Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Adv ; 7(10)2021 03.
Article in English | MEDLINE | ID: mdl-33674307

ABSTRACT

Cognitive function depends on frontal cortex development; however, the mechanisms driving this process are poorly understood. Here, we identify that dynamic regulation of the nicotinic cholinergic system is a key driver of attentional circuit maturation associated with top-down frontal neurons projecting to visual cortex. The top-down neurons receive robust cholinergic inputs, but their nicotinic tone decreases following adolescence by increasing expression of a nicotinic brake, Lynx1 Lynx1 shifts a balance between local and long-range inputs onto top-down frontal neurons following adolescence and promotes the establishment of attentional behavior in adulthood. This key maturational process is disrupted in a mouse model of fragile X syndrome but was rescued by a suppression of nicotinic tone through the introduction of Lynx1 in top-down projections. Nicotinic signaling may serve as a target to rebalance local/long-range balance and treat cognitive deficits in neurodevelopmental disorders.


Subject(s)
Nicotine , Visual Cortex , Animals , Attention/physiology , Cholinergic Agents , Mice , Neurons/physiology , Visual Cortex/physiology
2.
Neural Plast ; 2020: 1673897, 2020.
Article in English | MEDLINE | ID: mdl-32454811

ABSTRACT

The tens of thousands of industrial and synthetic chemicals released into the environment have an unknown but potentially significant capacity to interfere with neurodevelopment. Consequently, there is an urgent need for systematic approaches that can identify disruptive chemicals. Little is known about the impact of environmental chemicals on critical periods of developmental neuroplasticity, in large part, due to the challenge of screening thousands of chemicals. Using an integrative bioinformatics approach, we systematically scanned 2001 environmental chemicals and identified 50 chemicals that consistently dysregulate two transcriptional signatures of critical period plasticity. These chemicals included pesticides (e.g., pyridaben), antimicrobials (e.g., bacitracin), metals (e.g., mercury), anesthetics (e.g., halothane), and other chemicals and mixtures (e.g., vehicle emissions). Application of a chemogenomic enrichment analysis and hierarchical clustering across these diverse chemicals identified two clusters of chemicals with one that mimicked an immune response to pathogen, implicating inflammatory pathways and microglia as a common chemically induced neuropathological process. Thus, we established an integrative bioinformatics approach to systematically scan thousands of environmental chemicals for their ability to dysregulate molecular signatures relevant to critical periods of development.


Subject(s)
Brain/growth & development , Environmental Monitoring/methods , Environmental Pollutants/analysis , Immunity/genetics , Neuronal Plasticity/genetics , Transcriptome/genetics , Animals , Brain/metabolism , Computational Biology , Gene Expression Profiling , Genomics , Mice, Inbred C57BL
3.
Nat Commun ; 11(1): 1003, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32081848

ABSTRACT

Social isolation during the juvenile critical window is detrimental to proper functioning of the prefrontal cortex (PFC) and establishment of appropriate adult social behaviors. However, the specific circuits that undergo social experience-dependent maturation to regulate social behavior are poorly understood. We identify a specific activation pattern of parvalbumin-positive interneurons (PVIs) in dorsal-medial PFC (dmPFC) prior to an active bout, or a bout initiated by the focal mouse, but not during a passive bout when mice are explored by a stimulus mouse. Optogenetic and chemogenetic manipulation reveals that brief dmPFC-PVI activation triggers an active social approach to promote sociability. Juvenile social isolation decouples dmPFC-PVI activation from subsequent active social approach by freezing the functional maturation process of dmPFC-PVIs during the juvenile-to-adult transition. Chemogenetic activation of dmPFC-PVI activity in the adult animal mitigates juvenile isolation-induced social deficits. Therefore, social experience-dependent maturation of dmPFC-PVI is linked to long-term impacts on social behavior.


Subject(s)
Parvalbumins/physiology , Prefrontal Cortex/physiology , Social Behavior , Animals , Interneurons/physiology , Interpersonal Relations , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Models, Psychological , Optogenetics , Parvalbumins/genetics , Prefrontal Cortex/cytology , Prefrontal Cortex/growth & development , Social Isolation
4.
Schizophr Res ; 207: 12-21, 2019 05.
Article in English | MEDLINE | ID: mdl-30442475

ABSTRACT

Childhood critical periods of experience-dependent plasticity are essential for the development of environmentally appropriate behavior and cognition. Disruption of critical periods can alter development of normal function and confer risk for neurodevelopmental disorders. While genes and their expression relevant to neurodevelopment are associated with schizophrenia, the molecular relationship between schizophrenia and critical periods has not been assessed systematically. Here, we apply a transcriptome-based bioinformatics approach to assess whether genes associated with the human critical period for visual cortex plasticity, a well-studied model of cortical critical periods, are aberrantly expressed in schizophrenia and bipolar disorder. Across two dozen datasets encompassing 522 cases and 374 controls, we find that the majority show aberrations in expression of genes associated with the critical period. We observed both hyper- and hypo-critical period plasticity phenotypes at the transcriptome level, which partially mapped to drug candidates that reverse the disorder signatures in silico. Our findings indicate plasticity aberrations in schizophrenia and their treatment may need to be considered in the context of subpopulations with elevated and others reduced plasticity. Future work should leverage ongoing consortia RNA-sequencing efforts to tease out the sources of plasticity-related transcriptional aberrations seen in schizophrenia, including true biological heterogeneity, interaction between normal development/aging and the disorder, and medication history. Our study also urges innovation towards direct assessment of visual cortex plasticity in humans with schizophrenia to precisely deconstruct the role of plasticity in this disorder.


Subject(s)
Bipolar Disorder/genetics , Cerebral Cortex/metabolism , Human Development , Neuronal Plasticity/genetics , Schizophrenia/genetics , Transcriptome , Animals , Bipolar Disorder/drug therapy , Cerebral Cortex/drug effects , Computational Biology , Datasets as Topic , Drug Repositioning , Haplorhini , Humans , Mice , Neuronal Plasticity/drug effects , Schizophrenia/drug therapy , Time Factors
5.
Sci Rep ; 8(1): 16388, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30401819

ABSTRACT

Given that thousands of chemicals released into the environment have the potential capacity to harm neurodevelopment, there is an urgent need to systematically evaluate their toxicity. Neurodevelopment is marked by critical periods of plasticity wherein neural circuits are refined by the environment to optimize behavior and function. If chemicals perturb these critical periods, neurodevelopment can be permanently altered. Focusing on 214 human neurotoxicants, we applied an integrative bioinformatics approach using publically available data to identify dozens of neurotoxicant signatures that disrupt a transcriptional signature of a critical period for brain plasticity. This identified lead (Pb) as a critical period neurotoxicant and we confirmed in vivo that Pb partially suppresses critical period plasticity at a time point analogous to exposure associated with autism. This work demonstrates the utility of a novel informatics approach to systematically identify neurotoxicants that disrupt childhood neurodevelopment and can be extended to assess other environmental chemicals.


Subject(s)
Brain/drug effects , Brain/physiology , Computational Biology , Lead/toxicity , Neuronal Plasticity/drug effects , Neurotoxins/toxicity , Animals , Brain/growth & development , Male , Mice , Mice, Inbred C57BL
6.
BMC Med Inform Decis Mak ; 18(Suppl 3): 79, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30255805

