Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Curr Opin Neurobiol ; 86: 102858, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38547564

ABSTRACT

Advances in autism spectrum disorder (ASD) genetics have identified many genetic causes, reflecting remarkable progress while at the same time identifying challenges such as heterogeneity and pleiotropy, which complicate attempts to connect genetic risk to mechanisms. High-throughput functional genomic approaches have yielded progress by defining a molecular pathology in the brain of individuals with ASD and in identifying convergent biological pathways through which risk genes are predicted to act. These studies indicate that ASD genetic risk converges in early brain development, primarily during the period of cortical neurogenesis. Over development, genetic perturbations in turn lead to broad neuronal signaling dysregulation, most prominent in glutamatergic cortical-cortical projecting neurons and somatostatin positive interneurons, which is accompanied by glial dyshomeostasis throughout the cerebral cortex. Connecting these developmental perturbations to disrupted neuronal and glial function in the postnatal brain is an important direction in current research. Coupling functional genomic approaches with advances in induced pluripotent stem cell-derived neural organoid development provides a promising avenue for connecting brain pathology to developmental mechanisms.


Subject(s)
Autism Spectrum Disorder , Genomics , Humans , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/metabolism , Genomics/methods , Animals , Brain/pathology , Brain/metabolism
2.
Mol Psychiatry ; 23(4): 993-1000, 2018 04.
Article in English | MEDLINE | ID: mdl-28533516

ABSTRACT

Autism spectrum disorder (ASD) is a behaviorally defined condition that manifests in infancy or early childhood as deficits in communication skills and social interactions. Often, restricted and repetitive behaviors (RRBs) accompany this disorder. ASD is polygenic and genetically complex, so we hypothesized that focusing analyses on intermediate core component phenotypes, such as RRBs, can reduce genetic heterogeneity and improve statistical power. Applying this approach, we mined Caucasian genome-wide association studies (GWAS) data from two of the largest ASD family cohorts, the Autism Genetics Resource Exchange and Autism Genome Project (AGP). Of the 12 RRBs measured by the Autism Diagnostic Interview-Revised, seven were found to be significantly familial and substantially variable, and hence, were tested for genome-wide association in 3104 ASD-affected children from 2045 families. Using a stringent significance threshold (P<7.1 × 10-9), GWAS in the AGP revealed an association between 'the degree of the repetitive use of objects or interest in parts of objects' and rs2898883 (P<6.8 × 10-9), which resides within the sixth intron of PHB. To identify the candidate target genes of the associated single-nucleotide polymorphisms at that locus, we applied chromosome conformation studies in developing human brains and implicated three additional genes: SLC35B1, CALCOCO2 and DLX3. Gene expression, brain imaging and fetal brain expression quantitative trait locus studies prioritize SLC35B1 and PHB. These analyses indicate that GWAS of single heritable features of genetically complex disorders followed by chromosome conformation studies in relevant tissues can be successful in revealing novel risk genes for single core features of ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Chromosomes, Human, Pair 17 , Compulsive Behavior/genetics , Adolescent , Adult , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/psychology , Brain/metabolism , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Gestational Age , Homeodomain Proteins/genetics , Humans , Male , Middle Aged , Monosaccharide Transport Proteins/genetics , Multifactorial Inheritance , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Prohibitins , Quantitative Trait Loci , Repressor Proteins/genetics , Transcription Factors/genetics , Transcriptome
3.
Mol Psychiatry ; 22(8): 1134-1139, 2017 08.
Article in English | MEDLINE | ID: mdl-27843152

