Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Heliyon ; 7(7): e07570, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34377851

ABSTRACT

Brain-derived neurotrophic factor (Bdnf) expression is tightly controlled at the transcriptional and post-transcriptional levels. Previously, we showed that inhibition of noncoding Bdnf antisense (Bdnf-AS) RNA upregulates Bdnf protein. Here, we generated a Bdnf-antisense knockout (Bdnf-AS KO) mouse model by deleting 6 kilobases upstream of Bdnf-AS. After verifying suppression of Bdnf-AS, baseline behavioral tests indicated no significant difference in knockout and wild type mice, except for enhanced cognitive function in the knockout mice in the Y-maze. Following acute involuntary exercise, Bdnf-AS KO mice were re-assessed and a significant increase in Bdnf mRNA and protein were observed. Following long-term involuntary exercise, we observed a significant increase in nonspatial and spatial memory in novel object recognition and Barnes maze tests in young and aged Bdnf-AS KO mice. Our data provides evidence for the beneficial effects of endogenous Bdnf upregulation and the synergistic effect of Bdnf-AS knockout on exercise and memory retention.

3.
Cancer Res Commun ; 1(1): 1-16, 2021 10.
Article in English | MEDLINE | ID: mdl-35528192

ABSTRACT

A comprehensive constellation of somatic non-silent mutations and copy number (CN) variations in ocular adnexa marginal zone lymphoma (OAMZL) is unknown. By utilizing whole-exome sequencing in 69 tumors we define the genetic landscape of OAMZL. Mutations and CN changes in CABIN1 (30%), RHOA (26%), TBL1XR1 (22%), and CREBBP (17%) and inactivation of TNFAIP3 (26%) were among the most common aberrations. Candidate cancer driver genes cluster in the B-cell receptor (BCR), NFkB, NOTCH and NFAT signaling pathways. One of the most commonly altered genes is CABIN1, a calcineurin inhibitor acting as a negative regulator of the NFAT and MEF2B transcriptional activity. CABIN1 deletions enhance BCR-stimulated NFAT and MEF2B transcriptional activity, while CABIN1 mutations enhance only MEF2B transcriptional activity by impairing binding of mSin3a to CABIN1. Our data provide an unbiased identification of genetically altered genes that may play a role in the molecular pathogenesis of OAMZL and serve as therapeutic targets.


Subject(s)
Eye Neoplasms , Lymphoma, B-Cell, Marginal Zone , Humans , Lymphoma, B-Cell, Marginal Zone/genetics , Eye Neoplasms/genetics , Mutation/genetics , Signal Transduction/genetics , NF-kappa B/genetics , MEF2 Transcription Factors/genetics
4.
Mol Neurobiol ; 57(5): 2279-2289, 2020 May.
Article in English | MEDLINE | ID: mdl-32008165

ABSTRACT

Despite its heterogeneity, autism is characterized by a defined behavioral phenotype, suggesting that the molecular pathology affects specific neural substrates to cause behavioral dysfunction. Previous studies identified genes dysregulated in autism cortex but did not address their cell-type specificity. Moreover, it is unknown whether there is a core of genes dysregulated across multiple neocortical regions. We applied RNA sequencing to postmortem brain tissue samples from autism patients and neurologically normal controls and combined our data with previously published datasets. We then identified genes, pathways, and alternative splicing events which are dysregulated in five neocortical regions in autism. To gain information about cell-type specificity of the dysregulated genes, we analyzed single-nuclei RNA sequencing data of adult human cortex and intersected cell-type-specific gene signatures with genes dysregulated in autism in specific cortical regions. We found that autism-associated gene expression changes across 4 frontal and temporal cortex regions converge on 27 genes related to immune response and enriched in human astrocytes, microglia, and brain endothelium. Shared splicing changes, however, are found in genes predominantly associated with synaptic function and adult interneurons and projection neurons. Finally, we demonstrate that regions of DNA differentially methylated in autism overlap genes associated with development and enriched in human cortical oligodendrocytes. Our study identifies signatures of autism molecular pathology shared across neocortical regions, as well as neural cell types enriched for common dysregulated genes, thus paving way for assessing cell-type-specific mechanisms of autism pathology.


Subject(s)
Autism Spectrum Disorder/genetics , Neocortex/metabolism , RNA, Messenger/analysis , Alternative Splicing , Autism Spectrum Disorder/pathology , DNA Methylation , Gene Expression Regulation , Gene Ontology , Humans , Immunity/genetics , Metabolic Networks and Pathways/genetics , Neocortex/pathology , Neuroglia/metabolism , Neurons/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Single-Cell Analysis , Synapses/metabolism , Temporal Lobe/metabolism , Temporal Lobe/pathology , Transcriptome
5.
Leukemia ; 34(2): 553-566, 2020 02.
Article in English | MEDLINE | ID: mdl-31570756

ABSTRACT

Regulating B-cell receptor (BCR) signaling after antigenic stimulation is essential to properly control immune responses. Currently known mechanisms of inhibiting BCR signaling are via co-receptor stimulation and downstream immunoreceptor tyrosine-based inhibition motif (ITIM) phosphorylation. Herein we demonstrate that BCR stimulation induces rapid and reversible palmitoylation of the SCF-FBXO10 ubiquitin E3 ligase. This results in FBXO10 relocation to the cell membrane, where it targets the human germinal center-associated lymphoma (HGAL) protein for ubiquitylation and degradation, leading to decreases in both BCR-induced calcium influx and phosphorylation of proximal BCR effectors. Importantly, FBXO10 recognition and degradation of HGAL is phosphorylation independent and instead relies on a single evolutionarily conserved HGAL amino acid residue (H91) and FBXO10 relocalization to the cytoplasmic membrane. Together our findings demonstrate the first evidence of negative BCR signaling regulation from direct BCR stimulation and define the temporospatial functions of the FBXO10-HGAL axis. FBXO10 is infrequently mutated in DLBCL but some of these mutations deregulate BCR signaling. These observations may have important implications on lymphomagenesis and other immune processes.


Subject(s)
F-Box Proteins/metabolism , Germinal Center/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, Antigen, B-Cell/metabolism , B-Lymphocytes/metabolism , Cell Membrane/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Neoplasm Proteins/metabolism , Phosphorylation/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism
6.
Blood Adv ; 3(15): 2286-2297, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31362927

ABSTRACT

Human germinal center (GC)-associated lymphoma (HGAL) is an adaptor protein expressed in GC B cells. HGAL regulates cell motility and B-cell receptor (BCR) signaling, processes that are central for the successful completion of the GC reaction. Herein, we demonstrate phosphorylation of HGAL by Syk and Lyn kinases at tyrosines Y80, Y86, Y106Y107, Y128, and Y148. The HGAL YEN motif (amino acids 107-109) is similar to the phosphopeptide motif pYXN used as a binding site to the growth factor receptor-bound protein 2 (Grb2). We demonstrate by biochemical and molecular methodologies that HGAL directly interacts with Grb2. Concordantly, microscopy studies demonstrate HGAL-Grb2 colocalization in the membrane central supramolecular activation clusters (cSMAC) following BCR activation. Mutation of the HGAL putative binding site to Grb2 abrogates the interaction between these proteins. Further, this HGAL mutant localizes exclusively in the peripheral SMAC and decreases the rate and intensity of BCR accumulation in the cSMAC. Furthermore, we demonstrate that Grb2, HGAL, and Syk interact in the same complex, but Grb2 does not modulate the effects of HGAL on Syk kinase activity. Overall, the interplay between the HGAL and Grb2 regulates the magnitude of BCR signaling and synapse formation.


