlases as Topic , Brain Mapping , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cerebellum/diagnostic imaging , Cerebellum/metabolism , Cerebellum/pathology , Databases, Factual , Female , Frontal Lobe/diagnostic imaging , Frontal Lobe/metabolism , Frontal Lobe/pathology , Gene Expression , Humans , Limbic Lobe/diagnostic imaging , Limbic Lobe/metabolism , Limbic Lobe/pathology , Magnetic Resonance Imaging , Middle Aged , Occipital Lobe/diagnostic imaging , Occipital Lobe/metabolism , Occipital Lobe/pathology , Parietal Lobe/diagnostic imaging , Parietal Lobe/metabolism , Parietal Lobe/pathology , Retrospective Studies
12.
Mol Autism ; 8: 30, 2017.
Article in English | MEDLINE | ID: mdl-28649315

ABSTRACT

BACKGROUND: People with fragile X syndrome (FXS) often have deficits in social behavior, and a substantial portion meet criteria for autism spectrum disorder. Though the genetic cause of FXS is known to be due to the silencing of FMR1, and the Fmr1 null mouse model representing this lesion has been extensively studied, the contributions of this gene and its protein product, FMRP, to social behavior are not well understood. METHODS: Fmr1 null mice and wildtype littermates were exposed to a social or non-social stimulus. In one experiment, subjects were assessed for expression of the inducible transcription factor c-Fos in response to the stimulus, to detect brain regions with social-specific activity. In a separate experiment, tissue was taken from those brain regions showing differential activity, and RNA sequencing was performed. RESULTS: Immunohistochemistry revealed a significantly greater number of c-Fos-positive cells in the lateral amygdala and medial amygdala in the brains of mice exposed to a social stimulus, compared to a non-social stimulus. In the prelimbic cortex, there was no significant effect of social stimulus; although the number of c-Fos-positive cells was lower in the social condition compared to the non-social condition, and negatively correlated with c-Fos in the amygdala. RNA sequencing revealed differentially expressed genes enriched for molecules known to interact with FMRP and also for autism-related genes identified in the Simons Foundation Autism Research Initiative gene database. Ingenuity Pathway Analysis detected enrichment of differentially expressed genes in networks and pathways related to neuronal development, intracellular signaling, and inflammatory response. CONCLUSIONS: Using the Fmr1 null mouse model of fragile X syndrome, we have identified brain regions, gene networks, and molecular pathways responsive to a social stimulus. These findings, and future experiments following up on the role of specific gene networks, may shed light on the neural mechanisms underlying dysregulated social behaviors in fragile X syndrome and more broadly.


Subject(s)
Amygdala/metabolism , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Limbic Lobe/metabolism , Amygdala/physiopathology , Animals , Behavior, Animal , Biomarkers/analysis , Brain Mapping , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/diagnosis , Fragile X Syndrome/physiopathology , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Gene Regulatory Networks , Humans , Interpersonal Relations , Limbic Lobe/physiopathology , Male , Mice , Mice, Knockout , Molecular Sequence Annotation , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Sequence Analysis, RNA , Signal Transduction
13.
Article in English | MEDLINE | ID: mdl-28351548

ABSTRACT

Schizophrenia is associated with significant pathophysiological changes to interneurons within the prefrontal cortex (PFC), with mRNA and protein changes associated with the GABA network localized to specific interneuron subtypes. Methamphetamine is a commonly abused psychostimulant that can induce chronic psychosis and symptoms that are similar to schizophrenia, suggesting that chronic METH induced psychosis may be associated with similar brain pathology to schizophrenia in the PFC. The aim of this study, therefore, was to examine mRNA expression of interneuron markers across two regions of the PFC (prelimbic (PRL) and orbitofrontal cortices (OFC)) following METH sensitization, an animal model of METH psychosis. We also studied the association between GABA mRNA expression and interneuronal mRNA expression to identify whether particular changes to the GABA network could be localized to a specific inhibitory cellular phenotype. METH sensitization increased the transcriptional expression of calbindin, calretinin, somatostatin, cholecyctokinin and vasoactive intestinal peptide in the PRL while parvalbumin, calbindin, cholectokinin and vasoactive intestinal peptide were upregulated in the OFC. Based on our previous findings, we also found significant correlations between GAD67, GAT1 and parvalbumin while GAD67, GAD65 and GAT1 were positively correlated with cholecystokinin in the PRL of METH sensitized rats. Within the OFC, the expression of GABAAα1 was positively correlated with somatostatin while GABAAα5 was negatively associated with somatostatin and calbindin. These findings suggest that METH sensitization differentially changes the expression of mRNAs encoding for multiple peptides and calcium binding proteins across the PRL and the OFC. Furthermore, these findings support that changes to the GABA network may also occur within specific cell types. These results, therefore, provide the first evidence that METH sensitization mediates differential interneuronal pathology across the PRL and OFC and such changes could have profound consequences on behavior and cognitive output.


Subject(s)
Central Nervous System Sensitization , Interneurons/metabolism , Interneurons/pathology , Limbic Lobe/metabolism , Methamphetamine/pharmacology , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Animals , GABA Plasma Membrane Transport Proteins/metabolism , Glutamate Decarboxylase/metabolism , Limbic Lobe/pathology , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Prefrontal Cortex/pathology , Rats , Receptors, GABA-A/biosynthesis , gamma-Aminobutyric Acid/metabolism
14.
Transl Psychiatry ; 7(2): e1038, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28221365

ABSTRACT

Ketamine and deep brain stimulation produce rapid antidepressant effects in humans and rodents. An increased AMPA receptor (AMPA-R) signaling in medial prefrontal cortex (mPFC) has been suggested to mediate these responses. However, little research has addressed the direct effects of enhancing glutamate tone or AMPA-R stimulation in mPFC subdivisions. The current study investigates the behavioral and neurochemical consequences of glutamate transporter-1 (GLT-1) blockade or s-AMPA microinfusion in the infralimbic (IL) and prelimbic (PrL) cortex. Owing to the connectivity between the mPFC and raphe nuclei, the role of serotonin is also explored. The bilateral microinfusion of the depolarizing agent veratridine into IL -but not PrL- of rats evoked immediate antidepressant-like responses. The same regional selectivity was observed after microinfusion of dihydrokainic acid (DHK), a selective inhibitor of GLT-1, present in astrocytes. The DHK-evoked antidepressant-like responses appear to be mediated by an AMPA-R-driven enhancement of serotonergic activity, as (i) they were prevented by NBQX 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide disodium salt) and mimicked by s-AMPA; (ii) DHK and s-AMPA elevated similarly extracellular glutamate in IL and PrL, although extracellular 5-HT and c-fos expression in the midbrain dorsal raphe increased only when these agents were applied in IL; and (iii) DHK antidepressant-like responses were prevented by 5-HT synthesis inhibition and mimicked by citalopram microinfusion in IL. These results indicate that an acute increase of glutamatergic neurotransmission selectively in IL triggers immediate antidepressant-like responses in rats, likely mediated by the activation of IL-raphe pathways, which then results in a fast increase of serotonergic activity.


Subject(s)
Affect/drug effects , Behavior, Animal/drug effects , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Kainic Acid/analogs & derivatives , Neuroglia/metabolism , Veratridine/pharmacology , Animals , Citalopram/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Kainic Acid/pharmacology , Limbic Lobe/cytology , Limbic Lobe/metabolism , Male , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Quinoxalines/pharmacology , Raphe Nuclei/metabolism , Rats , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
15.
Brain Res ; 1661: 88-99, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28214522

ABSTRACT

Neural mechanism underlying memory retrieval has been extensively studied in the hippocampus and amygdala. However, little is known about the role of medial prefrontal cortex in long-term memory retrieval. We evaluate this issue in one-trial step-through inhibitory avoidance (IA) paradigm. Our results showed that, 1) inactivation of mPFC by local infusion of GABAA-receptor agonist muscimol caused severe deficits in retrieval of 1-day and 7-day but had no effects on 2-h inhibitory avoidance memory; 2) the protein level of phosphorylated-ERK1/2 in mPFC were significantly increased following retrieval of 1-day and 7-day IA memory, so did the numbers of phosphorylated-ERK (pERK) and phosphorylated-CREB (pCREB) labeled neurons; 3) intra-mPFC infusion of ERK kinase inhibitor PD98095 significantly reduced phosphorylated ERK1/2 levels and phosphorylated-ERK1/2 and phosphorylated-CREB labeled cells, and severely impaired retrieval of 7-day IA memory when the drugs were administrated 30min prior to test. The present study provides evidence that retrieval of long-lasting memory for inhibitory avoidance requires mPFC and involves the ERK-CREB signaling cascade.


Subject(s)
Memory, Long-Term/physiology , Prefrontal Cortex/physiology , Amygdala/physiology , Animals , Avoidance Learning/drug effects , CREB-Binding Protein , Hippocampus/metabolism , Limbic Lobe/metabolism , MAP Kinase Signaling System , Male , Memory/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Muscimol/adverse effects , Muscimol/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
16.
Neurotox Res ; 31(2): 289-297, 2017 02.
Article in English | MEDLINE | ID: mdl-27832448

ABSTRACT

During adolescence, the medial prefrontal cortex (mPFC) is still developing. We have previously shown that developmental cocaine exposure alters mPFC's ability to cope with challenging events. In this manuscript, we exposed rats developmentally treated with cocaine to a novelty task and analyzed the molecular changes of mPFC. Rats were exposed to cocaine from post-natal day (PND) 28 to PND 42 and sacrificed at PND 43, immediately after the novel object recognition (NOR) test. Cocaine-treated rats spent more time exploring the novel object than saline-treated counterparts, suggesting an increased response to novelty. The messenger RNA (mRNA) and protein levels of the immediate early gene Arc/Arg3.1 were reduced in both infralimbic (IL) and prelimbic (PL) cortices highlighting a baseline reduction of mPFC neuronal activity as a consequence of developmental exposure to cocaine. Intriguingly, significant molecular changes were observed in the IL, but not PL, cortex in response to the combination of cocaine exposure and test such as a marked upregulation of both Arc/Arg3.1 mRNA and protein levels only in cocaine-treated rats. As for proteins, such increase was observed only in the post-synaptic density and not in the whole homogenate, suggesting psychostimulant-induced changes in trafficking of Arc/Arg3.1 or an increased local translation. Notably, the same profile of Arc/Arg3.1 was observed for post-synaptic density (PSD)-95 leading to the possibility that Arc/Arg3.1 and PSD-95 bridge together to promote aberrant synaptic connectivity in IL cortex following repeated exposure to cocaine during brain development.


Subject(s)
Cocaine/adverse effects , Cytoskeletal Proteins/biosynthesis , Limbic Lobe/metabolism , Nerve Tissue Proteins/biosynthesis , Prefrontal Cortex/metabolism , Recognition, Psychology/drug effects , Animals , Disks Large Homolog 4 Protein , Exploratory Behavior/drug effects , Intracellular Signaling Peptides and Proteins/biosynthesis , Limbic Lobe/drug effects , Male , Membrane Proteins/biosynthesis , Post-Synaptic Density/metabolism , Prefrontal Cortex/drug effects , Rats , Up-Regulation/drug effects
17.
Sci Rep ; 6: 30298, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27444269

ABSTRACT

Temporal lobe epilepsy or limbic epilepsy lacks effective therapies due to a void in understanding the cellular and molecular mechanisms that set in motion aberrant neuronal network formations during the course of limbic epileptogenesis (LE). Here we show in in vivo rodent models of LE that the phospholipid mediator platelet-activating factor (PAF) increases in LE and that PAF receptor (PAF-r) ablation mitigates its progression. Synthetic PAF-r antagonists, when administered intraperitoneally in LE, re-establish hippocampal dendritic spine density and prevent formation of dysmorphic dendritic spines. Concomitantly, hippocampal interictal spikes, aberrant oscillations, and neuronal hyper-excitability, evaluated 15-16 weeks after LE using multi-array silicon probe electrodes implanted in the dorsal hippocampus, are reduced in PAF-r antagonist-treated mice. We suggest that over-activation of PAF-r signaling induces aberrant neuronal plasticity in LE and leads to chronic dysfunctional neuronal circuitry that mediates epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Platelets/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Limbic Lobe/metabolism , Limbic Lobe/pathology , Mice , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Platelet Activating Factor/genetics , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics
18.
Behav Brain Res ; 306: 170-7, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26956153

ABSTRACT

High frequency deep brain stimulation (DBS) of the lateral habenula (LHb) reduces symptoms of depression in severely treatment-resistant individuals. Despite the observed therapeutic effects, the molecular underpinnings of DBS are poorly understood. This study investigated the efficacy of high frequency LHb DBS (130Hz; 200µA; 90µs) in an animal model of tricyclic antidepressant resistance. Further, we reported DBS mediated changes in Ca(2+)/calmodulin-dependent protein kinase (CaMKIIα/ß), glycogen synthase kinase 3 (GSK3α/ß) and AMP-activated protein kinase (AMPK) both locally and in the infralimbic cortex (IL). Protein expressions were then correlated to immobility time during the forced swim test (FST). Antidepressant actions were quantified via FST. Treatment groups comprised of animals treated with adrenocorticotropic hormone alone (ACTH; 100µg/day, 14days, n=7), ACTH with active DBS (n=7), sham DBS (n=8), surgery only (n=8) or control (n=8). Active DBS significantly reduced immobility in ACTH-treated animals (p<0.05). For this group, western blot results demonstrated phosphorylation status of LHb CaMKIIα/ß and GSK3α/ß significantly correlated to immobility time in the FST. Concurrently, we observed phosphorylation status of CaMKIIα/ß, GSK3α/ß, and AMPK in the IL to be negatively correlated with antidepressant actions of DBS. These findings suggest that activity dependent phosphorylation of CaMKIIα/ß, and GSK3α/ß in the LHb together with the downregulation of CaMKIIα/ß, GSK3α/ß, and AMPK in the IL, contribute to the antidepressant actions of DBS.


Subject(s)
Deep Brain Stimulation/methods , Depressive Disorder, Treatment-Resistant/therapy , Habenula/physiology , Limbic Lobe/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Adrenocorticotropic Hormone/therapeutic use , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Glycogen Synthase Kinase 3/metabolism , Immobility Response, Tonic/drug effects , Immobility Response, Tonic/physiology , Limbic Lobe/drug effects , Male , Phosphorylation , Rats , Rats, Wistar , Signal Transduction/drug effects , Swimming/psychology
19.
Proc Natl Acad Sci U S A ; 113(16): 4500-5, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27001846

ABSTRACT

Anxiety disorders peak in incidence during adolescence, a developmental window that is marked by dynamic changes in gene expression, endocannabinoid signaling, and frontolimbic circuitry. We tested whether genetic alterations in endocannabinoid signaling related to a common polymorphism in fatty acid amide hydrolase (FAAH), which alters endocannabinoid anandamide (AEA) levels, would impact the development of frontolimbic circuitry implicated in anxiety disorders. In a pediatric imaging sample of over 1,000 3- to 21-y-olds, we show effects of the FAAH genotype specific to frontolimbic connectivity that emerge by ∼12 y of age and are paralleled by changes in anxiety-related behavior. Using a knock-in mouse model of the FAAH polymorphism that controls for genetic and environmental backgrounds, we confirm phenotypic differences in frontoamygdala circuitry and anxiety-related behavior by postnatal day 45 (P45), when AEA levels begin to decrease, and also, at P75 but not before. These results, which converge across species and level of analysis, highlight the importance of underlying developmental neurobiology in the emergence of genetic effects on brain circuitry and function. Moreover, the results have important implications for the identification of risk for disease and precise targeting of treatments to the biological state of the developing brain as a function of developmental changes in gene expression and neural circuit maturation.


Subject(s)
Endocannabinoids/metabolism , Frontal Lobe/metabolism , Limbic Lobe/metabolism , Nerve Net/metabolism , Signal Transduction/physiology , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Frontal Lobe/cytology , Humans , Limbic Lobe/cytology , Male , Mice , Mice, Transgenic , Nerve Net/cytology , Species Specificity
20.
Addict Biol ; 21(3): 603-12, 2016 May.
Article in English | MEDLINE | ID: mdl-25899624

ABSTRACT

Orexins (hypocretins) are hypothalamic neuropeptides that innervate the entire neuraxis, including the prelimbic cortex and ventral tegmental area and have been implicated in ethanol-seeking behaviour. The present study aimed to use the orexin-1 (OX1 ) receptor antagonist SB-334867 to examine the role of prelimbic cortex and ventral tegmental area OX1 receptors in cue-induced reinstatement of ethanol-seeking. Ethanol-preferring rats (iP) rats were trained to self-administer ethanol (10 percent v/v, FR3) or sucrose (0.2-1 percent w/v, FR3) in the presence of reward-associated cues before being implanted with indwelling guide cannulae. Rats then underwent extinction training for 11 days. On test days, rats were given a microinjection of vehicle or SB-334867 (3 µg/side) and presented with reward-associated cues to precipitate reinstatement. Results show SB-334867 infused into the prelimbic cortex attenuated cue-induced reinstatement of ethanol-seeking, but not sucrose-seeking. OX1 antagonism in the ventral tegmental area also attenuated cue-induced reinstatement of ethanol-seeking. These findings suggest that OX1 receptors located in the prelimbic cortex and ventral tegmental area are part of a circuit driving cue-mediated ethanol-seeking behaviour.


Subject(s)
Central Nervous System Depressants/pharmacology , Conditioning, Psychological , Drug-Seeking Behavior/drug effects , Ethanol/pharmacology , Extinction, Psychological , Orexin Receptors/drug effects , Animals , Behavior, Animal/drug effects , Benzoxazoles/pharmacology , Cues , Limbic Lobe/metabolism , Male , Naphthyridines , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Rats , Rats, Inbred Strains , Signal Transduction , Urea/analogs & derivatives , Urea/pharmacology , Ventral Tegmental Area/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...