ABSTRACT

Several common alleles in the oxytocin receptor gene (OXTR) are associated with altered brain function in reward circuitry in neurotypical adults and may increase risk for autism spectrum disorders (ASD). Yet, it is currently unknown how variation in the OXTR relates to brain functioning in individuals with ASD, and, critically, whether neural endophenotypes vary as a function of aggregate genetic risk. Here, for we believe the first time, we use a multi-locus approach to examine how genetic variation across several OXTR single-nucleotide polymorphisms (SNPs) affect functional connectivity of the brain's reward network. Using data from 41 children with ASD and 41 neurotypical children, we examined functional connectivity of the nucleus accumbens (NAcc) - a hub of the reward network - focusing on how connectivity varies with OXTR risk-allele dosage. Youth with ASD showed reduced NAcc connectivity with other areas in the reward circuit as a function of increased OXTR risk-allele dosage, as well as a positive association between risk-allele dosage and symptom severity, whereas neurotypical youth showed increased NAcc connectivity with frontal brain regions involved in mentalizing. In addition, we found that increased NAcc-frontal cortex connectivity in typically developing youth was related to better scores on a standardized measure of social functioning. Our results indicate that cumulative genetic variation on the OXTR impacts reward system connectivity in both youth with ASD and neurotypical controls. By showing differential genetic effects on neuroendophenotypes, these pathways elucidate mechanisms of vulnerability versus resilience in carriers of disease-associated risk alleles.


Subject(s)
Autism Spectrum Disorder/genetics , Receptors, Oxytocin/genetics , Adolescent , Alleles , Autistic Disorder/genetics , Brain , Case-Control Studies , Child , Female , Frontal Lobe , Gene Dosage/genetics , Gene Frequency/genetics , Genetic Variation , Humans , Male , Neuroimaging/methods , Nucleus Accumbens/physiopathology , Oxytocin/metabolism , Polymorphism, Single Nucleotide/genetics , Receptors, Oxytocin/metabolism , Reward , Social Behavior
4.
Transl Psychiatry ; 6(8): e864, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27483382

ABSTRACT

Genes for autism spectrum disorders (ASDs) are also implicated in fragile X syndrome (FXS), intellectual disabilities (ID) or schizophrenia (SCZ), and converge on neuronal function and differentiation. The SH-SY5Y neuroblastoma cell line, the most widely used system to study neurodevelopment, is currently discussed for its applicability to model cortical development. We implemented an optimal neuronal differentiation protocol of this system and evaluated neurodevelopment at the transcriptomic level using the CoNTeXT framework, a machine-learning algorithm based on human post-mortem brain data estimating developmental stage and regional identity of transcriptomic signatures. Our improved model in contrast to currently used SH-SY5Y models does capture early neurodevelopmental processes with high fidelity. We applied regression modelling, dynamic time warping analysis, parallel independent component analysis and weighted gene co-expression network analysis to identify activated gene sets and networks. Finally, we tested and compared these sets for enrichment of risk genes for neuropsychiatric disorders. We confirm a significant overlap of genes implicated in ASD with FXS, ID and SCZ. However, counterintuitive to this observation, we report that risk genes affect pathways specific for each disorder during early neurodevelopment. Genes implicated in ASD, ID, FXS and SCZ were enriched among the positive regulators, but only ID-implicated genes were also negative regulators of neuronal differentiation. ASD and ID genes were involved in dendritic branching modules, but only ASD risk genes were implicated in histone modification or axonal guidance. Only ID genes were over-represented among cell cycle modules. We conclude that the underlying signatures are disorder-specific and that the shared genetic architecture results in overlaps across disorders such as ID in ASD. Thus, adding developmental network context to genetic analyses will aid differentiating the pathophysiology of neuropsychiatric disorders.


Subject(s)
Autism Spectrum Disorder/genetics , Fragile X Syndrome/genetics , Gene Expression Regulation, Developmental , Intellectual Disability/genetics , Neurogenesis/genetics , Schizophrenia/genetics , Transcriptome , Algorithms , Brain/growth & development , Cell Line, Tumor , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Machine Learning , Neuronal Plasticity/genetics , RNA, Messenger/metabolism , Regression Analysis
6.
Mol Psychiatry ; 20(1): 118-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25199916

