Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mol Psychiatry ; 20(6): 677-684, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25824307

ABSTRACT

The human genome project has revolutionized our understanding of the underlying mechanisms in psychiatric disease. It is now abundantly clear that neurobehavioral phenotypes are epigenetically controlled by noncoding RNAs (ncRNAs). The microRNA (miRNA) class of ncRNAs are ubiquitously expressed throughout the brain and govern all major neuronal pathways. The attractive therapeutic potential of miRNAs is underscored by their pleiotropic capacities, putatively targeting multiple pathways within a single neuron. Many psychiatric diseases stem from a multifactorial origin, thus conventional drug targeting of single proteins may not prove most effective. In this exciting post-genome sequencing era, many new epigenetic targets are emerging for therapeutic investigation. Here we review the reported roles of miRNAs, as well as other ncRNA classes, in the pathology of psychiatric disorders; there are both common and unique ncRNA mechanisms that influence the various diagnoses. Collectively, these potent epigenetic regulators may clarify the disrupted signaling networks in psychiatric phenotypes.


Subject(s)
Cognition Disorders/etiology , Mental Disorders/complications , Mental Disorders/genetics , Nervous System Diseases/etiology , RNA, Untranslated/genetics , Animals , Humans , RNA, Untranslated/metabolism
2.
Nature ; 466(7303): 197-202, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20613834

ABSTRACT

Cocaine addiction is characterized by a gradual loss of control over drug use, but the molecular mechanisms regulating vulnerability to this process remain unclear. Here we report that microRNA-212 (miR-212) is upregulated in the dorsal striatum of rats with a history of extended access to cocaine. Striatal miR-212 decreases responsiveness to the motivational properties of cocaine by markedly amplifying the stimulatory effects of the drug on cAMP response element binding protein (CREB) signalling. This action occurs through miR-212-enhanced Raf1 activity, resulting in adenylyl cyclase sensitization and increased expression of the essential CREB co-activator TORC (transducer of regulated CREB; also known as CRTC). Our findings indicate that striatal miR-212 signalling has a key role in determining vulnerability to cocaine addiction, reveal new molecular regulators that control the complex actions of cocaine in brain reward circuitries and provide an entirely new direction for the development of anti-addiction therapeutics based on the modulation of noncoding RNAs.


Subject(s)
Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Cocaine/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , MicroRNAs/metabolism , Neostriatum/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Animals , Cocaine/pharmacology , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/enzymology , MAP Kinase Kinase Kinases/metabolism , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neostriatum/drug effects , Proto-Oncogene Proteins c-raf , Rats , Rats, Wistar , Reward , Signal Transduction/drug effects , Transcription Factors/metabolism , Up-Regulation/drug effects
3.
BMC Neurosci ; 15: 36, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24581271

ABSTRACT

BACKGROUND: A two-year longitudinal study composed of morphometric MRI measures and cognitive behavioral evaluation was performed on a transgenic Huntington's disease (HD) monkey. rHD1, a transgenic HD monkey expressing exon 1 of the human gene encoding huntingtin (HTT) with 29 CAG repeats regulated by a human polyubiquitin C promoter was used together with four age-matched wild-type control monkeys. This is the first study on a primate model of human HD based on longitudinal clinical measurements. RESULTS: Changes in striatal and hippocampal volumes in rHD1 were observed with progressive impairment in motor functions and cognitive decline, including deficits in learning stimulus-reward associations, recognition memory and spatial memory. The results demonstrate a progressive cognitive decline and morphometric changes in the striatum and hippocampus in a transgenic HD monkey. CONCLUSIONS: This is the first study on a primate model of human HD based on longitudinal clinical measurements. While this study is based a single HD monkey, an ongoing longitudinal study with additional HD monkeys will be important for the confirmation of our findings. A nonhuman primate model of HD could complement other animal models of HD to better understand the pathogenesis of HD and future development of diagnostics and therapeutics through longitudinal assessment.


Subject(s)
Brain/metabolism , Brain/pathology , Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/pathology , Nerve Tissue Proteins/genetics , Aging/genetics , Aging/pathology , Animals , Animals, Genetically Modified , Humans , Huntingtin Protein , Longitudinal Studies , Macaca mulatta , Male , Organ Size/genetics , Tissue Distribution
4.
BMC Neurosci ; 14: 88, 2013 Aug 17.
Article in English | MEDLINE | ID: mdl-23957861

ABSTRACT

BACKGROUND: Huntington's Disease (HD) is a progressive neurodegenerative disorder caused by an expansion in the polyglutamine (polyQ) region of the Huntingtin (HTT) gene. The clinical features of HD are characterized by cognitive, psychological, and motor deficits. Molecular instability, a core component in neurological disease progression, can be comprehensively evaluated through longitudinal transcriptomic profiling. Development of animal models amenable to longitudinal examination enables distinct disease-associated mechanisms to be identified. RESULTS: Here we report the first longitudinal study of transgenic monkeys with genomic integration of various lengths of the human HTT gene and a range of polyQ repeats. With this unique group of transgenic HD nonhuman primates (HD monkeys), we profiled over 47,000 transcripts from peripheral blood collected over a 2 year timespan from HD monkeys and age-matched wild-type control monkeys. CONCLUSIONS: Messenger RNAs with expression patterns which diverged with disease progression in the HD monkeys considerably facilitated our search for transcripts with diagnostic or therapeutic potential in the blood of human HD patients, opening up a new avenue for clinical investigation.


