Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 184(7): 1740-1756.e16, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33705688

ABSTRACT

The core symptoms of many neurological disorders have traditionally been thought to be caused by genetic variants affecting brain development and function. However, the gut microbiome, another important source of variation, can also influence specific behaviors. Thus, it is critical to unravel the contributions of host genetic variation, the microbiome, and their interactions to complex behaviors. Unexpectedly, we discovered that different maladaptive behaviors are interdependently regulated by the microbiome and host genes in the Cntnap2-/- model for neurodevelopmental disorders. The hyperactivity phenotype of Cntnap2-/- mice is caused by host genetics, whereas the social-behavior phenotype is mediated by the gut microbiome. Interestingly, specific microbial intervention selectively rescued the social deficits in Cntnap2-/- mice through upregulation of metabolites in the tetrahydrobiopterin synthesis pathway. Our findings that behavioral abnormalities could have distinct origins (host genetic versus microbial) may change the way we think about neurological disorders and how to treat them.


Subject(s)
Gastrointestinal Microbiome , Locomotion , Social Behavior , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Biopterins/analogs & derivatives , Biopterins/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials , Fecal Microbiota Transplantation , Feces/microbiology , Limosilactobacillus reuteri/metabolism , Limosilactobacillus reuteri/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/microbiology , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/therapy , Principal Component Analysis , Psychomotor Agitation/pathology , Synaptic Transmission
2.
Cell ; 165(7): 1762-1775, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27315483

ABSTRACT

Maternal obesity during pregnancy has been associated with increased risk of neurodevelopmental disorders, including autism spectrum disorder (ASD), in offspring. Here, we report that maternal high-fat diet (MHFD) induces a shift in microbial ecology that negatively impacts offspring social behavior. Social deficits and gut microbiota dysbiosis in MHFD offspring are prevented by co-housing with offspring of mothers on a regular diet (MRD) and transferable to germ-free mice. In addition, social interaction induces synaptic potentiation (LTP) in the ventral tegmental area (VTA) of MRD, but not MHFD offspring. Moreover, MHFD offspring had fewer oxytocin immunoreactive neurons in the hypothalamus. Using metagenomics and precision microbiota reconstitution, we identified a single commensal strain that corrects oxytocin levels, LTP, and social deficits in MHFD offspring. Our findings causally link maternal diet, gut microbial imbalance, VTA plasticity, and behavior and suggest that probiotic treatment may relieve specific behavioral abnormalities associated with neurodevelopmental disorders. VIDEO ABSTRACT.


Subject(s)
Autism Spectrum Disorder/microbiology , Diet, High-Fat , Gastrointestinal Microbiome , Obesity/complications , Social Behavior , Animals , Dysbiosis/physiopathology , Female , Germ-Free Life , Housing, Animal , Limosilactobacillus reuteri , Male , Mice , Mice, Inbred C57BL , Oxytocin/analysis , Oxytocin/metabolism , Pregnancy , Ventral Tegmental Area
3.
Annu Rev Neurosci ; 37: 17-38, 2014.
Article in English | MEDLINE | ID: mdl-25032491

ABSTRACT

Activity-dependent changes in the strength of synaptic connections are fundamental to the formation and maintenance of memory. The mechanisms underlying persistent changes in synaptic strength in the hippocampus, specifically long-term potentiation and depression, depend on new protein synthesis. Such changes are thought to be orchestrated by engaging the signaling pathways that regulate mRNA translation in neurons. In this review, we discuss the key regulatory pathways that govern translational control in response to synaptic activity and the mRNA populations that are specifically targeted by these pathways. The critical contribution of regulatory control over new protein synthesis to proper cognitive function is underscored by human disorders associated with either silencing or mutation of genes encoding proteins that directly regulate translation. In light of these clinical implications, we also consider the therapeutic potential of targeting dysregulated translational control to treat cognitive disorders of synaptic dysfunction.


