Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Front Integr Neurosci ; 17: 1149159, 2023.
Article En | MEDLINE | ID: mdl-37255843

Prematurity is among the leading risks for poor neurocognitive outcomes. The brains of preterm infants show alterations in structure and electrical activity, but the underlying circuit mechanisms are unclear. To address this, we performed a cross-species study of the electrophysiological activity in the visual cortices of prematurely born infants and mice. Using electroencephalography (EEG) in a sample of healthy preterm (N = 29) and term (N = 28) infants, we found that the maturation of the aperiodic EEG component was accelerated in the preterm cohort, with a significantly flatter 1/f slope when compared to the term infants. The flatter slope was a result of decreased spectral power in the theta and alpha bands and was correlated with the degree of prematurity. To determine the circuit and cellular changes that potentially mediate the changes in 1/f slope after preterm birth, we used in vivo electrophysiology in preterm mice and found that, similar to infants, preterm birth results in a flattened 1/f slope. We analyzed neuronal activity in the visual cortex of preterm (N = 6) and term (N = 9) mice and found suppressed spontaneous firing of neurons. Using immunohistochemistry, we further found an accelerated maturation of inhibitory circuits. In both preterm mice and infants, the functional maturation of the cortex was accelerated, underscoring birth as a critical checkpoint in cortical maturation. Our study points to a potential mechanism of preterm birth-related changes in resting neural activity, highlighting the utility of a cross-species approach in studying the neural circuit mechanisms of preterm birth-related neurodevelopmental conditions.

2.
Nat Commun ; 14(1): 459, 2023 01 28.
Article En | MEDLINE | ID: mdl-36709330

Multiple trans-synaptic complexes organize synapse development, yet their roles in the mature brain and cooperation remain unclear. We analyzed the postsynaptic adhesion protein LRRTM1 in the prefrontal cortex (PFC), a region relevant to cognition and disorders. LRRTM1 knockout (KO) mice had fewer synapses, and we asked whether other synapse organizers counteract further loss. This determined that the immunoglobulin family member SynCAM 1 controls synapse number in PFC and was upregulated upon LRRTM1 loss. Combined LRRTM1 and SynCAM 1 deletion substantially lowered dendritic spine number in PFC, but not hippocampus, more than the sum of single KO impairments. Their cooperation extended presynaptically, and puncta of Neurexins, LRRTM1 partners, were less abundant in double KO (DKO) PFC. Electrophysiology and fMRI demonstrated aberrant neuronal activity in DKO mice. Further, DKO mice were impaired in social interactions and cognitive tasks. Our results reveal concerted roles of LRRTM1 and SynCAM 1 across synaptic, network, and behavioral domains.


Cell Adhesion Molecule-1 , Membrane Proteins , Nerve Tissue Proteins , Synapses , Animals , Mice , Cognition , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/metabolism , Synapses/metabolism , Cell Adhesion Molecule-1/genetics , Cell Adhesion Molecule-1/metabolism
3.
bioRxiv ; 2023 Apr 12.
Article En | MEDLINE | ID: mdl-36711801

Prematurity is among the leading risks for poor neurocognitive outcomes. The brains of preterm infants show alterations in structure and electrical activity, but the underlying circuit mechanisms are unclear. To address this, we performed a cross-species study of the electrophysiological activity in the visual cortices of prematurely born infants and mice. Using electroencephalography (EEG) in a sample of healthy preterm (N=29) and term (N=28) infants, we found that the maturation of the aperiodic EEG component was accelerated in the preterm cohort, with a significantly flatter 1/f slope when compared to the term infants. The flatter slope was a result of decreased spectral power in the theta and alpha bands and was correlated with the degree of prematurity. To determine the circuit and cellular changes that potentially mediate the changes in 1/f slope after preterm birth, we used in vivo electrophysiology in preterm mice and found that, similar to infants, preterm birth results in a flattened 1/f slope. We analyzed neuronal activity in the visual cortex of preterm mice (N=6 preterm and 9 term mice) and found suppressed spontaneous firing of neurons. Using immunohistochemistry, we further found an accelerated maturation of inhibitory circuits. In both preterm mice and infants, the functional maturation of the cortex was accelerated, underscoring birth as a critical checkpoint in cortical maturation. Our study points to a potential mechanism of preterm birth-related changes in resting neural activity, highlighting the utility of a cross-species approach in studying the neural circuit mechanisms of preterm birth-related neurodevelopmental conditions.

4.
Front Cell Neurosci ; 14: 76, 2020.
Article En | MEDLINE | ID: mdl-32372915

Plasticity is a fundamental property of the nervous system that enables its adaptations to the ever-changing environment. Heightened plasticity typical for developing circuits facilitates their robust experience-dependent functional maturation. This plasticity wanes during adolescence to permit the stabilization of mature brain function, but abundant evidence supports that adult circuits exhibit both transient and long-term experience-induced plasticity. Cortical plasticity has been extensively studied throughout the life span in sensory systems and the main distinction between development and adulthood arising from these studies is the concept that passive exposure to relevant information is sufficient to drive robust plasticity early in life, while higher-order attentional mechanisms are necessary to drive plastic changes in adults. Recent work in the primary visual and auditory cortices began to define the circuit mechanisms that govern these processes and enable continuous adaptation to the environment, with transient circuit disinhibition emerging as a common prerequisite for both developmental and adult plasticity. Drawing from studies in visual and auditory systems, this review article summarizes recent reports on the circuit and cellular mechanisms of experience-driven plasticity in the developing and adult brains and emphasizes the similarities and differences between them. The benefits of distinct plasticity mechanisms used at different ages are discussed in the context of sensory learning, as well as their relationship to maladaptive plasticity and neurodevelopmental brain disorders. Knowledge gaps and avenues for future work are highlighted, and these will hopefully motivate future research in these areas, particularly those about the learning of complex skills during development.

5.
Cereb Cortex ; 30(1): 226-240, 2020 01 10.
Article En | MEDLINE | ID: mdl-31034037

Brain development is likely impacted by micronutrients. This is supported by the effects of the ω-3 fatty acid docosahexaenoic acid (DHA) during early neuronal differentiation, when it increases neurite growth. Aiming to delineate DHA roles in postnatal stages, we selected the visual cortex due to its stereotypic maturation. Immunohistochemistry showed that young mice that received dietary DHA from birth exhibited more abundant presynaptic and postsynaptic specializations. DHA also increased density and size of synapses in a dose-dependent manner in cultured neurons. In addition, dendritic arbors of neurons treated with DHA were more complex. In agreement with improved connectivity, DHA enhanced physiological parameters of network maturation in vitro, including bursting strength and oscillatory behavior. Aiming to analyze functional maturation of the cortex, we performed in vivo electrophysiological recordings from awake mice to measure responses to patterned visual inputs. Dietary DHA robustly promoted the developmental increase in visual acuity, without altering light sensitivity. The visual acuity of DHA-supplemented animals continued to improve even after their cortex had matured and DHA abolished the acuity plateau. Our findings show that the ω-3 fatty acid DHA promotes synaptic connectivity and cortical processing. These results provide evidence that micronutrients can support the maturation of neuronal networks.


Docosahexaenoic Acids/administration & dosage , Neurons/drug effects , Neurons/physiology , Synapses/drug effects , Synapses/physiology , Visual Cortex/drug effects , Visual Cortex/growth & development , Animals , Cells, Cultured , Dendrites/drug effects , Dendrites/physiology , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/cytology , Visual Acuity/physiology
6.
Neural Plast ; 2019: 1538137, 2019.
Article En | MEDLINE | ID: mdl-31565044

Experience remodels cortical connectivity during developmental windows called critical periods. Experience-dependent regulation of synaptic strength during these periods establishes circuit functions that are stabilized as critical period plasticity wanes. These processes have been extensively studied in the developing visual cortex, where critical period opening and closure are orchestrated by the assembly, maturation, and strengthening of distinct synapse types. The synaptic specificity of these processes points towards the involvement of distinct molecular pathways. Attractive candidates are pre- and postsynaptic transmembrane proteins that form adhesive complexes across the synaptic cleft. These synapse-organizing proteins control synapse development and maintenance and modulate structural and functional properties of synapses. Recent evidence suggests that they have pivotal roles in the onset and closure of the critical period for vision. In this review, we describe roles of synapse-organizing adhesion molecules in the regulation of visual critical period plasticity and we discuss the potential they offer to restore circuit functions in amblyopia and other neurodevelopmental disorders.


Critical Period, Psychological , Neuronal Plasticity/physiology , Neurons/metabolism , Synapses/metabolism , Animals , Humans , Membrane Proteins/metabolism
7.
Cell Rep ; 26(2): 381-393.e6, 2019 01 08.
Article En | MEDLINE | ID: mdl-30625321

Cortical plasticity peaks early in life and tapers in adulthood, as exemplified in the primary visual cortex (V1), wherein brief loss of vision in one eye reduces cortical responses to inputs from that eye during the critical period but not in adulthood. The synaptic locus of cortical plasticity and the cell-autonomous synaptic factors determining critical periods remain unclear. We here demonstrate that the immunoglobulin protein Synaptic Cell Adhesion Molecule 1 (SynCAM 1/Cadm1) is regulated by visual experience and limits V1 plasticity. Loss of SynCAM 1 selectively reduces the number of thalamocortical inputs onto parvalbumin (PV+) interneurons, impairing the maturation of feedforward inhibition in V1. SynCAM 1 acts in PV+ interneurons to actively restrict cortical plasticity, and brief PV+-specific knockdown of SynCAM 1 in adult visual cortex restores juvenile-like plasticity. These results identify a synapse-specific, cell-autonomous mechanism for thalamocortical visual circuit maturation and closure of the visual critical period.


Cell Adhesion Molecule-1/metabolism , Neuronal Plasticity , Synapses/metabolism , Visual Cortex/metabolism , Animals , Cells, Cultured , Female , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Neurogenesis , Parvalbumins/genetics , Parvalbumins/metabolism , Rats , Rats, Sprague-Dawley , Synapses/physiology , Thalamus/growth & development , Thalamus/metabolism , Thalamus/physiology , Visual Cortex/cytology , Visual Cortex/growth & development , Visual Cortex/physiology
8.
Methods Mol Biol ; 1538: 277-289, 2017.
Article En | MEDLINE | ID: mdl-27943197

Understanding the rules of synapse dynamics in the context of development, learning, and nervous system disorders is an important part of several fields of neuroscience. Despite significant methodological advances, observations of structural dynamics of synapses still present a significant experimental challenge. In this chapter we describe a set of techniques that allow repetitive observations of synaptic structures in vitro in organotypic cultures of rodent hippocampus. We describe culturing of slices, transfection with reporter-carrying plasmids, repetitive imaging of dendritic spines with confocal laser scanning microscopy and analysis of spine morphology dynamics.


Dendritic Spines/physiology , Microscopy, Confocal , Neuronal Plasticity , Synapses/physiology , Animals , Gene Expression , Genes, Reporter , Hippocampus/cytology , Hippocampus/physiology , Neurons/cytology , Neurons/physiology , Organ Culture Techniques , Rats
9.
J Neurosci ; 36(28): 7464-75, 2016 07 13.
Article En | MEDLINE | ID: mdl-27413156

UNLABELLED: Select adhesion proteins control the development of synapses and modulate their structural and functional properties. Despite these important roles, the extent to which different synapse-organizing mechanisms act across brain regions to establish connectivity and regulate network properties is incompletely understood. Further, their functional roles in different neuronal populations remain to be defined. Here, we applied diffusion tensor imaging (DTI), a modality of magnetic resonance imaging (MRI), to map connectivity changes in knock-out (KO) mice lacking the synaptogenic cell adhesion protein SynCAM 1. This identified reduced fractional anisotropy in the hippocampal CA3 area in absence of SynCAM 1. In agreement, mossy fiber refinement in CA3 was impaired in SynCAM 1 KO mice. Mossy fibers make excitatory inputs onto postsynaptic specializations of CA3 pyramidal neurons termed thorny excrescences and these structures were smaller in the absence of SynCAM 1. However, the most prevalent targets of mossy fibers are GABAergic interneurons and SynCAM 1 loss unexpectedly reduced the number of excitatory terminals onto parvalbumin (PV)-positive interneurons in CA3. SynCAM 1 KO mice additionally exhibited lower postsynaptic GluA1 expression in these PV-positive interneurons. These synaptic imbalances in SynCAM 1 KO mice resulted in CA3 disinhibition, in agreement with reduced feedforward inhibition in this network in the absence of SynCAM 1-dependent excitatory drive onto interneurons. In turn, mice lacking SynCAM 1 were impaired in memory tasks involving CA3. Our results support that SynCAM 1 modulates excitatory mossy fiber inputs onto both interneurons and principal neurons in the hippocampal CA3 area to balance network excitability. SIGNIFICANCE STATEMENT: This study advances our understanding of synapse-organizing mechanisms on two levels. First, the data support that synaptogenic proteins guide connectivity and can function in distinct brain regions even if they are expressed broadly. Second, the results demonstrate that a synaptogenic process that controls excitatory inputs to both pyramidal neurons and interneurons can balance excitation and inhibition. Specifically, the study reveals that hippocampal CA3 connectivity is modulated by the synapse-organizing adhesion protein SynCAM 1 and identifies a novel, SynCAM 1-dependent mechanism that controls excitatory inputs onto parvalbumin-positive interneurons. This enables SynCAM 1 to regulate feedforward inhibition and set network excitability. Further, we show that diffusion tensor imaging is sensitive to these cellular refinements affecting neuronal connectivity.


CA3 Region, Hippocampal/cytology , Cell Adhesion Molecules/metabolism , Gene Expression Regulation/genetics , Immunoglobulins/metabolism , Neural Inhibition/physiology , Neural Pathways/physiology , Synapses/physiology , Animals , CA3 Region, Hippocampal/diagnostic imaging , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/genetics , Conditioning, Classical/drug effects , Fear/drug effects , Female , GABA Antagonists/pharmacology , Gene Expression Regulation/drug effects , Immunoglobulins/genetics , In Vitro Techniques , Male , Memory Disorders/diagnostic imaging , Memory Disorders/genetics , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/drug effects , Parvalbumins/metabolism , Pyridazines/pharmacology , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Time Factors
10.
Nat Commun ; 6: 8005, 2015 Aug 14.
Article En | MEDLINE | ID: mdl-26272629

Retinitis pigmentosa is a leading cause of inherited blindness, with no effective treatment currently available. Mutations primarily in genes expressed in rod photoreceptors lead to early rod death, followed by a slower phase of cone photoreceptor death. Rd1 mice provide an invaluable animal model to evaluate therapies for the disease. We previously reported that overexpression of histone deacetylase 4 (HDAC4) prolongs rod survival in rd1 mice. Here we report a key role of a short N-terminal domain of HDAC4 in photoreceptor protection. Expression of this domain suppresses multiple cell death pathways in photoreceptor degeneration, and preserves even more rd1 rods than the full-length HDAC4 protein. Expression of a short N-terminal domain of HDAC4 as a transgene in mice carrying the rd1 mutation also prolongs the survival of cone photoreceptors, and partially restores visual function. Our results may facilitate the design of a small protein therapy for some forms of retinitis pigmentosa.


Histone Deacetylases/metabolism , Histone Deacetylases/pharmacology , Photoreceptor Cells/drug effects , Repressor Proteins/metabolism , Repressor Proteins/pharmacology , Retinitis Pigmentosa/drug therapy , Vision, Ocular/drug effects , Animals , Electroretinography , Gene Deletion , Gene Expression Regulation , Genotype , HEK293 Cells , Histone Deacetylases/genetics , Humans , Mice , Mice, Transgenic , Protein Structure, Tertiary , Repressor Proteins/genetics
11.
J Comp Neurol ; 522(4): 900-20, 2014 Mar.
Article En | MEDLINE | ID: mdl-23982969

Adhesive interactions in the retina instruct the developmental specification of inner retinal layers. However, potential roles of adhesion in the development and function of photoreceptor synapses remain incompletely understood. This contrasts with our understanding of synapse development in the CNS, which can be guided by select adhesion molecules such as the Synaptic Cell Adhesion Molecule 1 (SynCAM 1/CADM1/nectin-like 2 protein). This immunoglobulin superfamily protein modulates the development and plasticity of classical excitatory synapses. We show here by immunoelectron microscopy and immunoblotting that SynCAM 1 is expressed on mouse rod photoreceptors and their terminals in the outer nuclear and plexiform layers in a developmentally regulated manner. Expression of SynCAM 1 on rods is low in early postnatal stages (P3-P7) but increases after eye opening (P14). In support of functional roles in the photoreceptors, electroretinogram recordings demonstrate impaired responses to light stimulation in SynCAM 1 knockout (KO) mice. In addition, the structural integrity of synapses in the OPL requires SynCAM 1. Quantitative ultrastructural analysis of SynCAM 1 KO retina measured fewer fully assembled, triadic rod ribbon synapses. Furthermore, rod synapse ribbons are shortened in KO mice, and protein levels of Ribeye, a major structural component of ribbons, are reduced in SynCAM 1 KO retina. Together, our results implicate SynCAM 1 in the synaptic organization of the rod visual pathway and provide evidence for novel roles of synaptic adhesion in the structural and functional integrity of ribbon synapses.


Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Retina/cytology , Retina/growth & development , Retinal Rod Photoreceptor Cells/ultrastructure , Synapses/metabolism , Alcohol Oxidoreductases , Analysis of Variance , Animals , Animals, Newborn , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/ultrastructure , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Electroretinography , Female , Gene Expression Regulation, Developmental/genetics , Immunoglobulins/genetics , Immunoglobulins/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/ultrastructure , Vesicular Glutamate Transport Protein 1/metabolism
12.
Behav Brain Res ; 224(1): 180-8, 2011 Oct 10.
Article En | MEDLINE | ID: mdl-21679729

Environmental disruptions can influence neurodevelopment during pre- and postnatal periods. Given such a large time window of opportunity for insult, the "double-hit hypothesis" proposes that exposure to an environmental challenge may impact development such that an individual becomes vulnerable to developing a psychopathology, which then manifests upon exposure to a second challenge later in life. The present study in male rats utilized the framework of the "double-hit hypothesis" to investigate potential compounding effects of maternal immune activation (MIA) during pregnancy and exposure of offspring to stress during juvenility on physiological and behavioural indications of anxiety in adulthood. We used an established rat model of MIA via maternal treatment with polyinosinic:polycytidylic acid (poly I:C) on gestation day 15 in combination with a model of juvenile stress (applied ages 27-29 d) in offspring to explore potential interacting/additive effects. First, we confirmed our employment of the MIA model by replicating previous findings that prenatal treatment with poly I:C caused deficits in sensorimotor gating in adult offspring, as measured by prepulse inhibition. Juvenile stress, on the other hand, had no effect on prepulse inhibition. In terms of anxiety-related behaviour and physiology, we found that prenatal poly I:C alone or in combination with juvenile stress had no effects on body weight, adrenal weight, and plasma concentration of corticosterone and cytokines in adult rats. MIA and juvenile stress increased anxiety-related behaviour on the elevated plus maze, but did so independently of each other. In all, our findings do not support an interaction between MIA and juvenile stress in terms of producing marked changes related to anxiety-like behaviour in adulthood.


Anxiety/etiology , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/complications , Acoustic Stimulation/adverse effects , Adrenal Glands/pathology , Analysis of Variance , Animals , Animals, Newborn , Anxiety/pathology , Body Weight/drug effects , Corticosterone/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Inhibition, Psychological , Male , Maze Learning/physiology , Poly I-C/adverse effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Sprague-Dawley , Reflex, Startle/physiology
13.
Behav Brain Funct ; 7: 1, 2011 Jan 04.
Article En | MEDLINE | ID: mdl-21205317

BACKGROUND: Several recent studies have highlighted the important role of immunity-related molecules in synaptic plasticity processes in the developing and adult mammalian brains. It has been suggested that neuronal MHCI (major histocompatibility complex class I) genes play a role in the refinement and pruning of synapses in the developing visual system. As a fast evolutionary rate may generate distinct properties of molecules in different mammalian species, we studied the expression of MHCI molecules in a nonhuman primate, the common marmoset monkey (Callithrix jacchus). METHODS AND RESULTS: Analysis of expression levels of MHCI molecules in the developing visual cortex of the common marmoset monkeys revealed a distinct spatio-temporal pattern. High levels of expression were detected very early in postnatal development, at a stage when synaptogenesis takes place and ocular dominance columns are formed. To determine whether the expression of MHCI molecules is regulated by retinal activity, animals were subjected to monocular enucleation. Levels of MHCI heavy chain subunit transcripts in the visual cortex were found to be elevated in response to monocular enucleation. Furthermore, MHCI heavy chain immunoreactivity revealed a banded pattern in layer IV of the visual cortex in enucleated animals, which was not observed in control animals. This pattern of immunoreactivity indicated that higher expression levels were associated with retinal activity coming from the intact eye. CONCLUSIONS: These data demonstrate that, in the nonhuman primate brain, expression of MHCI molecules is regulated by neuronal activity. Moreover, this study extends previous findings by suggesting a role for neuronal MHCI molecules during synaptogenesis in the visual cortex.


Genes, MHC Class I/physiology , Visual Cortex/growth & development , Visual Cortex/metabolism , Age Factors , Animals , Callithrix , Eye Enucleation/methods , Gene Expression Regulation, Developmental , Male , Neurons/metabolism , Neurons/physiology , Visual Cortex/physiology
14.
Cell Mol Neurobiol ; 30(6): 827-39, 2010 Aug.
Article En | MEDLINE | ID: mdl-20232136

Several recent studies suggested a role for neuronal major histocompatibility complex class I (MHCI) molecules in certain forms of synaptic plasticity in the hippocampus of rodents. Here, we report for the first time on the expression pattern and functional properties of MHCI molecules in the hippocampus of a nonhuman primate, the common marmoset monkey (Callithrix jacchus). We detected a presynaptic, mossy fiber-specific localization of MHCI proteins within the marmoset hippocampus. MHCI molecules were present in the large, VGlut1-positive, mossy fiber terminals, which provide input to CA3 pyramidal neurons. Furthermore, whole-cell recordings of CA3 pyramidal neurons in acute hippocampal slices of the common marmoset demonstrated that application of antibodies which specifically block MHCI proteins caused a significant decrease in the frequency, and a transient increase in the amplitude, of spontaneous excitatory postsynaptic currents (sEPSCs) in CA3 pyramidal neurons. These findings add to previous studies on neuronal MHCI molecules by describing their expression and localization in the primate hippocampus and by implicating them in plasticity-related processes at the mossy fiber-CA3 synapses. In addition, our results suggest significant interspecies differences in the localization of neuronal MHCI molecules in the hippocampus of mice and marmosets, as well as in their potential function in these species.


Callithrix/immunology , Histocompatibility Antigens Class I/immunology , Mossy Fibers, Hippocampal/immunology , Neurons/immunology , Synapses/immunology , Synaptic Transmission/immunology , Animals , Antibodies/immunology , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/immunology , Cell Line , Female , Humans , In Vitro Techniques , Male , Neurons/cytology , Presynaptic Terminals/metabolism , Protein Transport
15.
Cell Mol Neurobiol ; 28(3): 331-42, 2008 May.
Article En | MEDLINE | ID: mdl-18197473

Serotonin is implicated in stress-related psychopathologies. Two isoforms of the rate-limiting enzyme of serotonin biosynthesis, tryptophan hydroxylase, TPH1 and TPH2, are known. We show here that in the rat dorsal raphe nucleus (DRN), the nucleus that contains the highest number of 5-HT neurons in the brain, TPH1 mRNA reveals a low level of expression but is detectable both by quantitative real-time PCR and in situ hybridization whereas in the pineal gland (PiG), TPH1 mRNA is strongly expressed. To examine effects of stress on TPH expression we exposed male Wistar rats to daily restraint stress for 1 week. As shown by quantitative real-time PCR, TPH1 mRNA is 2.5-fold upregulated by the stress in DRN but not in PiG. Using 3'-RACE, we identified two TPH2 mRNA splice variants in the rat DRN which differ in the length of their 3'-untranslated regions (UTRs). TPH2b (with a short 3'-UTR) is the predominant variant in the DRN, whereas TPH2a (with a longer 3'-UTR) shows a low abundance in this nucleus. In the PiG, only TPH2b is detectable revealing a low level of expression. Expression of both TPH2 splice variants is not affected by stress, neither in DRN nor in the PiG. These data indicate that TPH1 in the serotonergic neurons of the DRN might be relevant for stress-induced psychopathologies.


Alternative Splicing/physiology , Raphe Nuclei/metabolism , Stress, Physiological/genetics , Tryptophan Hydroxylase/genetics , Adrenal Glands/anatomy & histology , Animals , Gene Expression Regulation , Male , Nucleic Acid Amplification Techniques , Organ Size , Pineal Gland/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Restraint, Physical/physiology , Stress, Physiological/metabolism , Tryptophan Hydroxylase/metabolism
...