Subject(s)
Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Transcriptome , Animals , Animals, Genetically Modified , Disease Models, Animal , Disease Progression , Female , Gene Expression Profiling , Humans , Huntingtin Protein , Huntington Disease/blood , Longitudinal Studies , Macaca mulatta , Male , RNA, Messenger/metabolism
5.
Proc Natl Acad Sci U S A ; 106(9): 3507-12, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19196972

ABSTRACT

N-methyl-D-aspartate (NMDA) glutamate receptors are regulators of fast neurotransmission and synaptic plasticity in the brain. Disruption of NMDA-mediated glutamate signaling has been linked to behavioral deficits displayed in psychiatric disorders such as schizophrenia. Recently, noncoding RNA molecules such as microRNAs (miRNAs) have emerged as critical regulators of neuronal functions. Here we show that pharmacological (dizocilpine) or genetic (NR1 hypomorphism) disruption of NMDA receptor signaling reduces levels of a brain-specific miRNA, miR-219, in the prefrontal cortex (PFC) of mice. Consistent with a role for miR-219 in NMDA receptor signaling, we identify calcium/calmodulin-dependent protein kinase II gamma subunit (CaMKIIgamma), a component of the NMDA receptor signaling cascade, as a target of miR-219. In vivo inhibition of miR-219 by specific antimiR in the murine brain significantly modulated behavioral responses associated with disrupted NMDA receptor transmission. Furthermore, pretreatment with the antipsychotic drugs haloperidol and clozapine prevented dizocilpine-induced effects on miR-219. Taken together, these data support an integral role for miR-219 in the expression of behavioral aberrations associated with NMDA receptor hypofunction.


Subject(s)
Genetic Therapy , MicroRNAs/genetics , MicroRNAs/therapeutic use , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Behavior, Animal/drug effects , Biological Transport , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line, Tumor , Dizocilpine Maleate/pharmacology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Nervous System Diseases/physiopathology , Nervous System Diseases/therapy , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction
6.
Cells ; 11(12)2022 06 09.
Article in English | MEDLINE | ID: mdl-35741010

ABSTRACT

The change from viewing noncoding RNA as "junk" in the genome to seeing it as a critical epigenetic regulator in almost every human condition or disease has forced a paradigm shift in biomedical and clinical research. Small and long noncoding RNA transcripts are now routinely evaluated as putative diagnostic or therapeutic agents. A prominent role for noncoding microRNAs in the central nervous system has uncovered promising new clinical candidates for dementia-related disorders, treatments for which currently remain elusive even as the percentage of diagnosed patients increases significantly. Cognitive decline is a core neurodegenerative process in Alzheimer's Disease, Frontotemporal Dementia, Lewy body dementia, vascular dementia, Huntington's Disease, Creutzfeldt-Jakob disease, and a significant portion of Parkinson's Disease patients. This review will discuss the microRNA-associated networks which influence these pathologies, including inflammatory and viral-mediated pathways (such as the novel SARS-CoV-2 virus implicated in COVID-19), and their current status in clinical trials.


Subject(s)
Cognition , Dementia , MicroRNAs , COVID-19 , Dementia/genetics , Humans , MicroRNAs/genetics
7.
BMC Genomics ; 12: 527, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-22032330

ABSTRACT

BACKGROUND: Frontotemporal lobar degeneration (FTLD) is a progressive neurodegenerative disorder that can be triggered through genetic or sporadic mechanisms. MicroRNAs (miRNAs) have become a major therapeutic focus as their pervasive expression and powerful regulatory roles in disease pathogenesis become increasingly apparent. Here we examine the role of miRNAs in FTLD patients with TAR DNA-binding protein 43 pathology (FTLD-TDP) caused by genetic mutations in the progranulin (PGRN) gene. RESULTS: Using miRNA array profiling, we identified the 20 miRNAs that showed greatest evidence (unadjusted P < 0.05) of dysregulation in frontal cortex of eight FTLD-TDP patients carrying PGRN mutations when compared to 32 FTLD-TDP patients with no apparent genetic abnormalities. Quantitative real-time PCR (qRT-PCR) analyses provided technical validation of the differential expression for 9 of the 20 miRNAs in frontal cortex. Additional qRT-PCR analyses showed that 5 out of 9 miRNAs (miR-922, miR-516a-3p, miR-571, miR-548b-5p, and miR-548c-5p) were also significantly dysregulated (unadjusted P < 0.05) in cerebellar tissue samples of PGRN mutation carriers, consistent with a systemic reduction in PGRN levels. We developed a list of gene targets for the 5 candidate miRNAs and found 18 genes dysregulated in a reported FTLD mRNA study to exhibit anti-correlated miRNA-mRNA patterns in affected cortex and cerebellar tissue. Among the targets is brain-specific angiogenesis inhibitor 3, which was recently identified as an important player in synapse biology. CONCLUSIONS: Our study suggests that miRNAs may contribute to the pathogenesis of FTLD-TDP caused by PGRN mutations and provides new insight into potential future therapeutic options.