ABSTRACT

Autism is a heritable disorder, with over 250 associated genes identified to date, yet no single gene accounts for >1-2% of cases. The clinical presentation, behavioural symptoms, imaging and histopathology findings are strikingly heterogeneous. A more complete understanding of autism can be obtained by examining multiple genetic or behavioural mouse models of autism using magnetic resonance imaging (MRI)-based neuroanatomical phenotyping. Twenty-six different mouse models were examined and the consistently found abnormal brain regions across models were parieto-temporal lobe, cerebellar cortex, frontal lobe, hypothalamus and striatum. These models separated into three distinct clusters, two of which can be linked to the under and over-connectivity found in autism. These clusters also identified previously unknown connections between Nrxn1α, En2 and Fmr1; Nlgn3, BTBR and Slc6A4; and also between X monosomy and Mecp2. With no single treatment for autism found, clustering autism using neuroanatomy and identifying these strong connections may prove to be a crucial step in predicting treatment response.


Subject(s)
Autistic Disorder/pathology , Brain/pathology , Disease Models, Animal , Multigene Family/genetics , Animals , Autistic Disorder/genetics , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Transgenic
7.
Mol Psychiatry ; 20(9): 1069-78, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25311365

ABSTRACT

Rare maternally inherited duplications at 15q11-13 are observed in ~1% of individuals with an autism spectrum disorder (ASD), making it among the most common causes of ASD. 15q11-13 comprises a complex region, and as this copy number variation encompasses many genes, it is important to explore individual genotype-phenotype relationships. Cytoplasmic FMR1-interacting protein 1 (CYFIP1) is of particular interest because of its interaction with Fragile X mental retardation protein (FMRP), its upregulation in transformed lymphoblastoid cell lines from patients with duplications at 15q11-13 and ASD and the presence of smaller overlapping deletions of CYFIP1 in patients with schizophrenia and intellectual disability. Here, we confirm that CYFIP1 is upregulated in transformed lymphoblastoid cell lines and demonstrate its upregulation in the post-mortem brain from 15q11-13 duplication patients for the first time. To investigate how increased CYFIP1 dosage might predispose to neurodevelopmental disease, we studied the consequence of its overexpression in multiple systems. We show that overexpression of CYFIP1 results in morphological abnormalities including cellular hypertrophy in SY5Y cells and differentiated mouse neuronal progenitors. We validate these results in vivo by generating a BAC transgenic mouse, which overexpresses Cyfip1 under the endogenous promotor, observing an increase in the proportion of mature dendritic spines and dendritic spine density. Gene expression profiling on embryonic day 15 suggested the dysregulation of mammalian target of rapamycin (mTOR) signaling, which was confirmed at the protein level. Importantly, similar evidence of mTOR-related dysregulation was seen in brains from 15q11-13 duplication patients with ASD. Finally, treatment of differentiated mouse neuronal progenitors with an mTOR inhibitor (rapamycin) rescued the morphological abnormalities resulting from CYFIP1 overexpression. Together, these data show that CYFIP1 overexpression results in specific cellular phenotypes and implicate modulation by mTOR signaling, further emphasizing its role as a potential convergent pathway in some forms of ASD.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Dendritic Cells/physiology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Cells, Cultured , Chromosomes, Human, Pair 15 , DNA Copy Number Variations , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dendritic Spines/genetics , Dendritic Spines/pathology , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Up-Regulation
11.
Mol Psychiatry ; 18(2): 226-35, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22105621

ABSTRACT

Autism Spectrum Disorder (ASD) has a heterogeneous etiology that is genetically complex. It is defined by deficits in communication and social skills and the presence of restricted and repetitive behaviors. Genetic analyses of heritable quantitative traits that correlate with ASD may reduce heterogeneity. With this in mind, deficits in nonverbal communication (NVC) were quantified based on items from the Autism Diagnostic Interview Revised. Our previous analysis of 228 families from the Autism Genetics Research Exchange (AGRE) repository reported 5 potential quantitative trait loci (QTL). Here we report an NVC QTL replication study in an independent sample of 213 AGRE families. One QTL was replicated (P<0.0004). It was investigated using a targeted-association analysis of 476 haplotype blocks with 708 AGRE families using the Family Based Association Test (FBAT). Blocks in two QTL genes were associated with NVC with a P-value of 0.001. Three associated haplotype blocks were intronic to the Nerve Growth Factor (NGF) gene (P=0.001, 0.001, 0.002), and one was intronic to KCND3 (P=0.001). Individual haplotypes within the associated blocks drove the associations (0.003, 0.0004 and 0.0002) for NGF and 0.0001 for KCND3. Using the same methods, these genes were tested for association with NVC in an independent sample of 1517 families from an Autism Genome Project (AGP). NVC was associated with a haplotype in an adjacent NGF block (P=0.0005) and one 46 kb away from the associated block in KCND3 (0.008). These analyses illustrate the value of QTL and targeted association studies for genetically complex disorders such as ASD. NGF is a promising risk gene for NVC deficits.


