Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Adv Genet ; 90: 103-131, 2015.
Article in English | MEDLINE | ID: mdl-26296934

ABSTRACT

MicroRNAs (miRNAs) are small regulatory RNAs that modulate the translation of mRNA. They have emerged over the past few years as indispensable entities in the transcriptional regulation of genes. Their discovery has added additional layers of complexity to regulatory networks that control cellular homeostasis. Also, their dysregulated pattern of expression is now well demonstrated in myriad diseases and pathogenic processes. In the current review, we highlight the role of miRNAs in Lesch-Nyhan disease (LND), a rare neurogenetic syndrome caused by mutations in the purine metabolic gene encoding the hypoxanthine-guanine phosphoribosyltransferase (HPRT) enzyme. We describe how experimental and biocomputational approaches have helped to unravel genetic and signaling pathways that provide mechanistic understanding of some of the molecular and cellular basis of this ill-defined neurogenetic disorder. Through miRNA-based target predictions, we have identified signaling pathways that may be of significance in guiding biological therapeutic discovery for this incurable neurological disorder. We also propose a model to explain how a gene such as HPRT, mostly known for its housekeeping metabolic functions, can have pleiotropic effects on disparate genes and signal transduction pathways. Our hypothetical model suggests that HPRT mRNA transcripts may be acting as competitive endogenous RNAs (ceRNAs) intertwined in multiregulatory cross talk between key neural transcripts and miRNAs. Overall, this approach of using miRNA-based genomic approaches to elucidate the molecular and cellular basis of LND and guide biological target identification might be applicable to other ill-defined rare inborn-error metabolic diseases.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/therapy , MicroRNAs/therapeutic use , Animals , Gene Expression Regulation , Humans , Lesch-Nyhan Syndrome/genetics , RNA, Messenger/genetics
2.
PLoS One ; 9(5): e96575, 2014.
Article in English | MEDLINE | ID: mdl-24804781

ABSTRACT

Lesch-Nyhan Syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). This syndrome is characterized by an array of severe neurological impairments that in part originate from striatal dysfunctions. However, the molecular and cellular mechanisms underlying these dysfunctions remain largely unidentified. In this report, we demonstrate that HPRT-deficiency causes dysregulated expression of key genes essential for striatal patterning, most notably the striatally-enriched transcription factor B-cell leukemia 11b (Bcl11b). The data also reveal that the down-regulated expression of Bcl11b in HPRT-deficient immortalized mouse striatal (STHdh) neural stem cells is accompanied by aberrant expression of some of its transcriptional partners and other striatally-enriched genes, including the gene encoding dopamine- and cAMP-regulated phosphoprotein 32, (DARPP-32). Furthermore, we demonstrate that components of the BDNF/TrkB signaling, a known activator of DARPP-32 striatal expression and effector of Bcl11b transcriptional activation are markedly increased in HPRT-deficient cells and in the striatum of HPRT knockout mouse. Consequently, the HPRT-deficient cells display superior protection against reactive oxygen species (ROS)-mediated cell death upon exposure to hydrogen peroxide. These findings suggest that the purine metabolic defect caused by HPRT-deficiency, while it may provide neuroprotection to striatal neurons, affects key genes and signaling pathways that may underlie the neuropathogenesis of LNS.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Corpus Striatum/pathology , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/genetics , Receptor, trkB/genetics , Signal Transduction/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Death/genetics , Cell Differentiation/genetics , Corpus Striatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Lesch-Nyhan Syndrome/metabolism , Lesch-Nyhan Syndrome/pathology , Mice , Mice, Knockout , Neurons/metabolism , Reactive Oxygen Species/metabolism , Receptor, trkB/metabolism
3.
Hum Mol Genet ; 22(22): 4502-15, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23804752

ABSTRACT

Lesch-Nyhan syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). A series of motor, cognitive and neurobehavioral anomalies characterize this disease phenotype, which is still poorly understood. The clinical manifestations of this syndrome are believed to be the consequences of deficiencies in neurodevelopmental pathways that lead to disordered brain function. We have used microRNA array and gene ontology analysis to evaluate the gene expression of differentiating HPRT-deficient human neuron-like cell lines. We set out to identify dysregulated genes implicated in purine-based cellular functions. Our approach was based on the premise that HPRT deficiency affects preeminently the expression and the function of purine-based molecular complexes, such as guanine nucleotide exchange factors (GEFs) and small GTPases. We found that several microRNAs from the miR-17 family cluster and genes encoding GEF are dysregulated in HPRT deficiency. Most notably, our data show that the expression of the exchange protein activated by cAMP (EPAC) is blunted in HPRT-deficient human neuron-like cell lines and fibroblast cells from LNS patients, and is altered in the cortex, striatum and midbrain of HPRT knockout mouse. We also show a marked impairment in the activation of small GTPase RAP1 in the HPRT-deficient cells, as well as differences in cytoskeleton dynamics that lead to increased motility for HPRT-deficient neuron-like cell lines relative to control. We propose that the alterations in EPAC/RAP1 signaling and cell migration in HPRT deficiency are crucial for neuro-developmental events that may contribute to the neurological dysfunctions in LNS.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Lesch-Nyhan Syndrome/genetics , MicroRNAs/genetics , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Movement/physiology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Cytoskeleton/metabolism , Gene Ontology , Guanine Nucleotide Exchange Factors/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Male , Mesencephalon/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , Multigene Family , Oligonucleotide Array Sequence Analysis , Signal Transduction , rap1 GTP-Binding Proteins/genetics
4.
PLoS One ; 8(5): e63333, 2013.
Article in English | MEDLINE | ID: mdl-23691025

ABSTRACT

Lesch-Nyhan Disease (LND) is the result of mutations in the X-linked gene encoding the purine metabolic enzyme, hypoxanthine guanine phosphoribosyl transferase (HPRT). LND gives rise to severe neurological anomalies including mental retardation, dystonia, chorea, pyramidal signs and a compulsive and aggressive behavior to self injure. The neurological phenotype in LND has been shown to reflect aberrant dopaminergic signaling in the basal ganglia, however there are little data correlating the defect in purine metabolism to the neural-related abnormalities. In the present studies, we find that HPRT-deficient neuronal cell lines have reduced CREB (cAMP response element-binding protein) expression and intracellular cyclic AMP (cAMP), which correlates with attenuated CREB-dependent transcriptional activity and a reduced phosphorylation of protein kinase A (PKA) substrates such as synapsin (p-syn I). Of interest, we found increased expression of phosphodiesterase 10A (PDE10A) in HPRT-deficient cell lines and that the PDE10 inhibitor papaverine and PDE10A siRNA restored cAMP/PKA signaling. Furthermore, reconstitution of HPRT expression in mutant cells partly increased cAMP signaling synapsin phosphorylation. In conclusion, our data show that HPRT-deficiency alters cAMP/PKA signaling pathway, which is in part due to the increased of PDE10A expression and activity. These findings suggest a mechanistic insight into the possible causes of LND and highlight PDE10A as a possible therapeutic target for this intractable neurological disease.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation, Enzymologic , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/drug therapy , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Knockdown Techniques , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Lesch-Nyhan Syndrome/enzymology , Lesch-Nyhan Syndrome/pathology , MicroRNAs/genetics , Molecular Targeted Therapy , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapsins/genetics , Transcription, Genetic
5.
Hum Mol Genet ; 21(3): 609-22, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22042773

ABSTRACT

Mutations in the gene encoding the purine biosynthetic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) cause the intractable neurodevelopmental Lesch-Nyhan disease (LND) associated with aberrant development of brain dopamine pathways. In the current study, we have identified an increased expression of the microRNA miR181a in HPRT-deficient human dopaminergic SH-SY5Y neuroblastoma cells. Among the genes potentially regulated by miR181a are several known to be required for neural development, including Engrailed1 (En1), Engrailed2 (En2), Lmx1a and Brn2. We demonstrate that these genes are down-regulated in HPRT-deficient SH-SY5Y cells and that over-expression of miR181a significantly reduces endogenous expression of these genes and inhibits translation of luciferase plasmids bearing the En1/2 or Lmx1a 3'UTR miRNA-binding elements. Conversely, inhibition of miR181a increases the expression of these genes and enhances translation of luciferase constructs bearing the En1/2 and Lmx1a 3'UTR miRNA-binding sequences. We also demonstrate that key neurodevelopmental genes (e.g. Nurr1, Pitx3, Wnt1 and Mash1) known to be functional partners of Lmx1a and Brn2 are also markedly down-regulated in SH-SY5Y cells over-expressing miR181a and in HPRT-deficient cells. Our findings in SH-SY5Y cells demonstrate that HPRT deficiency is accompanied by dysregulation of some of the important pathways that regulate the development of dopaminergic neurons and dopamine pathways and that this defect is associated with and possibly due at least partly to aberrant expression of miR181a. Because aberrant expression of miR181a is not as apparent in HPRT-deficient LND fibroblasts, the relevance of the SH-SY5Y neuroblastoma cells to human disease remains to be proven. Nevertheless, we propose that these pleiotropic neurodevelopment effects of miR181a may play a role in the pathogenesis of LND.


Subject(s)
Dopaminergic Neurons/metabolism , Gene Expression Regulation , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/genetics , MicroRNAs/metabolism , 3' Untranslated Regions , Cell Line, Tumor , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/antagonists & inhibitors , Hypoxanthine Phosphoribosyltransferase/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Lesch-Nyhan Syndrome/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
PLoS One ; 6(1): e16572, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305049

ABSTRACT

We have used microarray-based methods of global gene expression together with quantitative PCR and Western blot analysis to identify dysregulation of genes and aberrant cellular processes in human fibroblasts and in SH-SY5Y neuroblastoma cells made HPRT-deficient by transduction with a retrovirus stably expressing an shRNA targeted against HPRT. Analysis of the microarray expression data by Gene ontology (GO) and Gene Set Enrichment Analysis (GSEA) as well as significant pathway analysis by GeneSpring GX10 and Panther Classification System reveal that HPRT deficiency is accompanied by aberrations in a variety of pathways known to regulate neurogenesis or to be implicated in neurodegenerative disease, including the canonical Wnt/ß-catenin and the Alzheimer's disease/presenilin signaling pathways. Dysregulation of the Wnt/ß-catenin pathway is confirmed by Western blot demonstration of cytosolic sequestration of ß-catenin during in vitro differentiation of the SH-SY5Y cells toward the neuronal phenotype. We also demonstrate that two key transcription factor genes known to be regulated by Wnt signaling and to be vital for the generation and function of dopaminergic neurons; i.e., Lmx1a and Engrailed 1, are down-regulated in the HPRT knockdown SH-SY5Y cells. In addition to the Wnt signaling aberration, we found that expression of presenilin-1 shows severely aberrant expression in HPRT-deficient SH-SY5Y cells, reflected by marked deficiency of the 23 kDa C-terminal fragment of presenilin-1 in knockdown cells. Western blot analysis of primary fibroblast cultures from two LND patients also shows dysregulated presenilin-1 expression, including aberrant proteolytic processing of presenilin-1. These demonstrations of dysregulated Wnt signaling and presenilin-1 expression together with impaired expression of dopaminergic transcription factors reveal broad pleitropic neuro-regulatory defects played by HPRT expression and suggest new directions for investigating mechanisms of aberrant neurogenesis and neuropathology in LND and potential new targets for restoration of effective signaling in this neuro-developmental defect.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Neurogenesis/genetics , Presenilin-1/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , Cell Line, Tumor , Dopamine , Gene Expression Profiling/methods , Humans , Lesch-Nyhan Syndrome , Transcription Factors
7.
Mol Ther ; 18(1): 54-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19672249

ABSTRACT

Neuronal transcription factors play vital roles in the specification and development of neurons, including dopaminergic (DA) neurons. Mutations in the gene encoding the purine biosynthetic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT) cause the resulting intractable and largely untreatable neurological impairment of Lesch-Nyhan disease (LND). The disorder is associated with a defect in basal ganglia DA pathways. The mechanisms connecting the purine metabolic defect and the central nervous system (CNS) phenotype are poorly understood but have been presumed to reflect a developmental defect of DA neurons. We have examined the effect of HPRT deficiency on the differentiation of neurons in the well-established human (NT2) embryonic carcinoma neurogenesis model. We have used a retrovirus expressing a small hairpin RNA (shRNA) to knock down HPRT gene expression and have examined the expression of a number of transcription factors essential for neuronal differentiation and marker genes involved in DA biosynthetic pathway. HPRT-deficient NT2 cells demonstrate aberrant expression of several transcription factors and DA markers. Although differentiated HPRT-deficient neurons also demonstrate a striking deficit in neurite outgrowth during differentiation, resulting neurons demonstrate wild-type electrophysiological properties. These results represent direct experimental evidence for aberrant neurogenesis in HPRT deficiency and suggest developmental roles for other housekeeping genes in neurodevelopmental disease.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/metabolism , Neurogenesis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Electrophysiology , Humans , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Neurites/metabolism , Neurons/cytology , Neurons/metabolism
8.
J Virol Methods ; 148(1-2): 277-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18207578

ABSTRACT

The current study reports the production of baculoviral-virosomal vectors consisting of lipoplexes and of the viral glycoprotein (GP64) of baculovirus Autographa californica multiple nucleopolyhdrovirus (AcMNPV). This study demonstrates that such complexes have an increased transfection capability in a number of cells, including undifferentiated H9 human embryonic stem H9hES cells compared to lipoplexes alone. The GP64-mediated enhancement of gene transfer of lipoplexes is inhibited by the addition of anti-GP64 neutralizing antibody and by a modified GP64 protein, but is however less potent than vesicular stomatitis virus glycoprotein (VSV-G)-mediated enhancement of gene transfer of lipoplexes. This difference may be explained in part by the dissimilarity in the fusogenic properties of their respective viral glycoprotein.


Subject(s)
Liposomes/metabolism , Nucleopolyhedroviruses/genetics , Transfection/methods , Viral Envelope Proteins/biosynthesis , Virosomes/biosynthesis , Cell Line , Humans , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL