Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.204
Filter
1.
J Clin Invest ; 134(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352378

ABSTRACT

Social deficits represent a core symptom domain of autism spectrum disorder (ASD), which is often comorbid with sleep disturbances. In this issue of the JCI, Sun et al. explored a medial septum (MS) circuit linking these behaviors in a neuroligin 3 conditional knockout model of autism. They identified GABAergic neuron hyperactivity following neuroligin 3 deletion in the MS. This hyperactivity resulted in the inhibition of the downstream preoptic area (POA) and hippocampal CA2 region, resulting in sleep loss and social memory deficits, respectively. Inactivating the hyperactive MS GABA neurons or activating the POA or CA2 rescued the behavioral deficits. Together, these findings deepen our understanding of neural circuits underlying social and sleep deficits in ASD.


Subject(s)
Cell Adhesion Molecules, Neuronal , Disease Models, Animal , GABAergic Neurons , Membrane Proteins , Nerve Tissue Proteins , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/deficiency , Mice, Knockout , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Septal Nuclei/metabolism , Septal Nuclei/physiopathology , Septal Nuclei/pathology , Social Behavior
2.
Nat Commun ; 15(1): 7909, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256359

ABSTRACT

Members of the leucine rich repeat (LRR) and PDZ domain (LAP) protein family are essential for animal development and histogenesis. Densin-180, encoded by LRRC7, is the only LAP protein selectively expressed in neurons. Densin-180 is a postsynaptic scaffold at glutamatergic synapses, linking cytoskeletal elements with signalling proteins such as the α-subunit of Ca2+/calmodulin-dependent protein kinase II. We have previously observed an association between high impact variants in LRRC7 and Intellectual Disability; also three individual cases with variants in LRRC7 had been described. We identify here 33 individuals (one of them previously described) with a dominant neurodevelopmental disorder due to heterozygous missense or loss-of-function variants in LRRC7. The clinical spectrum involves intellectual disability, autism, ADHD, aggression and, in several cases, hyperphagia-associated obesity. A PDZ domain variant interferes with synaptic targeting of Densin-180 in primary cultured neurons. Using in vitro systems (two hybrid, BioID, coimmunoprecipitation of tagged proteins from 293T cells) we identified new candidate interaction partners for the LRR domain, including protein phosphatase 1 (PP1), and observed that variants in the LRR reduced binding to these proteins. We conclude that LRRC7 encodes a major determinant of intellectual development and behaviour.


Subject(s)
Aggression , Autistic Disorder , Intellectual Disability , Adolescent , Adult , Animals , Child , Child, Preschool , Female , Humans , Male , Young Adult , Autistic Disorder/genetics , Autistic Disorder/metabolism , HEK293 Cells , Intellectual Disability/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , PDZ Domains/genetics , Synapses/metabolism
3.
Biomolecules ; 14(9)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39334949

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with a highly variable expression of phenotypes (restricted interest or activity and repetitive behavior in communication and social interactions), genes (mutation), markers (alteration of transcription) and pathways. Loss of function of the CC2D1A gene appears to primarily affect the brain, leading to a range of behavioral problems in humans. In our study published in 2020, we found that the expressions of miR-19a-3p, miR-361-5p, miR-150-5p, miR-3613-3p, miR-126-3p and miR-499a-5p were downregulated in the serum samples of autistic patients, their families and mouse models (Cc2d1a +/- and valproic acid treated males). Here, acquired non-Mendelian hereditary character in a genetically defined mouse model of autism (Cc2d1a +/-) correlates with the transcriptional alteration of five miRNAs. We seek to test the hypothesis that miRNA levels vary by changes in RNA/DNA structure during development, thereby creating transcription alteration and cell memory. Behavioral tests were conducted on the offspring of Cc2d1a (+/-) mutant and control mice, such as novel object, social interaction, marble burying and tail suspension behavior. Two RNA fractions were isolated from mouse hippocampal tissues and sperm cells via standard TRIzol extraction: free RNA and the fraction of RNA bound to DNA in the form of a DNA/RNA hybrid (R-loop). The expression levels of miR-19a-3p, miR-361-5p, miR-150-5p, miR-126-3p and miR-499a-5p were investigated by quantitative real-time RT-PCR. We report differences in the distribution of five miRNAs in the hippocampus between male and female mice, particularly in colonies of Cc2d1a (+/-) mice. Furthermore, the number of miRNAs engaged in the DNA/RNA hybrid fraction is generally higher in the mutant pedigree than in the control group. On the other hand, in sperm, both fractions are at lower levels than in controls. R-loops contribute to the physiology and pathology of organisms including human disease. Here, we report a variation in five miRNA levels between gender and tissue. Our results suggest that the transcription levels of these five miRNAs are directly regulated by their RNA.


Subject(s)
Heterozygote , MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Male , Female , Disease Models, Animal , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Humans , DNA/genetics , Behavior, Animal
4.
Cell Rep ; 43(9): 114733, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39269903

ABSTRACT

Mutations in SYNGAP1 are a common genetic cause of intellectual disability (ID) and a risk factor for autism. SYNGAP1 encodes a synaptic GTPase-activating protein (GAP) that has both signaling and scaffolding roles. Most pathogenic variants of SYNGAP1 are predicted to result in haploinsufficiency. However, some affected individuals carry missense mutations in its calcium/lipid binding (C2) and GAP domains, suggesting that many clinical features result from loss of functions carried out by these domains. To test this hypothesis, we targeted the exons encoding the C2 and GAP domains of SYNGAP. Rats heterozygous for this deletion exhibit reduced exploration and fear extinction, altered social investigation, and spontaneous seizures-key phenotypes shared with Syngap heterozygous null rats. Together, these findings indicate that the reduction of SYNGAP C2/GAP domain function is a main feature of SYNGAP haploinsufficiency. This rat model provides an important system for the study of ID, autism, and epilepsy.


Subject(s)
ras GTPase-Activating Proteins , Animals , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Rats , Protein Domains , Haploinsufficiency , Male , Intellectual Disability/genetics , Intellectual Disability/metabolism , Humans , Seizures/metabolism , Seizures/genetics , Heterozygote , Fear/physiology , Autistic Disorder/genetics , Autistic Disorder/metabolism , Disease Models, Animal
5.
PLoS One ; 19(9): e0308224, 2024.
Article in English | MEDLINE | ID: mdl-39226293

ABSTRACT

Currently, there are no reliable biomarkers for autism diagnosis. The heterogeneity of autism and several co-occurring conditions are key challenges to establishing these. Here, we used untargeted mass spectrometry-based urine metabolomics to investigate metabolic differences for autism diagnosis and autistic traits in a well-characterized twin cohort (N = 105). We identified 208 metabolites in the urine samples of the twins. No clear, significant metabolic drivers for autism diagnosis were detected when controlling for other neurodevelopmental conditions. However, we identified nominally significant changes for several metabolites. For instance, phenylpyruvate (p = 0.019) and taurine (p = 0.032) were elevated in the autism group, while carnitine (p = 0.047) was reduced. We furthermore accounted for the shared factors, such as genetics within the twin pairs, and report additional metabolite differences. Based on the nominally significant metabolites for autism diagnosis, the arginine and proline metabolism pathway (p = 0.024) was enriched. We also investigated the association between quantitative autistic traits, as measured by the Social Responsiveness Scale 2nd Edition, and metabolite differences, identifying a greater number of nominally significant metabolites and pathways. A significant positive association between indole-3-acetate and autistic traits was observed within the twin pairs (adjusted p = 0.031). The utility of urine biomarkers in autism, therefore, remains unclear, with mixed findings from different study populations.


Subject(s)
Autistic Disorder , Biomarkers , Metabolomics , Humans , Autistic Disorder/urine , Autistic Disorder/metabolism , Autistic Disorder/genetics , Male , Female , Metabolomics/methods , Child , Biomarkers/urine , Adolescent , Metabolome , Adult , Twins, Monozygotic , Child, Preschool
6.
Biochemistry (Mosc) ; 89(8): 1509-1518, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39245458

ABSTRACT

A large body of evidence implies the involvement of brain-derived neurotrophic factor (BDNF) in the pathogenesis of autism spectrum disorders (ASDs). A deficiency of BDNF in the hippocampus and frontal cortex of BTBR mice (a model of autism) has been noted in a number of studies. Earlier, we showed that induction of BDNF overexpression in the hippocampus of BTBR mice reduced anxiety and severity of stereotyped behavior, but did not affect social interest. Here, we induced BDNF overexpression in the frontal cortex neurons of BTBR mice using an adeno-associated viral vector, which resulted in a significant increase in the social interest in the three-chamber social test. At the same time, the stereotypy, exploratory behavior, anxiety-like behavior, and novel object recognition were not affected. Therefore, we have shown for the first time that the presence of BDNF in the frontal cortex is critical for the expression of social interest in BTBR mice, since compensation for its deficiency in this structure eliminated the autism-like deficiencies in the social behavior characteristic for these animals.


Subject(s)
Autistic Disorder , Brain-Derived Neurotrophic Factor , Disease Models, Animal , Frontal Lobe , Social Behavior , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Frontal Lobe/metabolism , Mice , Autistic Disorder/metabolism , Autistic Disorder/genetics , Male , Behavior, Animal , Mice, Inbred C57BL
7.
CNS Neurosci Ther ; 30(9): e70015, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218796

ABSTRACT

INTRODUCTION: Neuroinflammation and microglial activation-related dendritic injury contribute to the pathogenesis of Autism Spectrum Disorder (ASD). Previous studies show that Progranulin (PGRN) is a growth factor associated with inflammation and synaptic development, but the role of PGRN in autism and the mechanisms underlying changes in PGRN expression remain unclear. AIMS: To investigate the impact of PGRN in autism, we stereotactically injected recombinant PGRN into the hippocampus of ASD model rats. Additionally, we explored the possibility that sortilin may be the factor behind the alterations in PGRN by utilizing SORT1 knockdown. Ultimately, we aimed to identify potential targets for the treatment of autism. RESULTS: PGRN could alleviate inflammatory responses, protect neuronal dendritic spines, and ameliorate autism-like behaviors. Meanwhile, elevated expression of sortilin and decreased levels of PGRN were observed in both ASD patients and rats. Enhanced sortilin levels facilitated PGRN internalization into lysosomes. Notably, suppressing SORT1 expression amplified PGRN levels, lessened microglial activation, and mitigated inflammation, thereby alleviating autism-like behaviors. CONCLUSION: Collectively, our findings highlight elevated sortilin levels in ASD rat brains, exacerbating dendrite impairment by affecting PGRN expression. PGRN supplementation and SORT1 knockdown hold potential as therapeutic strategies for ASD.


Subject(s)
Adaptor Proteins, Vesicular Transport , Autistic Disorder , Progranulins , Valproic Acid , Animals , Female , Humans , Male , Rats , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Autistic Disorder/metabolism , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Dendritic Spines/drug effects , Dendritic Spines/pathology , Dendritic Spines/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Microglia/metabolism , Microglia/drug effects , Progranulins/genetics , Rats, Sprague-Dawley , Valproic Acid/pharmacology
8.
Neuroreport ; 35(15): 980-986, 2024 Oct 16.
Article in English | MEDLINE | ID: mdl-39166394

ABSTRACT

Dysregulated appetite is common in autism spectrum disorder (ASD) and it includes excessive interest in tasty foods. Overconsumption of palatable fluids has been found in the valproic acid-induced ASD rat. Though ASD has a strong genetic component, the link between ASD-related genes and appetite for palatable foods remains elusive. We focused on the CNTNAP2 gene whose deletion in mice recapitulates human ASD symptoms. We investigated whether Cntnap2-/- male mice consume greater amounts of palatable 10% sucrose, 0.1% saccharin, and 4.1% intralipid solutions offered in episodic meals either in a no-choice paradigm or a two-bottle choice test. We examined how sucrose intake affects c-Fos immunoreactivity in feeding-related brain areas. Finally, we determined doses at which intraperitoneal oxytocin decreases sucrose intake in mutants. In the single-bottle tests, Cntnap2-/- mice drank more sucrose, saccharin, and intralipid compared to WTs. Given a choice between two tastants, Cntnap2-/- mice had a higher preference for sucrose than intralipid. While the standard 1 mg/kg oxytocin dose reduced sucrose intake in WTs, a low oxytocin dose (0.1 mg/kg) decreased sucrose intake in Cntnap2-/- mice. Sucrose intake induced a more robust c-Fos response in wild-type (WT) than Cntnap2-/- mice in the reward and hypothalamic sites and it increased the percentage of Fos-immunoreactivity oxytocin neurons in WTs, but not in mutants. We conclude that Cntnap2-/- mice overconsume palatable solutions, especially sucrose, beyond levels seen in WTs. This excessive consumption is associated with blunted c-Fos immunoreactivity in feeding-related brain sites, and it can be reversed by low-dose oxytocin.


Subject(s)
Disease Models, Animal , Membrane Proteins , Mice, Knockout , Nerve Tissue Proteins , Oxytocin , Saccharin , Animals , Oxytocin/metabolism , Male , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Saccharin/administration & dosage , Mice , Sucrose/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Phospholipids/metabolism , Mice, Inbred C57BL , Soybean Oil/pharmacology , Soybean Oil/administration & dosage , Autistic Disorder/metabolism , Autistic Disorder/genetics , Food Preferences/drug effects , Food Preferences/physiology , Feeding Behavior/physiology , Feeding Behavior/drug effects , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Emulsions
9.
Brain Behav Immun ; 122: 527-546, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39182588

ABSTRACT

Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder caused by the interaction of multiple pathogenic factors. Epidemiological studies and animal experiments indicate that maternal immune activation (MIA) is closely related to the development of ASD in offspring. A large number of pro-inflammatory cytokines are transferred from the placenta to the fetal brain during MIA, which impedes fetal neurodevelopment and is accompanied by activation of immune cells and microglia. Programmed cell death protein 1 (PD-1) can be highly expressed on the surface of various activated immune cells, when combined with programmed cell death-ligand 1 (PD-L1), it can activate the PD-1/PD-L1 pathway and exert powerful immunosuppressive effects, suggesting that this immune checkpoint may have the potential to treat MIA-induced ASD. This study combined bioinformatics analysis and experimental validation to explore the efficacy of Fc-fused PD-L1 (PD-L1-Fc) in treating MIA-induced ASD. Bioinformatics analysis results showed that in human placental inflammation, IL-6 was upregulated, T cells proliferated significantly, and the PD-1/PD-L1 pathway was significantly enriched. The experimental results showed that intraperitoneal injection of poly(I:C) induced MIA in pregnant mice resulted in significant expression of IL-6 in their serum, placenta, and fetal brain. At the same time, the expression of PD-1 and PD-L1 in the placenta and fetal brain increased, CD4+ T cells in the spleen were significantly activated, and PD-1 expression increased. Their offspring mice exhibited typical ASD-like behaviors. In vitro experiments on primary microglia of offspring mice have confirmed that the expression of IL-6, PD-1, and PD-L1 is significantly increased, and PD-L1-Fc effectively reduced their expression levels. In the prefrontal cortex of MIA offspring mice, there was an increase in the expression of IL-6, PD-1, and PD-L1; activation of microglial cells, and colocalization with PD-1. Then we administered brain stereotaxic injections of PD-L1-Fc to MIA offspring mice and intraperitoneal injections to MIA pregnant mice. The results indicated that PD-L1-Fc effectively suppressed neuroinflammation in the frontal cortex of offspring mice and partially ameliorated ASD-like behaviors; MIA in pregnant mice was significantly alleviated, and the offspring mice they produced did not exhibit neuroinflammation or ASD-like behaviors. In summary, we have demonstrated the therapeutic ability of PD-L1-Fc for MIA-induced ASD, aiming to provide new strategies and insights for the treatment of ASD.


Subject(s)
Autism Spectrum Disorder , B7-H1 Antigen , Placenta , Programmed Cell Death 1 Receptor , Animals , Female , B7-H1 Antigen/metabolism , Pregnancy , Programmed Cell Death 1 Receptor/metabolism , Mice , Male , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/prevention & control , Humans , Placenta/metabolism , Disease Models, Animal , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Behavior, Animal , Mice, Inbred C57BL , Autistic Disorder/metabolism , Autistic Disorder/immunology , Inflammation/metabolism , Interleukin-6/metabolism , Brain/metabolism , Brain/drug effects
10.
Article in English | MEDLINE | ID: mdl-39209101

ABSTRACT

Chromogranin A (CgA), a âˆ¼ 49 kDa acidic secretory protein, is ubiquitously distributed in endocrine and neuroendocrine cells and neurons. As a propeptide, CgA is proteolytically cleaved to generate several peptides of biological importance, including pancreastatin (PST: hCgA250-301), Vasostatin 1 (VS1: hCgA1-76), and catestatin (CST: CgA 352-372). VS1 represents the most conserved fragment of CgA. A 20 amino acid domain within VS1 (CgA 47-66) exhibits potent antimicrobial and anti-inflammatory activities. Autism is known to be associated with inflammation. Therefore, we seek to test the hypothesis that VS1 modulates autism behaviors by reducing inflammation in the hippocampus. Treatment of C57BL/6 (B6) and BTBR (a mouse model of idiopathic autism) mice with VS1 revealed the following: BTBR mice showed a significant decrease in chamber time in the presence of a stranger or a novel object. Treatment with VS1 significantly increased chamber time in both cases, underscoring a crucial role for VS1 in improving behavioral deficits in BTBR mice. In contrast to chamber time, sniffing time in BTBR mice in the presence of a stranger was less compared to B6 control mice. VS1 did not improve this latter parameter. Surprisingly, sniffing time in BTBR mice in the presence of a novel object was comparable with B6 mice. Proinflammatory cytokines such as IL-6 and IL-1b, as well as other inflammatory markers, were elevated in BTBR mice, which were dramatically reduced after supplementation with VS1. Interestingly, even Beclin-1/p62, pAKT/AKT, and p-p70-S6K/p70-S6K ratios were notably reduced by VS1. We conclude that VS1 plays a crucial role in restoring autistic spectrum disorders (ASD) plausibly by attenuating neuroinflammation.


Subject(s)
Autistic Disorder , Chromogranin A , Disease Models, Animal , Hippocampus , Neuroinflammatory Diseases , Peptide Fragments , Animals , Male , Mice , Autistic Disorder/drug therapy , Autistic Disorder/metabolism , Chromogranin A/pharmacology , Chromogranin A/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Mice, Inbred C57BL , Neuroinflammatory Diseases/drug therapy , Peptide Fragments/pharmacology
11.
Nat Commun ; 15(1): 6801, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122707

ABSTRACT

One of the main drivers of autism spectrum disorder is risk alleles within hundreds of genes, which may interact within shared but unknown protein complexes. Here we develop a scalable genome-editing-mediated approach to target 14 high-confidence autism risk genes within the mouse brain for proximity-based endogenous proteomics, achieving the identification of high-specificity spatial proteomes. The resulting native proximity proteomes are enriched for human genes dysregulated in the brain of autistic individuals, and reveal proximity interactions between proteins from high-confidence risk genes with those of lower-confidence that may provide new avenues to prioritize genetic risk. Importantly, the datasets are enriched for shared cellular functions and genetic interactions that may underlie the condition. We test this notion by spatial proteomics and CRISPR-based regulation of expression in two autism models, demonstrating functional interactions that modulate mechanisms of their dysregulation. Together, these results reveal native proteome networks in vivo relevant to autism, providing new inroads for understanding and manipulating the cellular drivers underpinning its etiology.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Brain , Disease Models, Animal , Proteome , Proteomics , Animals , Proteome/metabolism , Mice , Humans , Brain/metabolism , Proteomics/methods , Autistic Disorder/genetics , Autistic Disorder/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Phenotype , Gene Editing , Male , Genetic Predisposition to Disease , Mice, Inbred C57BL , Female , CRISPR-Cas Systems
12.
Sci Rep ; 14(1): 19227, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164365

ABSTRACT

Maternal malnutrition has been associated with neurodevelopmental deficits and long-term implications on the offspring's health and behavior. Here, we investigated the effects of maternal low-protein diet (LPD) or obesity-inducing maternal high-fat diet (HFD) on dyadic social interactions, group organization and autism-related behaviors in mice. We found that maternal HFD induced an autism-related behavioral phenotype in the male offspring, including a robust decrease in sociability, increased aggression, cognitive rigidity and repetitive behaviors. Maternal LPD led to a milder yet significant effect on autism-related symptoms, with no effects on olfactory-mediated social behavior. Under naturalistic conditions in a group setting, this manifested in altered behavioral repertoires, increased magnitude in dominance relations, and reduced interactions with novel social stimuli in the HFD male offspring, but not in the LPD offspring. Finally, we found HFD-induced transcriptomic changes in the olfactory bulbs of the male offspring. Together, our findings show that maternal malnutrition induces long-lasting effects on aggression and autism-related behaviors in male offspring, and potential impairments in brain regions processing chemosensory signals.


Subject(s)
Autistic Disorder , Behavior, Animal , Diet, High-Fat , Diet, Protein-Restricted , Social Behavior , Animals , Diet, High-Fat/adverse effects , Female , Male , Mice , Autistic Disorder/etiology , Autistic Disorder/metabolism , Pregnancy , Diet, Protein-Restricted/adverse effects , Aggression , Prenatal Exposure Delayed Effects/metabolism , Mice, Inbred C57BL , Maternal Nutritional Physiological Phenomena , Olfactory Bulb/metabolism , Disease Models, Animal , Obesity/metabolism , Obesity/etiology
13.
Cell Rep ; 43(8): 114612, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39110592

ABSTRACT

Atypical sensory processing is common in autism, but how neural coding is disrupted in sensory cortex is unclear. We evaluate whisker touch coding in L2/3 of somatosensory cortex (S1) in Cntnap2-/- mice, which have reduced inhibition. This classically predicts excess pyramidal cell spiking, but this remains controversial, and other deficits may dominate. We find that c-fos expression is elevated in S1 of Cntnap2-/- mice under spontaneous activity conditions but is comparable to that of control mice after whisker stimulation, suggesting normal sensory-evoked spike rates. GCaMP8m imaging from L2/3 pyramidal cells shows no excess whisker responsiveness, but it does show multiple signs of degraded somatotopic coding. This includes broadened whisker-tuning curves, a blurred whisker map, and blunted whisker point representations. These disruptions are greater in noisy than in sparse sensory conditions. Tuning instability across days is also substantially elevated in Cntnap2-/-. Thus, Cntnap2-/- mice show no excess sensory-evoked activity, but a degraded and unstable tactile code in S1.


Subject(s)
Autistic Disorder , Disease Models, Animal , Membrane Proteins , Nerve Tissue Proteins , Somatosensory Cortex , Vibrissae , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiopathology , Mice , Autistic Disorder/physiopathology , Autistic Disorder/genetics , Autistic Disorder/metabolism , Mice, Knockout , Touch/physiology , Mice, Inbred C57BL , Pyramidal Cells/metabolism , Male , Proto-Oncogene Proteins c-fos/metabolism
14.
Neuroscience ; 558: 92-104, 2024 Oct 18.
Article in English | MEDLINE | ID: mdl-39168175

ABSTRACT

This study aimed to evaluate the potential benefits of acetyl-L-carnitine (ALCAR) in the context of valproate-induced autism. After prenatal exposure to valproate (VPA; 600 mg/kg, i.p.) on embryonic day 12.5, followed by ALCAR treatment (300 mg/kg on postnatal days 21-49, p.o.), assessment of oxidative stress, mitochondrial membrane potential (MMP), mitochondrial biogenesis, parvalbumin interneurons, and hippocampal volume was conducted. These assessments were carried out subsequent to the evaluation of autism-like behaviors. Hippocampal analysis of oxidative factors (reactive oxygen species and malondialdehyde) and antioxidants (superoxide dismutase, catalase, and glutathione) revealed a burden of oxidative stress in VPA rats. Additionally, mitochondrial biogenesis and MMP were elevated, while the number of parvalbumin interneurons decreased. These changes were accompanied by autism-like behaviors observed in the three-chamber maze, marble burring test, and Y-maze, as well as a learning deficit in the Barnes maze. In contrast, administrating ALCAR attenuated behavioral deficits, reduced oxidative stress, improved parvalbumin-positive neuronal population, and properly modified MMP and mitochondrial biogenesis. Collectively, our results indicate that oral administration of ALCAR ameliorates autism-like behaviors, partly through its targeting oxidative stress and mitochondrial biogenesis. This suggests that ALCAR may have potential benefits ASD managing.


Subject(s)
Acetylcarnitine , Autistic Disorder , Hippocampus , Mitochondria , Oxidative Stress , Valproic Acid , Animals , Valproic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Acetylcarnitine/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Autistic Disorder/metabolism , Female , Male , Pregnancy , Membrane Potential, Mitochondrial/drug effects , Rats , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Rats, Sprague-Dawley , Disease Models, Animal , Behavior, Animal/drug effects
15.
Proc Natl Acad Sci U S A ; 121(35): e2406928121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39178233

ABSTRACT

Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.


Subject(s)
Valproic Acid , gamma-Aminobutyric Acid , Animals , Female , Mice , Male , Pregnancy , Valproic Acid/toxicity , gamma-Aminobutyric Acid/metabolism , Interneurons/metabolism , Animals, Newborn , Behavior, Animal , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Autistic Disorder/etiology , Autistic Disorder/metabolism , Glutamic Acid/metabolism , Neurotransmitter Agents/metabolism , Poly I-C , Prefrontal Cortex/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/pathology , Cholecystokinin/metabolism , Parvalbumins/metabolism , Mice, Inbred C57BL , Stereotyped Behavior/drug effects
16.
PLoS One ; 19(8): e0308792, 2024.
Article in English | MEDLINE | ID: mdl-39146282

ABSTRACT

BACKGROUND: The neurobiological underpinnings of Autism Spectrum Disorder (ASD) are diverse and likely multifactorial. One possible mechanism is increased oxidative stress leading to altered neurodevelopment and brain function. However, this hypothesis has mostly been tested in post-mortem studies. So far, available in vivo studies in autistic individuals have reported no differences in glutathione (GSH) levels in frontal, occipital, and subcortical regions. However, these studies were limited by the technically challenging quantification of GSH, the main brain antioxidant molecule. This study aimed to overcome previous studies' limitations by using a GSH-tailored spectroscopy sequence and optimised quantification methodology to provide clarity on GSH levels in autistic adults. METHODS: We used spectral editing proton-magnetic resonance spectroscopy (1H-MRS) combined with linear combination model fitting to quantify GSH in the dorsomedial prefrontal cortex (DMPFC) and medial occipital cortex (mOCC) of autistic and non-autistic adults (male and female). We compared GSH levels between groups. We also examined correlations between GSH and current autism symptoms, measured using the Autism Quotient (AQ). RESULTS: Data were available from 31 adult autistic participants (24 males, 7 females) and 40 non-autistic participants (21 males, 16 females); the largest sample to date. The GSH levels did not differ between groups in either region. No correlations with AQ were observed. CONCLUSION: GSH levels as measured using 1H-MRS are unaltered in the DMPFC and mOCC regions of autistic adults, suggesting that oxidative stress in these cortical regions is not a marked neurobiological signature of ASD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Glutathione , Occipital Lobe , Humans , Male , Female , Glutathione/metabolism , Glutathione/analysis , Adult , Occipital Lobe/metabolism , Occipital Lobe/diagnostic imaging , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Young Adult , Proton Magnetic Resonance Spectroscopy , Frontal Lobe/metabolism , Oxidative Stress , Middle Aged , Prefrontal Cortex/metabolism , Prefrontal Cortex/diagnostic imaging
18.
Article in English | MEDLINE | ID: mdl-39032854

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that is marked by impaired social interactions, and increased repetitive behaviors. There is evidence of genetic changes in ASD, and several of these altered genes are linked to the process of DNA repair. Therefore, individuals with ASD must have improved DNA repair efficiency to mitigate risks associated with ASD. Despite numerous milestones in ASD research, the disease remains incurable, with a high occurrence rate and substantial financial burdens. This motivates scientists to search for new drugs to manage the disease. Disruption of glucagon-like peptide-1 (GLP-1) signaling, a regulator in neuronal development and maintains homeostasis, has been associated with the pathogenesis and progression of several neurological disorders, such as ASD. Our study aimed to assess the impact of semaglutide, a new GLP-1 analog antidiabetic medication, on behavioral phenotypes and DNA repair efficiency in the BTBR autistic mouse model. Furthermore, we elucidated the underlying mechanism(s) responsible for the ameliorative effects of semaglutide against behavioral problems and DNA repair deficiency in BTBR mice. The current results demonstrate that repeated treatment with semaglutide efficiently decreased autism-like behaviors in BTBR mice without affecting motor performance. Semaglutide also mitigated spontaneous DNA damage and enhanced DNA repair efficiency in the BTBR mice as determined by comet assay. Moreover, administering semaglutide recovered oxidant-antioxidant balance in BTBR mice. Semaglutide restored the disrupted DNA damage/repair pathways in the BTBR mice by reducing Gadd45a expression and increasing Ogg1 and Xrcc1 expression at both the mRNA and protein levels. This suggests that semaglutide holds great potential as a novel therapeutic candidate for treating ASD traits.


Subject(s)
DNA Repair , Glucagon-Like Peptides , Animals , Male , Glucagon-Like Peptides/pharmacology , DNA Repair/drug effects , Mice , Disease Models, Animal , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Gene Expression/drug effects , Hypoglycemic Agents/pharmacology , Autistic Disorder/drug therapy , Autistic Disorder/genetics , Autistic Disorder/metabolism , Behavior, Animal/drug effects
19.
Cells ; 13(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38994948

ABSTRACT

Excessive inflammatory reactions and oxidative stress are well-recognized molecular findings in autism and these processes can affect or be affected by the epigenetic landscape. Nonetheless, adequate therapeutics are unavailable, as patient-specific brain molecular markers for individualized therapies remain challenging. METHODS: We used iPSC-derived neurons and astrocytes of patients with autism vs. controls (5/group) to examine whether they replicate the postmortem brain expression/epigenetic alterations of autism. Additionally, DNA methylation of 10 postmortem brain samples (5/group) was analyzed for genes affected in PSC-derived cells. RESULTS: We found hyperexpression of TGFB1, TGFB2, IL6 and IFI16 and decreased expression of HAP1, SIRT1, NURR1, RELN, GPX1, EN2, SLC1A2 and SLC1A3 in the astrocytes of patients with autism, along with DNA hypomethylation of TGFB2, IL6, TNFA and EN2 gene promoters and a decrease in HAP1 promoter 5-hydroxymethylation in the astrocytes of patients with autism. In neurons, HAP1 and IL6 expression trended alike. While HAP1 promoter was hypermethylated in neurons, IFI16 and SLC1A3 promoters were hypomethylated and TGFB2 exhibited increased promoter 5-hydroxymethlation. We also found a reduction in neuronal arborization, spine size, growth rate, and migration, but increased astrocyte size and a reduced growth rate in autism. In postmortem brain samples, we found DNA hypomethylation of TGFB2 and IFI16 promoter regions, but DNA hypermethylation of HAP1 and SLC1A2 promoters in autism. CONCLUSION: Autism-associated expression/epigenetic alterations in iPSC-derived cells replicated those reported in the literature, making them appropriate surrogates to study disease pathogenesis or patient-specific therapeutics.


Subject(s)
Astrocytes , Autistic Disorder , Brain , DNA Methylation , Epigenesis, Genetic , Induced Pluripotent Stem Cells , Neurons , Humans , Astrocytes/metabolism , Astrocytes/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Autistic Disorder/genetics , Autistic Disorder/pathology , Autistic Disorder/metabolism , Neurons/metabolism , Neurons/pathology , DNA Methylation/genetics , Brain/pathology , Brain/metabolism , Male , Female , Promoter Regions, Genetic/genetics , Cell Shape , Child , Gene Expression Regulation , Reelin Protein
20.
Bull Exp Biol Med ; 177(1): 47-50, 2024 May.
Article in English | MEDLINE | ID: mdl-38955852

ABSTRACT

Ectonucleotidases play an important role in regulating the level of extracellular nucleotides and nucleosides and are an important part of the regulation of the effects of adenosine and ATP on adenosine and P2 receptors, respectively. We have previously established the ambiguous effect of P2 receptor agonists on the contractile activity of smooth muscle tissue in rats with the valproate model of autism. In this work, HPLC was used to evaluate the activity of ectonucleotidases in the smooth muscle tissues of the internal organs of rats with a valproate model of autism. The activity of ectonucleotidases was significantly higher in the smooth muscle tissues of the duodenum, vas deferens, and bladder, but lower in the ileum and uterus. The results obtained make it possible to compare the activity of ectonucleotidases identified here with changes in P2 receptor-mediated contractility of smooth muscle tissues revealed in our previous experiments.


Subject(s)
Autistic Disorder , Muscle Contraction , Muscle, Smooth , Urinary Bladder , Valproic Acid , Vas Deferens , Animals , Rats , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Valproic Acid/pharmacology , Autistic Disorder/metabolism , Autistic Disorder/chemically induced , Autistic Disorder/drug therapy , Male , Female , Vas Deferens/drug effects , Vas Deferens/metabolism , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/enzymology , Muscle Contraction/drug effects , Uterus/drug effects , Uterus/metabolism , Ileum/drug effects , Ileum/metabolism , Ileum/enzymology , Disease Models, Animal , Rats, Wistar , Receptors, Purinergic P2/metabolism , Adenosine Triphosphatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL