Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 25(4): 598-606, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38565970

ABSTRACT

The intricate relationship between immune dysregulation and neurodevelopmental disorders (NDDs) has been observed across the stages of both prenatal and postnatal development. In this Review, we provide a comprehensive overview of various maternal immune conditions, ranging from infections to chronic inflammatory conditions, that impact the neurodevelopment of the fetus during pregnancy. Furthermore, we examine the presence of immunological phenotypes, such as immune-related markers and coexisting immunological disorders, in individuals with NDDs. By delving into these findings, we shed light on the potential underlying mechanisms responsible for the high occurrence of immune dysregulation alongside NDDs. We also discuss current mouse models of NDDs and their contributions to our understanding of the immune mechanisms underlying these diseases. Additionally, we discuss how neuroimmune interactions contribute to shaping the manifestation of neurological phenotypes in individuals with NDDs while also exploring potential avenues for mitigating these effects.


Subject(s)
Neurodevelopmental Disorders , Neuroimmunomodulation , Pregnancy , Animals , Female , Mice , Neurodevelopmental Disorders/genetics , Disease Models, Animal
2.
medRxiv ; 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38370801

ABSTRACT

Pregnancy is a risk factor for increased severity of SARS-CoV-2 and other respiratory infections. The mechanisms underlying this risk have not been well-established, partly due to a limited understanding of how pregnancy shapes immune responses. To gain insight into the role of pregnancy in modulating immune responses at steady state and upon perturbation, we collected peripheral blood mononuclear cells (PBMC), plasma, and stool from 226 women, including 152 pregnant individuals (n = 96 with SARS-CoV-2 infection and n = 56 healthy controls) and 74 non-pregnant women (n = 55 with SARS-CoV-2 and n = 19 healthy controls). We found that SARS-CoV-2 infection was associated with altered T cell responses in pregnant compared to non-pregnant women. Differences included a lower percentage of memory T cells, a distinct clonal expansion of CD4-expressing CD8 + T cells, and the enhanced expression of T cell exhaustion markers, such as programmed cell death-1 (PD-1) and T cell immunoglobulin and mucin domain-3 (Tim-3), in pregnant women. We identified additional evidence of immune dysfunction in severely and critically ill pregnant women, including a lack of expected elevation in regulatory T cell (Treg) levels, diminished interferon responses, and profound suppression of monocyte function. Consistent with earlier data, we found maternal obesity was also associated with altered immune responses to SARS-CoV-2 infection, including enhanced production of inflammatory cytokines by T cells. Certain gut bacterial species were altered in pregnancy and upon SARS-CoV-2 infection in pregnant individuals compared to non-pregnant women. Shifts in cytokine and chemokine levels were also identified in the sera of pregnant individuals, most notably a robust increase of interleukin-27 (IL-27), a cytokine known to drive T cell exhaustion, in the pregnant uninfected control group compared to all non-pregnant groups. IL-27 levels were also significantly higher in uninfected pregnant controls compared to pregnant SARS-CoV-2-infected individuals. Using two different preclinical mouse models of inflammation-induced fetal demise and respiratory influenza viral infection, we found that enhanced IL-27 protects developing fetuses from maternal inflammation but renders adult female mice vulnerable to viral infection. These combined findings from human and murine studies reveal nuanced pregnancy-associated immune responses, suggesting mechanisms underlying the increased susceptibility of pregnant individuals to viral respiratory infections.

3.
Curr Opin Neurobiol ; 80: 102708, 2023 06.
Article in English | MEDLINE | ID: mdl-36947942

ABSTRACT

Cytokines are small, secreted proteins that are known for their roles in the immune system. An accumulating body of evidence indicates that cytokines also work as neuromodulators in the central nervous system (CNS). Cytokines can access the CNS through multiple routes to directly impact neurons. The neuromodulatory effects of cytokines maintain the overall homeostasis of neural networks. In addition, cytokines regulate a diverse repertoire of behaviors both at a steady state and in inflammatory conditions by acting on discrete brain regions and neural networks. In this review, we discuss recent findings that provide insight into how combinatorial codes of cytokines might mediate neuro-immune communications to orchestrate functional responses of the brain to changes in immunological milieus.


Subject(s)
Central Nervous System , Cytokines , Brain/physiology , Neurons , Neuroimmunomodulation
4.
Neuron ; 111(3): 299-301, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36731428

ABSTRACT

In this issue of Neuron, Kim et al.1 demonstrate that theta oscillations between the right ACC and BLA are critical for observational fear and identify a role for the hippocampus in modulating these oscillations. They further show that theta oscillations are specifically involved in vicarious rather than directly experienced fear.


Subject(s)
Hippocampus , Theta Rhythm , Theta Rhythm/physiology , Hippocampus/physiology , Fear/physiology
5.
Immunity ; 55(4): 586-588, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35417672

ABSTRACT

The impact of intestinal fungi on host physiology and their mechanisms of interaction are incompletely understood. In a recent issue of Cell, Leonardi et al. (2022) showed that mucosal fungi induce intestinal Th17 cells to produce IL-22 and IL-17A. IL-22 acts on the gut epithelium to protect barrier integrity, whereas IL-17 acts on IL-17RA+ neurons to enhance sociability.


Subject(s)
Receptors, Interleukin-17 , Th17 Cells , Fungi
6.
Trends Immunol ; 43(3): 230-244, 2022 03.
Article in English | MEDLINE | ID: mdl-35131181

ABSTRACT

Exposure to heightened inflammation in pregnancy caused by infections or other inflammatory insults has been associated with the onset of neurodevelopmental and psychiatric disorders in children. Rodent models have provided unique insights into how this maternal immune activation (MIA) disrupts brain development. Here, we discuss the key immune factors involved, highlight recent advances in determining the molecular and cellular pathways of MIA, and review how the maternal immune system affects fetal development. We also examine the roles of microbiomes in shaping maternal immune function and the development of autism-like phenotypes. A comprehensive understanding of the gut bacteria-immune-neuro interaction in MIA is essential for developing diagnostic and therapeutic measures for high-risk pregnant women and identifying targets for treating inflammation-induced neurodevelopmental disorders.


Subject(s)
Microbiota , Prenatal Exposure Delayed Effects , Animals , Disease Models, Animal , Female , Humans , Immune System , Inflammation/immunology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology
7.
Immunity ; 55(1): 145-158.e7, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34879222

ABSTRACT

Children with autism spectrum disorders often display dysregulated immune responses and related gastrointestinal symptoms. However, the underlying mechanisms leading to the development of both phenotypes have not been elucidated. Here, we show that mouse offspring exhibiting autism-like phenotypes due to prenatal exposure to maternal inflammation were more susceptible to developing intestinal inflammation following challenges later in life. In contrast to its prenatal role in neurodevelopmental phenotypes, interleukin-17A (IL-17A) generated immune-primed phenotypes in offspring through changes in the maternal gut microbiota that led to postnatal alterations in the chromatin landscape of naive CD4+ T cells. The transfer of stool samples from pregnant mice with enhanced IL-17A responses into germ-free dams produced immune-primed phenotypes in offspring. Our study provides mechanistic insights into why children exposed to heightened inflammation in the womb might have an increased risk of developing inflammatory diseases in addition to neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder/immunology , CD4-Positive T-Lymphocytes/immunology , Chromatin/metabolism , Gastrointestinal Microbiome/immunology , Inflammation/immunology , Interleukin-17/metabolism , Intestines/immunology , Neurodevelopmental Disorders/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Autism Spectrum Disorder/microbiology , Child , Disease Models, Animal , Fecal Microbiota Transplantation , Female , Humans , Immunization , Inflammation/microbiology , Mice , Neurodevelopmental Disorders/microbiology , Pregnancy , Prenatal Exposure Delayed Effects/microbiology
8.
Nat Commun ; 12(1): 3435, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103511

ABSTRACT

To understand the underlying mechanisms of progressive neurophysiological phenomena, neural interfaces should interact bi-directionally with brain circuits over extended periods of time. However, such interfaces remain limited by the foreign body response that stems from the chemo-mechanical mismatch between the probes and the neural tissues. To address this challenge, we developed a multifunctional sensing and actuation platform consisting of multimaterial fibers intimately integrated within a soft hydrogel matrix mimicking the brain tissue. These hybrid devices possess adaptive bending stiffness determined by the hydration states of the hydrogel matrix. This enables their direct insertion into the deep brain regions, while minimizing tissue damage associated with the brain micromotion after implantation. The hydrogel hybrid devices permit electrophysiological, optogenetic, and behavioral studies of neural circuits with minimal foreign body responses and tracking of stable isolated single neuron potentials in freely moving mice over 6 months following implantation.


Subject(s)
Biosensing Techniques , Hydrogels/chemistry , Molecular Probes/chemistry , Neurons/physiology , Action Potentials/physiology , Animals , Behavior, Animal , Biological Assay , Brain/physiology , Electrophysiological Phenomena , Foreign-Body Reaction/physiopathology , Male , Mice, Inbred C57BL , Optogenetics , Stress, Mechanical , Time Factors
9.
Nature ; 593(7857): 114-118, 2021 05.
Article in English | MEDLINE | ID: mdl-33790466

ABSTRACT

Innate social behaviours, such as mating and fighting, are fundamental to animal reproduction and survival1. However, social engagements can also put an individual at risk2. Little is known about the neural mechanisms that enable appropriate risk assessment and the suppression of hazardous social interactions. Here we identify the posteromedial nucleus of the cortical amygdala (COApm) as a locus required for the suppression of male mating when a female mouse is unhealthy. Using anatomical tracing, functional imaging and circuit-level epistatic analyses, we show that suppression of mating with an unhealthy female is mediated by the COApm projections onto the glutamatergic population of the medial amygdalar nucleus (MEA). We further show that the role of the COApm-to-MEA connection in regulating male mating behaviour relies on the neuromodulator thyrotropin-releasing hormone (TRH). TRH is expressed in the COApm, whereas the TRH receptor (TRHR) is found in the postsynaptic MEA glutamatergic neurons. Manipulating neural activity of TRH-expressing neurons in the COApm modulated male mating behaviour. In the MEA, activation of the TRHR pathway by ligand infusion inhibited mating even towards healthy female mice, whereas genetic ablation of TRHR facilitated mating with unhealthy individuals. In summary, we reveal a neural pathway that relies on the neuromodulator TRH to modulate social interactions according to the health status of the reciprocating individual. Individuals must balance the cost of social interactions relative to the benefit, as deficits in the ability to select healthy mates may lead to the spread of disease.


Subject(s)
Amygdala/cytology , Amygdala/physiology , Mating Preference, Animal/physiology , Neural Pathways/physiology , Social Behavior , Animals , Copulation/physiology , Corticomedial Nuclear Complex/cytology , Corticomedial Nuclear Complex/metabolism , Female , Glutamic Acid/metabolism , Health , Ligands , Lipopolysaccharides/pharmacology , Male , Mice , Neurons/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism
10.
Nat Neurosci ; 24(2): 204-213, 2021 02.
Article in English | MEDLINE | ID: mdl-33361822

ABSTRACT

Maternal infection and inflammation during pregnancy are associated with neurodevelopmental disorders in offspring, but little is understood about the molecular mechanisms underlying this epidemiologic phenomenon. Here, we leveraged single-cell RNA sequencing to profile transcriptional changes in the mouse fetal brain in response to maternal immune activation (MIA) and identified perturbations in cellular pathways associated with mRNA translation, ribosome biogenesis and stress signaling. We found that MIA activates the integrated stress response (ISR) in male, but not female, MIA offspring in an interleukin-17a-dependent manner, which reduced global mRNA translation and altered nascent proteome synthesis. Moreover, blockade of ISR activation prevented the behavioral abnormalities as well as increased cortical neural activity in MIA male offspring. Our data suggest that sex-specific activation of the ISR leads to maternal inflammation-associated neurodevelopmental disorders.


Subject(s)
Brain/immunology , Fetus/immunology , Immunity, Innate/genetics , Proteostasis/genetics , Animals , Behavior, Animal , Developmental Disabilities/genetics , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Pregnancy , Protein Biosynthesis/genetics , Proteome/biosynthesis , RNA/biosynthesis , RNA/genetics , RNA, Small Interfering , Sex Characteristics , Signal Transduction , Stress, Psychological/genetics , Stress, Psychological/psychology
11.
Nature ; 577(7789): 249-253, 2020 01.
Article in English | MEDLINE | ID: mdl-31853066

ABSTRACT

A subset of children with autism spectrum disorder appear to show an improvement in their behavioural symptoms during the course of a fever, a sign of systemic inflammation1,2. Here we elucidate the molecular and neural mechanisms that underlie the beneficial effects of inflammation on social behaviour deficits in mice. We compared an environmental model of neurodevelopmental disorders in which mice were exposed to maternal immune activation (MIA) during embryogenesis3,4 with mouse models that are genetically deficient for contactin-associated protein-like 2 (Cntnap2)5, fragile X mental retardation-1 (Fmr1)6 or Sh3 and multiple ankyrin repeat domains 3 (Shank3)7. We establish that the social behaviour deficits in offspring exposed to MIA can be temporarily rescued by the inflammatory response elicited by the administration of lipopolysaccharide (LPS). This behavioural rescue was accompanied by a reduction in neuronal activity in the primary somatosensory cortex dysgranular zone (S1DZ), the hyperactivity of which was previously implicated in the manifestation of behavioural phenotypes associated with offspring exposed to MIA8. By contrast, we did not observe an LPS-induced rescue of social deficits in the monogenic models. We demonstrate that the differences in responsiveness to the LPS treatment between the MIA and the monogenic models emerge from differences in the levels of cytokine production. LPS treatment in monogenic mutant mice did not induce amounts of interleukin-17a (IL-17a) comparable to those induced in MIA offspring; bypassing this difference by directly delivering IL-17a into S1DZ was sufficient to promote sociability in monogenic mutant mice as well as in MIA offspring. Conversely, abrogating the expression of IL-17 receptor subunit a (IL-17Ra) in the neurons of the S1DZ eliminated the ability of LPS to reverse the sociability phenotypes in MIA offspring. Our data support a neuroimmune mechanism that underlies neurodevelopmental disorders in which the production of IL-17a during inflammation can ameliorate the expression of social behaviour deficits by directly affecting neuronal activity in the central nervous system.


Subject(s)
Interleukin-17/immunology , Neurodevelopmental Disorders/immunology , Animals , Behavior, Animal , Disease Models, Animal , Female , Fragile X Mental Retardation Protein , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects , Social Behavior
12.
Neuron ; 98(3): 462-463, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29723498

ABSTRACT

In this issue of Neuron, Keum et al. (2018) identify a Nrxn3 variant that produces an enhancement of observational fear learning. Results suggest that Nrxn3 loss of function, specifically within somatostatin-positive interneurons of the anterior cingulate cortex, is responsible.


Subject(s)
Empathy , Fear , Animals , Gyrus Cinguli , Interneurons , Learning , Mice
13.
Neuron ; 98(4): 736-742.e3, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29706585

ABSTRACT

Neurons in piriform cortex receive input from a random collection of glomeruli, resulting in odor representations that lack the stereotypic organization of the olfactory bulb. We have performed in vivo optical imaging and mathematical modeling to demonstrate that correlations are retained in the transformation from bulb to piriform cortex, a feature essential for generalization across odors. Random connectivity also implies that the piriform representation of a given odor will differ among different individuals and across brain hemispheres in a single individual. We show that these different representations can nevertheless support consistent agreement about odor quality across a range of odors. Our model also demonstrates that, whereas odor discrimination and categorization require far fewer neurons than reside in piriform cortex, consistent generalization may require the full complement of piriform neurons.


Subject(s)
Neurons/physiology , Olfactory Bulb/physiology , Olfactory Perception/physiology , Piriform Cortex/physiology , Animals , Calcium/metabolism , Drosophila , Functional Laterality , Generalization, Psychological , Intravital Microscopy , Mice , Models, Theoretical , Mushroom Bodies/cytology , Mushroom Bodies/metabolism , Mushroom Bodies/physiology , Neurons/cytology , Neurons/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Olfactory Pathways/physiology , Optical Imaging , Piriform Cortex/cytology , Piriform Cortex/metabolism
14.
Nature ; 549(7673): 482-487, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28902835

ABSTRACT

Viral infection during pregnancy is correlated with increased frequency of neurodevelopmental disorders, and this is studied in mice prenatally subjected to maternal immune activation (MIA). We previously showed that maternal T helper 17 cells promote the development of cortical and behavioural abnormalities in MIA-affected offspring. Here we show that cortical abnormalities are preferentially localized to a region encompassing the dysgranular zone of the primary somatosensory cortex (S1DZ). Moreover, activation of pyramidal neurons in this cortical region was sufficient to induce MIA-associated behavioural phenotypes in wild-type animals, whereas reduction in neural activity rescued the behavioural abnormalities in MIA-affected offspring. Sociability and repetitive behavioural phenotypes could be selectively modulated according to the efferent targets of S1DZ. Our work identifies a cortical region primarily, if not exclusively, centred on the S1DZ as the major node of a neural network that mediates behavioural abnormalities observed in offspring exposed to maternal inflammation.


Subject(s)
Behavior, Animal , Inflammation/physiopathology , Mental Disorders/etiology , Pregnancy Complications, Infectious/physiopathology , Prenatal Exposure Delayed Effects/psychology , Th17 Cells , Animals , Female , Male , Mental Disorders/psychology , Mice , Mothers , Phenotype , Pregnancy , Pyramidal Cells/pathology , Pyramidal Cells/physiology , Social Behavior , Somatosensory Cortex/abnormalities , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Th17 Cells/physiology
15.
Nature ; 549(7673): 528-532, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28902840

ABSTRACT

Maternal immune activation (MIA) contributes to behavioural abnormalities associated with neurodevelopmental disorders in both primate and rodent offspring. In humans, epidemiological studies suggest that exposure of fetuses to maternal inflammation increases the likelihood of developing autism spectrum disorder. In pregnant mice, interleukin-17a (IL-17a) produced by T helper 17 (TH17) cells (CD4+ T helper effector cells involved in multiple inflammatory conditions) induces behavioural and cortical abnormalities in the offspring exposed to MIA. However, it is unclear whether other maternal factors are required to promote MIA-associated phenotypes. Moreover, the underlying mechanisms by which MIA leads to T cell activation with increased IL-17a in the maternal circulation are not well understood. Here we show that MIA phenotypes in offspring require maternal intestinal bacteria that promote TH17 cell differentiation. Pregnant mice that had been colonized with mouse commensal segmented filamentous bacteria or human commensal bacteria that induce intestinal TH17 cells were more likely to produce offspring with MIA-associated abnormalities. We also show that small intestine dendritic cells from pregnant, but not from non-pregnant, females secrete IL-1ß, IL-23 and IL-6 and stimulate T cells to produce IL-17a upon exposure to MIA. Overall, our data suggest that defined gut commensal bacteria with a propensity to induce TH17 cells may increase the risk of neurodevelopmental disorders in the offspring of pregnant mothers undergoing immune system activation owing to infections or autoinflammatory syndromes.


Subject(s)
Gastrointestinal Microbiome/immunology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/microbiology , Animals , Behavior, Animal , Dendritic Cells/immunology , Female , Inflammation/immunology , Inflammation/microbiology , Interleukin-17/immunology , Interleukin-1beta/immunology , Interleukin-23/immunology , Interleukin-6/immunology , Intestine, Small/cytology , Intestine, Small/immunology , Intestine, Small/microbiology , Male , Mice , Phenotype , Pregnancy , Symbiosis , Th17 Cells/cytology , Th17 Cells/immunology
16.
Nat Neurosci ; 20(4): 612-619, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28218915

ABSTRACT

Optogenetic interrogation of neural pathways relies on delivery of light-sensitive opsins into tissue and subsequent optical illumination and electrical recording from the regions of interest. Despite the recent development of multifunctional neural probes, integration of these modalities in a single biocompatible platform remains a challenge. We developed a device composed of an optical waveguide, six electrodes and two microfluidic channels produced via fiber drawing. Our probes facilitated injections of viral vectors carrying opsin genes while providing collocated neural recording and optical stimulation. The miniature (<200 µm) footprint and modest weight (<0.5 g) of these probes allowed for multiple implantations into the mouse brain, which enabled opto-electrophysiological investigation of projections from the basolateral amygdala to the medial prefrontal cortex and ventral hippocampus during behavioral experiments. Fabricated solely from polymers and polymer composites, these flexible probes minimized tissue response to achieve chronic multimodal interrogation of brain circuits with high fidelity.


Subject(s)
Electrodes, Implanted , Hippocampus/physiology , Neurons/physiology , Optical Fibers , Optogenetics/instrumentation , Polymers , Animals , Basolateral Nuclear Complex/physiology , Brain/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Channelrhodopsins , Evoked Potentials/physiology , Male , Mice , Mice, Transgenic , Motor Activity/physiology , Neural Pathways/physiology , Opsins/genetics , Photic Stimulation , Prefrontal Cortex/physiology
17.
Nat Commun ; 7: 12238, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27426965

ABSTRACT

The ability to target subclasses of neurons with defined connectivity is crucial for uncovering neural circuit functions. The olfactory (piriform) cortex is thought to generate odour percepts and memories, and odour information encoded in piriform is routed to target brain areas involved in multimodal sensory integration, cognition and motor control. However, it remains unknown if piriform outputs are spatially organized, and if distinct output channels are delineated by different gene expression patterns. Here we identify genes selectively expressed in different layers of the piriform cortex. Neural tracing experiments reveal that these layer-specific piriform genes mark different subclasses of neurons, which project to distinct target areas. Interestingly, these molecular signatures of connectivity are maintained in reeler mutant mice, in which neural positioning is scrambled. These results reveal that a predictive link between a neuron's molecular identity and connectivity in this cortical circuit is determined independent of its spatial position.


Subject(s)
Gene Expression Profiling , Nerve Net/metabolism , Olfactory Cortex/metabolism , Amygdala/metabolism , Animals , Male , Mice, Inbred C57BL , Neurons/metabolism , Olfactory Bulb/metabolism , Piriform Cortex/metabolism , Prefrontal Cortex/metabolism
18.
Science ; 351(6276): 933-9, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26822608

ABSTRACT

Viral infection during pregnancy has been correlated with increased frequency of autism spectrum disorder (ASD) in offspring. This observation has been modeled in rodents subjected to maternal immune activation (MIA). The immune cell populations critical in the MIA model have not been identified. Using both genetic mutants and blocking antibodies in mice, we show that retinoic acid receptor-related orphan nuclear receptor gamma t (RORγt)-dependent effector T lymphocytes [for example, T helper 17 (TH17) cells] and the effector cytokine interleukin-17a (IL-17a) are required in mothers for MIA-induced behavioral abnormalities in offspring. We find that MIA induces an abnormal cortical phenotype, which is also dependent on maternal IL-17a, in the fetal brain. Our data suggest that therapeutic targeting of TH17 cells in susceptible pregnant mothers may reduce the likelihood of bearing children with inflammation-induced ASD-like phenotypes.


Subject(s)
Autism Spectrum Disorder/immunology , Cerebral Cortex/abnormalities , Cerebral Cortex/immunology , Interleukin-17/immunology , Maternal-Fetal Exchange/immunology , Prenatal Exposure Delayed Effects/immunology , Th17 Cells/immunology , Animals , Antibodies, Blocking/immunology , Antibodies, Blocking/therapeutic use , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/prevention & control , Behavior, Animal , Behavioral Symptoms/immunology , Cerebral Cortex/drug effects , Female , Interleukin-17/biosynthesis , Interleukin-17/pharmacology , Male , Mice , Mutation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Phenotype , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/immunology , Signal Transduction , Th17 Cells/drug effects , Retinoic Acid Receptor gamma
19.
Neuron ; 87(1): 152-63, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26139372

ABSTRACT

Meaningful social interactions modify behavioral responses to sensory stimuli. The neural mechanisms underlying the entrainment of neutral sensory stimuli to salient social cues to produce social learning remain unknown. We used odor-driven behavioral paradigms to ask if oxytocin, a neuropeptide implicated in various social behaviors, plays a crucial role in the formation of learned associations between odor and socially significant cues. Through genetic, optogenetic, and pharmacological manipulations, we show that oxytocin receptor signaling is crucial for entrainment of odor to social cues but is dispensable for entrainment to nonsocial cues. Furthermore, we demonstrate that oxytocin directly impacts the piriform, the olfactory sensory cortex, to mediate social learning. Lastly, we provide evidence that oxytocin plays a role in both appetitive and aversive social learning. These results suggest that oxytocin conveys saliency of social stimuli to sensory representations in the piriform cortex during odor-driven social learning.


Subject(s)
Association Learning/physiology , Olfactory Cortex/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Piriform Cortex/metabolism , Receptors, Oxytocin/metabolism , Smell/physiology , Social Perception , Animals , Cues , Female , Learning/physiology , Male , Mice , Neurons/metabolism , Optogenetics
20.
Cell ; 146(6): 1004-15, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925321

ABSTRACT

Anatomic and physiologic studies have suggested a model in which neurons of the piriform cortex receive convergent input from random collections of glomeruli. In this model, odor representations can only be afforded behavioral significance upon experience. We have devised an experimental strategy that permits us to ask whether the activation of an arbitrarily chosen subpopulation of neurons in piriform cortex can elicit different behavioral responses dependent upon learning. Activation of a small subpopulation of piriform neurons expressing channelrhodopsin at multiple loci in the piriform cortex, when paired with reward or shock, elicits either appetitive or aversive behavior. Moreover, we demonstrate that different subpopulations of piriform neurons expressing ChR2 can be discriminated and independently entrained to elicit distinct behaviors. These observations demonstrate that the piriform cortex is sufficient to elicit learned behavioral outputs in the absence of sensory input. These data imply that the piriform does not use spatial order to map odorant identity or behavioral output.


Subject(s)
Behavior, Animal , Neurons/physiology , Olfactory Pathways/cytology , Smell , Animals , Appetitive Behavior , Channelrhodopsins , Conditioning, Psychological , Mice , Neurons/cytology , Odorants , Olfactory Pathways/physiology
SELECTION OF CITATIONS
SEARCH DETAIL