Subject(s)
Communication Disorders/genetics , Genetic Predisposition to Disease , Nerve Growth Factor/genetics , Nonverbal Communication/physiology , Quantitative Trait, Heritable , Child , Child Development Disorders, Pervasive/complications , Child Development Disorders, Pervasive/genetics , Communication Disorders/etiology , Family Health , Female , Follow-Up Studies , Genetic Association Studies , Genetic Linkage , Genotype , Humans , Male , Shal Potassium Channels/genetics
12.
Mol Psychiatry ; 18(10): 1090-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23044707

ABSTRACT

Copy number variants (CNVs) have a major role in the etiology of autism spectrum disorders (ASD), and several of these have reached statistical significance in case-control analyses. Nevertheless, current ASD cohorts are not large enough to detect very rare CNVs that may be causative or contributory (that is, risk alleles). Here, we use a tiered approach, in which clinically significant CNVs are first identified in large clinical cohorts of neurodevelopmental disorders (including but not specific to ASD), after which these CNVs are then systematically identified within well-characterized ASD cohorts. We focused our initial analysis on 48 recurrent CNVs (segmental duplication-mediated 'hotspots') from 24 loci in 31 516 published clinical cases with neurodevelopmental disorders and 13 696 published controls, which yielded a total of 19 deletion CNVs and 11 duplication CNVs that reached statistical significance. We then investigated the overlap of these 30 CNVs in a combined sample of 3955 well-characterized ASD cases from three published studies. We identified 73 deleterious recurrent CNVs, including 36 deletions from 11 loci and 37 duplications from seven loci, for a frequency of 1 in 54; had we considered the ASD cohorts alone, only 58 CNVs from eight loci (24 deletions from three loci and 34 duplications from five loci) would have reached statistical significance. In conclusion, until there are sufficiently large ASD research cohorts with enough power to detect very rare causative or contributory CNVs, data from larger clinical cohorts can be used to infer the likely clinical significance of CNVs in ASD.


Subject(s)
Child Development Disorders, Pervasive/genetics , Gene Dosage , Autistic Disorder/epidemiology , Autistic Disorder/genetics , Causality , Child Development Disorders, Pervasive/epidemiology , Congenital Abnormalities/epidemiology , Congenital Abnormalities/genetics , Data Mining , Developmental Disabilities/epidemiology , Developmental Disabilities/genetics , Gene Deletion , Gene Duplication , Genetic Association Studies , Genetic Heterogeneity , Genetic Predisposition to Disease , Homologous Recombination , Humans , Prevalence , Sample Size
13.
Mol Psychiatry ; 17(1): 62-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21079609

ABSTRACT

Approximately 40-50% of individuals affected by tuberous sclerosis (TSC) develop autism spectrum disorders (ASDs). One possible explanation for this partial penetrance is an interaction between TSC gene mutations and other risk factors such as gestational immune activation. In this study, we report the interactive effects of these two ASD risk factors in a mouse model of TSC. Combined, but not single, exposure had adverse effects on intrauterine survival. Additionally, provisional results suggest that these factors synergize to disrupt social approach behavior in adult mice. Moreover, studies in human populations are consistent with an interaction between high seasonal flu activity in late gestation and TSC mutations in ASD. Taken together, our studies raise the possibility of a gene × environment interaction between heterozygous TSC gene mutations and gestational immune activation in the pathogenesis of TSC-related ASD.