Subject(s)
B-Lymphocytes/metabolism , GRB2 Adaptor Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , B-Lymphocytes/immunology , Cell Line , Mice , Models, Biological , Phosphorylation , Protein Binding , Syk Kinase/metabolism , src-Family Kinases/metabolism
7.
Neurobiol Dis ; 119: 149-158, 2018 11.
Article in English | MEDLINE | ID: mdl-30099093

ABSTRACT

BACKGROUND: With the capacity to modulate gene networks in an environmentally-sensitive manner, the role of epigenetic systems in mental disorders has come under intense investigation. Dysregulation of epigenetic effectors, including microRNAs and histone-modifying enzymes, may better explain the role of environmental risk factors and the observed heritability rate that cannot be fully attributed to known genetic risk alleles. Here, we aimed to identify novel epigenetic targets of the schizophrenia-associated microRNA 132 (miR-132). METHODS: Histone modifications were quantified by immunodetection in response to viral-mediated overexpression of miR-132 while a luminescent reporter system was used to validate targets of miR-132 in vitro. Genome-wide profiling, quantitative PCR and NanoSting were used to quantify gene expression in post-mortem human brains, neuronal cultures and prefrontal cortex (PFC) of mice chronically exposed to antipsychotics. Following viral-mediated depletion of Enhancer of Zeste 1 (EZH1) in the murine PFC, behaviors including sociability and motivation were assessed using a 3-chambered apparatus and forced-swim test, respectively. RESULTS: Overexpression of miR-132 decreased global histone 3 lysine 27 tri-methylation (H3K27me3), a repressive epigenetic mark. Moreover, the polycomb-associated H3K27 methyltransferase, EZH1, is regulated by miR-132 and upregulated in the PFC of schizophrenics. Unlike its homolog EZH2, expression of EZH1 in the murine PFC decreased following chronic exposure to antipsychotics. Viral-mediated depletion of EZH1 in the mouse PFC attenuated sociability, enhanced motivational behaviors, and affected gene expression pathways related to neurotransmission and behavioral phenotypes. CONCLUSIONS: EZH1 is dysregulated in schizophrenia, sensitive to antipsychotic medications, and a brain-enriched miR-132 target that controls neurobehavioral phenotypes.


Subject(s)
Antipsychotic Agents/therapeutic use , Epigenesis, Genetic/physiology , Motivation/physiology , Polycomb Repressive Complex 2/biosynthesis , Schizophrenia/metabolism , Social Behavior , Adult , Aged , Animals , Antipsychotic Agents/pharmacology , Cell Line, Tumor , Cohort Studies , Epigenesis, Genetic/drug effects , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Motivation/drug effects , Polycomb Repressive Complex 2/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Schizophrenia/drug therapy , Schizophrenia/genetics
8.
Mol Cell Neurosci ; 85: 183-189, 2017 12.
Article in English | MEDLINE | ID: mdl-29055697

ABSTRACT

Natural antisense transcripts (NATs) are an abundant class of long noncoding RNAs that have recently been shown to be key regulators of chromatin dynamics and gene expression in nervous system development and neurological disorders. However, it is currently unclear if NAT-based mechanisms also play a role in drug-induced neuroadaptations. Aberrant regulation of gene expression is one critical factor underlying the long-lasting behavioral abnormalities that characterize substance use disorder, and it is possible that some drug-induced transcriptional responses are mediated, in part, by perturbations in NAT activity. To test this hypothesis, we used an automated algorithm that mines the NCBI AceView transcriptomics database to identify NAT overlapping genes linked to addiction. We found that 22% of the genes examined contain NATs and that expression of Homer1 natural antisense transcript (Homer1-AS) was altered in the nucleus accumbens (NAc) of mice 2h and 10days following repeated cocaine administration. In in vitro studies, depletion of Homer1-AS lead to an increase in the corresponding sense gene expression, indicating a potential regulatory mechanisms of Homer1 expression by its corresponding antisense transcript. Future in vivo studies are needed to definitely determine a role for Homer1-AS in cocaine-induced behavioral and molecular adaptations.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Homer Scaffolding Proteins/drug effects , Nucleus Accumbens/drug effects , RNA, Antisense/biosynthesis , Animals , Gene Expression Regulation/genetics , Homer Scaffolding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Antisense/drug effects
9.
Nat Commun ; 8: 15622, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28555645