Subject(s)
Cognition Disorders/genetics , Cognition Disorders/physiopathology , Neuronal Plasticity/genetics , Protein Biosynthesis/genetics , Animals , Hippocampus/physiology , Humans , Models, Biological , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction/genetics
4.
Hum Mol Genet ; 24(9): 2662-72, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25634563

ABSTRACT

Rett syndrome (RTT) is a severe neurodevelopmental disorder that is usually caused by mutations in Methyl-CpG-binding Protein 2 (MECP2). Four of the eight common disease causing mutations in MECP2 are nonsense mutations and are responsible for over 35% of all cases of RTT. A strategy to overcome disease-causing nonsense mutations is treatment with nonsense mutation suppressing drugs that allow expression of full-length proteins from mutated genes with premature in-frame stop codons. To determine if this strategy is useful in RTT, we characterized a new mouse model containing a knock-in nonsense mutation (p.R255X) in the Mecp2 locus (Mecp2(R255X)). To determine whether the truncated gene product acts as a dominant negative allele and if RTT-like phenotypes could be rescued by expression of wild-type protein, we genetically introduced an extra copy of MECP2 via an MECP2 transgene. The addition of MECP2 transgene to Mecp2(R255X) mice abolished the phenotypic abnormalities and resulted in near complete rescue. Expression of MECP2 transgene Mecp2(R255X) allele also rescued mTORC1 signaling abnormalities discovered in mice with loss of function and overexpression of Mecp2. Finally, we treated Mecp2(R255X) embryonic fibroblasts with the nonsense mutation suppressing drug gentamicin and we were able to induce expression of full-length MeCP2 from the mutant p.R255X allele. These data provide proof of concept that the p.R255X mutation of MECP2 is amenable to the nonsense suppression therapeutic strategy and provide guidelines for the extent of rescue that can be expected by re-expressing MeCP2 protein.


Subject(s)
Alleles , Genetic Association Studies , Methyl-CpG-Binding Protein 2/genetics , Mutation , Phenotype , Amino Acid Substitution , Animals , Behavior, Animal , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Gentamicins/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Transgenic , Multiprotein Complexes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rett Syndrome/diagnosis , Rett Syndrome/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transgenes
5.
J Neurosci ; 33(14): 6191-202, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554500

ABSTRACT

The axon initial segment (AIS) and nodes of Ranvier are the sites of action potential initiation and regeneration in axons. Although the basic molecular architectures of AIS and nodes, characterized by dense clusters of Na(+) and K(+) channels, are similar, firing patterns vary among cell types. Neuronal firing patterns are established by the collective activity of voltage-gated ion channels and can be modulated through interaction with auxiliary subunits. Here, we report the neuronal expression pattern and subcellular localization of Navß4, the modulatory Na(+) channel subunit thought to underlie resurgent Na(+) current. Immunostaining of rat tissues revealed that Navß4 is strongly enriched at the AIS of a select set of neuron types, including many characterized by high-frequency firing, and at nodes of Ranvier in the PNS and some nodes in the CNS. By introducing full-length and mutant GFP-tagged Navß4 into cultured neurons, we determined that the AIS and nodal localization of Navß4 depends on its direct interaction with Na(+) channel α subunits through an extracellular disulfide bond. Based on these results, we propose that differences in the specific composition of the Na(+) channel complexes enriched at the AIS and nodes contribute to the diverse physiologies observed among cell types.


Subject(s)
Axons/metabolism , Brain/cytology , Neurons/cytology , Ranvier's Nodes/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/metabolism , Action Potentials/genetics , Animals , Ankyrins/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cysteine/metabolism , Embryo, Mammalian , Female , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Male , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Optical Imaging , Pregnancy , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Transfection , Voltage-Gated Sodium Channel beta Subunits/genetics , Voltage-Gated Sodium Channel beta Subunits/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/genetics
6.
medRxiv ; 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38562743