Subject(s)
Child Development Disorders, Pervasive , Haploinsufficiency/genetics , Immunity, Active/physiology , Pregnancy Complications/physiopathology , Social Behavior , Tumor Suppressor Proteins/deficiency , Age Factors , Animals , Animals, Newborn , Behavior, Animal , Child Development Disorders, Pervasive/etiology , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/immunology , Disease Models, Animal , Embryo, Mammalian , Exploratory Behavior , Female , Humans , Immunity, Active/drug effects , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/adverse effects , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/immunology , Pregnancy Complications/mortality , Tuberous Sclerosis Complex 2 Protein
15.
Mol Psychiatry ; 17(2): 202-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21647150

ABSTRACT

Human neural progenitors from a variety of sources present new opportunities to model aspects of human neuropsychiatric disease in vitro. Such in vitro models provide the advantages of a human genetic background combined with rapid and easy manipulation, making them highly useful adjuncts to animal models. Here, we examined whether a human neuronal culture system could be utilized to assess the transcriptional program involved in human neural differentiation and to model some of the molecular features of a neurodevelopmental disorder, such as autism. Primary normal human neuronal progenitors (NHNPs) were differentiated into a post-mitotic neuronal state through addition of specific growth factors and whole-genome gene expression was examined throughout a time course of neuronal differentiation. After 4 weeks of differentiation, a significant number of genes associated with autism spectrum disorders (ASDs) are either induced or repressed. This includes the ASD susceptibility gene neurexin 1, which showed a distinct pattern from neurexin 3 in vitro, and which we validated in vivo in fetal human brain. Using weighted gene co-expression network analysis, we visualized the network structure of transcriptional regulation, demonstrating via this unbiased analysis that a significant number of ASD candidate genes are coordinately regulated during the differentiation process. As NHNPs are genetically tractable and manipulable, they can be used to study both the effects of mutations in multiple ASD candidate genes on neuronal differentiation and gene expression in combination with the effects of potential therapeutic molecules. These data also provide a step towards better understanding of the signaling pathways disrupted in ASD.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/pathology , Gene Expression Regulation, Developmental/genetics , Genomics , Models, Genetic , Neurons/metabolism , Cell Differentiation/physiology , Cells, Cultured , Fetus , Gene Expression Profiling , Genotype , Gestational Age , Humans , Ki-67 Antigen/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Oligonucleotide Array Sequence Analysis
16.
Neurology ; 76(5): 467-74, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21178100

ABSTRACT

OBJECTIVES: To determine whether TMEM106B single nucleotide polymorphisms (SNPs) are associated with frontotemporal lobar degeneration (FTLD) in patients with and without mutations in progranulin (GRN) and to determine whether TMEM106B modulates GRN expression. METHODS: We performed a case-control study of 3 SNPs in TMEM106B in 482 patients with clinical and 80 patients with pathologic FTLD-TAR DNA-binding protein 43 without GRN mutations, 78 patients with FTLD with GRN mutations, and 822 controls. Association analysis of TMEM106B with GRN plasma levels was performed in 1,013 controls and TMEM106B and GRN mRNA expression levels were correlated in peripheral blood samples from 33 patients with FTLD and 150 controls. RESULTS: In our complete FTLD patient cohort, nominal significance was identified for 2 TMEM106B SNPs (top SNP rs1990622, p(allelic) = 0.036). However, the most significant association with risk of FTLD was observed in the subgroup of GRN mutation carriers compared to controls (corrected p(allelic) = 0.0009), where there was a highly significant decrease in the frequency of homozygote carriers of the minor alleles of all TMEM106B SNPs (top SNP rs1990622, CC genotype frequency 2.6% vs 19.1%, corrected p(recessive) = 0.009). We further identified a significant association of TMEM106B SNPs with plasma GRN levels in controls (top SNP rs1990622, corrected p = 0.002) and in peripheral blood samples a highly significant correlation was observed between TMEM106B and GRN mRNA expression in patients with FTLD (r = -0.63, p = 7.7 × 10(-5)) and controls (r = -0.49, p = 2.2 × 10(-10)). CONCLUSIONS: In our study, TMEM106B SNPs significantly reduced the disease penetrance in patients with GRN mutations, potentially by modulating GRN levels. These findings hold promise for the development of future protective therapies for FTLD.


Subject(s)
Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Penetrance , Polymorphism, Single Nucleotide/genetics , Protein Precursors/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cohort Studies , Female , Frontotemporal Lobar Degeneration/blood , Frontotemporal Lobar Degeneration/diagnosis , Genetic Association Studies , Genetic Carrier Screening , Genetic Predisposition to Disease/genetics , Humans , Intercellular Signaling Peptides and Proteins/blood , Male , Membrane Proteins/blood , Middle Aged , Nerve Tissue Proteins/blood , Progranulins , Protein Precursors/blood
17.
Mol Psychiatry ; 15(10): 996-1005, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19455149

ABSTRACT

Chromosome 17q11-q21 is a region of the genome likely to harbor susceptibility to autism (MIM(209850)) based on earlier evidence of linkage to the disorder. This linkage is specific to multiplex pedigrees containing only male probands (MO) within the Autism Genetic Resource Exchange (AGRE). Earlier, Stone et al.(1) completed a high-density single nucleotide polymorphism association study of 13.7 Mb within this interval, but common variant association was not sufficient to account for the linkage signal. Here, we extend this single nucleotide polymorphism-based association study to complete the coverage of the two-LOD support interval around the chromosome 17q linkage peak by testing the majority of common alleles in 284 MO trios. Markers within an interval containing the gene, CACNA1G, were found to be associated with Autism Spectrum Disorder at a locally significant level (P=1.9 × 10(-5)). While establishing CACNA1G as a novel candidate gene for autism, these alleles do not contribute a sufficient genetic effect to explain the observed linkage, indicating that there is substantial genetic heterogeneity despite the clear linkage signal. The region thus likely harbors a combination of multiple common and rare alleles contributing to the genetic risk. These data, along with earlier studies of chromosomes 5 and 7q3, suggest few if any major common risk alleles account for Autism Spectrum Disorder risk under major linkage peaks in the AGRE sample. This provides important evidence for strategies to identify Autism Spectrum Disorder genes, suggesting that they should focus on identifying rare variants and common variants of small effect.


Subject(s)
Autistic Disorder/genetics , Calcium Channels, T-Type/genetics , Chromosomes, Human, Pair 17 , Polymorphism, Single Nucleotide , Autistic Disorder/epidemiology , Child , Female , Follow-Up Studies , Gene Dosage , Genetic Markers , Genetic Predisposition to Disease/epidemiology , Haplotypes , Humans , Linkage Disequilibrium , Lod Score , Male , Risk Factors
18.
Physiol Genomics ; 38(3): 362-71, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19531577

ABSTRACT

Type I interferons (IFNs) are pleiotropic cytokines that modulate both innate and adaptive immune responses. They have been used to treat autoimmune disorders, cancers, and viral infection and have been demonstrated to elicit differential responses within cells, despite sharing a single receptor. The molecular basis for such differential responses has remained elusive. To identify the mechanisms underlying differential type I IFN signaling, we used whole genome microarrays to measure longitudinal transcriptional events within human CD4(+) T cells treated with IFN-alpha(2b) or IFN-beta(1a). We identified differentially regulated genes, analyzed them for the enrichment of known promoter elements and pathways, and constructed a network module based on weighted gene coexpression network analysis (WGCNA). WGCNA uses advanced statistical measures to find interconnected modules of correlated genes. Overall, differential responses to IFN in CD4(+) T cells related to three dominant themes: migration, antigen presentation, and the cytotoxic response. For migration, WGCNA identified subtype-specific regulation of pre-mRNA processing factor 4 homolog B and eukaryotic translation initiation factor 4A2, which work at various levels within the cell to affect the expression of the chemokine CCL5. WGCNA also identified sterile alpha-motif domain-containing 9-like (SAMD9L) as critical in subtype-independent effects of IFN treatment. RNA interference of SAMD9L expression enhanced the migratory phenotype of activated T cells treated with IFN-beta compared with controls. Through the analysis of the dynamic transcriptional events after differential IFN treatment, we were able to identify specific signatures and to uncover novel genes that may underpin the type I IFN response.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Interferon Type I/pharmacology , Adult , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Cluster Analysis , Gene Regulatory Networks , Humans , Interferon alpha-2 , Interferon beta-1a , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Intracellular Signaling Peptides and Proteins , Male , Models, Genetic , Oligonucleotide Array Sequence Analysis , Proteins/genetics , RNA Interference , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction
19.
Cell Death Differ ; 16(6): 807-25, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19282867

ABSTRACT

Insights into early human development are fundamental for our understanding of human biology. Efficient differentiation of human embryonic stem cells (hESCs) into neural precursor cells is critical for future cell-based therapies. Here, using defined conditions, we characterized a new method for rapid and uniform differentiation of hESCs into committed neural precursor cells (designated C-NPCs). Dynamic gene expression analysis identified several distinct stages of ESC neuralization and revealed functional modules of coregulated genes and pathways. The first wave of gene expression changes, likely corresponding to the transition through primitive ectoderm, started at day 3, preceding the formation of columnar neuroepithelial rosettes. The second wave started at day 5, coinciding with the formation of rosettes. The majority of C-NPCs were positive for both anterior and posterior markers of developing neuroepithelium. In culture, C-NPCs became electrophysiologically functional neurons; on transplantation into neonatal mouse brains, C-NPCs integrated into the cortex and olfactory bulb, acquiring appropriate neuronal morphologies and markers. Compared to rosette-NPCs,(1) C-NPCs exhibited limited in vitro expansion capacity and did not express potent oncogenes such as PLAG1 or RSPO3. Concordantly, we never detected tumors or excessive neural proliferation after transplantation of C-NPCs into mouse brains. In conclusion, our study provides a framework for future analysis of molecular signaling during ESC neuralization.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neurons/cytology , Animals , Animals, Newborn , Brain/metabolism , Cell Line , Ectoderm/metabolism , Electrophysiology , Gene Expression Profiling , Humans , Mice , Mice, Inbred ICR , Proteomics , Rosette Formation , Transplantation, Heterologous
20.
Endocrinology ; 150(6): 2822-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19213835

ABSTRACT

Iron is essential for normal cellular homeostasis but in excess promotes free radical formation and is detrimental. Therefore, iron metabolism is tightly regulated. Here, we show that mechanisms regulating systemic iron metabolism may also control iron release into the brain at the blood-choroid plexus-cerebrospinal fluid (CSF) barrier. Intraperitoneal administration of lipopolysaccharide (LPS) in mice triggers a transient transcription of the gene encoding for hepcidin, a key regulator of iron homeostasis, in the choroid plexus, which correlated with increased detection of pro-hepcidin in the CSF. Similarly, the expression of several other iron-related genes is influenced in the choroid plexus by the inflammatory stimulus. Using primary cultures of rat choroid plexus epithelial cells, we show that this response is triggered not only directly by LPS but also by molecules whose expression increases in the blood in response to inflammation, such as IL-6. Intracellular conveyors of these signaling molecules include signal transducer and activator of transcription 3, which becomes phosphorylated, and SMAD family member 4, whose mRNA levels increase soon after LPS administration. This novel role for the choroid plexus-CSF barrier in regulating iron metabolism may be particularly relevant to restrict iron availability for microorganism growth, and in neurodegenerative diseases in which an inflammatory underlying component has been reported.


Subject(s)
Choroid Plexus/metabolism , Homeostasis/physiology , Inflammation/metabolism , Iron/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , Choroid Plexus/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hepcidins , Inflammation/chemically induced , Inflammation/pathology , Injections, Intraperitoneal , Interleukin-6/blood , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Rats , Rats, Wistar , STAT3 Transcription Factor/metabolism , Smad4 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...