ABSTRACT

Long noncoding RNAs are emerging players in the epigenetic machinery with key roles in development and diseases. Here we uncover a complex network comprising a promoter-associated noncoding RNA (paRNA), microRNA and epigenetic regulators that controls transcription of the tumour suppressor E-cadherin in epithelial cancers. E-cadherin silencing relies on the formation of a complex between the paRNA and microRNA-guided Argonaute 1 that, together, recruit SUV39H1 and induce repressive chromatin modifications in the gene promoter. A single nucleotide polymorphism (rs16260) linked to increased cancer risk alters the secondary structure of the paRNA, with the risk allele facilitating the assembly of the microRNA-guided Argonaute 1 complex and gene silencing. Collectively, these data demonstrate the role of a paRNA in E-cadherin regulation and the impact of a noncoding genetic variant on its function. Deregulation of paRNA-based epigenetic networks may contribute to cancer and other diseases making them promising targets for drug discovery.


Subject(s)
Argonaute Proteins/genetics , Cadherins/genetics , Eukaryotic Initiation Factors/genetics , Gene Silencing , Methyltransferases/genetics , Neoplasms/genetics , Prostatic Neoplasms/genetics , Repressor Proteins/genetics , Alleles , Antigens, CD , Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Male , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism
10.
Methods Mol Biol ; 1543: 197-208, 2017.
Article in English | MEDLINE | ID: mdl-28349428

ABSTRACT

Transcriptomic as well as in vivo studies have revealed the importance of several lncRNAs in many complex diseases including cancer, cardiovascular, and neurological disorders. In this protocol, we describe how to perform RNAseq data analysis to identify lncRNAs associated with disease states utilizing the open-source software CANEapp (application for Comprehensive automated Analysis of Next-generation sequencing Experiments).


Subject(s)
Computational Biology/methods , Genetic Association Studies , Genetic Predisposition to Disease , RNA, Long Noncoding/genetics , Software , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Association Studies/methods , Humans , Transcriptome , Web Browser
11.
JCI Insight ; 2(6): e91782, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28352664

ABSTRACT

Molecular mechanisms underlying learning and memory remain imprecisely understood, and restorative interventions are lacking. We report that intranasal administration of siRNAs can be used to identify targets important in cognitive processes and to improve genetically impaired learning and memory. In mice modeling the intellectual deficiency of Fragile X syndrome, intranasally administered siRNA targeting glycogen synthase kinase-3ß (GSK3ß), histone deacetylase-1 (HDAC1), HDAC2, or HDAC3 diminished cognitive impairments. In WT mice, intranasally administered brain-derived neurotrophic factor (BDNF) siRNA or HDAC4 siRNA impaired learning and memory, which was partially due to reduced insulin-like growth factor-2 (IGF2) levels because the BDNF siRNA- or HDAC4 siRNA-induced cognitive impairments were ameliorated by intranasal IGF2 administration. In Fmr1-/- mice, hippocampal IGF2 was deficient, and learning and memory impairments were ameliorated by IGF2 intranasal administration. Therefore intranasal siRNA administration is an effective means to identify mechanisms regulating cognition and to modulate therapeutic targets.


Subject(s)
Cognition Disorders/genetics , Fragile X Syndrome/genetics , Insulin-Like Growth Factor II/genetics , RNA, Small Interfering/administration & dosage , Administration, Intranasal , Animals , Cognition Disorders/psychology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/psychology , Glycogen Synthase Kinase 3 beta/genetics , Histone Deacetylases/genetics , Mice , Mice, Knockout
12.
World J Biol Psychiatry ; 18(6): 445-456, 2017 09.
Article in English | MEDLINE | ID: mdl-27723376

ABSTRACT

OBJECTIVES: We examined mechanisms that contribute to the rapid antidepressant effect of ketamine in mice that is dependent on glycogen synthase kinase-3 (GSK3) inhibition. METHODS: We measured serotonergic (5HT)-2C-receptor (5HTR2C) cluster microRNA (miRNA) levels in mouse hippocampus after administering an antidepressant dose of ketamine (10 mg/kg) in wild-type and GSK3 knockin mice, after GSK3 inhibition with L803-mts, and in learned helpless mice. RESULTS: Ketamine up-regulated cluster miRNAs 448-3p, 764-5p, 1264-3p, 1298-5p and 1912-3p (2- to 11-fold). This up-regulation was abolished in GSK3 knockin mice that express mutant constitutively active GSK3. The GSK3 specific inhibitor L803-mts was antidepressant in the learned helplessness and novelty suppressed feeding depression-like behaviours and up-regulated the 5HTR2C miRNA cluster in mouse hippocampus. After administration of the learned helplessness paradigm mice were divided into cohorts that were resilient (non-depressed) or were susceptible (depressed) to learned helplessness. The resilient, but not depressed, mice displayed increased hippocampal levels of miRNAs 448-3p and 1264-3p. Administration of an antagonist to miRNA 448-3p diminished the antidepressant effect of ketamine in the learned helplessness paradigm, indicating that up-regulation of miRNA 448-3p provides an antidepressant action. CONCLUSIONS: These findings identify a new outcome of GSK3 inhibition by ketamine that may contribute to antidepressant effects.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hippocampus/drug effects , Hippocampus/metabolism , Introns/drug effects , Ketamine/pharmacology , MicroRNAs/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, Serotonin, 5-HT2C/drug effects , Animals , Antidepressive Agents/administration & dosage , Depression/drug therapy , Disease Models, Animal , Helplessness, Learned , Ketamine/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Protein Kinase Inhibitors/administration & dosage , Up-Regulation
13.
Eur J Neurosci ; 44(10): 2858-2870, 2016 11.
Article in English | MEDLINE | ID: mdl-27564458

ABSTRACT

Astrocytes are a morphologically and functionally heterogeneous population of cells that play critical roles in neurodevelopment and in the regulation of central nervous system homeostasis. Studies of human astrocytes have been hampered by the lack of specific molecular markers and by the difficulties associated with purifying and culturing astrocytes from adult human brains. Human neural progenitor cells (NPCs) with self-renewal and multipotent properties represent an appealing model system to gain insight into the developmental genetics and function of human astrocytes, but a comprehensive molecular characterization that confirms the validity of this cellular system is still missing. Here we used an unbiased transcriptomic analysis to characterize in vitro culture of human NPCs and to define the gene expression programs activated during the differentiation of these cells into astrocytes using FBS or the combination of CNTF and BMP4. Our results demonstrate that in vitro cultures of human NPCs isolated during the gliogenic phase of neurodevelopment mainly consist of radial glial cells (RGCs) and glia-restricted progenitor cells. In these cells the combination of CNTF and BMP4 activates the JAK/STAT and SMAD signaling cascades, leading to the inhibition of oligodendrocytes lineage commitment and activation of astrocytes differentiation. On the other hand, FBS-derived astrocytes have properties of reactive astrocytes. Our work suggests that in vitro culture of human NPCs represents a valuable cellular system to study human disorders characterized by impairment of astrocytes development and function. Our datasets represent an important resource for researchers studying human astrocytes development and might set the basis for the discovery of novel human-specific astrocyte markers.


Subject(s)
Astrocytes/metabolism , Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , Transcriptome , Astrocytes/cytology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cells, Cultured , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Neural Stem Cells/cytology , Neurogenesis , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism
14.
Curr Alzheimer Res ; 13(9): 985-95, 2016.
Article in English | MEDLINE | ID: mdl-27117003

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by welldefined neuropathological brain changes including amyloid plaques, neurofibrillary tangles and the presence of chronic neuroinflammation. OBJECTIVE: The brain penetrant BET bromodomain inhibitor JQ1 has been shown to regulate inflammation responses in vitro and in vivo, but its therapeutic potential in AD is currently unknown. METHOD: Three-month-old 3xTg mice were injected once a day with JQ1 (50 mg/kg) or vehicle for 15 weeks. At the end of the treatment learning and memory was assessed using the modified Barnes maze and the Y maze behavioral tests. Tissue from the brain and other organs was collected for molecular evaluation of neuroinflammation tau pathology and amyloid ß. RESULTS: JQ1 treatment reduced splenomegaly and neuroinflammation in the brain of treated mice where we observed a reduction in the expression of the pro-inflammatory modulators Il-1b, Il-6, Tnfa, Ccl2, Nos2 and Ptgs2. Additionally, JQ1-treated mice showed a reduction of tau phosphorylation at Ser396 in the hippocampus and frontal cortex while total levels of tau remained unaffected. On the other hand, JQ1 did not ameliorate learning and memory deficits in 7-month-old 3xTg mice. CONCLUSION: Taken together, our data suggest that BET bromodomain inhibitors hold the promise to be used for the treatment of neurological disorders characterized by neuroinflammation.


Subject(s)
Alzheimer Disease/drug therapy , Azepines/pharmacology , Brain/drug effects , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Triazoles/pharmacology , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Brain/immunology , Brain/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Inflammation/metabolism , Inflammation/pathology , Learning/drug effects , Male , Memory/drug effects , Mice, Transgenic , Organ Size , Peptide Fragments/metabolism , Phosphorylation/drug effects , Spleen/drug effects , Spleen/immunology , Spleen/pathology
15.
Mol Cell Neurosci ; 74: 49-57, 2016 07.
Article in English | MEDLINE | ID: mdl-27001315

ABSTRACT

Triplet repeat expansions in the Fragile X mental retardation 1 (FMR1) gene cause either intellectual disability and autism, or adult-onset neurodegeneration, with poorly understood variability in presentation. Previous studies have identified several long noncoding RNAs (lncRNAs) at the FMR1 locus, including FMR4. Similarly to FMR1, FMR4 is silenced by large-repeat expansions that result in enrichment of DNA and histone methylation within the shared promoter and repeat sequence, suggesting a possible role for this noncoding RNA in the pathophysiology of Fragile X. We therefore assessed the functional role of FMR4 to gain further insight into the molecular processes in Fragile X-associated disorders. Previous work showed that FMR4 does not exhibit cis-regulation of FMR1. Here, we found that FMR4 is a chromatin-associated transcript and, using genome-wide chromatin immunoprecipitation experiments, showed that FMR4 alters the chromatin state and the expression of several hundred genes in trans. Among the genes regulated by FMR4, we found enrichment for those involved in neural development and cellular proliferation. S-phase marker assays further demonstrated that FMR4 may promote cellular proliferation, rather than differentiation, of human neural precursor cells (hNPCs). By establishing this novel function for FMR4 in hNPCs, we lend support to existing evidence of the epigenetic involvement of lncRNA in nervous system development, and increase our understanding of the complex pathogenesis underlying neurological disorders associated with FMR1 repeat expansions.


Subject(s)
Cell Proliferation , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , RNA, Long Noncoding/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Genes, Developmental , HEK293 Cells , Humans , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , RNA, Long Noncoding/metabolism
16.
BMC Genomics ; 17: 49, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758513

ABSTRACT

BACKGROUND: Next generation sequencing (NGS) technologies are indispensable for molecular biology research, but data analysis represents the bottleneck in their application. Users need to be familiar with computer terminal commands, the Linux environment, and various software tools and scripts. Analysis workflows have to be optimized and experimentally validated to extract biologically meaningful data. Moreover, as larger datasets are being generated, their analysis requires use of high-performance servers. RESULTS: To address these needs, we developed CANEapp (application for Comprehensive automated Analysis of Next-generation sequencing Experiments), a unique suite that combines a Graphical User Interface (GUI) and an automated server-side analysis pipeline that is platform-independent, making it suitable for any server architecture. The GUI runs on a PC or Mac and seamlessly connects to the server to provide full GUI control of RNA-sequencing (RNA-seq) project analysis. The server-side analysis pipeline contains a framework that is implemented on a Linux server through completely automated installation of software components and reference files. Analysis with CANEapp is also fully automated and performs differential gene expression analysis and novel noncoding RNA discovery through alternative workflows (Cuffdiff and R packages edgeR and DESeq2). We compared CANEapp to other similar tools, and it significantly improves on previous developments. We experimentally validated CANEapp's performance by applying it to data derived from different experimental paradigms and confirming the results with quantitative real-time PCR (qRT-PCR). CANEapp adapts to any server architecture by effectively using available resources and thus handles large amounts of data efficiently. CANEapp performance has been experimentally validated on various biological datasets. CANEapp is available free of charge at http://psychiatry.med.miami.edu/research/laboratory-of-translational-rna-genomics/CANE-app . CONCLUSIONS: We believe that CANEapp will serve both biologists with no computational experience and bioinformaticians as a simple, timesaving but accurate and powerful tool to analyze large RNA-seq datasets and will provide foundations for future development of integrated and automated high-throughput genomics data analysis tools. Due to its inherently standardized pipeline and combination of automated analysis and platform-independence, CANEapp is an ideal for large-scale collaborative RNA-seq projects between different institutions and research groups.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , RNA/genetics , Software , Genomics , Internet , User-Computer Interface
17.
Front Genet ; 6: 263, 2015.
Article in English | MEDLINE | ID: mdl-26322075

ABSTRACT

CGG repeat expansions in the Fragile X mental retardation 1 (FMR1) gene are responsible for a family of associated disorders characterized by either intellectual disability and autism Fragile X Syndrome (FXS), or adult-onset neurodegeneration Fragile X-associated Tremor/Ataxia Syndrome. However, the FMR1 locus is complex and encodes several long non-coding RNAs, whose expression is altered by repeat expansion mutations. The role of these lncRNAs is thus far unknown; therefore we investigated the functionality of FMR4, which we previously identified. "Full"-length expansions of the FMR1 triplet repeat cause silencing of both FMR1 and FMR4, thus we are interested in potential loss-of-function that may add to phenotypic manifestation of FXS. Since the two transcripts do not exhibit cis-regulation of one another, we examined the potential for FMR4 to regulate target genes at distal genomic loci using gene expression microarrays. We identified FMR4-responsive genes, including the methyl-CpG-binding domain protein 4 (MBD4). Furthermore, we found that in differentiating human neural precursor cells, FMR4 expression is developmentally regulated in opposition to expression of both FMR1 (which is expected to share a bidirectional promoter with FMR4) and MBD4. We therefore propose that FMR4's function is as a gene-regulatory lncRNA and that this transcript may function in normal development. Closer examination of FMR4 increases our understanding of the role of regulatory lncRNA and the consequences of FMR1 repeat expansions.

18.
J Alzheimers Dis ; 48(3): 647-65, 2015.
Article in English | MEDLINE | ID: mdl-26402107

ABSTRACT

The underlying genetic variations of late-onset Alzheimer's disease (LOAD) cases remain largely unknown. A combination of genetic variations with variable penetrance and lifetime epigenetic factors may converge on transcriptomic alterations that drive LOAD pathological process. Transcriptome profiling using deep sequencing technology offers insight into common altered pathways regardless of underpinning genetic or epigenetic factors and thus represents an ideal tool to investigate molecular mechanisms related to the pathophysiology of LOAD. We performed directional RNA sequencing on high quality RNA samples extracted from hippocampi of LOAD and age-matched controls. We further validated our data using qRT-PCR on a larger set of postmortem brain tissues, confirming downregulation of the gene encoding substance P (TAC1) and upregulation of the gene encoding the plasminogen activator inhibitor-1 (SERPINE1). Pathway analysis indicates dysregulation in neural communication, cerebral vasculature, and amyloid-ß clearance. Beside protein coding genes, we identified several annotated and non-annotated long noncoding RNAs that are differentially expressed in LOAD brain tissues, three of them are activity-dependent regulated and one is induced by Aß(1-42) exposure of human neural cells. Our data provide a comprehensive list of transcriptomics alterations in LOAD hippocampi and warrant holistic approach including both coding and non-coding RNAs in functional studies aimed to understand the pathophysiology of LOAD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , RNA, Long Noncoding/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Female , Gene Expression Profiling , Hippocampus/metabolism , Homeostasis/genetics , Homeostasis/physiology , Humans , Male , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Transcriptome
19.
J Vasc Res ; 52(2): 71-80, 2015.
Article in English | MEDLINE | ID: mdl-26088024

ABSTRACT

Adenosine and uridine triphosphate (ATP and UTP) can act as extracellular signalling molecules, playing important roles in vascular biology and disease. ATP and UTP acting via the P2Y2-receptor have, for example, been shown to regulate endothelial dilatation, inflammation and angiogenesis. MicroRNAs (miRNAs), a class of regulatory, short, non-coding RNAs, have been shown to be important regulators of these biological processes. In this study, we used RNA deep-sequencing to explore changes in miRNA expression in the human microvascular endothelial cell line HMEC-1 upon UTP treatment. The expression of miR-22, which we have previously shown to target ICAM-1 mRNA in HMEC-1, increased significantly after stimulation. Up-regulation of miR-22 and down-regulation of cell surface ICAM-1 were confirmed with qRT-PCR and flow cytometry, respectively. siRNA-mediated knockdown of the P2Y2-receptor abolished the effect of UTP on miR-22 transcription. Leukocyte adhesion was significantly inhibited in HMEC-1 following miR-22 overexpression and treatment with UTP/ATP. In conclusion, extracellular UTP and ATP can attenuate ICAM-1 expression and leukocyte adhesion in endothelial cells through miR-22.


Subject(s)
Adenosine Triphosphate/pharmacology , Anti-Inflammatory Agents/pharmacology , Endothelial Cells/drug effects , Inflammation/prevention & control , Intercellular Adhesion Molecule-1/metabolism , MicroRNAs/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Uridine Triphosphate/pharmacology , Cell Adhesion/drug effects , Cell Line , Coculture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Flow Cytometry , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Intercellular Adhesion Molecule-1/immunology , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , MicroRNAs/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA Interference , Receptors, Purinergic P2Y2/drug effects , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
20.
Cell Rep ; 11(6): 967-976, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25937287

ABSTRACT

Long non-coding RNAs (lncRNAs), including natural antisense transcripts (NATs), are expressed more extensively than previously anticipated and have widespread roles in regulating gene expression. Nevertheless, the molecular mechanisms of action of the majority of NATs remain largely unknown. Here, we identify a NAT of low-density lipoprotein receptor-related protein 1 (Lrp1), referred to as Lrp1-AS, that negatively regulates Lrp1 expression. We show that Lrp1-AS directly binds to high-mobility group box 2 (Hmgb2) and inhibits the activity of Hmgb2 to enhance Srebp1a-dependent transcription of Lrp1. Short oligonucleotides targeting Lrp1-AS inhibit the interaction of antisense transcript and Hmgb2 protein and increase Lrp1 expression by enhancing Hmgb2 activity. Quantitative RT-PCR analysis of brain tissue samples from Alzheimer's disease patients and aged-matched controls revealed upregulation of LRP1-AS and downregulation of LRP1. Our data suggest a regulatory mechanism whereby a NAT interacts with a ubiquitous chromatin-associated protein to modulate its activity in a locus-specific fashion.


Subject(s)
Chromatin/metabolism , HMGB2 Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , RNA, Antisense/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Genome , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Models, Biological , Protein Binding , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...