Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 737
Filter
1.
J Integr Neurosci ; 23(9): 165, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39344236

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social interaction, communication, repetitive behaviors, and narrow interests. This study aimed to investigate the impact of the Hypoxia-inducible factor-1 alpha (HIF-1α) inhibitor (PX-478) on ASD-like behaviors in rat offspring exposed to prenatal hypoxia (PH). METHODS: Pregnant rats were randomly assigned to control or PH groups, with the latter experiencing six hours of hypoxia on the 17th day of gestation. Offspring were further treated with PX-478 treatment initiated at one week (+1 w) or three weeks (+3 w) after birth. Hippocampal histology was assessed using hematoxylin and eosin (HE) staining, while protein levels of HIF-1α and phosphatase and tensin homolog (PTEN) were analyzed via western blotting. The concentration of vascular endothelial growth factor (VEGF) was measured using an Enzyme-Linked Immunosorbent Assay (ELISA) kit. RESULTS: PX-478 treatment significantly improved spatial memory, learning, and social ability, while reducing anxiety-like behavior in PH-exposed offspring rats. HE staining revealed that PX-478 treatment decreased the number of hippocampal neurons necrosis in offspring. However, PX-478 treatment at one week post-birth led to decreased body weight and elevated levels of alkaline phosphatase (ALP) and Alanine aminotransferase (ALT) in offspring rats, whereas no significant effect was observed after three weeks of treatment. Additionally, PX-478 treatment resulted in reduced HIF-1α protein levels in the hippocampus and VEGF concentration in the serum of PH-exposed offspring rats, along with elevated PTEN protein levels. CONCLUSIONS: The findings suggest that PX-478 treatment attenuated autism-like behavior in offspring. HIF-1α might play an important role in autism-like behavior induced by prenatal hypoxia, which may be realized by inhibiting PTEN activity.


Subject(s)
Autism Spectrum Disorder , Hypoxia-Inducible Factor 1, alpha Subunit , Prenatal Exposure Delayed Effects , Rats, Sprague-Dawley , Animals , Pregnancy , Female , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/drug therapy , Prenatal Exposure Delayed Effects/metabolism , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Disease Models, Animal , Behavior, Animal/drug effects , PTEN Phosphohydrolase/metabolism , Male , Hypoxia/complications , Disease Progression , Vascular Endothelial Growth Factor A/metabolism , Social Behavior
2.
BMC Med ; 22(1): 393, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39278907

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder that is globally increasing in prevalence. The rise of ASD can be partially attributed to diagnostic expansion and advocacy efforts; however, the interplay between genetic predisposition and modern environmental exposures is likely driving a true increase in incidence. A range of evidence indicates that prenatal exposures are critical. Infection during pregnancy, gestational diabetes, and maternal obesity are established risk factors for ASD. Emerging areas of research include the effects of maternal use of selective serotonin reuptake inhibitors, antibiotics, and exposure to toxicants during pregnancy on brain development and subsequent ASD. The underlying pathways of these risk factors remain uncertain, with varying levels of evidence implicating immune dysregulation, mitochondrial dysfunction, oxidative stress, gut microbiome alterations, and hormonal disruptions. This narrative review assesses the evidence of contributing prenatal environmental factors for ASD and associated mechanisms as potential targets for novel prevention strategies.


Subject(s)
Autism Spectrum Disorder , Prenatal Exposure Delayed Effects , Humans , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/epidemiology , Pregnancy , Risk Factors , Female , Environmental Exposure/adverse effects , Maternal Exposure/adverse effects
3.
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
4.
Med Sci Monit ; 30: e945087, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39180197

ABSTRACT

Fetal brain development is an important determinant of neuropsychological performance in children. Any alterations in the intrauterine environment at different stages of pregnancy, such as maternal metabolic disorders, can lead to the development of chronic conditions in the offspring. Therefore, maternal diabetes, especially gestational diabetes mellitus, is an important factor in the development of pathological changes, such as miscarriage, fetal macrosomia, or neurodevelopmental disorders. During pregnancy, the hyperglycemic intrauterine environment adversely affects fetal brain development. A growing body of scientific research indicates that prenatal environmental factors, by affecting fetal brain development, can contribute to the appearance of autism spectrum disorders. According to the latest estimates from the International Diabetes Federation (2021), approximately 21.1 million live births worldwide (16.7%) have been affected by some form of hyperglycemia during pregnancy. The condition is more prevalent in low- and middle-income countries, where access to obstetric care is limited. The following factors have been identified as potential risk factors for gestational diabetes: advanced maternal age, overweight and obesity, family history of diabetes, and any form of diabetes. The purpose of this review is to summarize recent studies evaluating the effect of prenatal and maternal risk factors such as maternal pre-pregnancy diabetes, gestational diabetes, and obesity on the risk of developing autism spectrum disorder in offspring.


Subject(s)
Autism Spectrum Disorder , Diabetes, Gestational , Obesity , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/epidemiology , Female , Obesity/complications , Risk Factors
5.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125662

ABSTRACT

Evidence shows that Autism Spectrum Disorder (ASD) stems from an interplay of genetic and environmental factors, which may include propionic acid (PPA), a microbial byproduct and food preservative. We previously reported that in vitro treatment of neural stem cells with PPA leads to gliosis and neuroinflammation. In this study, mice were exposed ad libitum to a PPA-rich diet for four weeks before mating. The same diet was maintained through pregnancy and administered to the offspring after weaning. The brains of the offspring were studied at 1 and 5 months postpartum. Glial fibrillary acidic protein (astrocytic marker) was significantly increased (1.53 ± 0.56-fold at 1 M and 1.63 ± 0.49-fold at 5 M) in the PPA group brains. Tubulin IIIß (neuronal marker) was significantly decreased in the 5 M group. IL-6 and TNF-α expression were increased in the brain of the PPA group (IL-6: 2.48 ± 1.25-fold at 5 M; TNF-α: 2.84 ± 1.16-fold at 1 M and 2.64 ± 1.42-fold, at 5 M), while IL-10 was decreased. GPR41 and p-Akt were increased, while PTEN (p-Akt inhibitor) was decreased in the PPA group. The data support the role of a PPA-rich diet in glia over-proliferation and neuro-inflammation mediated by the GPR41 receptor and PTEN/Akt pathway. These findings strongly support our earlier study on the role of PPA in ASD.


Subject(s)
Autism Spectrum Disorder , Disease Models, Animal , Gliosis , Propionates , Animals , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/pathology , Mice , Gliosis/metabolism , Gliosis/pathology , Female , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Glial Fibrillary Acidic Protein/metabolism , Male , Diet/adverse effects , Brain/metabolism , Brain/pathology , Pregnancy , Mice, Transgenic
6.
Int J Mol Sci ; 25(16)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39201625

ABSTRACT

Increasing evidence suggests an association between atopic dermatitis, the most chronic inflammatory disease of the skin, and autism spectrum disorders, which are a group of neurodevelopmental diseases. Inflammation and immune dysregulation associated with genetic and environmental factors seem to characterize the pathophysiological mechanisms of both conditions. We conducted a literature review of the PubMed database aimed at identifying the clinical features and alleged risk factors that could be used in clinical practice to predict the onset of ASD and/or AD or worsen their prognosis in the context of comorbidities.


Subject(s)
Autism Spectrum Disorder , Dermatitis, Atopic , Dermatitis, Atopic/etiology , Dermatitis, Atopic/pathology , Humans , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/epidemiology , Risk Factors , Comorbidity
7.
Gut Microbes ; 16(1): 2385117, 2024.
Article in English | MEDLINE | ID: mdl-39120056

ABSTRACT

Autism spectrum disorders (ASD) are highly heritable, heterogeneous neurodevelopmental disorders characterized by clinical presentation of atypical social, communicative, and repetitive behaviors. Over the past 25 years, hundreds of ASD risk genes have been identified. Many converge on key molecular pathways, from translational control to those regulating synaptic structure and function. Despite these advances, therapeutic approaches remain elusive. Emerging data unearthing the relationship between genetics, microbes, and immunity in ASD suggest an integrative physiology approach could be paramount to delivering therapeutic breakthroughs. Indeed, the advent of large-scale multi-OMIC data acquisition, analysis, and interpretation is yielding an increasingly mechanistic understanding of ASD and underlying risk factors, revealing how genetic susceptibility interacts with microbial genetics, metabolism, epigenetic (re)programming, and immunity to influence neurodevelopment and behavioral outcomes. It is now possible to foresee exciting advancements in the treatment of some forms of ASD that could markedly improve quality of life and productivity for autistic individuals. Here, we highlight recent work revealing how gene X maternal exposome interactions influence risk for ASD, with emphasis on the intrauterine environment and fetal neurodevelopment, host-microbe interactions, and the evolving therapeutic landscape for ASD.


Subject(s)
Autism Spectrum Disorder , Humans , Autism Spectrum Disorder/microbiology , Autism Spectrum Disorder/etiology , Female , Pregnancy , Exposome , Gastrointestinal Microbiome , Risk Factors , Genetic Predisposition to Disease , Animals , Autistic Disorder/etiology , Autistic Disorder/microbiology
8.
J Korean Med Sci ; 39(30): e218, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39106887

ABSTRACT

BACKGROUND: Recent studies have reported the burden of attention deficit hyperactivity disorder [ADHD], autism spectrum disorder [ASD], and depressive disorder. Also, there is mounting evidence on the effects of environmental factors, such as ambient air pollution, on these disorders among children and adolescents. However, few studies have evaluated the burden of mental disorders attributable to air pollution exposure in children and adolescents. METHODS: We estimated the risk ratios of major mental disorders (ADHD, ASD, and depressive disorder) associated with air pollutants among children and adolescents using time-series data (2011-2019) obtained from a nationwide air pollution monitoring network and healthcare utilization claims data in the Republic of Korea. Based on the estimated risk ratios, we determined the population attributable fraction (PAF) and calculated the medical costs of major mental disorders attributable to air pollution. RESULTS: A total of 33,598 patients were diagnosed with major mental disorders during 9 years. The PAFs for all the major mental disorders were estimated at 6.9% (particulate matter < 10 µm [PM10]), 3.7% (PM2.5), and 2.2% (sulfur dioxide [SO2]). The PAF of PM10 was highest for depressive disorder (9.2%), followed by ASD (8.4%) and ADHD (5.2%). The direct medical costs of all major mental disorders attributable to PM10 and SO2 decreased during the study period. CONCLUSION: This study assessed the burden of major mental disorders attributable to air pollution exposure in children and adolescents. We found that PM10, PM2.5, and SO2 attributed 7%, 4%, and 2% respectively, to the risk of major mental disorders among children and adolescents.


Subject(s)
Air Pollution , Attention Deficit Disorder with Hyperactivity , Particulate Matter , Humans , Child , Adolescent , Republic of Korea/epidemiology , Air Pollution/adverse effects , Particulate Matter/adverse effects , Particulate Matter/analysis , Male , Female , Attention Deficit Disorder with Hyperactivity/epidemiology , Attention Deficit Disorder with Hyperactivity/etiology , Environmental Exposure/adverse effects , Mental Disorders/epidemiology , Mental Disorders/etiology , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Air Pollutants/adverse effects , Air Pollutants/analysis , Sulfur Dioxide/adverse effects , Sulfur Dioxide/analysis , Child, Preschool , Risk Factors , Health Care Costs
9.
Front Endocrinol (Lausanne) ; 15: 1389947, 2024.
Article in English | MEDLINE | ID: mdl-39157677

ABSTRACT

Objective: The relationship between diabetes mellitus (DM) and autism spectrum disorder (ASD) remains controversial. This study aimed to analyze the causal relationship between different types of DM and ASD by bidirectional Mendelian randomization (MR). Methods: Single nucleotide polymorphisms for type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), gestational diabetes mellitus (GDM), and ASD were obtained from genome-wide association studies. Subsequently, inverse variance weighted, MR-Egger, and weighted median were used to test the exposure-outcome causality. Finally, MR-Egger's intercept, Cochran's Q, and leave-one-out method were used to assess horizontal pleiotropy, heterogeneity, and sensitivity of the results, respectively. Results: The positive analysis showed that T2DM was associated with an increased risk of ASD, whereas neither T1DM nor GDM was associated with the risk of ASD. The reverse analysis showed that ASD was associated with an increased risk of T2DM, while it was not associated with the risk of either T1DM or GDM. MR-Egger intercept showed no horizontal pleiotropy (p > 0.05) for these results. Cochran's Q showed no heterogeneity expect for the results of T1DM on the risk of ASD, and leave-one-out sensitivity analysis showed these results were robust. Conclusion: This MR analysis suggests that T2DM and ASD are reciprocal risk factors and that they may create an intergenerational risk cycling in female patients. Aggressive prevention and treatment of T2DM and ASD help to break the trap of this risk cycling. Additionally, this study does not support a causal relationship between T1DM and ASD, as well as GDM and ASD. And more studies are needed in the future to continue to explore the interactions and underlying mechanisms between different types of DM and ASD.


Subject(s)
Autism Spectrum Disorder , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Diabetes, Gestational , Genome-Wide Association Study , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Humans , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Female , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/complications , Pregnancy , Diabetes, Gestational/genetics , Diabetes, Gestational/epidemiology , Risk Factors , Genetic Predisposition to Disease , Male
10.
Adv Nutr ; 15(9): 100279, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39059765

ABSTRACT

Pregnancy is a time of high metabolic coordination, as maternal metabolism adapts to support the growing fetus. Many of these changes are coordinated by the placenta, a critical fetal endocrine organ and the site of maternal-fetal crosstalk. Dysregulation in maternal and placental metabolism during pregnancy has been linked to adverse outcomes, including altered neurodevelopment. Autism spectrum disorder (ASD) is a neurodevelopmental disorder linked to metabolic alterations in both children and their mothers. Prenatal environmental exposures have been linked to risk of ASD through dysregulated maternal, placental, and fetal metabolism. In this review, we focus on recent studies investigating the associations between prenatal metabolism in the maternal-placental-fetal unit and the impact of prenatal environmental exposures to phthalates and micronutrients on ASD risk. By identifying the mechanisms through which phthalates and other ubiquitous endocrine disrupting chemicals influence development, and how nutritional interventions can impact those mechanisms, we can identify promising ways to prevent suboptimal neurodevelopment.


Subject(s)
Autism Spectrum Disorder , Micronutrients , Phthalic Acids , Placenta , Prenatal Exposure Delayed Effects , Humans , Autism Spectrum Disorder/etiology , Pregnancy , Female , Placenta/metabolism , Endocrine Disruptors/adverse effects , Endocrine Disruptors/toxicity , Maternal Exposure/adverse effects , Environmental Exposure/adverse effects , Child
11.
Neurosci Biobehav Rev ; 164: 105815, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053787

ABSTRACT

Exposure to environmental pollutants, such as metals, pesticides, and air pollutants during early life, is a risk factor for neurodevelopmental disorders (NDDs), including Autism Spectrum Disorder (ASD). Our systematic review aimed to select and summarize more recent case-control studies that examined the association between prenatal and early postnatal exposure to environmental pollutants and NDDs. We searched five databases (Web of Science, PubMed, Embase, Scopus, Ovid), screened 2261 records, and included 24 eligible case-control studies. Meta-analyses were conducted on subgroups of at least three studies that shared both the outcome and the exposure. A noteworthy discovery from this literature review is the existence of non-linear or non-monotonic dose-response relationships between the exposure to certain metals and the risk of ASD. The meta-analysis revealed a significant association between exposure to particular matter (PM)10 during the first year of life and the risk of ASD. Overall, studies included in our systematic review indicate that exposure to several pollutants within the first three years of life was significantly associated with the risk of NDDs.


Subject(s)
Environmental Pollutants , Neurodevelopmental Disorders , Humans , Environmental Pollutants/adverse effects , Environmental Pollutants/toxicity , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/epidemiology , Neurodevelopmental Disorders/etiology , Case-Control Studies , Prenatal Exposure Delayed Effects/chemically induced , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Environmental Exposure/adverse effects , Pregnancy , Autistic Disorder/chemically induced , Autistic Disorder/epidemiology , Autistic Disorder/etiology , Female
12.
Sci Rep ; 14(1): 17159, 2024 07 26.
Article in English | MEDLINE | ID: mdl-39060326

ABSTRACT

The burden of autism spectrum disorder (ASD) is increasing worldwide with genetic, epigenetic, and environmental factors being possibly responsible for the observed epidemiological figures. In the setting of environmental exposure, the city of Taranto, in Southern Italy, represents an interesting case study as it hosts well inside the city one of the biggest steel plants in Europe. This is a cross-sectional ecological study carried out in the year 2020 in the province of Taranto designed to estimate the burden of ASD in the municipalities of Taranto and Statte, classified as high environmental risk areas (Contaminated Site of National Interest-SIN), compared to the other 27 municipalities of the same province. Differences have been evaluated using the Chi Square Test. Children aged 6-11 years identified in SIN municipalities had a statistically significant higher prevalence of ASD than children of other municipalities (9.58 vs. 6.66/1000 respectively, p = 0.002). No statistically significant difference was observed for the 12-18 years group (3.41 vs. 2.54/1000, p = 0.12). The findings observed in this study are suggestive of the association between urban residential proximity to industrial facilities emitting air pollutants and higher ASD prevalence.


Subject(s)
Autism Spectrum Disorder , Environmental Exposure , Humans , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Child , Male , Female , Prevalence , Italy/epidemiology , Cross-Sectional Studies , Environmental Exposure/adverse effects , Adolescent , Air Pollutants/adverse effects , Air Pollutants/analysis , Air Pollution/adverse effects , Environmental Pollution/adverse effects
13.
BMC Pediatr ; 24(1): 465, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033272

ABSTRACT

BACKGROUND: The nutritional status and growth of children with Autism spectrum disorders (ASD) is influenced significantly by two factors; food selectivity behaviors due to their consumption of a limited variety of food and the high incidence of gastrointestinal (GIT) disorders. AIM: This study aimed to assess the nutritional adequacy and growth pattern of ASD children aged three to twelve years compared to their healthy developing peers. METHODS: A national comparative, facility-based cross-sectional study was conducted in eight Egyptian governorates on 285 Egyptian children diagnosed with ASD and 224 children who are their relatives as healthy developing peers. Anthropometric measurements were obtained, including weight, height, head circumference, and mid-upper arm circumference. Body Mass Index (BMI) was calculated and all numbers were plotted on WHO growth charts. Assessment of food preferences, and nutrient intake adequacy of children was done using the Food preference questionnaire, and the Dietary Reference Intakes (DRIs) of Egyptian children. RESULTS: Calorie-dense food and sugar intake were higher among ASD children than their healthy developing peers. ASD children omit some important protein sources such as dairy (COR = 5.2, 95% CI:2.7-9.9), meat, and poultry (COR = 2.7, 95% CI: 1.6-4.7), and a lower intake of fruits and vegetables than their healthy developing peers. For children with ASD in all age groups, a deficiency in the range of 50-60% was detected for vitamins (C, D, B6, thiamine, riboflavin, niacin) and minerals (iron). A deficiency in the range of 60-70% was detected for folate and calcium. A deficiency of vitamin C calcium and iron was also detected for both children with ASD and their healthy developing relatives aged 6 to 12 years. GIT disorders were common among ASD children compared to healthy developing peers (COR = 2.8 to 10.3). Children with ASD had four-fold higher odds of stunting (COR = 4.1, CI: 1.7-10.1), threefold higher odds of being overweight (COR = 3.3, CI: 1.48-7.32), and nearly eleven-fold higher odds of obesity (COR = 11.4, CI: 4.05-32.17) compared to their healthy developing peers. CONCLUSION: ASD children are prone to overweight and protein malnutrition. Their intake of fruits and vegetables is inadequate and hence their intake of vitamins and minerals is insufficient, contributing to stunting.


Subject(s)
Autism Spectrum Disorder , Growth Disorders , Pediatric Obesity , Humans , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Egypt/epidemiology , Male , Female , Cross-Sectional Studies , Child, Preschool , Child , Pediatric Obesity/epidemiology , Growth Disorders/epidemiology , Growth Disorders/etiology , Nutritional Status , Case-Control Studies , Food Preferences , Body Mass Index , Diet
14.
Int J Mol Sci ; 25(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39062930

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social interaction and communication, anxiety, hyperactivity, and interest restricted to specific subjects. In addition to the genetic factors, multiple environmental factors have been related to the development of ASD. Animal models can serve as crucial tools for understanding the complexity of ASD. In this study, a chemical model of ASD has been developed in zebrafish by exposing embryos to valproic acid (VPA) from 4 to 48 h post-fertilization, rearing them to the adult stage in fish water. For the first time, an integrative approach combining behavioral analysis and neurotransmitters profile has been used for determining the effects of early-life exposure to VPA both in the larval and adult stages. Larvae from VPA-treated embryos showed hyperactivity and decreased visual and vibrational escape responses, as well as an altered neurotransmitters profile, with increased glutamate and decreased acetylcholine and norepinephrine levels. Adults from VPA-treated embryos exhibited impaired social behavior characterized by larger shoal sizes and a decreased interest for their conspecifics. A neurotransmitter analysis revealed a significant decrease in dopamine and GABA levels in the brain. These results support the potential predictive validity of this model for ASD research.


Subject(s)
Behavior, Animal , Disease Models, Animal , Valproic Acid , Zebrafish , Animals , Valproic Acid/toxicity , Valproic Acid/adverse effects , Behavior, Animal/drug effects , Neurotransmitter Agents/metabolism , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/etiology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Larva/drug effects , Social Behavior , Brain/drug effects , Brain/metabolism , Brain/growth & development
15.
Sci Rep ; 14(1): 14759, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926504

ABSTRACT

ADHD and ASD are highly heritable and show a high co-occurrence and persistence into adulthood. This study aimed to identify pre and perinatal risk factors, and early psychosocial exposures related to later diagnosis of ADHD, ASD, and their co-occurrence. 16,365 children born 1997-1999 and their families, involved in the prospective population-based ABIS study (All Babies in Southeast Sweden), were included in this sub-study. Pre and perinatal factors and early environmental psychosocial exposures were collected from parental-questionnaires at birth and 1-year follow-up. Diagnoses from birth up to 23 years of age were obtained from the Swedish National Diagnosis Register in 2020. The cumulative incidence of ADHD, ASD, and their co-occurrence in the ABIS-cohort Study were 4.6%, 1.7%, and 1.1%, respectively. Being male was associated with an increased risk for ADHD, ASD, and their co-occurrence (aOR 1.30, 1.56, and 1.91, respectively), while higher household income reduced it (aOR 0.82, 0.73, and 0.64). Serious life events during pregnancy (aOR 1.40) and maternal smoking (aOR 1.51) increased the risk of ADHD, while older maternal age (aOR 0.96), higher parental education (aOR 0.72 maternal and aOR 0.74 paternal) and longer exclusive breastfeeding (aOR 0.72) reduced it. Non-Swedish paternal nationality (aOR 0.40) and higher maternal education (aOR 0.74) were associated with a lower risk of ASD, while a family history of autoimmune diseases increased the risk of the co-occurrence of both disorders (aOR 1.62). Obtained results suggest that the etiology of ADHD, ASD, and their co-occurrence is independently associated with environmental psychosocial predictors. The co-occurrence seems to overlap the etiology of ADHD, in which psychosocial determinants have a larger role, however, it is also independently influenced by a family history of autoimmune diseases.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Humans , Attention Deficit Disorder with Hyperactivity/epidemiology , Attention Deficit Disorder with Hyperactivity/etiology , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Female , Male , Prospective Studies , Child , Sweden/epidemiology , Risk Factors , Child, Preschool , Pregnancy , Infant , Adolescent , Adult , Infant, Newborn , Young Adult , Incidence , Prenatal Exposure Delayed Effects/epidemiology
16.
J Neurodev Disord ; 16(1): 26, 2024 May 26.
Article in English | MEDLINE | ID: mdl-38796448

ABSTRACT

BACKGROUND: Synthetic oxytocin (sOT) is frequently administered during parturition. Studies have raised concerns that fetal exposure to sOT may be associated with altered brain development and risk of neurodevelopmental disorders. In a large and diverse sample of children with data about intrapartum sOT exposure and subsequent diagnoses of two prevalent neurodevelopmental disorders, i.e., attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD), we tested the following hypotheses: (1) Intrapartum sOT exposure is associated with increased odds of child ADHD or ASD; (2) associations differ across sex; (3) associations between intrapartum sOT exposure and ADHD or ASD are accentuated in offspring of mothers with pre-pregnancy obesity. METHODS: The study sample comprised 12,503 participants from 44 cohort sites included in the Environmental Influences on Child Health Outcomes (ECHO) consortium. Mixed-effects logistic regression analyses were used to estimate the association between intrapartum sOT exposure and offspring ADHD or ASD (in separate models). Maternal obesity (pre-pregnancy BMI ≥ 30 kg/m2) and child sex were evaluated for effect modification. RESULTS: Intrapartum sOT exposure was present in 48% of participants. sOT exposure was not associated with increased odds of ASD (adjusted odds ratio [aOR] 0.86; 95% confidence interval [CI], 0.71-1.03) or ADHD (aOR 0.89; 95% CI, 0.76-1.04). Associations did not differ by child sex. Among mothers with pre-pregnancy obesity, sOT exposure was associated with lower odds of offspring ADHD (aOR 0.72; 95% CI, 0.55-0.96). No association was found among mothers without obesity (aOR 0.97; 95% CI, 0.80-1.18). CONCLUSIONS: In a large, diverse sample, we found no evidence of an association between intrapartum exposure to sOT and odds of ADHD or ASD in either male or female offspring. Contrary to our hypothesis, among mothers with pre-pregnancy obesity, sOT exposure was associated with lower odds of child ADHD diagnosis.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Body Mass Index , Oxytocin , Prenatal Exposure Delayed Effects , Humans , Female , Pregnancy , Male , Attention Deficit Disorder with Hyperactivity/epidemiology , Attention Deficit Disorder with Hyperactivity/etiology , Child , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Adult , Obesity, Maternal/epidemiology , Child, Preschool , Cohort Studies , Obesity/epidemiology
17.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(5): 535-540, 2024 May 15.
Article in Chinese | MEDLINE | ID: mdl-38802917

ABSTRACT

Autism spectrum disorder (ASD) is a pervasive neurodevelopmental disorder with onset in infancy or early childhood. Mendelian randomization (MR) is a statistical method used to infer causal relationships between exposures and outcomes. This article summarizes MR studies related to ASD. Existing research supports a causal relationship between maternal inflammatory bowel disease in children with ASD, parental education levels, screen time exposure, obesity, insomnia, serum transferrin, decreased blood selenium, abnormal signals in brain functional MRI, interleukin-6, phosphodiesterase 2A, mitogen-activated protein kinase kinase 3, mitochondrial ribosomal protein L33, serotonin, and ASD. However, it does not support a causal relationship between parental rheumatoid arthritis, systemic lupus erythematosus, neonatal jaundice in children with ASD, cytomegalovirus infection, asthma, oral ulcers, vitamin D levels, and ASD. This article reviews the etiological factors related to ASD and MR studies, aiming to explore and deepen the understanding of the pathophysiology of ASD. It provides strong statistical support for the prevention, diagnosis, and treatment of ASD, and offers new methods and strategies for the etiological analysis of complex traits.


Subject(s)
Autism Spectrum Disorder , Mendelian Randomization Analysis , Humans , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/etiology
18.
Psychiatry Res ; 337: 115971, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788554

ABSTRACT

This study aimed to investigate the association between pre-pregnancy, prenatal and perinatal exposures to cannabis use disorder (CUD) and the risk of autism spectrum disoder (ASD) in offspring. Data were drawn from the New South Wales (NSW) Perinatal Data Collection (PDC), population-based, linked administrative health data encompassing all-live birth cohort from January 2003 to December 2005. This study involved 222 534 mother-offspring pairs. . The exposure variable (CUD) and the outcome of interest (ASD) were identified using the 10th international disease classification criteria, Australian Modified (ICD-10-AM). We found a three-fold increased risk of ASD in the offspring of mothers with maternal CUD compared to non-exposed offspring. In our sensitivity analyses, male offspring have a higher risk of ASD associated with maternal CUD than their female counterparts. In conclusion, exposure to maternal CUD is linked to a higher risk of ASD in offspring, with a stronger risk in male offspring. Further research is needed to understand these gender-specific effects and the relationship between maternal CUD and ASD risk in children.


Subject(s)
Autism Spectrum Disorder , Marijuana Abuse , Prenatal Exposure Delayed Effects , Humans , Female , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/etiology , Male , Pregnancy , Prenatal Exposure Delayed Effects/epidemiology , Adult , Cohort Studies , Marijuana Abuse/epidemiology , New South Wales/epidemiology , Child , Young Adult , Information Storage and Retrieval , Child, Preschool , Sex Factors
19.
Front Endocrinol (Lausanne) ; 15: 1381180, 2024.
Article in English | MEDLINE | ID: mdl-38752179

ABSTRACT

Background: The prevalence of autism spectrum disorder (ASD) has significantly risen in the past three decades, prompting researchers to explore the potential contributions of environmental factors during pregnancy to ASD development. One such factor of interest is gestational hypothyroxinemia (HTX), a frequent condition in pregnancy associated with cognitive impairments in the offspring. While retrospective human studies have linked gestational HTX to autistic traits, the cellular and molecular mechanisms underlying the development of ASD-like phenotypes remain poorly understood. This study used a mouse model of gestational HTX to evaluate ASD-like phenotypes in the offspring. Methods: To induce gestational HTX, pregnant mice were treated with 2-mercapto-1-methylimidazole (MMI), a thyroid hormones synthesis inhibitor, in the tap-drinking water from embryonic days (E) 10 to E14. A separate group received MMI along with a daily subcutaneous injection of T4, while the control group received regular tap water during the entire pregnancy. Female and male offspring underwent assessments for repetitive, anxious, and social behaviors from postnatal day (P) 55 to P64. On P65, mice were euthanized for the evaluation of ASD-related inflammatory markers in blood, spleen, and specific brain regions. Additionally, the expression of glutamatergic proteins (NLGN3 and HOMER1) was analyzed in the prefrontal cortex and hippocampus. Results: The HTX-offspring exhibited anxious-like behavior, a subordinate state, and impaired social interactions. Subsequently, both female and male HTX-offspring displayed elevated proinflammatory cytokines in blood, including IL-1ß, IL-6, IL-17A, and TNF-α, while only males showed reduced levels of IL-10. The spleen of HTX-offspring of both sexes showed increased Th17/Treg ratio and M1-like macrophages. In the prefrontal cortex and hippocampus of male HTX-offspring, elevated levels of IL-17A and reduced IL-10 were observed, accompanied by increased expression of hippocampal NLGN3 and HOMER1. All these observations were compared to those observed in the Control-offspring. Notably, the supplementation with T4 during the MMI treatment prevents the development of the observed phenotypes. Correlation analysis revealed an association between maternal T4 levels and specific ASD-like outcomes. Discussion: This study validates human observations, demonstrating for the first time that gestational HTX induces ASD-like phenotypes in the offspring, highlighting the need of monitoring thyroid function during pregnancy.


Subject(s)
Autism Spectrum Disorder , Prenatal Exposure Delayed Effects , Animals , Female , Pregnancy , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , Mice , Male , Prenatal Exposure Delayed Effects/metabolism , Phenotype , Behavior, Animal , Hypothyroidism/metabolism , Thyroxine/blood , Biomarkers/metabolism , Mice, Inbred C57BL , Pregnancy Complications/metabolism , Disease Models, Animal , Inflammation/metabolism , Social Behavior
20.
PLoS One ; 19(5): e0299380, 2024.
Article in English | MEDLINE | ID: mdl-38748694

ABSTRACT

Autism Spectrum Disorder (ASD) is a neurodevelopmental behavioral disorder characterized by social, communicative, and motor deficits. There is no single etiological cause for ASD, rather, there are various genetic and environmental factors that increase the risk for ASD. It is thought that some of these factors influence the same underlying neural mechanisms, and that an interplay of both genetic and environmental factors would better explain the pathogenesis of ASD. To better appreciate the influence of genetic-environment interaction on ASD-related behaviours, rats lacking a functional copy of the ASD-linked gene Cntnap2 were exposed to maternal immune activation (MIA) during pregnancy and assessed in adolescence and adulthood. We hypothesized that Cntnap2 deficiency interacts with poly I:C MIA to aggravate ASD-like symptoms in the offspring. In this double-hit model, we assessed attention, a core deficit in ASD due to prefrontal cortical dysfunction. We employed a well-established attentional paradigm known as the 5-choice serial reaction time task (5CSRTT). Cntnap2-/- rats exhibited greater perseverative responses which is indicative of repetitive behaviors. Additionally, rats exposed to poly I:C MIA exhibited premature responses, a marker of impulsivity. The rats exposed to both the genetic and environmental challenge displayed an increase in impulsive activity; however, this response was only elicited in the presence of an auditory distractor. This implies that exacerbated symptomatology in the double-hit model may situation-dependent and not generally expressed.


Subject(s)
Attention , Autism Spectrum Disorder , Disease Models, Animal , Gene-Environment Interaction , Nerve Tissue Proteins , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/etiology , Rats , Female , Attention/physiology , Pregnancy , Nerve Tissue Proteins/genetics , Male , Membrane Proteins/genetics , Poly I-C , Behavior, Animal , Prenatal Exposure Delayed Effects/genetics
SELECTION OF CITATIONS
SEARCH DETAIL