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.
PLoS One ; 8(7): e66629, 2013.
Article in English | MEDLINE | ID: mdl-23874394

ABSTRACT

LifeMap Discovery™ provides investigators with an integrated database of embryonic development, stem cell biology and regenerative medicine. The hand-curated reconstruction of cell ontology with stem cell biology; including molecular, cellular, anatomical and disease-related information, provides efficient and easy-to-use, searchable research tools. The database collates in vivo and in vitro gene expression and guides translation from in vitro data to the clinical utility, and thus can be utilized as a powerful tool for research and discovery in stem cell biology, developmental biology, disease mechanisms and therapeutic discovery. LifeMap Discovery is freely available to academic nonprofit institutions at http://discovery.lifemapsc.com.


Subject(s)
Embryonic Development , Regenerative Medicine , Animals , Cell Differentiation , Data Mining , Gene Expression , Humans , Protein Biosynthesis , Stem Cells/cytology
2.
J Cell Mol Med ; 16(3): 555-68, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21507200

ABSTRACT

Exercise and inherited factors both affect recovery from stroke and head injury, but the underlying mechanisms and interconnections between them are yet unknown. Here, we report that similar cation channels mediate the protective effect of exercise and specific genetic background in a kainate injection model of cerebellar stroke. Microinjection to the cerebellum of the glutamatergic agonist, kainate, creates glutamatergic excito\xE2\x80\x90toxicity characteristic of focal stroke, head injury or alcoholism. Inherited protection and prior exercise were both accompanied by higher cerebellar expression levels of the Kir6.1 ATP-dependent potassium channel in adjacent Bergmann glia, and voltage-gated KVbeta2 and cyclic nucleotide-gated cation HCN1 channels in basket cells. Sedentary FVB/N and exercised C57BL/6 mice both expressed higher levels of these cation channels compared to sedentary C57BL/6 mice, and were both found to be less sensitive to glutamate toxicity. Moreover, blocking ATP-dependent potassium channels with Glibenclamide enhanced kainate-induced cell death in cerebellar slices from the resilient sedentary FVB/N mice. Furthermore, exercise increased the number of acetylcholinesterase-positive fibres in the molecular layer, reduced cerebellar cytokine levels and suppressed serum acetylcholinesterase activity, suggesting anti-inflammatory protection by enhanced cholinergic signalling. Our findings demonstrate for the first time that routine exercise and specific genetic backgrounds confer protection from cerebellar glutamatergic damages by similar molecular mechanisms, including elevated expression of cation channels. In addition, our findings highlight the involvement of the cholinergic anti-inflammatory pathway in insult-inducible cerebellar processes. These mechanisms are likely to play similar roles in other brain regions and injuries as well, opening new venues for targeted research efforts.


Subject(s)
Cerebellum/metabolism , Cyclic Nucleotide-Gated Cation Channels/metabolism , KATP Channels/metabolism , Potassium Channels/metabolism , Shaker Superfamily of Potassium Channels/metabolism , Stroke/metabolism , Animals , Cerebellum/drug effects , Cerebellum/physiopathology , Cyclic Nucleotide-Gated Cation Channels/genetics , Excitatory Amino Acid Agonists/toxicity , Gene Expression Profiling , Glutamates/metabolism , Glyburide/pharmacology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Injections, Intraventricular , KATP Channels/antagonists & inhibitors , KATP Channels/genetics , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal , Potassium Channels/genetics , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Shaker Superfamily of Potassium Channels/genetics , Stroke/genetics , Stroke/physiopathology , Up-Regulation
3.
Neurodegener Dis ; 9(2): 87-98, 2012.
Article in English | MEDLINE | ID: mdl-22042332

ABSTRACT

BACKGROUND/OBJECTIVE: Environmental exposure to anti-acetylcholinesterases (AChEs) aggravates the risk of Parkinsonism due to currently unclear mechanism(s). We explored the possibility that the brain's capacity to induce a widespread adaptive alternative splicing response to such exposure may be involved. METHODS: Following exposure to the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), brain region transcriptome profiles were tested. RESULTS: Changes in transcript profiles, alternative splicing patterns and splicing-related gene categories were identified. Engineered mice over-expressing the protective AChE-R splice variant showed less total changes but more splicing-related ones than hypersensitive AChE-S over-expressors with similarly increased hydrolytic activities. Following MPTP exposure, the substantia nigra and prefrontal cortex (PFC) of both strains showed a nuclear increase in the splicing factor ASF/SF2 protein. Furthermore, intravenous injection with highly purified recombinant human AChE-R changed transcript profiles in the striatum. CONCLUSIONS: Our findings are compatible with the working hypothesis that inherited or acquired alternative splicing deficits may promote parkinsonism, and we propose adaptive alternative splicing as a strategy for attenuating its progression.


Subject(s)
Acetylcholinesterase/genetics , Alternative Splicing/genetics , Parkinsonian Disorders/genetics , Adaptation, Biological , Animals , Cluster Analysis , Environmental Exposure/adverse effects , Gene Expression Profiling , Humans , Immunohistochemistry , Isoenzymes , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Parkinsonian Disorders/enzymology , Real-Time Polymerase Chain Reaction , Recombinant Proteins/genetics , Risk Factors
4.
Biochim Biophys Acta ; 1823(2): 368-78, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22093924

ABSTRACT

Cholinergic signaling suppresses inflammation in blood and brain and attenuates apoptosis in other tissues, but whether it blocks inflammation in skeletal muscle under toxicant exposure, injuries and diseases remained unexplored. Here, we report nicotinic attenuation of inflammation and alteration of apoptotic protein expression pattern in murine muscle tissue and cultured myotubes, involving the RNA-binding protein, Tristetraprolin, and the anti-apoptotic protein, Mcl-1. In muscles and C2C12 myotubes, cholinergic excitation by exposure to nicotine or the organophosphorous pesticide, Paraoxon, induced Tristetraprolin overproduction while reducing pro-inflammatory transcripts such as IL-6, CXCL1 (KC) and CCL2 (MCP-1). Furthermore, nicotinic excitation under exposure to the bacterial endotoxin LPS attenuated over-expression of the CCL2 and suppressed the transcriptional activity of NF-ĸB and AP-1. Tristetraprolin was essential for this anti-inflammatory effect of nicotine in basal conditions. However, its knockdown also impaired the pro-inflammatory response to LPS. Finally, in vivo administration of Paraoxon or recombinant Acetylcholinesterase, leading respectively to either gain or loss of cholinergic signaling, modified muscle expression of key mRNA processing factors and several of their apoptosis-related targets. Specifically, cholinergic imbalances enhanced the kinase activators of the Serine-Arginine splicing kinases, Clk1 and Clk3. Moreover, Paraoxon raised the levels of the anti-apoptotic protein, Mcl-1, through a previously unrecognized polyadenylation site selection mechanism, producing longer, less stable Mcl-1 mRNA transcripts. Together, our findings demonstrate that in addition to activating muscle function, acetylcholine regulates muscle inflammation and cell survival, and point to Tristetraprolin and the choice of Mcl-1 mRNA polyadenylation sites as potential key players in muscle reactions to insults.


Subject(s)
Inflammation/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Nicotine/pharmacology , Tristetraprolin/metabolism , Animals , Apoptosis/physiology , Cell Line , Cholinesterase Inhibitors/pharmacology , Cytokines/metabolism , Gene Expression Profiling , Inflammation/metabolism , Male , Mice , Microarray Analysis , Myeloid Cell Leukemia Sequence 1 Protein , Nicotinic Agonists/pharmacology , Paraoxon/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tristetraprolin/genetics
5.
Neurobiol Dis ; 40(1): 348-59, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20600907

ABSTRACT

Down syndrome (DS) associates with impaired brain functions, but the underlying mechanism(s) are yet unclear. The "gene dosage" hypothesis predicts that in DS, overexpression of a single gene can impair multiple brain functions through a signal amplification effect due to impaired regulatory mechanism(s). Here, we report findings attributing to impairments in the splicing process such a regulatory role. We have used DS fetal brain samples in search for initial evidence and employed engineered mice with MMU16 partial trisomy (Ts65Dn) or direct excess of the splicing-associated nuclear kinase Dyrk1A, overdosed in DS for further analyses. We present specific albeit modest changes in the DS brain's splicing machinery with subsequently amplified effects in target transcripts; and we demonstrate that engineered excess of Dyrk1A can largely recapitulate these changes. Specifically, in both the fetal DS brains and the Dyrk1A overdose models, we found ample modestly modified splicing-associated transcripts which apparently induced secondary enhancement in exon inclusion of key synaptic transcripts. Thus, DS-reduced levels of the dominant-negative TRKBT1 transcript, but not other TRKB mRNA transcripts, were accompanied by corresponding decreases in BDNF. In addition, the DS brains and Dyrk1A overdosage models showed selective changes in the transcripts composition of neuroligin mRNAs as well as reductions in the "synaptic" acetylcholinesterase variant AChE-S mRNA and corresponding increases in the stress-inducible AChE-R mRNA variant, yielding key synaptic proteins with unusual features. In cotransfected cells, Dyrk1A overdosage caused parallel changes in the splicing pattern of an AChE mini-gene, suggesting that Dyrk1A overdosage is both essential and sufficient to induce the observed change in the composition of AChE mRNA variants. Furthermore, the Dyrk1A overdosage animal models showed pronounced changes in the structure of neuronal nuclear speckles, where splicing events take place and in SR proteins phosphorylation known to be required for the splicing process. Together, our findings demonstrate DS-like brain splicing machinery malfunctioning in Dyrk1A overexpressing mice. Since individual splicing choices may alter cell fate determination, axon guidance, and synaptogenesis, these findings suggest the retrieval of balanced splicing as a goal for DS therapeutic manipulations early in DS development.


Subject(s)
Alternative Splicing/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Down Syndrome/genetics , Down Syndrome/metabolism , Gene Dosage/genetics , Genetic Engineering/methods , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Acetylcholinesterase/genetics , Animals , Cells, Cultured , Cerebral Cortex/embryology , Down Syndrome/embryology , Female , Gene Expression Regulation, Developmental/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , RNA Splicing/genetics , Dyrk Kinases
6.
J Biol Chem ; 284(21): 14303-15, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19282290

ABSTRACT

Almost every protein-coding gene undergoes pre-mRNA splicing, and the majority of these pre-mRNAs are alternatively spliced. Alternative exon usage is regulated by the transient formation of protein complexes on the pre-mRNA that typically contain heterogeneous nuclear ribonucleoproteins (hnRNPs). Here we characterize hnRNP G, a member of the hnRNP class of proteins. We show that hnRNP G is a nuclear protein that is expressed in different concentrations in various tissues and that interacts with other splicing regulatory proteins. hnRNP G is part of the supraspliceosome, where it regulates alternative splice site selection in a concentration-dependent manner. Its action on alternative exons can occur without a functional RNA-recognition motif by binding to other splicing regulatory proteins. The RNA-recognition motif of hnRNP G binds to a loose consensus sequence containing a CC(A/C) motif, and hnRNP G preferentially regulates alternative exons where this motif is clustered in close proximity. The X-chromosomally encoded hnRNP G regulates different RNAs than its Y-chromosomal paralogue RNA-binding motif protein, Y-linked (RBMY), suggesting that differences in alternative splicing, evoked by the sex-specific expression of hnRNP G and RBMY, could contribute to molecular sex differences in mammals.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA Precursors/genetics , RNA Splice Sites/genetics , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Exons/genetics , Genes, Reporter , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , RNA Splicing/genetics , Rats , Spliceosomes/metabolism
7.
J Neurochem ; 97 Suppl 1: 24-34, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16635247

ABSTRACT

Alternative splicing configurations and acetylcholinesterase (AChE) gene expression are both modified in neurons under stress. However, it is unclear if these phenomena are functionally interrelated. Using a home-made spotted microarray focused on splicing-associated transcripts, we tested the effects of excess 3' splice variants of human AChE on splicing-related gene expression in semi-differentiated neuronal P19 cells. Of the tested transcripts, 17.3% and 20.2% showed modified expression levels (log2 of the ratio<-0.3 or>0.3) in transfected P19 cells overexpressing the stress-inducible AChE-R variant or the synaptic AChE-S protein, respectively. Multiple transcripts encoding serine-arginine rich (SR) and SR-related splicing regulators were suppressed in cells expressing either of these variants, whereas the gene groups including splicing-related helicases and transcripts involved in apoptosis displayed variant-specific changes. Our findings are compatible with the assumption that both neuronal overexpression and alternative splicing of pre-AChE mRNA may be causally involved in initiating global changes in neuronal alternative splicing, causing subsequent modifications in the expression patterns of numerous target genes.


Subject(s)
Acetylcholinesterase/genetics , Alternative Splicing/genetics , Gene Expression/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Embryo, Mammalian , Enzyme Precursors/genetics , Humans , Mice , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Spliceosomes/genetics , Transfection , Vesicular Acetylcholine Transport Proteins/genetics
8.
FASEB J ; 19(8): 910-22, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923401

ABSTRACT

Cholinergic imbalances occur after traumatic effects and in the initial stages of neurodegenerative diseases, but their long-lasting effects remained largely unexplained. To address this, we used TgS transgenic mice constitutively overexpressing synaptic acetylcholinesterase (AChE-S) and presenting a complex phenotype of progressive neurodeterioration. T1- and T2-weighted magnetic resonance (MR) brain images appeared similar. However, diffusion-weighted MRI showed decreased baseline water apparent diffusion coefficient in the brains of TgS animals. Furthermore, contrast-enhanced MRI after gadolinium diethylenetriaminepentaacetic acid (Gd-DTPA) injection demonstrated slower recovery of normal signals in the TgS brains than with controls. Perfusion MR imaging and difference T1 maps calculated from pre- postcontrast T1-weighted MR images indicated accumulation of more Gd-DTPA molecules in the TgS brains than in the parent strain, reflecting impaired blood-brain barrier (BBB) functioning in these transgenic mice. To explore the molecular mechanism(s) underlying these global phenotypes, we performed microarray analysis in the stress-controlling prefrontal cortex of TgS vs. strain-matched wild-type animals. Profound overexpression of numerous ion channels, transporters, and adhesion genes was confirmed by real time RT-PCR tests. Immunohistochemical and immunoblot analyses revealed corresponding increases in the level and cellular distributions of the chloride channel CLCN3 and the water channel AQP4, both of which contribute to BBB maintenance. Our study attributes to balanced cholinergic neurotransmission, a central role in the brain's maintenance of water diffusion and ion transport, and indicates that chronic impairments in this maintenance facilitate neurodeterioration through interference with BBB function.


Subject(s)
Acetylcholinesterase/physiology , Brain/metabolism , Neurodegenerative Diseases/etiology , Acetylcholine/metabolism , Acetylcholinesterase/genetics , Animals , Aquaporin 4/analysis , Biological Transport , Blood-Brain Barrier , Chloride Channels/analysis , Diffusion , Female , Gene Expression , Ion Transport , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Neurotransmitter Agents , Oligonucleotide Array Sequence Analysis , Prefrontal Cortex/chemistry , RNA, Messenger/analysis , Receptors, Cholinergic/physiology , Synapses/enzymology , Water/metabolism
9.
Gene ; 344: 1-20, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15656968

ABSTRACT

Alternative splicing is one of the most important mechanisms to generate a large number of mRNA and protein isoforms from the surprisingly low number of human genes. Unlike promoter activity, which primarily regulates the amount of transcripts, alternative splicing changes the structure of transcripts and their encoded proteins. Together with nonsense-mediated decay (NMD), at least 25% of all alternative exons are predicted to regulate transcript abundance. Molecular analyses during the last decade demonstrate that alternative splicing determines the binding properties, intracellular localization, enzymatic activity, protein stability and posttranslational modifications of a large number of proteins. The magnitude of the effects range from a complete loss of function or acquisition of a new function to very subtle modulations, which are observed in the majority of cases reported. Alternative splicing factors regulate multiple pre-mRNAs and recent identification of physiological targets shows that a specific splicing factor regulates pre-mRNAs with coherent biological functions. Therefore, evidence is now accumulating that alternative splicing coordinates physiologically meaningful changes in protein isoform expression and is a key mechanism to generate the complex proteome of multicellular organisms.


Subject(s)
Alternative Splicing/genetics , Animals , Binding Sites/genetics , Databases, Genetic , Humans , Internet , Mutation/genetics , Protein Processing, Post-Translational/genetics , RNA Precursors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...