ABSTRACT

Type 2 diabetes (T2D) is a common forerunner of neurodegeneration and dementia, including Alzheimer's Disease (AD), yet the underlying mechanisms remain unresolved. Individuals of Mexican descent living in South Texas have increased prevalence of comorbid T2D and early onset AD, despite low incidence of the predisposing APOE-e4 variant and an absence of the phenotype among relatives residing in Mexico - suggesting a role for environmental factors in coincident T2D and AD susceptibility. Here, in a small clinical trial, we show dysbiosis of the human gut microbiome could contribute to neuroinflammation and risk for AD in this population. Divergent Gastrointestinal Symptom Rating Scale (GSRS) responses, despite no differences in expressed dietary preferences, provided the first evidence for altered gut microbial ecology among T2D subjects (sT2D) versus population-matched healthy controls (HC). Metataxonomic 16S rRNA sequencing of participant stool revealed a decrease in alpha diversity of sT2D versus HC gut communities and identified BMI as a driver of gut community structure. Linear discriminant analysis effect size (LEfSe) identified a significant decrease in the relative abundance of the short-chain fatty acid-producing taxa Lachnospiraceae, Faecalibacterium, and Alistipes and an increase in pathobionts Escherichia-Shigella, Enterobacter, and Clostridia innocuum among sT2D gut microbiota, as well as differentially abundant gene and metabolic pathways. These results suggest characterization of the gut microbiome of individuals with T2D could identify key actors among "disease state" microbiota which may increase risk for or accelerate the onset of neurodegeneration. Furthermore, they identify candidate microbiome-targeted approaches for prevention and treatment of neuroinflammation in AD.

7.
Prog Neurobiol ; 237: 102612, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642602

ABSTRACT

Recurrent seizures lead to accumulation of the activity-dependent transcription factor ∆FosB in hippocampal dentate granule cells in both mouse models of epilepsy and mouse models of Alzheimer's disease (AD), which is also associated with increased incidence of seizures. In patients with AD and related mouse models, the degree of ∆FosB accumulation corresponds with increasing severity of cognitive deficits. We previously found that ∆FosB impairs spatial memory in mice by epigenetically regulating expression of target genes such as calbindin that are involved in synaptic plasticity. However, the suppression of calbindin in conditions of neuronal hyperexcitability has been demonstrated to provide neuroprotection to dentate granule cells, indicating that ∆FosB may act over long timescales to coordinate neuroprotective pathways. To test this hypothesis, we used viral-mediated expression of ∆JunD to interfere with ∆FosB signaling over the course of several months in transgenic mice expressing mutant human amyloid precursor protein (APP), which exhibit spontaneous seizures and develop AD-related neuropathology and cognitive deficits. Our results demonstrate that persistent ∆FosB activity acts through discrete modes of hippocampal target gene regulation to modulate neuronal excitability, limit recurrent seizure activity, and provide neuroprotection to hippocampal dentate granule cells in APP mice.


Subject(s)
Amyloid beta-Protein Precursor , Dentate Gyrus , Mice, Transgenic , Proto-Oncogene Proteins c-fos , Seizures , Animals , Dentate Gyrus/metabolism , Mice , Seizures/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Neuroprotection/physiology , Disease Models, Animal , Alzheimer Disease/metabolism , Male , Mice, Inbred C57BL , Humans
8.
Mol Cell Neurosci ; 50(1): 1-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22445657

ABSTRACT

The inhibitor of NF-κB alpha (IκBα) protein is an important regulator of the transcription factor NF-κB. In neurons, IκBα has been shown to play a role in neurite outgrowth and cell survival. Recently, a phosphorylated form of IκBα (pIκBα Ser32/36) was reported to be highly enriched at the axon initial segment (AIS) and was proposed to function upstream of ankyrinG in AIS assembly, including ion channel recruitment. However, we report here that the AIS clustering of ankyrinG and Na(+) channels in the brains of IκBα knockout (Nfkbia(-/-)) mice is comparable to that in wild-type littermates. Furthermore, we found that multiple phospho-specific antibodies against pIκBα Ser32/36 non-specifically label AIS in Nfkbia(-/-) cortex and AIS in dissociated Nfkbia(-/-) hippocampal neurons. With the exception of ankyrinG, shRNA-mediated knockdown of known AIS proteins in cultured hippocampal neurons did not eliminate the AIS labeling with pIκBα antibodies. Instead, the pIκBα antibodies cross-react with a phosphorylated epitope of a protein associated with the microtubule-based AIS cytoskeleton that is not integrated into the AIS membrane complex organized by ankyrinG. Our results indicate that pIκBα is neither enriched at the AIS nor required for AIS assembly.


Subject(s)
Axons/metabolism , I-kappa B Proteins/metabolism , Animals , Ankyrins/antagonists & inhibitors , Ankyrins/metabolism , Cells, Cultured , I-kappa B Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha , Neurons/cytology , Neurons/metabolism , Phosphorylation , RNA, Small Interfering , Rats , Rats, Sprague-Dawley
9.
STAR Protoc ; 4(3): 102386, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37379217

ABSTRACT

Dysbiosis of the gut microbiome is implicated in the growing burden of non-communicable chronic diseases, including neurodevelopmental disorders, and both preclinical and clinical studies highlight the potential for precision probiotic therapies in their prevention and treatment. Here, we present an optimized protocol for the preparation and administration of Limosilactobacillus reuteri MM4-1A (ATCC-PTA-6475) to adolescent mice. We also describe steps for performing downstream analysis of metataxonomic sequencing data with careful assessment of sex-specific effects on microbiome composition and structure. For complete details on the use and execution of this protocol, please refer to Di Gesù et al.1.


Subject(s)
Gastrointestinal Microbiome , Limosilactobacillus reuteri , Microbiota , Probiotics , Male , Female , Animals , Mice , Probiotics/pharmacology , Probiotics/therapeutic use
10.
J Neurosci ; 31(48): 17637-48, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-22131424

ABSTRACT

The axon initial segment (AIS) is the site of action potential initiation in neurons. Recent studies have demonstrated activity-dependent regulation of the AIS, including homeostatic changes in AIS length, membrane excitability, and the localization of voltage-gated Na(+) channels. The neurodevelopmental disorder Angelman syndrome (AS) is usually caused by the deletion of small portions of the maternal copy of chromosome 15, which includes the UBE3A gene. A mouse model of AS has been generated and these mice exhibit multiple neurological abnormalities similar to those observed in humans. We examined intrinsic properties of pyramidal neurons in hippocampal area CA1 from AS model mice and observed alterations in resting membrane potential, threshold potential, and action potential amplitude. The altered intrinsic properties in the AS mice were correlated with significant increases in the expression of the α1 subunit of Na/K-ATPase (α1-NaKA), the Na(+) channel NaV1.6, and the AIS anchoring protein ankyrin-G, as well as an increase in length of the AIS. These findings are the first evidence for pathology of intrinsic membrane properties and AIS-specific changes in AS, a neurodevelopmental disorder associated with autism.


Subject(s)
Action Potentials/physiology , Angelman Syndrome/physiopathology , Axons/physiology , Membrane Potentials/physiology , Pyramidal Cells/physiology , Action Potentials/genetics , Angelman Syndrome/genetics , Animals , Disease Models, Animal , Hippocampus/physiopathology , Membrane Potentials/genetics , Mice
11.
Cell Rep ; 41(2): 111461, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36223744

ABSTRACT

Dysbiosis of the maternal gut microbiome during pregnancy is associated with adverse neurodevelopmental outcomes. We previously showed that maternal high-fat diet (MHFD) in mice induces gut dysbiosis, social dysfunction, and underlying synaptic plasticity deficits in male offspring (F1). Here, we reason that, if HFD-mediated changes in maternal gut microbiota drive offspring social deficits, then MHFD-induced dysbiosis in F1 female MHFD offspring would likewise impair F2 social behavior. Metataxonomic sequencing reveals reduced microbial richness among female F1 MHFD offspring. Despite recovery of microbial richness among MHFD-descendant F2 mice, they display social dysfunction. Post-weaning Limosilactobacillus reuteri treatment increases the abundance of short-chain fatty acid-producing taxa and rescues MHFD-descendant F2 social deficits. L. reuteri exerts a sexually dimorphic impact on gut microbiota configuration, increasing discriminant taxa between female cohorts. Collectively, these results show multigenerational impacts of HFD-induced dysbiosis in the maternal lineage and highlight the potential of maternal microbiome-targeted interventions for neurodevelopmental disorders.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Animals , Diet, High-Fat/adverse effects , Dysbiosis , Female , Male , Mice , Pregnancy , Social Behavior
12.
Eur J Neurosci ; 34(10): 1609-19, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22103418

ABSTRACT

The axon initial segment (AIS), with its dense clusters of voltage-gated ion channels decorating the axonal membrane, regulates action potential initiation and modulation. The AIS also functions as a barrier to maintain axodendritic polarity, and its precise axonal location contributes to the fine-tuning of neuronal excitability. Therefore, it is not surprising that mutations in AIS-related genes, disruption of the molecular organization of the AIS and altered AIS ion channel expression, function, location and/or density are emerging as key players in neurological disorders. Here, we consider the role of the AIS in nervous system disease and injury.


Subject(s)
Axons , Mental Disorders , Nervous System Diseases , Nervous System , Action Potentials/physiology , Aging/physiology , Animals , Ankyrins/metabolism , Axons/physiology , Axons/ultrastructure , Cell Polarity , Humans , Ion Channels/metabolism , Mental Disorders/pathology , Mental Disorders/physiopathology , Nervous System/pathology , Nervous System/physiopathology , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Neurons/ultrastructure
13.
Neurosci Res ; 168: 3-19, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33992660

ABSTRACT

The maternal gut microbiome plays a critical role in fetal and early postnatal development, shaping fundamental processes including immune maturation and brain development, among others. Consequently, it also contributes to fetal programming of health and disease. Over the last decade, epidemiological studies and work in preclinical animal models have begun to uncover a link between dysbiosis of the maternal gut microbiome and neurodevelopmental disorders in offspring. Neurodevelopmental disorders are caused by both genetic and environmental factors, and their interactions; however, clinical heterogeneity, phenotypic variability, and comorbidities make identification of underlying mechanisms difficult. Among environmental factors, exposure to maternal obesity in utero confers a significant increase in risk for neurodevelopmental disorders. Obesogenic diets in humans, non-human primates, and rodents induce functional modifications in maternal gut microbiome composition, which animal studies suggest are causally related to adverse mental health outcomes in offspring. Here, we review evidence linking maternal diet-induced gut dysbiosis to neurodevelopmental disorders and discuss how it could affect pre- and early postnatal brain development. We are hopeful that this burgeoning field of research will revolutionize antenatal care by leading to accessible prophylactic strategies, such as prenatal probiotics, to improve mental health outcomes in children affected by maternal diet-induced obesity.


Subject(s)
Gastrointestinal Microbiome , Neurodevelopmental Disorders , Animals , Diet , Dysbiosis , Female , Humans , Pregnancy , Primates
14.
Front Microbiol ; 12: 745469, 2021.
Article in English | MEDLINE | ID: mdl-34899632

ABSTRACT

Background: Bacteroidetes are the most common bacterial phylum in the mammalian intestine and the effects of several Bacteroides spp. on multiple facets of host physiology have been previously described. Of the Bacteroides spp., Bacteroides ovatus has recently garnered attention due to its beneficial effects in the context of intestinal inflammation. In this study, we aimed to examine model host intestinal physiological conditions and dietary modifications to characterize their effects on B. ovatus growth. Methods and Results: Using Biolog phenotypic microarrays, we evaluated 62 primary carbon sources and determined that B. ovatus ATCC 8384 can use the following carbohydrates as primary carbon sources: 10 disaccharides, 4 trisaccharides, 4 polysaccharides, 4 polymers, 3 L-linked sugars, 6 D-linked sugars, 5 amino-sugars, 6 alcohol sugars, and 15 organic acids. Proteomic profiling of B. ovatus bacteria revealed that a significant portion of the B. ovatus proteome contains proteins important for metabolism. Among the proteins, we found glycosyl hydrolase (GH) familes GH2, GH5, GH20, GH 43, GH88, GH92, and GH95. We also identified multiple proteins with antioxidant properties and reasoned that these proteins may support B. ovatus growth in the GI tract. Upon further testing, we showed that B. ovatus grew robustly in various pH, osmolarity, bile, ethanol, and H2O2 concentrations; indicating that B. ovatus is a well-adapted gut microbe. Conclusion: Taken together, we have demonstrated that key host and diet-derived changes in the intestinal environment influence B. ovatus growth. These data provide the framework for future work toward understanding how diet and lifestyle interventions may promote a beneficial environment for B. ovatus growth.

15.
Neuroscience ; 463: 272-287, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33811940

ABSTRACT

Opioid use by women during pregnancy has risen dramatically since 2004, accompanied by a striking increase in the prevalence of neonatal opioid withdrawal syndrome (NOWS) and other long-term neurological deficits. However, the mechanisms underlying the impact of prenatal opioid exposure on fetal neurodevelopment are largely unknown. To translate from the clinical presentation, we developed a novel mouse model to study the neurodevelopmental consequences of maternal opioid use and management. Female mice were treated with oxycodone (OXY) before mating to mimic opioid use disorder (OUD) in humans. Following pregnancy confirmation, dams were switched to buprenorphine (BUP) via oral administration, simulating medication management of OUD (MOUD) in pregnant women. Here, we document critical changes in fetal brain development including reduced cortical thickness, altered corticogenesis, and ventriculomegaly in embryos from dams that were treated with opioids before and throughout pregnancy. Maternal care giving behavior was slightly altered without affecting gross growth of offspring. However, adolescent offspring exposed to maternal opioid use during pregnancy exhibited hyperactivity in late adolescence. Remarkably, we also show increased generation of dopaminergic neurons within the ventral tegmental area (VTA) of mice exposed to prenatal opioids. These data provide critical evidence of teratogenic effects of opioid use during pregnancy and suggest a causal relationship between maternal opioid use and neurodevelopmental/behavioral anomalies in adolescence.


Subject(s)
Buprenorphine , Neonatal Abstinence Syndrome , Opioid-Related Disorders , Prenatal Exposure Delayed Effects , Adolescent , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/toxicity , Animals , Buprenorphine/therapeutic use , Female , Humans , Infant, Newborn , Mice , Neonatal Abstinence Syndrome/drug therapy , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy
16.
Neuron ; 101(2): 246-259.e6, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30522820

ABSTRACT

Currently, there are no medications that effectively treat the core symptoms of Autism Spectrum Disorder (ASD). We recently found that the bacterial species Lactobacillus (L.) reuteri reverses social deficits in maternal high-fat-diet offspring. However, whether the effect of L. reuteri on social behavior is generalizable to other ASD models and its mechanism(s) of action remains unknown. Here, we found that treatment with L. reuteri selectively rescues social deficits in genetic, environmental, and idiopathic ASD models. Interestingly, the effects of L. reuteri on social behavior are not mediated by restoring the composition of the host's gut microbiome, which is altered in all of these ASD models. Instead, L. reuteri acts in a vagus nerve-dependent manner and rescues social interaction-induced synaptic plasticity in the ventral tegmental area of ASD mice, but not in oxytocin receptor-deficient mice. Collectively, treatment with L. reuteri emerges as promising non-invasive microbial-based avenue to combat ASD-related social dysfunction.


Subject(s)
Autism Spectrum Disorder/complications , Limosilactobacillus reuteri/physiology , Social Behavior Disorders/etiology , Social Behavior Disorders/therapy , Action Potentials/drug effects , Action Potentials/genetics , Animals , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/genetics , Benzoxazines/administration & dosage , Capillary Permeability/drug effects , Capillary Permeability/genetics , Central Nervous System Stimulants/pharmacology , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Exploratory Behavior/physiology , Gastrointestinal Microbiome/physiology , Humans , Interpersonal Relations , Limosilactobacillus reuteri/genetics , Male , Mice , Mice, Transgenic , Microfilament Proteins , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Picrotoxin/pharmacology , Piperidines/administration & dosage , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Smell/physiology , Valproic Acid/toxicity
17.
Neuron ; 104(4): 665-679.e8, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31585809

ABSTRACT

In humans, disruption of nonsense-mediated decay (NMD) has been associated with neurodevelopmental disorders (NDDs) such as autism spectrum disorder and intellectual disability. However, the mechanism by which deficient NMD leads to neurodevelopmental dysfunction remains unknown, preventing development of targeted therapies. Here we identified novel protein-coding UPF2 (UP-Frameshift 2) variants in humans with NDD, including speech and language deficits. In parallel, we found that mice lacking Upf2 in the forebrain (Upf2 fb-KO mice) show impaired NMD, memory deficits, abnormal long-term potentiation (LTP), and social and communication deficits. Surprisingly, Upf2 fb-KO mice exhibit elevated expression of immune genes and brain inflammation. More importantly, treatment with two FDA-approved anti-inflammatory drugs reduced brain inflammation, restored LTP and long-term memory, and reversed social and communication deficits. Collectively, our findings indicate that impaired UPF2-dependent NMD leads to neurodevelopmental dysfunction and suggest that anti-inflammatory agents may prove effective for treatment of disorders with impaired NMD.


Subject(s)
Learning/physiology , Memory/physiology , Nonsense Mediated mRNA Decay/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Animals , Child , Drosophila , Female , Humans , Language Development Disorders/genetics , Male , Mice , Mice, Knockout , RNA-Binding Proteins/metabolism
19.
Nat Neurosci ; 17(8): 1073-82, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24974795

ABSTRACT

At hippocampal synapses, activation of group I metabotropic glutamate receptors (mGluRs) induces long-term depression (LTD), which requires new protein synthesis. However, the underlying mechanism remains elusive. Here we describe the translational program that underlies mGluR-LTD and identify the translation factor eIF2α as its master effector. Genetically reducing eIF2α phosphorylation, or specifically blocking the translation controlled by eIF2α phosphorylation, prevented mGluR-LTD and the internalization of surface AMPA receptors (AMPARs). Conversely, direct phosphorylation of eIF2α, bypassing mGluR activation, triggered a sustained LTD and removal of surface AMPARs. Combining polysome profiling and RNA sequencing, we identified the mRNAs translationally upregulated during mGluR-LTD. Translation of one of these mRNAs, oligophrenin-1, mediates the LTD induced by eIF2α phosphorylation. Mice deficient in phospho-eIF2α-mediated translation are impaired in object-place learning, a behavioral task that induces hippocampal mGluR-LTD in vivo. Our findings identify a new model of mGluR-LTD, which promises to be of value in the treatment of mGluR-LTD-linked cognitive disorders.


Subject(s)
Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Learning/physiology , Long-Term Synaptic Depression/genetics , Protein Biosynthesis , Receptors, AMPA/physiology , Receptors, Metabotropic Glutamate/metabolism , Animals , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/genetics , Receptors, AMPA/deficiency , Space Perception/physiology
20.
Cell Rep ; 4(3): 405-12, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911285

ABSTRACT

Angelman syndrome (AS) is associated with symptoms that include autism, intellectual disability, motor abnormalities, and epilepsy. We recently showed that AS model mice have increased expression of the alpha1 subunit of Na/K-ATPase (α1-NaKA) in the hippocampus, which was correlated with increased expression of axon initial segment (AIS) proteins. Our developmental analysis revealed that the increase in α1-NaKA expression preceded that of the AIS proteins. Therefore, we hypothesized that α1-NaKA overexpression drives AIS abnormalities and that by reducing its expression these and other phenotypes could be corrected in AS model mice. Herein, we report that the genetic normalization of α1-NaKA levels in AS model mice corrects multiple hippocampal phenotypes, including alterations in the AIS, aberrant intrinsic membrane properties, impaired synaptic plasticity, and memory deficits. These findings strongly suggest that increased expression of α1-NaKA plays an important role in a broad range of abnormalities in the hippocampus of AS model mice.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/pathology , Hippocampus/metabolism , Hippocampus/pathology , Sodium-Potassium-Exchanging ATPase/genetics , Angelman Syndrome/enzymology , Angelman Syndrome/metabolism , Animals , Ankyrins/biosynthesis , Disease Models, Animal , Female , Hippocampus/enzymology , Male , Mice , Mice, Inbred C57BL , NAV1.6 Voltage-Gated Sodium Channel/biosynthesis , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Protein Subunits , Sodium-Potassium-Exchanging ATPase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL