Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Autism ; 12(1): 74, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911565

ABSTRACT

BACKGROUND: The neurocognitive mechanisms underlying autism spectrum disorder (ASD) remain unclear. Progress has been largely hampered by small sample sizes, variable age ranges and resulting inconsistent findings. There is a pressing need for large definitive studies to delineate the nature and extent of key case/control differences to direct research towards fruitful areas for future investigation. Here we focus on perception of biological motion, a promising index of social brain function which may be altered in ASD. In a large sample ranging from childhood to adulthood, we assess whether biological motion preference differs in ASD compared to neurotypical participants (NT), how differences are modulated by age and sex and whether they are associated with dimensional variation in concurrent or later symptomatology. METHODS: Eye-tracking data were collected from 486 6-to-30-year-old autistic (N = 282) and non-autistic control (N = 204) participants whilst they viewed 28 trials pairing biological (BM) and control (non-biological, CTRL) motion. Preference for the biological motion stimulus was calculated as (1) proportion looking time difference (BM-CTRL) and (2) peak look duration difference (BM-CTRL). RESULTS: The ASD group showed a present but weaker preference for biological motion than the NT group. The nature of the control stimulus modulated preference for biological motion in both groups. Biological motion preference did not vary with age, gender, or concurrent or prospective social communicative skill within the ASD group, although a lack of clear preference for either stimulus was associated with higher social-communicative symptoms at baseline. LIMITATIONS: The paired visual preference we used may underestimate preference for a stimulus in younger and lower IQ individuals. Our ASD group had a lower average IQ by approximately seven points. 18% of our sample was not analysed for various technical and behavioural reasons. CONCLUSIONS: Biological motion preference elicits small-to-medium-sized case-control effects, but individual differences do not strongly relate to core social autism associated symptomatology. We interpret this as an autistic difference (as opposed to a deficit) likely manifest in social brain regions. The extent to which this is an innate difference present from birth and central to the autistic phenotype, or the consequence of a life lived with ASD, is unclear.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Adolescent , Biomarkers , Case-Control Studies , Child , Humans , Severity of Illness Index , Young Adult
2.
J Autism Dev Disord ; 48(7): 2490-2505, 2018 07.
Article in English | MEDLINE | ID: mdl-29468576

ABSTRACT

Research on sex-related differences in Autism Spectrum Disorder (ASD) has been impeded by small samples. We pooled 28 datasets from 18 sites across nine European countries to examine sex differences in the ASD phenotype on the ADI-R (376 females, 1763 males) and ADOS (233 females, 1187 males). On the ADI-R, early childhood restricted and repetitive behaviours were lower in females than males, alongside comparable levels of social interaction and communication difficulties in females and males. Current ADI-R and ADOS scores showed no sex differences for ASD severity. There were lower socio-communicative symptoms in older compared to younger individuals. This large European ASD sample adds to the literature on sex and age variations of ASD symptomatology.


Subject(s)
Autism Spectrum Disorder/epidemiology , Adolescent , Adult , Age Factors , Autism Spectrum Disorder/diagnosis , Child , Child, Preschool , Female , Humans , Infant , Interpersonal Relations , Male , Phenotype , Sex Characteristics
3.
Clin Genet ; 94(3-4): 283-295, 2018 10.
Article in English | MEDLINE | ID: mdl-28139835

ABSTRACT

Kabuki syndrome (KS) is a rare genetic syndrome characterized by a typical facial gestalt, variable degrees of intellectual disability, organ malformations, postnatal growth retardation and skeletal abnormalities. So far, KMT2D or KDM6A mutation has been identified as the main cause of KS, accounting for 56%-75% and 3%-8% of cases, respectively. Patients without mutations in 1 of the 2 causative KS genes are often referred to as affected by Kabuki-like syndrome. Overall, they represent approximately 30% of KS cases, pointing toward substantial genetic heterogeneity for this condition. Here, we review all currently available literature describing KS-like phenotypes (or phenocopies) associated with genetic variants located in loci different from KMT2D and KDM6A . We also report on a new KS phenocopy harboring a 5 Mb de novo deletion in chr10p11.22-11.21. An enrichment analysis aimed at identifying functional Gene Ontology classes shared by the 2 known KS causative genes and by new candidate genes currently associated with KS-like phenotypes primarily converges upon abnormal chromatin remodeling and transcriptional dysregulation as pivotal to the pathophysiology of KS phenotypic hallmarks. The identification of mutations in genes belonging to the same functional pathways of KMT2D and KDM6A can help design molecular screenings targeted to KS-like phenotypes.


Subject(s)
Abnormalities, Multiple/genetics , Face/abnormalities , Genetic Predisposition to Disease , Hematologic Diseases/genetics , Vestibular Diseases/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/physiopathology , Chromosomes, Human, Pair 11 , Diagnosis, Differential , Face/physiopathology , Genetic Heterogeneity , Hematologic Diseases/diagnosis , Hematologic Diseases/physiopathology , Humans , Phenotype , Vestibular Diseases/diagnosis , Vestibular Diseases/physiopathology
4.
Neurogenetics ; 16(2): 77-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25501253

ABSTRACT

In the last two decades, migraine research has greatly advanced our current knowledge of the genetic contributions and the pathophysiology of this common and debilitating disorder. Nonetheless, this knowledge still needs to grow further and to translate into more effective treatments. To date, several genes involved in syndromic and monogenic forms of migraine have been identified, allowing the generation of animal models which have significantly contributed to current knowledge of the mechanisms underlying these rare forms of migraine. Common forms of migraine are instead posing a greater challenge, as they may most often stem from complex interactions between multiple common genetic variants, with environmental triggers. This paper reviews our current understanding of migraine genetics, moving from syndromic and monogenic forms to oligogenic/polygenic migraines most recently addressed with some success through genome-wide association studies. Methodological issues in study design and future perspectives opened by biomarker research will also be briefly addressed.


Subject(s)
Migraine Disorders/genetics , Animals , Genetic Predisposition to Disease , Humans
5.
Brain Behav Immun ; 38: 91-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24389156

ABSTRACT

Circulating 45 and 62kDa antibodies targeting the cerebellum were previously associated with Autism Spectrum Disorder (ASD), lower adaptive/cognitive function and aberrant behaviors. Moreover, 37, 39 and 73kDa maternal antibodies (mAb) targeting the fetal brain were previously correlated with broad autism spectrum, irritability, abnormal brain enlargement and impaired expressive language. The present study aims towards clinically characterizing individuals with brain-targeted IgG and/or exposed to maternal antibrain antibodies in a large sample of Italian autistic children (N=355), their unaffected siblings (N=142) and mothers (N=333). The presence of patient- and mother-produced anti-brain antibodies does not confer increased risk of autism within the same sibship. However, the 45 and 62kDa antibodies are correlated with autism severity: the 45kDa Ab is associated with cognitive impairment and lower scores at the Vineland Adaptive Behavior Scales, the 62kDa Ab with motor stereotypies, while both correlate with larger head circumference (all P<0.05). On the other hand, maternal 37, 39 and 73kDa antibrain antibodies, either alone or in combination, are correlated with impaired verbal and non-verbal language development, neurodevelopmental delay and sleep/wake cycle disturbances in their autistic children (P<0.05). Presence of the 62kDa autoAb in the child is significantly associated with presence of the 39 and/or 73kDa antibodies in his/her mother. Our results confirm and extend previous observations in an ethnically distinct sample, providing further evidence of a pathomorphic role for anti-brain antibodies in autism while demonstrating their familial clustering.


Subject(s)
Autoantibodies/blood , Brain/immunology , Child Development Disorders, Pervasive/immunology , Adolescent , Adult , Autoantibodies/immunology , Autoimmunity , Child , Child, Preschool , Female , Humans , Italy , Male , Middle Aged , Young Adult
6.
Mol Psychiatry ; 15(1): 38-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-18607376

ABSTRACT

Autism is a severe developmental disorder, whose pathogenetic underpinnings are still largely unknown. Temporocortical gray matter from six matched patient-control pairs was used to perform post-mortem biochemical and genetic studies of the mitochondrial aspartate/glutamate carrier (AGC), which participates in the aspartate/malate reduced nicotinamide adenine dinucleotide shuttle and is physiologically activated by calcium (Ca(2+)). AGC transport rates were significantly higher in tissue homogenates from all six patients, including those with no history of seizures and with normal electroencephalograms prior to death. This increase was consistently blunted by the Ca(2+) chelator ethylene glycol tetraacetic acid; neocortical Ca(2+) levels were significantly higher in all six patients; no difference in AGC transport rates was found in isolated mitochondria from patients and controls following removal of the Ca(2+)-containing postmitochondrial supernatant. Expression of AGC1, the predominant AGC isoform in brain, and cytochrome c oxidase activity were both increased in autistic patients, indicating an activation of mitochondrial metabolism. Furthermore, oxidized mitochondrial proteins were markedly increased in four of the six patients. Variants of the AGC1-encoding SLC25A12 gene were neither correlated with AGC activation nor associated with autism-spectrum disorders in 309 simplex and 17 multiplex families, whereas some unaffected siblings may carry a protective gene variant. Therefore, excessive Ca(2+) levels are responsible for boosting AGC activity, mitochondrial metabolism and, to a more variable degree, oxidative stress in autistic brains. AGC and altered Ca(2+) homeostasis play a key interactive role in the cascade of signaling events leading to autism: their modulation could provide new preventive and therapeutic strategies.


Subject(s)
Aggrecans/metabolism , Calcium/metabolism , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/metabolism , Homeostasis/physiology , Adolescent , Aggrecans/genetics , Aspartic Acid/metabolism , Brain/drug effects , Brain/metabolism , Case-Control Studies , Chelating Agents/pharmacology , Child , Child Development Disorders, Pervasive/pathology , Child, Preschool , Egtazic Acid/pharmacology , Electron Transport Complex IV/metabolism , Family Health , Female , Gene Expression Regulation/physiology , Genotype , Glutamic Acid/metabolism , Homeostasis/drug effects , Humans , Linkage Disequilibrium , Male , Mitochondria/metabolism , Neocortex/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Serotonin/blood , Young Adult
7.
J Med Genet ; 46(1): 1-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18728070

ABSTRACT

Autism spectrum disorders represent a group of developmental disorders with strong genetic underpinnings. Several cytogenetic abnormalities or de novo mutations able to cause autism have recently been uncovered. In this study, the literature was reviewed to highlight genotype-phenotype correlations between causal gene mutations or cytogenetic abnormalities and behavioural or morphological phenotypes. Based on this information, a set of practical guidelines is proposed to help clinical geneticists pursue targeted genetic testing for patients with autism whose clinical phenotype is suggestive of a specific genetic or genomic aetiology.


Subject(s)
Autistic Disorder/genetics , Genetic Testing , Autistic Disorder/metabolism , Carrier Proteins , Cell Adhesion Molecules, Neuronal , Genetic Counseling , Genetics, Medical , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phenotype , Tensins
8.
Mol Psychiatry ; 14(7): 705-18, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18317465

ABSTRACT

Protein kinase C enzymes play an important role in signal transduction, regulation of gene expression and control of cell division and differentiation. The fsI and betaII isoenzymes result from the alternative splicing of the PKCbeta gene (PRKCB1), previously found to be associated with autism. We performed a family-based association study in 229 simplex and 5 multiplex families, and a postmortem study of PRKCB1 gene expression in temporocortical gray matter (BA41/42) of 11 autistic patients and controls. PRKCB1 gene haplotypes are significantly associated with autism (P<0.05) and have the autistic endophenotype of enhanced oligopeptiduria (P<0.05). Temporocortical PRKCB1 gene expression was reduced on average by 35 and 31% for the PRKCB1-1 and PRKCB1-2 isoforms (P<0.01 and <0.05, respectively) according to qPCR. Protein amounts measured for the PKCbetaII isoform were similarly decreased by 35% (P=0.05). Decreased gene expression characterized patients carrying the 'normal' PRKCB1 alleles, whereas patients homozygous for the autism-associated alleles displayed mRNA levels comparable to those of controls. Whole genome expression analysis unveiled a partial disruption in the coordinated expression of PKCbeta-driven genes, including several cytokines. These results confirm the association between autism and PRKCB1 gene variants, point toward PKCbeta roles in altered epithelial permeability, demonstrate a significant downregulation of brain PRKCB1 gene expression in autism and suggest that it could represent a compensatory adjustment aimed at limiting an ongoing dysreactive immune process. Altogether, these data underscore potential PKCbeta roles in autism pathogenesis and spur interest in the identification and functional characterization of PRKCB1 gene variants conferring autism vulnerability.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/pathology , Gene Expression/genetics , Genetic Predisposition to Disease , Neocortex/metabolism , Protein Kinase C/genetics , Adolescent , Adult , Chi-Square Distribution , Child , Child, Preschool , Family Health , Female , Gene Frequency , Genome-Wide Association Study/methods , Genotype , Humans , Male , Polymorphism, Single Nucleotide/genetics , Protein Kinase C beta , Young Adult
9.
J Neural Transm (Vienna) ; 113(10): 1373-82, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16604303

ABSTRACT

The human gene encoding Reelin (RELN), a pivotal protein in neurodevelopment, includes a polymorphic GGC repeat in its 5' untranslated region (UTR). CHO cells transfected with constructs encompassing the RELN 5'UTR with 4-to-13 GGC repeats upstream of the luciferase reporter gene show declining luciferase activity with increasing GGC repeat number (P < 0.005), as predicted by computer-based simulations. Conversely, RELN 5'UTR sequences boost reporter gene expression above control levels in neuronal SN56 and N2A cell lines, but 12- and 13-repeat alleles still yield 50-60% less luciferase activity compared to the more common 8- and 10-repeat alleles (P < 0.0001). RELN "long" GGC alleles significantly blunt gene expression and may, through this effect, confer vulnerability to human disorders, such as schizophrenia and autism.


Subject(s)
5' Untranslated Regions/genetics , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Polymorphism, Genetic , Serine Endopeptidases/genetics , Trinucleotide Repeats , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Fibroblasts/metabolism , Gene Expression , Genes, Reporter , Humans , Neurons/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reelin Protein , Transfection
10.
Mol Psychiatry ; 10(11): 1006-16, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16027737

ABSTRACT

Organophosphates (OPs) are routinely used as pesticides in agriculture and as insecticides within the household. Our prior work on Reelin and APOE delineated a gene-environment interactive model of autism pathogenesis, whereby genetically vulnerable individuals prenatally exposed to OPs during critical periods in neurodevelopment could undergo altered neuronal migration, resulting in an autistic syndrome. Since household use of OPs is far greater in the USA than in Italy, this model was predicted to hold validity in North America, but not in Europe. Here, we indirectly test this hypothesis by assessing linkage/association between autism and variants of the paraoxonase gene (PON1) encoding paraoxonase, the enzyme responsible for OP detoxification. Three functional single nucleotide polymorphisms, PON1 C-108T, L55M, and Q192R, were assessed in 177 Italian and 107 Caucasian-American complete trios with primary autistic probands. As predicted, Caucasian-American and not Italian families display a significant association between autism and PON1 variants less active in vitro on the OP diazinon (R192), according to case-control contrasts (Q192R: chi2=6.33, 1 df, P<0.025), transmission/disequilibrium tests (Q192R: TDT chi2=5.26, 1 df, P<0.025), family-based association tests (Q192R and L55M: FBAT Z=2.291 and 2.435 respectively, P<0.025), and haplotype-based association tests (L55/R192: HBAT Z=2.430, P<0.025). These results are consistent with our model and provide further support for the hypothesis that concurrent genetic vulnerability and environmental OP exposure may possibly contribute to autism pathogenesis in a sizable subgroup of North American individuals.


Subject(s)
Aryldialkylphosphatase/genetics , Autistic Disorder/enzymology , Autistic Disorder/genetics , Aryldialkylphosphatase/metabolism , Autistic Disorder/etiology , Base Sequence , Case-Control Studies , Child , DNA/genetics , DNA Mutational Analysis , Environment , Female , Genetic Variation , Humans , Insecticides/metabolism , Italy , Linkage Disequilibrium , Male , Models, Biological , Organophosphates/metabolism , Peptides/urine , Polymorphism, Single Nucleotide , Reelin Protein , Serotonin/blood , United States
11.
Psychiatr Genet ; 14(2): 73-82, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15167692

ABSTRACT

We have previously described linkage/association between reelin gene polymorphisms and autistic disorder. APOE also participates in the Reelin signaling pathway, by competitively antagonizing Reelin binding to APOE receptor 2 and to very-low-density lipoprotein receptors. The APOE2 protein variant displays the lowest receptor binding affinity compared with APOE3 and APOE4. In this study, we assess linkage/association between primary autism and APOE alleles in 223 complete trios, from 119 simplex Italian families and 44 simplex and 29 multiplex Caucasian-American families. Statistically significant disequilibrium favors the transmission of epsilon2 alleles to autistic offspring, over epsilon3 and epsilon4 (allele-wise transmission/disequilibrium test [TDT], chi2 = 6.16, 2 degrees of freedom [d.f.], P<0.05; genotype-wise TDT, chi2 = 10.68, 3 d.f., P<0.05). A novel epsilon3r allele was also discovered in an autistic child and his mother. Autistic patients do not differ significantly from unaffected siblings (allele-wise TDT comparing autistic patients versus unaffected sibs, chi2 = 1.83, 2 d.f., P<0.40, not significant). The major limitation of this study consists of our small sample size of trios including one unaffected sibling, currently not possessing the statistical power necessary to conclusively discriminate a specific association of epsilon2 with autism, from a distorted segregation pattern characterized by enhanced epsilon2 transmission rates both to affected and unaffected offspring. Our findings are thus compatible with either (a) pathogenetic contributions by epsilon2 alleles to autism spectrum vulnerability, requiring additional environmental and/or genetic factors to yield an autistic syndrome, and/or (b) a protective effect of epsilon2 alleles against the enhanced risk of miscarriage and infertility previously described among parents of autistic children.


Subject(s)
Apolipoproteins E/genetics , Autistic Disorder/genetics , Apolipoprotein E2 , Apolipoprotein E3 , Apolipoprotein E4 , Base Sequence , DNA Primers , Family , Genotype , Humans , Linkage Disequilibrium , Reelin Protein , White People
12.
Med Hypotheses ; 61(4): 446-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-13679010

ABSTRACT

Tinnitus and temporomandibular joint dysfunction (TMJD) are among the most common complaints encountered by physicians. Though the relationship between tinnitus and TMJD has attracted great interest during the past several years, theories attempting to explain this association are still few and inconsistent. Conceivably, TMJD could irritate auricolo-temporal nerve (ATN), triggering a somatosensory pathway-induced disinhibition of dorsal cochlear nucleus (DCN) activity in the auditory pathway. In genetically-predisposed TMJD patients, signals from cronically stimulated DCNs activating specific cortical neuronal networks, could yield plastic neural changes resulting in tinnitus. Based on current evidence of serotoninergic modulation of neural activity and plasticity in sensory pathways, reduced serotoninergic tone could promote plastic changes underlying tinnitus through diminished filtering of incoming signals. Therefore, the early establishment of specific treatments aimed at improving TMJD and/or boosting serotoninergic activity may be required to prevent the creation of 'tinnitus memory circuits'.


Subject(s)
Temporomandibular Joint Dysfunction Syndrome/diagnosis , Tinnitus/diagnosis , Acoustic Stimulation , Humans , Models, Theoretical , Serotonin/metabolism
13.
Mol Psychiatry ; 7(7): 795-800, 2002.
Article in English | MEDLINE | ID: mdl-12192626

ABSTRACT

Autism is a biologically-heterogeneous disease. Distinct subgroups of autistic patients may be marked by intermediate phenotypes, such as elevated serotonin (5-HT) blood levels, potentially associated with different underlying disease mechanisms. This could lead to inconsistent genetic association results, such as those of prior studies on serotonin transporter (5-HTT) gene promoter variants and autistic disorder. Contributions of 5-HTT gene promoter alleles to 5-HT blood levels were thus investigated in 134 autistic patients and 291 first-degree relatives. Mean 5-HT blood levels are 11% higher in autistic patients carrying the L/L genotype, compared to patients with the S/S or S/L genotype; this trend is not observed in first-degree relatives. The probability of inheriting L or S alleles is significantly enhanced in patients with 5-HT blood levels above or below the mean, respectively (P < 0.05), but quantitative TDT analyses yield a non-significant trend (P = 0.10), as this polymorphism explains only 2.5% of the variance in 5-HT blood levels of autistic patients. In conclusion, 5-HTT gene promoter variants seemingly exert a small effect on 5-HT blood levels in autistic children, which largely does not account for hyperserotoninemia. Nonetheless, the inconsistent outcome of prior association studies could partly stem from a selection bias of hyper- or hypo-serotoninemic probands.


Subject(s)
Autistic Disorder/blood , Autistic Disorder/genetics , Carrier Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Transport Proteins , Nerve Tissue Proteins , Serotonin/blood , Child , Family Health , Female , Genetic Variation , Haplotypes , Humans , Linkage Disequilibrium , Male , Promoter Regions, Genetic/genetics , Serotonin Plasma Membrane Transport Proteins
14.
J Neural Transm (Vienna) ; 109(5-6): 557-65, 2002 May.
Article in English | MEDLINE | ID: mdl-12111449

ABSTRACT

The serotonin transporter (5HTT) is the molecule responsible for the high-affinity reuptake of 5HT from the synaptic cleft. Mice lacking the 5HTT exhibit highly elevated extracellular concentrations of 5HT. We assessed whether the glutathione detoxification system is altered in 5HTT-deficient mice. While levels of reduced and oxidized glutathione were unchanged, glutathione metabolising enzymes showed a differential pattern of modulation. Glutathione peroxidase was reduced in frontal cortex, brainstem, and cerebellum of 5HTT-deficient mice, though not to a statistically significant extent, while a putative isoform of the detoxifying enzyme glutathione-S-transferase pi was decreased in a number of brain regions, especially in brainstem. At the level of the DNA, we found an increase of oxidative DNA adducts in the hippocampus of 5HTT-deficient mice. Given the importance of the hippocampus in learning and memory, this may be the most important neurochemical consequence of the absence of the 5HTT.


Subject(s)
Carrier Proteins/physiology , DNA/metabolism , Hippocampus/metabolism , Membrane Glycoproteins/physiology , Membrane Transport Proteins , Nerve Tissue Proteins , Animals , Brain/enzymology , Carrier Proteins/genetics , DNA Adducts/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Glutathione Peroxidase/metabolism , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout/genetics , Oxidation-Reduction , Oxidative Stress/physiology , RNA, Messenger/metabolism , Serotonin Plasma Membrane Transport Proteins , Tissue Distribution
15.
Psychiatr Genet ; 11(2): 99-103, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11525425

ABSTRACT

Plasmin, a serine protease, is involved in many physiologically relevant processes, including haemostasis, cellular recruitment during immune response, tumour growth, and also neuronal migration and synaptic remodelling. Both tissue-type and urokinase-type plasminogen activators can be efficiently inhibited by plasminogen activator inhibitor-1 (PAI-1), a protease inhibitor of the serpin family. The human PAI-1 gene is located on chromosome 7q, within or close to a region that has been linked to autism in several linkage studies. Autism seems to be characterized by altered neuronal cytoarchitecture, synaptogenesis and possibly also cellular immune responses. We began addressing the potential involvement of the PAI-1 gene in autistic disorder with this linkage/association study, assessing transmission patterns of the 4G/5G polymorphism in the PAI-1 gene promoter that was previously shown to significantly affect PAI-1 plasma levels. No linkage/association was found in 167 trios with autistic probands, recruited in Italy and in the USA. We thus found no evidence that this polymorphism, or putative functionally relevant gene variants in linkage disequilibrium with it, confer vulnerability to autistic disorder.


Subject(s)
Autistic Disorder/genetics , Chromosomes, Human, Pair 7/genetics , Plasminogen Activator Inhibitor 1/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Adolescent , Adult , Autistic Disorder/epidemiology , Case-Control Studies , Child , Child, Preschool , Chromosome Mapping , Female , Genetic Predisposition to Disease , Genotype , Humans , Italy/epidemiology , Italy/ethnology , Male , Netherlands/ethnology , Plasminogen Activator Inhibitor 1/blood , Polymerase Chain Reaction , United States/epidemiology
16.
J Neurosci ; 21(17): 6862-73, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11517274

ABSTRACT

Thalamocortical neurons innervating the barrel cortex in neonatal rodents transiently store serotonin (5-HT) in synaptic vesicles by expressing the plasma membrane serotonin transporter (5-HTT) and the vesicular monoamine transporter (VMAT2). 5-HTT knock-out (ko) mice reveal a nearly complete absence of 5-HT in the cerebral cortex by immunohistochemistry, and of barrels, both at P7 and adulthood. Quantitative electron microscopy reveals that 5-HTT ko affects neither the density of synapses nor the length of synaptic contacts in layer IV. VMAT2 ko mice, completely lacking activity-dependent vesicular release of monoamines including 5-HT, also show a complete lack of 5-HT in the cortex but display largely normal barrel fields, despite sometimes markedly reduced postnatal growth. Transient 5-HTT expression is thus required for barrel pattern formation, whereas activity-dependent vesicular 5-HT release is not.


Subject(s)
Biogenic Monoamines/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Neurons, Afferent/metabolism , Neuropeptides , Organic Anion Transporters , Somatosensory Cortex/metabolism , Synaptic Vesicles/metabolism , Thalamus/metabolism , Aging/metabolism , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Extracellular Space/metabolism , Fenclonine/pharmacology , GABA Plasma Membrane Transport Proteins , Immunohistochemistry , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons, Afferent/ultrastructure , Serotonin/analysis , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects , Synapses/metabolism , Synapses/ultrastructure , Thalamus/cytology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins , Vibrissae/innervation , Vibrissae/physiology
17.
Mol Psychiatry ; 6(2): 150-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11317216

ABSTRACT

Autistic disorder (MIM 209850) is currently viewed as a neurodevelopmental disease. Reelin plays a pivotal role in the development of laminar structures including the cerebral cortex, hippocampus, cerebellum and of several brainstem nuclei. Neuroanatomical evidence is consistent with Reelin involvement in autistic disorder. In this study, we describe several polymorphisms identified using RNA-SSCP and DNA sequencing. Association and linkage were assessed comparing 95 Italian patients to 186 ethnically-matched controls, and using the transmission/disequilibrium test and haplotype-based haplotype relative risk in 172 complete trios from 165 families collected in Italy and in the USA. Both case-control and family-based analyses yield a significant association between autistic disorder and a polymorphic GGC repeat located immediately 5' of the reelin gene (RELN) ATG initiator codon, as well as with specific haplotypes formed by this polymorphism with two single-base substitutions located in a splice junction in exon 6 and within exon 50. Triplet repeats located in 5' untranslated regions (5'UTRs) are indicative of strong transcriptional regulation. Our findings suggest that longer triplet repeats in the 5'UTR of the RELN gene confer vulnerability to autistic disorder.


Subject(s)
Autistic Disorder/epidemiology , Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Adult , Aged , Aged, 80 and over , Alleles , Brain Chemistry/genetics , Case-Control Studies , Exons , Family Health , Female , Genetic Markers , Genetic Predisposition to Disease , Haplotypes , Humans , Linkage Disequilibrium , Male , Middle Aged , Nerve Tissue Proteins , Point Mutation , Polymorphism, Single Nucleotide , RNA Splice Sites/genetics , Reelin Protein , Risk Factors , Serine Endopeptidases , Serotonin/physiology , Skull/anatomy & histology , Trinucleotide Repeats
18.
Eur Neuropsychopharmacol ; 10(6): 489-93, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115739

ABSTRACT

The serotonin (5HT) transporter (5HTT) removes 5HT from the synaptic cleft and is thus critical to the control of serotonergic neurotransmission. Mice with a targeted inactivation of the 5HTT represent a novel and unique tool to study serotonergic system functioning. Because the release of 5HT is regulated by adenosine, we investigated 5HTT-deficient mice for possible adaptive changes of adenosine A(1) and A(2A) receptors. A(1) and A(2A) receptors were studied by means of quantitative autoradiography using the radioligands [3H]8-cyclopentyl-1,3-dipropylxanthine and [3H]CGS 21680, respectively. A comparison of 5HTT knockout versus control mice revealed upregulation of A(1) receptors in the dorsal raphe nucleus (DRN, +21%), but not in any of the serotonergic projection areas, and downregulation of A(2A) receptors in basal ganglia. The adaptive changes of A(1) and A(2A) receptors in 5HTT-deficient mice are likely to represent a compensatory neuroprotective effect mediated by the adenosinergic modulatory system. For comparison, these receptors were also studied in monoamine oxidase A (MAOA) knockout mice and in 5HTT/MAOA double knockout mice. 5HTT/MAOA double knockout mice showed adaptive changes of adenosine A(1) and A(2A) receptors similar to 5HTT knockout mice, while investigation of MAOA-deficient mice revealed an upregulation of A(2A) receptors, which may relate to a role of both MAOA and adenosine A(2A) receptors in anxiety.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Monoamine Oxidase/metabolism , Nerve Tissue Proteins , Receptors, Purinergic P1/metabolism , Animals , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoamine Oxidase/deficiency , Receptor, Adenosine A2A , Serotonin Plasma Membrane Transport Proteins
19.
Am J Med Genet ; 96(6): 784-90, 2000 Dec 04.
Article in English | MEDLINE | ID: mdl-11121182

ABSTRACT

Adenosine deaminase (ADA) plays a relevant role in purine metabolism, immune responses, and peptidase activity, which may be altered in some autistic patients. Codominant ADA1 and ADA2 alleles code for ADA1 and ADA2 allozymes, the most frequent protein isoforms in the general population. Individuals carrying one copy of the ADA2 allele display 15 to 20% lower catalytic activity compared to ADA1 homozygotes. Recent preliminary data suggest that ADA2 alleles may be more frequent among autistic patients than healthy controls. The present study was undertaken to replicate these findings in a new case-control study, to test for linkage/association using a family-based design, and to characterize ADA2-carrying patients by serotonin blood levels, peptiduria, and head circumference. ADA2 alleles were significantly more frequent in 91 Caucasian autistic patients of Italian descent than in 152 unaffected controls (17.6% vs. 7.9%, P = 0.018), as well as among their fathers. Family-based tests involving these 91 singleton families, as well as 44 additional Caucasian-American trios, did not support significant linkage/association. However, the observed preferential maternal transmission of ADA2 alleles, if replicated, may point toward linkage disequilibrium between the ADA2 polymorphism and an imprinted gene variant located in its vicinity. Racial and ethnic differences in ADA allelic distributions, together with the low frequency of the ADA2 allele, may pose methodological problems to future linkage/association studies. Direct assessments of ADA catalytic activity in autistic individuals and unaffected siblings carrying ADA1/ADA1 vs ADA1/ADA2 genotypes may provide stronger evidence of ADA2 contributions to autistic disorder. Am. J. Med. Genet. (Neuropsychiatr. Genet.) 96:784-790, 2000.


Subject(s)
Adenosine Deaminase/genetics , Alleles , Autistic Disorder/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Autistic Disorder/blood , Autistic Disorder/urine , Case-Control Studies , Cephalometry , Child , Child, Preschool , DNA/genetics , Family Health , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Peptides/urine , Serotonin/blood
20.
Eur J Neurosci ; 12(7): 2281-90, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10947807

ABSTRACT

This study shows that unilateral transection of the infraorbital nerve (ION) in newborn (P0) rats induces apoptosis in the contralateral ventrobasal thalamic (VB) complex, as evidenced by terminal transferase-mediated deoxyuridine triphosphate-biotin nick end labelling (TUNEL) and electron miscroscopy. Double-labelling experiments using retrograde transport of labelled microspheres injected into the barrel cortex, followed by TUNEL staining, show that TUNEL-positive cells are thalamocortical neurons. The number of TUNEL-positive cells had begun to increase by 24 h postlesion, increased further 48 h after nerve section, and decreased to control levels after 120 h. Lesion-induced apoptosis in the VB complex is less pronounced if ION section is performed at P4, and disappears if the lesion is performed at P7. This time course closely matches the critical period of lesion-induced plasticity in the barrel cortex. Nerve growth factor (NGF) or brain-derived neurotrophic factor (BDNF), applied on the ION stump alone or in combination, are able to partially rescue thalamic neurons from apoptosis. Total cell counts in the VB complex of P7 animals that underwent ION section at P0 confirm the rescuing effect of BDNF and NGF. Blockade of axonal transport in the ION mimics the effect of ION section. These data suggest that survival-promoting signals from the periphery, maybe neurotrophins, are required for the survival of higher-order neurons in the somatosensory system during the period of fine-tuning of neuronal connections. We also propose that anterograde transneuronal degeneration in the neonatal rat trigeminal system may represent a new animal model for studying the pathways of programmed cell death in vivo.


Subject(s)
Apoptosis/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Critical Period, Psychological , Nerve Growth Factor/pharmacology , Neurons/cytology , Somatosensory Cortex/cytology , Thalamic Nuclei/cytology , Animals , Animals, Newborn , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Axonal Transport/drug effects , Axotomy , Cell Count , Cell Nucleus/ultrastructure , Chromatin/ultrastructure , In Situ Nick-End Labeling , Maxillary Nerve/cytology , Maxillary Nerve/physiology , Microscopy, Electron , Neural Pathways , Rats , Rats, Inbred Lew , Vinblastine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...