Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Hum Genet ; 111(2): 364-382, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38272033

ABSTRACT

The calcium/calmodulin-dependent protein kinase type 2 (CAMK2) family consists of four different isozymes, encoded by four different genes-CAMK2A, CAMK2B, CAMK2G, and CAMK2D-of which the first three have been associated recently with neurodevelopmental disorders. CAMK2D is one of the major CAMK2 proteins expressed in the heart and has been associated with cardiac anomalies. Although this CAMK2 isoform is also known to be one of the major CAMK2 subtypes expressed during early brain development, it has never been linked with neurodevelopmental disorders until now. Here we show that CAMK2D plays an important role in neurodevelopment not only in mice but also in humans. We identified eight individuals harboring heterozygous variants in CAMK2D who display symptoms of intellectual disability, delayed speech, behavioral problems, and dilated cardiomyopathy. The majority of the variants tested lead to a gain of function (GoF), which appears to cause both neurological problems and dilated cardiomyopathy. In contrast, loss-of-function (LoF) variants appear to induce only neurological symptoms. Together, we describe a cohort of individuals with neurodevelopmental disorders and cardiac anomalies, harboring pathogenic variants in CAMK2D, confirming an important role for the CAMK2D isozyme in both heart and brain function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cardiomyopathy, Dilated , Intellectual Disability , Neurodevelopmental Disorders , Animals , Humans , Mice , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart , Neurodevelopmental Disorders/genetics
2.
JCI Insight ; 6(23)2021 12 08.
Article in English | MEDLINE | ID: mdl-34877936

ABSTRACT

Tuberous sclerosis complex (TSC), caused by heterozygous mutations in TSC1 or TSC2, frequently results in intractable epilepsy. Here, we made use of an inducible Tsc1-knockout mouse model, allowing us to study electrophysiological and molecular changes of Tsc1-induced epileptogenesis over time. We recorded from pyramidal neurons in the hippocampus and somatosensory cortex (L2/L3) and combined this with an analysis of transcriptome changes during epileptogenesis. Deletion of Tsc1 resulted in hippocampus-specific changes in excitability and adaptation, which emerged before seizure onset and progressed over time. All phenotypes were rescued after early treatment with rapamycin, an mTOR inhibitor. Later in epileptogenesis, we observed a hippocampal increase of excitation-to-inhibition ratio. These cellular changes were accompanied by dramatic transcriptional changes, especially after seizure onset. Most of these changes were rescued upon rapamycin treatment. Of the genes encoding ion channels or belonging to the Gene Ontology term action potential, 27 were differentially expressed just before seizure onset, suggesting a potential driving role in epileptogenesis. Our data highlight the complex changes driving epileptogenesis in TSC, including the changed expression of multiple ion channels. Our study emphasizes inhibition of the TSC/mTOR signaling pathway as a promising therapeutic approach to target epilepsy in patients with TSC.


Subject(s)
Epilepsy/genetics , Tuberous Sclerosis/genetics , Animals , Disease Models, Animal , Humans , Ion Channels , Male , Mice , Mice, Knockout , Tuberous Sclerosis/pathology
3.
PLoS Biol ; 19(5): e3001279, 2021 05.
Article in English | MEDLINE | ID: mdl-34038402

ABSTRACT

Hyperactivation of the mammalian target of rapamycin (mTOR) pathway can cause malformation of cortical development (MCD) with associated epilepsy and intellectual disability (ID) through a yet unknown mechanism. Here, we made use of the recently identified dominant-active mutation in Ras Homolog Enriched in Brain 1 (RHEB), RHEBp.P37L, to gain insight in the mechanism underlying the epilepsy caused by hyperactivation of the mTOR pathway. Focal expression of RHEBp.P37L in mouse somatosensory cortex (SScx) results in an MCD-like phenotype, with increased mTOR signaling, ectopic localization of neurons, and reliable generalized seizures. We show that in this model, the mTOR-dependent seizures are caused by enhanced axonal connectivity, causing hyperexcitability of distally connected neurons. Indeed, blocking axonal vesicle release from the RHEBp.P37L neurons alone completely stopped the seizures and normalized the hyperexcitability of the distally connected neurons. These results provide new evidence of the extent of anatomical and physiological abnormalities caused by mTOR hyperactivity, beyond local malformations, which can lead to generalized epilepsy.


Subject(s)
Ras Homolog Enriched in Brain Protein/metabolism , Seizures/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Axons/metabolism , Brain/metabolism , Disease Models, Animal , Epilepsy/metabolism , Epilepsy/physiopathology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Seizures/physiopathology , Signal Transduction , Somatosensory Cortex/metabolism
4.
J Neurosci ; 41(26): 5579-5594, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34021041

ABSTRACT

Protein phosphatase 2B (PP2B) is critical for synaptic plasticity and learning, but the molecular mechanisms involved remain unclear. Here we identified different types of proteins that interact with PP2B, including various structural proteins of the postsynaptic densities (PSDs) of Purkinje cells (PCs) in mice. Deleting PP2B reduced expression of PSD proteins and the relative thickness of PSD at the parallel fiber to PC synapses, whereas reexpression of inactive PP2B partly restored the impaired distribution of nanoclusters of PSD proteins, together indicating a structural role of PP2B. In contrast, lateral mobility of surface glutamate receptors solely depended on PP2B phosphatase activity. Finally, the level of motor learning covaried with both the enzymatic and nonenzymatic functions of PP2B. Thus, PP2B controls synaptic function and learning both through its action as a phosphatase and as a structural protein that facilitates synapse integrity.SIGNIFICANCE STATEMENT Phosphatases are generally considered to serve their critical role in learning and memory through their enzymatic operations. Here, we show that protein phosphatase 2B (PP2B) interacts with structural proteins at the synapses of cerebellar Purkinje cells. Differentially manipulating the enzymatic and structural domains of PP2B leads to different phenotypes in cerebellar learning. We propose that PP2B is crucial for cerebellar learning via two complementary actions, an enzymatic and a structural operation.


Subject(s)
Calcineurin/metabolism , Learning/physiology , Neuronal Plasticity/physiology , Purkinje Cells/physiology , Animals , Eye Movements/physiology , Mice , Post-Synaptic Density/metabolism
5.
Brain Res Bull ; 171: 209-220, 2021 06.
Article in English | MEDLINE | ID: mdl-33774142

ABSTRACT

Neurodevelopmental disorders are a complex and heterogeneous group of neurological disorders characterized by their early-onset and estimated to affect more than 3% of children worldwide. The rapid advancement of sequencing technologies in the past years allowed the identification of hundreds of variants in several different genes causing neurodevelopmental disorders. Between those, new variants in the Calcium/calmodulin dependent protein kinase II (CAMK2) genes were recently linked to intellectual disability. Despite many years of research on CAMK2, this proves for the first time that this well-known and highly conserved molecule plays an important role in the human brain. In this review, we give an overview of the identified CAMK2 variants, and we speculate on potential mechanisms through which dysfunctions in CAMK2 result in neurodevelopmental disorders. Additionally, we discuss how the identification of CAMK2 variants might result in new exciting discoveries regarding the function of CAMK2 in the human brain.


Subject(s)
Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neurodevelopmental Disorders/metabolism , Neuronal Plasticity/physiology , Animals , Calcium/metabolism , Calmodulin/metabolism , Humans , Phosphorylation
7.
Ann Clin Transl Neurol ; 6(7): 1273-1291, 2019 07.
Article in English | MEDLINE | ID: mdl-31353861

ABSTRACT

OBJECTIVE: An epilepsy mouse model for Tuberous Sclerosis Complex (TSC) was developed and validated to investigate the mechanisms underlying epileptogenesis. Furthermore, the possible antiepileptogenic properties of commonly used antiepileptic drugs (AEDs) and new compounds were assessed. METHODS: Tsc1 deletion was induced in CAMK2A-expressing neurons of adult mice. The antiepileptogenic properties of commonly used AEDs and inhibitors of the mTOR pathways were assessed by EEG recordings and by molecular read outs. RESULTS: Mice developed epilepsy in a narrow time window (10 ± 2 days) upon Tsc1 gene deletion. Seizure frequency but not duration increased over time. Seizures were lethal within 18 days, were unpredictable, and did not correlate to seizure onset, length or frequency, reminiscent of sudden unexpected death in epilepsy (SUDEP). Tsc1 gene deletion resulted in a strong activation of the mTORC1 pathway, and both epileptogenesis and lethality could be entirely prevented by RHEB1 gene deletion or rapamycin treatment. However, other inhibitors of the mTOR pathway such as AZD8055 and PF4708671 were ineffective. Except for ketogenic diet, none of commonly used AEDs showed an effect on mTORC1 activity. Vigabatrin and ketogenic diet treatment were able to significantly delay seizure onset. In contrast, survival was shortened by lamotrigine. INTERPRETATION: This novel Tsc1 mouse model is highly suitable to assess the efficacy of antiepileptic and -epileptogenic drugs to treat mTORC1-dependent epilepsy. Additionally, it allows us to study the mechanisms underlying mTORC1-mediated epileptogenesis and SUDEP. We found that early treatment with vigabatrin was not able to prevent epilepsy, but significantly delayed seizure onset.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/physiology , Tuberous Sclerosis/physiopathology , Animals , Anticonvulsants , Brain/metabolism , Diet, Ketogenic , Epilepsy , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Ras Homolog Enriched in Brain Protein/deficiency , Ras Homolog Enriched in Brain Protein/physiology , Sirolimus/pharmacology , Tuberous Sclerosis Complex 1 Protein/deficiency , Tumor Suppressor Proteins , Vigabatrin
8.
J Neurosci ; 39(28): 5424-5439, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31064859

ABSTRACT

Ca2+/calmodulin-dependent protein kinase II (CAMK2) is a key player in synaptic plasticity and memory formation. Mutations in Camk2a or Camk2b cause intellectual disability in humans, and severe plasticity and learning deficits in mice, indicating unique functions for each isoform. However, considering the high homology between CAMK2A and CAMK2B, it is conceivable that for critical functions, one isoform compensates for the absence of the other, and that the full functional spectrum of neuronal CAMK2 remains to be revealed.Here we show that germline as well as adult deletion of both CAMK2 isoforms in male or female mice is lethal. Moreover, Ca2+-dependent activity as well as autonomous activity of CAMK2 is essential for survival. Loss of both CAMK2 isoforms abolished LTP, whereas synaptic transmission remained intact. The double-mutants showed no gross morphological changes of the brain, and in contrast to the long-considered role for CAMK2 in the structural organization of the postsynaptic density (PSD), deletion of both CAMK2 isoforms did not affect the biochemical composition of the PSD. Together, these results reveal an essential role for CAMK2 signaling in early postnatal development as well as the mature brain, and indicate that the full spectrum of CAMK2 requirements cannot be revealed in the single mutants because of partial overlapping functions of CAMK2A and CAMK2B.SIGNIFICANCE STATEMENT CAMK2A and CAMK2B have been studied for over 30 years for their role in neuronal functioning. However, most studies were performed using single knock-out mice. Because the two isoforms show high homology with respect to structure and function, it is likely that some redundancy exists between the two isoforms, meaning that for critical functions CAMK2B compensates for the absence of CAMK2A and vice versa, leaving these functions to uncover. In this study, we generated Camk2a/Camk2b double-mutant mice, and observed that loss of CAMK2, as well as the loss of Ca2+-dependent and Ca2+-independent activity of CAMK2 is lethal. These results indicate that despite 30 years of research the full spectrum of CAMK2 functioning in neurons remains to be unraveled.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Neurons/metabolism , Animals , Brain/growth & development , Brain/metabolism , Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Female , Gene Deletion , Germ-Line Mutation , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Neurogenesis , Neurons/cytology , Neurons/physiology , Post-Synaptic Density/metabolism
9.
Hum Mutat ; 39(12): 2008-2024, 2018 12.
Article in English | MEDLINE | ID: mdl-30184290

ABSTRACT

The abundantly expressed calcium/calmodulin-dependent protein kinase II (CAMK2), alpha (CAMK2A), and beta (CAMK2B) isoforms are essential for learning and memory formation. Recently, a de novo candidate mutation (p.Arg292Pro) in the gamma isoform of CAMK2 (CAMK2G) was identified in a patient with severe intellectual disability (ID), but the mechanism(s) by which this mutation causes ID is unknown. Here, we identified a second, unrelated individual, with a de novo CAMK2G p.Arg292Pro mutation, and used in vivo and in vitro assays to assess the impact of this mutation on CAMK2G and neuronal function. We found that knockdown of CAMK2G results in inappropriate precocious neuronal maturation. We further found that the CAMK2G p.Arg292Pro mutation acts as a highly pathogenic gain-of-function mutation, leading to increased phosphotransferase activity and impaired neuronal maturation as well as impaired targeting of the nuclear CAMK2G isoform. Silencing the catalytic site of the CAMK2G p.Arg292Pro protein reversed the pathogenic effect of the p.Arg292Pro mutation on neuronal maturation, without rescuing its nuclear targeting. Taken together, our results reveal an indispensable function of CAMK2G in neurodevelopment and indicate that the CAMK2G p.Arg292Pro protein acts as a pathogenic gain-of-function mutation, through constitutive activity toward cytosolic targets, rather than impaired targeting to the nucleus.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Gain of Function Mutation , Intellectual Disability/genetics , Amino Acid Substitution , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Catalytic Domain , Cells, Cultured , Disease Models, Animal , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Intellectual Disability/metabolism , Male , Mice
10.
Elife ; 62017 12 15.
Article in English | MEDLINE | ID: mdl-29243588

ABSTRACT

While research on the cerebellar cortex is crystallizing our understanding of its function in learning behavior, many questions surrounding its downstream targets remain. Here, we evaluate the dynamics of cerebellar interpositus nucleus (IpN) neurons over the course of Pavlovian eyeblink conditioning. A diverse range of learning-induced neuronal responses was observed, including increases and decreases in activity during the generation of conditioned blinks. Trial-by-trial correlational analysis and optogenetic manipulation demonstrate that facilitation in the IpN drives the eyelid movements. Adaptive facilitatory responses are often preceded by acquired transient inhibition of IpN activity that, based on latency and effect, appear to be driven by complex spikes in cerebellar cortical Purkinje cells. Likewise, during reflexive blinks to periocular stimulation, IpN cells show excitation-suppression patterns that suggest a contribution of climbing fibers and their collaterals. These findings highlight the integrative properties of subcortical neurons at the cerebellar output stage mediating conditioned behavior.


Subject(s)
Cerebellar Nuclei/physiology , Conditioning, Eyelid , Neurons/physiology , Animals , Mice , Optogenetics
11.
Am J Hum Genet ; 101(5): 768-788, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100089

ABSTRACT

Calcium/calmodulin-dependent protein kinase II (CAMK2) is one of the first proteins shown to be essential for normal learning and synaptic plasticity in mice, but its requirement for human brain development has not yet been established. Through a multi-center collaborative study based on a whole-exome sequencing approach, we identified 19 exceedingly rare de novo CAMK2A or CAMK2B variants in 24 unrelated individuals with intellectual disability. Variants were assessed for their effect on CAMK2 function and on neuronal migration. For both CAMK2A and CAMK2B, we identified mutations that decreased or increased CAMK2 auto-phosphorylation at Thr286/Thr287. We further found that all mutations affecting auto-phosphorylation also affected neuronal migration, highlighting the importance of tightly regulated CAMK2 auto-phosphorylation in neuronal function and neurodevelopment. Our data establish the importance of CAMK2A and CAMK2B and their auto-phosphorylation in human brain function and expand the phenotypic spectrum of the disorders caused by variants in key players of the glutamatergic signaling pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Intellectual Disability/genetics , Mutation/genetics , Animals , Brain/pathology , Cell Line , Exome/genetics , Female , Glutamic Acid/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Phosphorylation/genetics , Signal Transduction/genetics
12.
Neuron ; 89(3): 645-57, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26844836

ABSTRACT

Closed-loop circuitries between cortical and subcortical regions can facilitate precision of output patterns, but the role of such networks in the cerebellum remains to be elucidated. Here, we characterize the role of internal feedback from the cerebellar nuclei to the cerebellar cortex in classical eyeblink conditioning. We find that excitatory output neurons in the interposed nucleus provide efference-copy signals via mossy fibers to the cerebellar cortical zones that belong to the same module, triggering monosynaptic responses in granule and Golgi cells and indirectly inhibiting Purkinje cells. Upon conditioning, the local density of nucleocortical mossy fiber terminals significantly increases. Optogenetic activation and inhibition of nucleocortical fibers in conditioned animals increases and decreases the amplitude of learned eyeblink responses, respectively. Our data show that the excitatory nucleocortical closed-loop circuitry of the cerebellum relays a corollary discharge of premotor signals and suggests an amplifying role of this circuitry in controlling associative motor learning.


Subject(s)
Blinking/physiology , Cerebellar Cortex/physiology , Cerebellar Nuclei/physiology , Conditioning, Classical/physiology , Neural Pathways/physiology , Animals , Association Learning/physiology , Female , Male , Mice , Nerve Fibers/physiology , Optogenetics
13.
Sci Rep ; 5: 13257, 2015 08 25.
Article in English | MEDLINE | ID: mdl-26304458

ABSTRACT

Repeated exposure to Group-A ß-Haemolytic Streptococcus (GAS) may constitute a vulnerability factor in the onset and course of pediatric motor disturbances. GAS infections/colonization can stimulate the production of antibodies, which may cross the blood brain barrier, target selected brain areas (e.g. basal ganglia), and exacerbate motor alterations. Here, we exposed developing SJL male mice to four injections with a GAS homogenate and evaluated the following domains: motor coordination; general locomotion; repetitive behaviors; perseverative responses; and sensorimotor gating (pre-pulse inhibition, PPI). To demonstrate that behavioral changes were associated with immune-mediated brain alterations, we analyzed, in selected brain areas, the presence of infiltrates and microglial activation (immunohistochemistry), monoamines (HPLC), and brain metabolites (in vivo Magnetic Resonance Spectroscopy). GAS-exposed mice showed increased repetitive and perseverative behaviors, impaired PPI, and reduced concentrations of serotonin in prefrontal cortex, a brain area linked to the behavioral domains investigated, wherein they also showed remarkable elevations in lactate. Active inflammatory processes were substantiated by the observation of infiltrates and microglial activation in the white matter of the anterior diencephalon. These data support the hypothesis that repeated GAS exposure may elicit inflammatory responses in brain areas involved in motor control and perseverative behavior, and result in phenotypic abnormalities.


Subject(s)
Diencephalon/immunology , Gait Disorders, Neurologic/microbiology , Lameness, Animal/microbiology , Stereotypic Movement Disorder/microbiology , Streptococcal Infections/immunology , Streptococcus pyogenes , Animals , Behavior, Animal , Diencephalon/microbiology , Gait Disorders, Neurologic/immunology , Lameness, Animal/immunology , Male , Mice , Stereotypic Movement Disorder/immunology
14.
Neurotox Res ; 27(3): 275-83, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25516122

ABSTRACT

More than 10 % of children during school years suffer from a transient tic disorder, and 1 % has a particular type of tic disorder known as Tourette syndrome. At present, there is no available treatment that can improve tics without considerable side effects. Recent evidence indicates that tetrahydrocannabinol (THC), the principal psychoactive component of cannabis, reduced in mice the head twitch responses, a tic pharmacologically induced by the selective serotonin 5-HT2 receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI). THC has some considerable side effects that render its use problematic. In this view, cyclohexyl-carbamic acid 3'-carbamoyl-biphenyl-3-yl ester (URB597), an indirect cannabinoid agonist that enhances endogenous anandamide levels, can constitute a valid alternative to the use of direct CB1 receptor agonists. We investigated whether URB597 may reduce the exhibition of DOI-induced head twitch responses in mice. Moreover, to address whether the effects of URB597 on DOI-induced behavioral response constitute a general phenomenon, we evaluated four (ABH, C57BL/6N, SJL/J, CD-1) mouse strains. These strains have been selected in order to represent an ample spectrum of genetic background and phenotypic variation. Predictably, DOI induced consistent tic-like behaviors in all mice. While URB597 exerted slight sedation in C57BN/6L mice, this cannabinoid agonist remarkably mitigated the exhibition of DOI-induced head twitch in all strains. Present data may disclose novel avenues for the pharmacological treatment of tic disorders.


Subject(s)
Amphetamines/pharmacology , Benzamides/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Carbamates/pharmacology , Head Movements/drug effects , Serotonin Receptor Agonists/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Species Specificity
15.
Behav Brain Res ; 267: 95-105, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24675156

ABSTRACT

The preclinical study of human disorders associated with comorbidities and for which the aetiology is still unclear may substantially benefit from multi-strain studies conducted in mice. The latter can help isolating experimental populations (strains) exhibiting distinct facets in the parameters isomorphic to the symptoms of a given disorder. Through a reverse-translation approach, multi-strain studies can inform both natural predisposing factors and environmental modulators. Thus, mouse strains selected for a particular trait may be leveraged to generate hypothesis-driven studies aimed at clarifying the potential role played by the environment in modulating the exhibition of the symptoms of interest. Tourette's syndrome (TS) constitutes a paradigmatic example whereby: it is characterized by a core symptom (tics) often associated with comorbidities (attention-deficit-hyperactivity and obsessive-compulsive symptoms); it has a clear genetic origin though specific genes are, as yet, unidentified; its course (exacerbations and remissions) is under the influence of environmental factors. Based on these considerations, we tested four mouse strains (ABH, C57, CD1, and SJL) - varying along a plethora of behavioural, neurochemical, and immunological parameters - on a test battery tailored to address the following domains: tics (through the i.p. administration of the selective 5-HT2 receptor agonist DOI, 5mg/kg); locomotion (spontaneous locomotion in the home-cage); perseverative responding in an attentional set shifting task; and behavioural stereotypies in response to a single amphetamine (10mg/kg, i.p.) injection. Present data demonstrate that while ABH and SJL mice respectively exhibit selective increments in amphetamine-induced sniffing behaviour and DOI-induced tic-like behaviours, C57 and CD1 mice show a distinct phenotype, compared to other strains, in several parameters.


Subject(s)
Attention/physiology , Mice, Inbred Strains/physiology , Motor Activity/physiology , Stereotyped Behavior/physiology , Tics/physiopathology , Amphetamine/pharmacology , Amphetamines/adverse effects , Animals , Central Nervous System Stimulants/pharmacology , Circadian Rhythm/physiology , Disease Models, Animal , Dyskinesia, Drug-Induced/diagnosis , Dyskinesia, Drug-Induced/physiopathology , Male , Mice, Inbred C57BL/physiology , Psychological Tests , Serotonin Receptor Agonists/adverse effects , Species Specificity , Stereotyped Behavior/drug effects , Tics/chemically induced , Tics/diagnosis , Tourette Syndrome
16.
Neurosci Biobehav Rev ; 37(6): 1085-100, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23583771

ABSTRACT

In the present manuscript we review a substantial body of literature describing several pre-clinical animal models designed and developed with the purpose of investigating the biological determinants of Tourette syndrome (TS). In order to map the animal models onto the theoretical background upon which they have been devised, we first define phenomenological and etiological aspects of TS and then match this information to the available pre-clinical models. Thus, we first describe the characteristic symptoms exhibited by TS patients and then a series of hypotheses attempting to identify the multifactorial causes of TS. With respect to the former, we detail the phenomenology of abnormal repetitive behaviors (tics and stereotypies), obsessive-compulsive behaviors and aberrant sensory-motor gating. With respect to the latter, we describe both potential candidate vulnerability genes and environmental factors (difficult pregnancies, psychosocial stressors and infections). We then discuss how this evidence has been translated in pre-clinical research with respect to both dependent (symptoms) and independent (etiological factors) variables. Thus, while, on the one hand, we detail the methodologies adopted to measure abnormal repetitive and obsessive-compulsive behaviors, and sensory-motor gating, on the other hand, we describe genetic engineering studies and environmental modulations aimed at reproducing the proposed biological determinants in laboratory rodents. A special emphasis is placed upon "programming" events, occurring during critical stages of early development and exerting organizational delayed consequences. In the final section, we outline a heuristic model with the purpose of integrating clinical and pre-clinical evidence in the study of TS.


Subject(s)
Basal Ganglia/metabolism , Disease Models, Animal , Tourette Syndrome , Animals , Environment , Genetic Linkage , Humans , Neurotransmitter Agents/metabolism , Obsessive-Compulsive Disorder/diagnosis , Obsessive-Compulsive Disorder/etiology , Risk Factors , Tourette Syndrome/etiology , Tourette Syndrome/pathology , Tourette Syndrome/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...