ABSTRACT

BACKGROUND: Worldwide, over 14% of individuals hospitalized for psychiatric reasons have readmissions to hospitals within 30 days after discharge. Predicting patients at risk and leveraging accelerated interventions can reduce the rates of early readmission, a negative clinical outcome (i.e., a treatment failure) that affects the quality of life of patient. To implement individualized interventions, it is necessary to predict those individuals at highest risk for 30-day readmission. In this study, our aim was to conduct a data-driven investigation to find the pharmacological factors influencing 30-day all-cause, intra- and interdepartmental readmissions after an index psychiatric admission, using the compendium of prescription data (prescriptome) from electronic medical records (EMR). METHODS: The data scientists in the project received a deidentified database from the Mount Sinai Data Warehouse, which was used to perform all analyses. Data was stored in a secured MySQL database, normalized and indexed using a unique hexadecimal identifier associated with the data for psychiatric illness visits. We used Bayesian logistic regression models to evaluate the association of prescription data with 30-day readmission risk. We constructed individual models and compiled results after adjusting for covariates, including drug exposure, age, and gender. We also performed digital comorbidity survey using EMR data combined with the estimation of shared genetic architecture using genomic annotations to disease phenotypes. RESULTS: Using an automated, data-driven approach, we identified prescription medications, side effects (primary side effects), and drug-drug interaction-induced side effects (secondary side effects) associated with readmission risk in a cohort of 1275 patients using prescriptome analytics. In our study, we identified 28 drugs associated with risk for readmission among psychiatric patients. Based on prescription data, Pravastatin had the highest risk of readmission (OR = 13.10; 95% CI (2.82, 60.8)). We also identified enrichment of primary side effects (n = 4006) and secondary side effects (n = 36) induced by prescription drugs in the subset of readmitted patients (n = 89) compared to the non-readmitted subgroup (n = 1186). Digital comorbidity analyses and shared genetic analyses further reveals that cardiovascular disease and psychiatric conditions are comorbid and share functional gene modules (cardiomyopathy and anxiety disorder: shared genes (n = 37; P = 1.06815E-06)). CONCLUSIONS: Large scale prescriptome data is now available from EMRs and accessible for analytics that could improve healthcare outcomes. Such analyses could also drive hypothesis and data-driven research. In this study, we explored the utility of prescriptome data to identify factors driving readmission in a psychiatric cohort. Converging digital health data from EMRs and systems biology investigations reveal a subset of patient populations that have significant comorbidities with cardiovascular diseases are more likely to be readmitted. Further, the genetic architecture of psychiatric illness also suggests overlap with cardiovascular diseases. In summary, assessment of medications, side effects, and drug-drug interactions in a clinical setting as well as genomic information using a data mining approach could help to find factors that could help to lower readmission rates in patients with mental illness.


Subject(s)
Data Mining , Drug Interactions , Drug-Related Side Effects and Adverse Reactions/epidemiology , Mental Disorders/complications , Mental Disorders/drug therapy , Patient Readmission/statistics & numerical data , Adult , Aged , Bayes Theorem , Cohort Studies , Data Warehousing , Databases, Factual , Electronic Health Records , Female , Humans , Logistic Models , Male , Middle Aged , Quality of Life , Risk Factors , Time Factors
7.
Pac Symp Biocomput ; 23: 68-79, 2018.
Article in English | MEDLINE | ID: mdl-29218870

ABSTRACT

High and increasing prevalence of neurodevelopmental disorders place enormous personal and economic burdens on society. Given the growing realization that the roots of neurodevelopmental disorders often lie in early childhood, there is an urgent need to identify childhood risk factors. Neurodevelopment is marked by periods of heightened experience-dependent neuroplasticity wherein neural circuitry is optimized by the environment. If these critical periods are disrupted, development of normal brain function can be permanently altered, leading to neurodevelopmental disorders. Here, we aim to systematically identify human variants in neuroplasticity-related genes that confer risk for neurodevelopmental disorders. Historically, this knowledge has been limited by a lack of techniques to identify genes related to neurodevelopmental plasticity in a high-throughput manner and a lack of methods to systematically identify mutations in these genes that confer risk for neurodevelopmental disorders. Using an integrative genomics approach, we determined loss-of-function (LOF) variants in putative plasticity genes, identified from transcriptional profiles of brain from mice with elevated plasticity, that were associated with neurodevelopmental disorders. From five shared differentially expressed genes found in two mouse models of juvenile-like elevated plasticity (juvenile wild-type or adult Lynx1-/- relative to adult wild-type) that were also genotyped in the Mount Sinai BioMe Biobank we identified multiple associations between LOF genes and increased risk for neurodevelopmental disorders across 10,510 patients linked to the Mount Sinai Electronic Medical Records (EMR), including epilepsy and schizophrenia. This work demonstrates a novel approach to identify neurodevelopmental risk genes and points toward a promising avenue to discover new drug targets to address the unmet therapeutic needs of neurodevelopmental disease.


Subject(s)
Loss of Function Mutation , Neurodevelopmental Disorders/genetics , Neuronal Plasticity/genetics , Adaptor Proteins, Signal Transducing , Animals , Computational Biology , Epilepsy/genetics , Female , Genetic Predisposition to Disease , Genetic Variation , Glycoproteins/genetics , Humans , Lipocalin-2/genetics , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neurodevelopmental Disorders/etiology , Neuropeptides/deficiency , Neuropeptides/genetics , Phenotype , Risk Factors , Schizophrenia/genetics , Visual Cortex/metabolism
8.
eNeuro ; 3(6)2016.
Article in English | MEDLINE | ID: mdl-28101530

ABSTRACT

Throughout childhood and adolescence, periods of heightened neuroplasticity are critical for the development of healthy brain function and behavior. Given the high prevalence of neurodevelopmental disorders, such as autism, identifying disruptors of developmental plasticity represents an essential step for developing strategies for prevention and intervention. Applying a novel computational approach that systematically assessed connections between 436 transcriptional signatures of disease and multiple signatures of neuroplasticity, we identified inflammation as a common pathological process central to a diverse set of diseases predicted to dysregulate plasticity signatures. We tested the hypothesis that inflammation disrupts developmental cortical plasticity in vivo using the mouse ocular dominance model of experience-dependent plasticity in primary visual cortex. We found that the administration of systemic lipopolysaccharide suppressed plasticity during juvenile critical period with accompanying transcriptional changes in a particular set of molecular regulators within primary visual cortex. These findings suggest that inflammation may have unrecognized adverse consequences on the postnatal developmental trajectory and indicate that treating inflammation may reduce the burden of neurodevelopmental disorders.


Subject(s)
Cerebral Cortex/growth & development , Cerebral Cortex/immunology , Inflammation/metabolism , Neuronal Plasticity/physiology , Adaptor Proteins, Signal Transducing , Algorithms , Animals , Critical Period, Psychological , Dominance, Ocular/physiology , Escherichia coli , Lipopolysaccharides , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Microelectrodes , Neuropeptides/genetics , Neuropeptides/metabolism , Polymerase Chain Reaction , Sensory Deprivation/physiology , Transcription, Genetic , Transcriptome
9.
Neurosci Lett ; 513(2): 119-23, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22330750

ABSTRACT

The spinal nucleus of the bulbocavernosus (SNB) in rodents is a neuromuscular system consisting of lumbar motoneurons and the perineal muscles they innervate, the bulbocavernosus and levator ani. This system is present prenatally in both males and females but degenerates postnatally in females because of the lack of perinatal androgens. Whether androgens act on the motoneurons or muscles in the SNB system to promote survival is a longstanding question. Evidence in rats suggests androgens act primarily on the muscles in development, given that the muscles express androgen receptor (AR) before the critical period of androgen-dependent cell rescue, whereas motoneurons develop AR after this period. We now report, based on a novel AR-reporter mouse model, that AR is expressed in the bulbocavernosus muscles of C57/BL6(J) mice as early as embryonic day 15, while, based on AR-immunocytochemistry, SNB motoneurons do not express AR until postnatal day 4. These results indicate that the ontogeny of AR expression in the mouse SNB system resembles that found in rats, suggesting that androgens may also act on perineal muscles in mice to rescue the SNB system.


Subject(s)
Motor Neurons/metabolism , Muscle, Skeletal/innervation , Receptors, Androgen/metabolism , Animals , Male , Mice , Muscle, Skeletal/metabolism , Receptors, Androgen/genetics , Sex Differentiation/physiology , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...