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










Publication year range
1.
Rev Neurosci ; 35(1): 1-20, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-37415576

ABSTRACT

Autism spectrum disorders (ASDs) are among the most common neurodevelopmental diseases. These disorders are characterized by lack of social interaction, by repetitive behavior, and often anxiety and learning disabilities. The brain serotonin (5-HT) system is known to be crucially implicated in a wide range of physiological functions and in the control of different kinds of normal and pathological behavior. A growing number of studies indicate the involvement of the brain 5-HT system in the mechanisms underlying both ASD development and ASD-related behavioral disorders. There are some review papers describing the role of separate key players of the 5-HT system in an ASD and/or autistic-like behavior. In this review, we summarize existing data on the participation of all members of the brain 5-HT system, namely, 5-HT transporter, tryptophan hydroxylase 2, MAOA, and 5-HT receptors, in autism in human and various animal models. Additionally, we describe the most recent studies involving modern techniques for in vivo regulation of gene expression that are aimed at identifying exact roles of 5-HT receptors, MAOA, and 5-HT transporter in the mechanisms underlying autistic-like behavior. Altogether, results of multiple research articles show that the brain 5-HT system intimately partakes in the control of some types of ASD-related behavior, and that specific changes in a function of a certain 5-HT receptor, transporter, and/or enzyme may normalize this aberrant behavior. These data give hope that some of clinically used 5-HT-related drugs have potential for ASD treatment.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Humans , Serotonin/metabolism , Brain/metabolism , Autism Spectrum Disorder/metabolism , Receptors, Serotonin/metabolism , Membrane Transport Proteins/metabolism
2.
Biochemistry (Mosc) ; 88(6): 758-769, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37748872

ABSTRACT

The recombinant B6.CBA-D13Mit76C mouse strain is characterized by an altered sensitivity of 5-HT1A receptors and upregulated 5-HT1A gene transcription. Recently, we found that in B6.CBA-D13Mit76C mice, chronic fluoxetine treatment produced the pro-depressive effect in a forced swim test. Since 5-HT2A receptor blockade may be beneficial in treatment-resistant depression, we investigated the influence of chronic treatment (14 days, intraperitoneally) with selective 5-HT2A antagonist ketanserin (0.5 mg/kg), fluoxetine (20 mg/kg), or fluoxetine + ketanserin on the behavior, functional activity of 5-HT1A and 5-HT2A receptors, serotonin turnover, and transcription of principal genes of the serotonin system in the brain of B6.CBA-D13Mit76C mice. Ketanserin did not reverse the pro-depressive effect of fluoxetine, while fluoxetine, ketanserin, and fluoxetine + ketanserin decreased the functional activity of 5-HT1A receptors and Htr1a gene transcription in the midbrain and hippocampus. All tested drug regimens decreased the mRNA levels of Slc6a4 and Maoa in the midbrain. These changes were not accompanied by a significant shift in the levels of serotonin and its metabolite 5-HIAA. Notably, ketanserin upregulated enzymatic activity of tryptophan hydroxylase 2 (TPH2). Thus, despite some benefits (reduced Htr1a, Slc6a4, and Maoa transcription and increased TPH2 activity), prolonged blockade of 5-HT2A receptors failed to ameliorate the adverse effect of fluoxetine in the case of abnormal functioning of 5-HT1A receptors.


Subject(s)
Fluoxetine , Serotonin , Mice , Animals , Mice, Inbred CBA , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Ketanserin/pharmacology , Receptor, Serotonin, 5-HT1A/genetics
3.
Curr Alzheimer Res ; 20(7): 496-505, 2023.
Article in English | MEDLINE | ID: mdl-37641989

ABSTRACT

AIM: In this study, OXYS rats of three ages (1, 3, and 6 months), a proven model of Alzheimer's disease (AD), at various stages of disease progression were used to thoroughly study the effects of amisulpride on behavior and tau protein phosphorylation. BACKGROUND: With the growing number of patients with AD, the problem of finding a cure is very acute. Neurodegeneration in AD has various causes, one of which is hyperphosphorylation of tau protein. OBJECTIVE: This study aimed to investigate whether amisulpride would affect pathological tau phosphorylation in AD. METHODS: We assessed the influence of chronic administration of amisulpride (3 weeks, 3 mg/kg per day, intraperitoneally)-a 5-HT7 receptor inverse agonist-on behavior and tau hyperphosphorylation in OXYS rats (at ages of 1, 3, and 6 months). RESULTS: Chronic administration of amisulpride dramatically decreased tau phosphorylation in the frontal cortex and hippocampus of 3-month-old OXYS rats. Additionally, in 1- and 3-month-old rats' hippocampi, amisulpride diminished the mRNA level of the Cdk5 gene encoding one of the main tau kinases involved in the 5-HT7 receptor-induced effect on tau phosphorylation. CONCLUSION: Thus, We found that chronic administration of amisulpride could reduce pathological tau hyperphosphorylation while reducing anxiety. We propose amisulpride to have therapeutic potential against AD and that it can be the most effective in the early stages of the disease.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , Rats , Animals , Infant , tau Proteins/metabolism , Amisulpride/pharmacology , Amisulpride/therapeutic use , Rats, Wistar , Drug Inverse Agonism , Alzheimer Disease/metabolism , Brain/pathology , Hippocampus/metabolism , Phosphorylation , Disease Models, Animal
4.
Behav Brain Res ; 438: 114168, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36280010

ABSTRACT

Autism spectrum disorders (ASDs) are some of the most common neurodevelopmental disorders; however, the mechanisms underlying ASDs are still poorly understood. Serotonin (5-HT) and brain-derived neurotrophic factor (BDNF) are known as key players in brain and behavioral plasticity and interact with each other. 5-HT1A receptor is a principal regulator of the brain 5-HT system, which modulates normal and pathological behavior. Here we investigated effects of adeno-associated-virus-based 5-HT1A receptor overexpression in the hippocampus of BTBR mice (which are a model of autism) on various types of behavior and on the expression of 5-HT7 receptor, proBDNF, mature BDNF, and BDNF receptors (TrkB and p75NTR). The 5-HT1A receptor overexpression in BTBR mice reduced stereotyped behavior in the marble-burying test and extended the time spent in the center in the open field test. Meanwhile, this overexpression failed to affect social behavior in the three-chambered test, immobility time in the tail suspension test, locomotor activity in the open field test, and associative learning within the "operant wall" paradigm. The 5-HT1A receptor overexpression in the hippocampus raised hippocampal 5-HT7 receptor mRNA and protein levels. Additionally, the 5-HT1A receptor overexpression lowered both mRNA and protein levels of TrkB receptor but failed to affect proBDNF, mature BDNF, and p75NTR receptor expression in the hippocampus of BTBR mice. Thus, obtained results suggest the involvement of the 5-HT and BDNF systems' interaction mediated by 5-HT1A and TrkB receptors in the mechanisms underlying autistic-like behavior in BTBR mice.


Subject(s)
Autistic Disorder , Brain-Derived Neurotrophic Factor , Animals , Mice , Brain-Derived Neurotrophic Factor/metabolism , Serotonin/metabolism , Autistic Disorder/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Hippocampus/metabolism , Mice, Inbred Strains , RNA, Messenger/metabolism
5.
Biochemistry (Mosc) ; 88(12): 2023-2042, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38462447

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia worldwide that has an increasing impact on aging societies. Besides its critical role in the control of various physiological functions and behavior, brain serotonin (5-HT) system is involved in the regulation of migration, proliferation, differentiation, maturation, and programmed death of neurons. At the same time, a growing body of evidence indicates the involvement of 5-HT neurotransmission in the formation of insoluble aggregates of ß-amyloid and tau protein, the main histopathological signs of AD. The review describes the role of various 5-HT receptors and intracellular signaling cascades induced by them in the pathological processes leading to the development of AD, first of all, in protein aggregation. Changes in the functioning of certain types of 5-HT receptors or associated intracellular signaling mediators prevent accumulation of ß-amyloid plaques and tau protein neurofibrillary tangles. Based on the experimental data, it can be suggested that the use of 5-HT receptors as new drug targets will not only improve cognitive performance in AD, but will be also important in treating the causes of AD-related dementia.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , tau Proteins/metabolism , Serotonin , Amyloid beta-Peptides/metabolism , Receptors, Serotonin/therapeutic use
6.
Biochemistry (Mosc) ; 87(10): 1206-1218, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36273889

ABSTRACT

The mechanisms of autism are of extreme interest due to the high prevalence of this disorder in the human population. In this regard, special attention is given to the transcription factor Freud-1 (encoded by the Cc2d1a gene), which regulates numerous intracellular signaling pathways and acts as a silencer for 5-HT1A serotonin and D2 dopamine receptors. Disruption of the Freud-1 functions leads to the development of various psychopathologies. In this study, we found an increase in the expression of the Cc2d1a/Freud-1 gene in the hippocampus of BTBR mice (model of autistic-like behavior) in comparison with C57Bl/6J mice and examined how restoration of the Cc2d1a/Freud-1 expression in the hippocampus of BTBR mice affects their behavior, expression of 5-HT1A serotonin and D2 dopamine receptors, and CREB and NF-κB intracellular signaling pathways in these animals. Five weeks after administration of the adeno-associated viral vector (AAV) carrying the pAAV_H1-2_shRNA-Freud-1_Syn_EGFP plasmid encoding a small hairpin RNA (shRNA) that suppressed expression of the Cc2d1a/Freud-1 gene, we observed an elevation in the anxiety levels, as well as the increase in the escape latency and path length to the platform in the Morris water maze test, which was probably associated with a strengthening of the active stress avoidance strategy. However, the Cc2d1a/Freud-1 knockdown did not affect the spatial memory and phosphorylation of the CREB transcription factor, although such effect was found in C57Bl/6J mice in our previous study. These results suggest the impairments in the CREB-dependent effector pathway in BTBR mice, which may play an important role in the development of the autistic-like phenotype. The knockdown of Cc2d1a/Freud-1 in the hippocampus of BTBR mice did not affect expression of the 5-HT1A serotonin and D2 dopamine receptors and key NF-κB signaling genes (Nfkb1 and Rela). Our data suggest that the transcription factor Freud-1 plays a significant role in the pathogenesis of anxiety and active stress avoidance in autism.


Subject(s)
Autistic Disorder , Hippocampus , Animals , Humans , Mice , Autistic Disorder/genetics , Autistic Disorder/metabolism , Disease Models, Animal , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hippocampus/metabolism , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serotonin/genetics , Serotonin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism
7.
J Neurosci Res ; 100(7): 1506-1523, 2022 07.
Article in English | MEDLINE | ID: mdl-35443076

ABSTRACT

Heterodimerization between 5-HT7 and 5-HT1A receptors seems to play an important role in the mechanism of depression and antidepressant drug action. It was suggested that the shift of the ratio between 5-HT1A /5-HT7 hetero- and 5-HT1A /5-HT1A homodimers in presynaptic neurons toward 5-HT1A /5-HT1A homodimers is one of the reasons of depression. Consequently, the artificial elevation of 5-HT7 receptor number in presynaptic terminals might restore physiological homo-/heterodimer ratio resulting in antidepressive effect. Here we showed that adeno-associated virus (AAV)-based 5-HT7 receptor overexpression in the midbrain raphe nuclei area produced antidepressive effect in male mice of both C57Bl/6J and genetically predisposed to depressive-like behavior ASC (antidepressant sensitive cataleptics) strains. These changes were accompanied by the elevation of 5-HT7 receptor mRNA level in the frontal cortex of C57Bl/6J and its reduction in the hippocampus of ASC mice. The presence of engineered 5-HT7 receptor in the midbrain of both mouse strains was further demonstrated. Importantly that 5-HT7 receptor overexpression resulted in the reduction of 5-HT1A receptor level in the membrane protein fraction from the midbrain samples of C57Bl/6J, but not ASC, mice. 5-HT7 receptor overexpression caused an increase of 5-HIAA/5-HT ratio in the midbrain and the frontal cortex of C57Bl/6J and in all investigated brain structures of ASC mice. Thus, 5-HT7 receptor overexpression in the raphe nuclei area affects brain 5-HT system and causes antidepressive effect both in C57Bl/6J and in "depressive" ASC male mice. Obtained results indicate the involvement of 5-HT7 receptor in the mechanisms underlying depressive behavior.


Subject(s)
Raphe Nuclei , Receptors, Serotonin , Serotonin , Animals , Antidepressive Agents/metabolism , Brain/metabolism , Dependovirus , Genetic Vectors , Male , Mice , Mice, Inbred C57BL , Raphe Nuclei/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Serotonin/metabolism
8.
Int J Mol Sci ; 22(24)2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34948116

ABSTRACT

The serotonin 5-HT1A receptor is one of the most abundant and widely distributed brain serotonin (5-HT) receptors that play a major role in the modulation of emotions and behavior. The 5-HT1A receptor gene (Htr1a) is under the control of transcription factor Freud-1 (also known as Cc2d1a/Freud-1). Here, using adeno-associated virus (AAV) constructs in vivo, we investigated effects of a Cc2d1a/Freud-1 knockdown in the hippocampus of C57BL/6J mice on behavior, the brain 5-HT system, and brain-derived neurotrophic factor (BDNF). AAV particles carrying the pAAV_H1-2_shRNA-Freud-1_Syn_EGFP plasmid encoding a short-hairpin RNA targeting mouse Cc2d1a/Freud-1 mRNA had an antidepressant effect in the forced swim test 5 weeks after virus injection. The knockdown impaired spatiotemporal memory as assessed in the Morris water maze. pAAV_H1-2_shRNA-Freud-1_Syn_EGFP decreased Cc2d1a/Freud-1 mRNA and protein levels. Furthermore, the Cc2d1a/Freud-1 knockdown upregulated 5-HT and its metabolite 5-hydroxyindoleacetic acid but not their ratio. The Cc2d1a/Freud-1 knockdown failed to increase mRNA and protein levels of Htr1a but diminished a 5-HT1A receptor functional response. Meanwhile, the Cc2d1a/Freud-1 knockdown reduced Creb mRNA expression and CREB phosphorylation and upregulated cFos mRNA. The knockdown enhanced the expression of a BDNF precursor (proBDNF protein), which is known to play a crucial part in neuroplasticity. Our data indicate that transcription factor Cc2d1a/Freud-1 is implicated in the pathogenesis of depressive disorders not only via the 5-HT1A receptor and transcription factor CREB but also through an influence on BDNF.


Subject(s)
Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Maze Learning , Repressor Proteins/metabolism , Serotonin/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Gene Knockdown Techniques , Male , Mice , Repressor Proteins/genetics , Serotonin/genetics
9.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233644

ABSTRACT

The influence of genetic background on sensitivity to drugs represents a topical problem of personalized medicine. Here, we investigated the effect of chronic (20 mg/kg, 14 days, i.p.) antidepressant fluoxetine treatment on recombinant B6-M76C mice, differed from control B6-M76B mice by CBA-derived 102.73-110.56 Mbp fragment of chromosome 13 and characterized by altered sensitivity of 5-HT1A receptors to chronic 8-OH-DPAT administration and higher 5-HT1A receptor mRNA levels in the frontal cortex and hippocampus. Significant changes in the effects of fluoxetine treatment on behavior and brain 5-HT system in recombinant B6-M76C mice were revealed. In contrast to B6-M76B mice, in B6-M76C mice, fluoxetine produced pro-depressive effects, assessed in a forced swim test. Fluoxetine decreased 5-HT1A receptor mRNA levels in the cortex and hippocampus, reduced 5-HT1A receptor protein levels and increased receptor silencer Freud-1 protein levels in the hippocampus of B6-M76C mice. Fluoxetine increased mRNA levels of the gene encoding key enzyme for 5-HT synthesis in the brain, tryptophan hydroxylase-2, but decreased tryptophan hydroxylase-2 protein levels in the midbrain of B6-M76B mice. These changes were accompanied by increased expression of the 5-HT transporter gene. Fluoxetine reduced 5-HT and 5-HIAA levels in cortex, hippocampus and midbrain of B6-M76B and in cortex and midbrain of B6-M76C; mice. These data demonstrate that changes in genetic background may have a dramatic effect on sensitivity to classic antidepressants from the Selective Serotonin Reuptake Inhibitors family. Additionally, the results provide new evidence confirming our idea on the disrupted functioning of 5-HT1A autoreceptors in the brains of B6-M76C mice, suggesting these mice as a model of antidepressant resistance.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Cerebral Cortex/drug effects , Fluoxetine/pharmacology , Hippocampus/drug effects , Receptor, Serotonin, 5-HT1A/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred CBA , Receptor, Serotonin, 5-HT1A/metabolism
10.
Behav Brain Res ; 343: 102-110, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29425916

ABSTRACT

Brain-derived neurotrophic factor (BDNF), its precursor proBDNF, BDNF pro-peptide, BDNF mRNA levels, as well as TrkB and p75NTR receptors mRNA and protein levels, were studied in the brain of rats, selectively bred for more than 85 generations for either the high level or the lack of fear-induced aggressive behavior. Furthermore, we have found that rats of aggressive strain demonstrated both high level of aggression toward humans and increased amplitude of acoustic startle response compared to rats selectively bred for the lack of fear-induced aggression. Significant increase in the BDNF mRNA, mature BDNF and proBDNF protein levels in the raphe nuclei (RN), hippocampus (Hc), nucleus accumbens (NAcc), amygdala, striatum and hypothalamus (Ht) of aggressive rats was revealed. The BDNF/proBDNF ratio was significantly reduced in the Hc and NAcc of highly aggressive rats suggesting prevalence of the proBDNF in these structures. In the Hc and frontal cortex (FC) of aggressive rats, the level of the full-length TrkB (TrkB-FL) receptor form was decreased, whereas the truncated TrkB (TrkB-T) protein level was increased in the RN, FC, substantia nigra and Ht. The TrkB-FL/TrkB-T ratio was significantly decreased in highly aggressive rats suggesting TrkB-T is predominant in highly aggressive rats. The p75NTR expression was slightly changed in majority of studied brain structures of aggressive rats. The data indicate the BDNF system in the brain of aggressive and nonaggressive animals is extremely different at all levels, from transcription to reception, suggesting significant role of BDNF system in the development of highly aggressive phenotype.


Subject(s)
Aggression/physiology , Brain-Derived Neurotrophic Factor/metabolism , Diterpenes/metabolism , Fear/physiology , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Blotting, Western , Genetic Predisposition to Disease , Male , Nerve Tissue Proteins , RNA, Messenger/metabolism , Rats , Receptors, Growth Factor , Reflex, Startle/physiology , Species Specificity
11.
Behav Brain Res ; 310: 20-5, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27150226

ABSTRACT

Serotonin 5-HT1A receptor is known to play a crucial role in the mechanisms of genetically defined aggression. In its turn, 5-HT1A receptor functional state is under control of multiple factors. Among others, transcriptional factors Freud-1 and Freud-2 are known to be involved in the repression of 5-HT1A receptor gene expression. However, implication of these factors in the regulation of behavior is unclear. Here, we investigated the expression of 5-HT1A receptor and silencers Freud-1 and Freud-2 in the brain of rats selectively bred for 85 generations for either high level of fear-induced aggression or its absence. It was shown that Freud-1 and Freud-2 levels were different in aggressive and nonaggressive animals. Freud-1 protein level was decreased in the hippocampus, whereas Freud-2 protein level was increased in the frontal cortex of highly aggressive rats. There no differences in 5-HT1A receptor gene expression were found in the brains of highly aggressive and nonaggressive rats. However, 5-HT1A receptor protein level was decreased in the midbrain and increased in the hippocampus of highly aggressive rats. These data showed the involvement of Freud-1 and Freud-2 in the regulation of genetically defined fear-induced aggression. However, these silencers do not affect transcription of the 5-HT1A receptor gene in the investigated rats. Our data indicate the implication of posttranscriptional rather than transcriptional regulation of 5-HT1A receptor functional state in the mechanisms of genetically determined aggressive behavior. On the other hand, the implication of other transcriptional regulators for 5-HT1A receptor gene in the mechanisms of genetically defined aggression could be suggested.


Subject(s)
Aggression/physiology , Brain/metabolism , Fear/physiology , Receptor, Serotonin, 5-HT1A/metabolism , Repressor Proteins/metabolism , Animals , Blotting, Western , Gene Expression Regulation/physiology , Male , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Species Specificity
12.
Neural Plast ; 2015: 846589, 2015.
Article in English | MEDLINE | ID: mdl-26380122

ABSTRACT

In the present study behavioral effects of the 5-HT2C serotonin receptor were investigated in different mouse strains. The 5-HT2C receptor agonist MK-212 applied intraperitoneally induced significant dose-dependent reduction of distance traveled in the open field test in CBA/Lac mice. This effect was receptor-specific because it was inhibited by the 5-HT2C receptor antagonist RS102221. To study the role of genotype in 5-HT2C receptor-induced hypolocomotion, locomotor activity of seven inbred mouse strains was measured after MK-212 acute treatment. We found that the 5-HT2C receptor stimulation by MK-212 decreased distance traveled in the open field test in CBA/Lac, C57Bl/6, C3H/He, and ICR mice, whereas it failed to affect locomotor activity in DBA/2J, Asn, and Balb/c mice. We also compared the interstrain differences in functional response to 5-HT2C and 5-HT2A receptors activation measured by the quantification of receptor-mediated head-twitches. These experiments revealed significant positive correlation between 5-HT2C and 5-HT2A receptors functional responses for all investigated mouse strains. Moreover, we found that 5-HT2A receptor activation with DOI did not change locomotor activity in CBA/Lac mice. Taken together, our data indicate the implication of 5-HT2C receptors in regulation of locomotor activity and suggest the shared mechanism for functional responses mediated by 5-HT2C and 5-HT2A receptors.


Subject(s)
Motor Activity/genetics , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2C/genetics , Amphetamines/pharmacology , Animals , Dose-Response Relationship, Drug , Genotype , Head Movements/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Pyrazines/pharmacology , Receptor, Serotonin, 5-HT2C/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology , Species Specificity , Spiro Compounds/pharmacology , Sulfonamides/pharmacology
13.
Behav Brain Res ; 290: 45-50, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25934485

ABSTRACT

The brain-derived neurotrophic factor (BDNF), its precursor (proBDNF) and BDNF mRNA levels were studied in the brain of wild rats selectively bred for more than 70 generations for either high level or for the lack of affective aggressiveness towards man. Significant increase of BDNF mRNA level in the frontal cortex and increase of BDNF level in the hippocampus of aggressive rats was revealed. In the midbrain and hippocampus of aggressive rats proBDNF level was increased, whereas BDNF/proBDNF ratio was reduced suggesting the prevalence and increased influence of proBDNF in highly aggressive rats. In the frontal cortex, proBDNF level in aggressive rats was decreased. Thus, considerable structure-specific differences in BDNF and proBDNF levels as well as in BDNF gene expression between highly aggressive and nonaggressive rats were shown. The data suggested the implication of BDNF and its precursor proBDNF in the mechanism of aggressiveness and in the creation of either aggressive or nonaggressive phenotype.


Subject(s)
Aggression/physiology , Brain-Derived Neurotrophic Factor/metabolism , Frontal Lobe/metabolism , Hippocampus/metabolism , Mesencephalon/metabolism , Protein Precursors/metabolism , Animals , Fear , Male , RNA, Messenger/metabolism , Rats
14.
Behav Brain Res ; 274: 1-9, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25101543

ABSTRACT

The effect of glial cell line-derived neurotrophic factor (GDNF) on behavior and brain dopamine system in predisposed to depressive-like behavior ASC (Antidepressant Sensitive Cataleptics) mice in comparison with the parental "nondepressive" CBA mice was studied. In 7days after administration (800ng, i.c.v.) GDNF decreased escape latency time and the path traveled to reach hidden platform in Morris water maze in ASC mice. GDNF enhanced depressive-like behavioral traits in both "nondepressive" CBA and "depressive" ASC mice. In CBA mice, GDNF decreased functional response to agonists of D1 (chloro-APB hydrobromide) and D2 (sumanirole maleate) receptors in tail suspension test, reduced D2 receptor gene expression in the substantia nigra and increased monoamine oxydase A (MAO A) gene expression in the striatum. GDNF increased D1 and D2 receptor genes expression in the nucleus accumbens of ASC mice but failed to alter expression of catechol-O-methyltransferase, dopamine transporter, MAO B and tyrosine hydroxylase genes in both investigated mouse strains. Thus, GDNF produced long-term genotype-dependent effect on behavior and the brain dopamine system. GDNF pretreatment (1) reduced D1 and D2 receptors functional responses and D2 receptor gene expression in s. nigra of CBA mice; (2) increased D1 and D2 receptor genes expression in n. accumbens of ASC mice and (3) improved spatial learning in ASC mice. GDNF enhanced depressive-like behavior both in CBA and ASC mice. The data suggest that genetically defined variance in the cross-talk between GDNF and brain dopamine system contributes to the variability of GDNF-induced responses and might be responsible for controversial GDNF effects.


Subject(s)
Behavioral Symptoms , Brain/drug effects , Dopamine/metabolism , Genetic Predisposition to Disease , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Spatial Behavior/drug effects , Animals , Behavioral Symptoms/genetics , Behavioral Symptoms/metabolism , Behavioral Symptoms/pathology , Brain/metabolism , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Dopamine/genetics , Dopamine Agents/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Hindlimb Suspension , Male , Maze Learning/drug effects , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Reaction Time/drug effects , Swimming/psychology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
15.
J Neurosci Res ; 92(8): 1035-43, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24706292

ABSTRACT

Experiments were made on a congenic AKR.CBA-D13Mit76C (76C) mouse strain created by transferring a chromosome 13 fragment containing the 5-HT1A receptor gene from a CBA strain to an AKR background. It was shown that 76C mice differed from AKR mice by decreased 5-HT1A receptor and tryptophan hydroxylase-2 (tph-2) genes expression in the midbrain. Functional activity of 5-HT2A receptors and 5-HT(2A) receptor mRNA levels in the midbrain and hippocampus of 76C mice were decreased compared with AKR mice. Central brain-derived neurotrophic factor (BDNF) administration (300 ng i.c.v.) reduced 5-HT1A and 5-HT(2A) receptor mRNA levels in the frontal cortex and tph-2 mRNA level in the midbrain of AKR mice. However, BDNF failed to produce any effect on the expression of 5-HT(1A) , 5-HT(2A) , and tph-2 genes in 76C mice but decreased functional activity of 5-HT(2A) receptors in 76C mice and increased it in AKR mice. BDNF restored social deficiency in 76C mice but produced asocial behavior (aggressive attacks towards young mice) in AKR mice. The data indicate that a small genetic variation altered the response to BDNF and show an important role of 5-HT(1A) receptor gene in the 5-HT system response to BDNF treatment and in behavioral effects of BDNF.


Subject(s)
Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Hippocampus/metabolism , Mesencephalon/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Aggression/drug effects , Aggression/physiology , Animals , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Male , Mesencephalon/drug effects , Mice , Mice, Inbred AKR , Mice, Inbred CBA , Receptor, Serotonin, 5-HT1A/metabolism , Social Behavior
16.
J Neurosci Res ; 91(12): 1628-38, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24105724

ABSTRACT

The effect of glial cell line-derived neurotrophic factor (GDNF) on behavior and on the serotonin (5-HT) system of a mouse strain predisposed to depressive-like behavior, ASC/Icg (Antidepressant Sensitive Cataleptics), in comparison with the parental "nondepressive" CBA/Lac mice was studied. Within 7 days after acute administration, GDNF (800 ng, i.c.v.) decreased cataleptic immobility but increased depressive-like behavioral traits in both investigated mouse strains and produced anxiolytic effects in ASC mice. The expression of the gene encoding the key enzyme for 5-HT biosynthesis in the brain, tryptophan hydroxylase-2 (Tph-2), and 5-HT1A receptor gene in the midbrain as well as 5-HT2A receptor gene in the frontal cortex were increased in GDNF-treated ASC mice. At the same time, GDNF decreased 5-HT1A and 5-HT2A receptor gene expression in the hippocampus of ASC mice. GDNF failed to change Tph2, 5-HT1A , or 5-HT2A receptor mRNA levels in CBA mice as well as 5-HT transporter gene expression and 5-HT1A and 5-HT2A receptor functional activity in both investigated mouse strains. The results show 1) a GDNF-induced increase in the expression of key genes of the brain 5-HT system, Tph2, 5-HT1A , and 5-HT2A receptors, and 2) significant genotype-dependent differences in the 5-HT system response to GDNF treatment. The data suggest that genetically defined cross-talk between neurotrophic factors and the brain 5-HT system underlies the variability in behavioral response to GDNF.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Animals , Genetic Predisposition to Disease , Genotype , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , Male , Mice , Mice, Inbred CBA , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT2A/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/genetics , Serotonin/metabolism
17.
Neurosci Lett ; 522(1): 52-6, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22705909

ABSTRACT

Among serotonin (5-HT) receptors, the 5-HT(3) receptor is the only ligand-gated ion-channel. Little is known about the interaction between the 5-HT(3) receptor and other 5-HT receptors and influence of 5-HT(3) chronic activation on other 5-HT receptors and the expression of key genes of 5-HT system. Chronic activation of 5-HT(3) receptor with intracerebroventricularly administrated selective agonist 1-(3-chlorophenyl)biguanide hydrochloride (m-CPBG) (14 days, 40 nmol, i.c.v.) produced significant desensitization of 5-HT(3) and 5-HT(1A) receptors. The hypothermic responses produced by acute administration of selective agonist of 5-HT(3) receptor (m-CPBG, 40 nmol, i.c.v.) or selective agonist of 5-HT(1A) receptor (8-hydroxy-2-(di-n-propylamino)tetralin) (8-OH-DPAT, 1mg/kg, i.p.) was significantly lower in m-CPBG treated mice compared with the mice of control groups. Chronic m-CPBG administration failed to induce any significant change in the 5-HT(2A) receptor functional activity and in the expression of the gene encoding 5-HT(2A) receptor. Chronic activation of 5-HT(3) receptor produced no considerable effect on the expression on 5-HT(3), 5-HT(1A), and 5-HT transporter (5-HTT) and tryptophan hydroxylase-2 (TPH-2) genes - the key genes of brain 5-HT system, in the midbrain, frontal cortex and hippocampus. In conclusion, chronic activation of ionotropic 5-HT(3) receptor produced significant desensitization of 5-HT(3) and postsynaptic 5-HT(1A) receptors but caused no considerable changes in the expression of key genes of the brain 5-HT system.


Subject(s)
Biguanides/pharmacology , Brain/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Receptor, Serotonin, 5-HT2A/physiology , Receptors, Serotonin, 5-HT3/physiology , Serotonin 5-HT3 Receptor Agonists/pharmacology , Animals , Mice , Mice, Inbred Strains , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Serotonin, 5-HT3/genetics
18.
Neuropharmacology ; 61(8): 1360-5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21884711

ABSTRACT

Intracerebroventricular administration of selective agonist of serotonin 5-HT(7) receptor LP44 (4-[2-(methylthio)phenyl]-N-(1,2,3,4-tetrahydro-1-naphthalenyl)-1-pyperasinehexanamide hydrochloride; 10.3, 20.5 or 41.0 nmol) produced considerable hypothermic response in CBA/Lac mice. LP44-induced (20.5 nmol) hypothermia was significantly attenuated by the selective 5-HT(7) receptor antagonist SB 269970 (16.1 fmol, i.c.v.) pretreatment. At the same time, intraperitoneal administration of LP44 in a wide range of doses 1.0, 2.0 or 10.0 mg/kg (2.0, 4.0, 20.0 µmol/kg) did not cause considerable hypothermic response. These findings indicate the implication of central, rather than peripheral 5-HT(7) receptors in the regulation of hypothermia. The comparison of LP44-induced (20.5 nmol) hypothermic reaction in eight inbred mouse strains (DBA/2J, CBA/Lac, C57BL/6, BALB/c, ICR, AKR/J, C3H and Asn) was performed and a significant effect of genotype was found. In the same eight mouse strains, functional activity of 5-HT(1A) and 5-HT(3) receptors was studied. The comparison of hypothermic responses produced by 5-HT(7) receptor agonist LP44 (20.5 nmol, i.c.v.) and 5-HT(1A) receptor agonist 8-OH-DPAT 1.0 mg/kg, i.p. (3.0 µmol/kg), 5-HT(3) receptor agonist m-CPBG (40.0 nmol, i.c.v.) did not reveal considerable interstrain correlations between 5-HT(7) and 5-HT(1A) or 5-HT(3) receptor-induced hypothermia. The selective 5-HT(7) receptor antagonist SB 269970 (16.1 fmol, i.c.v.) failed to attenuate the hypothermic effect of 8-OH-DPAT 1.0 mg/kg, i.p. (3.0 µmol/kg) and m-CPBG (40.0 nmol, i.c.v.) indicating that the brain 5-HT(7) receptor is not involved in the hypothermic effects of 8-OH-DPAT or m-CPBG. The obtained results suggest that the central 5-HT(7) receptor plays an essential role in the mediation of thermoregulation independent of 5-HT(1A) and 5-HT(3) receptors.


Subject(s)
Brain/metabolism , Hypothermia/pathology , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin/metabolism , Analysis of Variance , Animals , Body Temperature/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Hypothermia/chemically induced , Male , Mice , Mice, Inbred Strains , Serotonin/adverse effects , Serotonin Agents/pharmacology , Species Specificity
19.
J Neurosci Res ; 89(2): 267-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21162133

ABSTRACT

Freezing reaction (catalepsy) is a natural passive defensive strategy in animals. An exaggerated form of catalepsy is a symptom of grave brain dysfunction. Catalepsy in mice was shown to be linked to the Map3k1, Il6st, Gzmk, and Hspb3 genes as potential candidates for a high predisposition to catalepsy. The study sought to test the hypothesis of an association between catalepsy and expression of these genes in the brain. Thegenes' mRNA levels were measured in the hypothalamus, hippocampus, frontal cortex, striatum, and midbrain of catalepsy-resistant AKR/J strain and catalepsy-prone strains CBA/Lac, ASC (antidepressant-sensitive cataleptic) and the congenic line AKR.CBA-D13M76C. No association between expression of any investigated genes and predisposition to catalepsy was found. At the same time, multivariate analysis revealed interactions among the expressions of Map3k1, Il6st, Gzmk, and Hspb3 genes in the brain structures. A factor analysis of all variables produced two independent factors explaining 76.2% of the total variance. The catalepsy-resistant AKR strain was distinguished from the catalepsy-prone strains CBA, ASC, and AKR.CBA-D13M76C by factor 1. It was suggested that a high predisposition to catalepsy in mice can be defined by the Map3k1, Il6st, Gzmk, and Hspb3 genes' coexpression network.


Subject(s)
Cytokine Receptor gp130/biosynthesis , Freezing Reaction, Cataleptic/physiology , Gene Regulatory Networks/genetics , Granzymes/biosynthesis , Heat-Shock Proteins/biosynthesis , MAP Kinase Kinase Kinase 1/biosynthesis , Animals , Brain/metabolism , Cytokine Receptor gp130/genetics , Gene Expression , Genetic Predisposition to Disease/genetics , Genotype , Granzymes/genetics , Heat-Shock Proteins/genetics , MAP Kinase Kinase Kinase 1/genetics , Male , Mice , Polymerase Chain Reaction , RNA, Messenger/analysis
20.
Neurosci Lett ; 465(1): 50-4, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19735696

ABSTRACT

The selective agonist of serotonin 5-HT(3) receptor 1-(3-chlorophenyl)biguanide hydrochloride (m-CPBG) administered intracerebroventricularly (40, 80 or 160 nmol) produced long-lasting dose-dependent hypothermic response in AKR/2J mice. m-CPBG (160 nmol i.c.v.) induced profound hypothermia (delta t=-4 degrees C) that lasted up to 7 h. m-CPBG (40 nmol i.c.v.)-induced hypothermia was attenuated by 5-HT(3) receptor antagonist ondansetron pretreatment. At the same time, intraperitoneal administration of m-CPBG in a wide range of doses (0.5, 1.0, 5.0 or 10.0 mg/kg) did not affect the body temperature. These findings indicate: (1) the implication of central, rather than peripheral 5-HT(3) receptor in the thermoregulation; (2) the inability of m-CPBG to cross blood-brain barrier in mice. The comparison of brain 5-HT(3)-induced hypothermic reaction in six inbred mouse strains (DBA/2J, CBA/Lac, C57BL/6, BALB/c, ICR, AKR/J) was performed and two highly sensitive to m-CPBG strains (CBA/Lac and C57BL/6) were found. In the same six mouse strains the functional activity of 5-HT(1A) receptor was studied. The comparison of hypothermic reactions produced by 5-HT(1A) receptor agonist 8-OH-DPAT (1.0 mg/kg i.p.) and m-CPBG revealed significant correlation between 5-HT(3) and 5-HT(1A)-induced hypothermia in five out of six investigated mouse strains. 5-HT(1A) receptor antagonist p-MPPI pretreatment (1 mg/kg i.p.) diminished hypothermia produced by centrally administered m-CPBG (40 nmol i.c.v.). The data suggest the cross-talk between 5-HT(1A) and 5-HT(3) receptors in the mechanism of 5-HT-related hypothermia.


Subject(s)
Hypothermia/chemically induced , Serotonin 5-HT1 Receptor Antagonists , Serotonin 5-HT3 Receptor Agonists , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Aminopyridines/pharmacology , Animals , Biguanides/administration & dosage , Biguanides/pharmacology , Blood-Brain Barrier/drug effects , Body Temperature/drug effects , Dose-Response Relationship, Drug , Hypothermia/physiopathology , Male , Mice , Mice, Inbred Strains , Ondansetron/administration & dosage , Ondansetron/pharmacology , Piperazines/pharmacology , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Serotonin Antagonists/administration & dosage , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/administration & dosage , Serotonin Receptor Agonists/pharmacology , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...