Subject(s)
DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , MicroRNAs/metabolism , Aged , Aged, 80 and over , Cerebellum/metabolism , Cerebellum/pathology , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Frontotemporal Lobar Degeneration/pathology , Gene Expression Profiling , Humans , Male , MicroRNAs/genetics , Middle Aged , Mutation , Oligonucleotide Array Sequence Analysis , Progranulins , Protein Precursors/genetics
8.
Hum Mutat ; 31(5): E1377-89, 2010 May.
Article in English | MEDLINE | ID: mdl-20232451

ABSTRACT

Mutations in the gene encoding fused in sarcoma (FUS) were recently identified as a novel cause of amyotrophic lateral sclerosis (ALS), emphasizing the genetic heterogeneity of ALS. We sequenced the genes encoding superoxide dismutase (SOD1), TAR DNA-binding protein 43 (TARDBP) and FUS in 99 sporadic and 17 familial ALS patients ascertained at Mayo Clinic. We identified two novel mutations in FUS in two out of 99 (2.0%) sporadic ALS patients and established the de novo occurrence of one FUS mutation. In familial patients, we identified three (17.6%) SOD1 mutations, while FUS and TARDBP mutations were excluded. The de novo FUS mutation (g.10747A>G; IVS13-2A>G) affects the splice-acceptor site of FUS intron 13 and was shown to induce skipping of FUS exon 14 leading to the C-terminal truncation of FUS (p.G466VfsX14). Subcellular localization studies showed a dramatic increase in the cytoplasmic localization of FUS and a reduction of normal nuclear expression in cells transfected with truncated compared to wild-type FUS. We further identified a novel in-frame insertion/deletion mutation in FUS exon 12 (p.S402_P411delinsGGGG) which is predicted to expand a conserved poly-glycine motif. Our findings extend the mutation spectrum in FUS leading to ALS and describe the first de novo mutation in FUS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , RNA-Binding Protein FUS/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/metabolism , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Male , Middle Aged , Mutation , Pedigree , RNA-Binding Protein FUS/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Young Adult
9.
Front Genet ; 10: 478, 2019.
Article in English | MEDLINE | ID: mdl-31156715

ABSTRACT

As FDA-approved small RNA drugs start to enter clinical medicine, ongoing studies for the microRNA (miRNA) class of small RNAs expand its preclinical and clinical research applications. A growing number of reports suggest a significant utility of miRNAs as biomarkers for pathogenic conditions, modulators of drug resistance, and/or as drugs for medical intervention in almost all human health conditions. The pleiotropic nature of this class of nonprotein-coding RNAs makes them particularly attractive drug targets for diseases with a multifactorial origin and no current effective treatments. As candidate miRNAs begin to proceed toward initiation and completion of potential phase 3 and 4 trials in the future, the landscape of both diagnostic and interventional medicine will arguably continue to evolve. In this mini-review, we discuss miRNA drug discovery development and their current status in clinical trials.

10.
Front Mol Biosci ; 3: 69, 2016.
Article in English | MEDLINE | ID: mdl-27826551

ABSTRACT

Dopaminergic neurotransmission mediates a majority of the vital central nervous system functions. Disruption of these synaptic events provokes a multitude of neurological pathologies, including Parkinson's, schizophrenia, depression, and addiction. Growing evidence supports a key role for noncoding RNA (ncRNA) regulation in the synapse. This review will discuss the role of both short and long ncRNAs in dopamine signaling, including bioinformatic examination of the pathways they target. Specifically, we focus on the contribution of ncRNAs to dopaminergic dysfunction in neurodegenerative as well as psychiatric disease.

11.
Mutat Res ; 552(1-2): 19-34, 2004 Aug 18.
Article in English | MEDLINE | ID: mdl-15288539

ABSTRACT

Effluents from pulp and paper mills that historically have used elemental chlorine in the bleaching process have been implicated in inhibiting reproduction in fish. Compounds with estrogenic and androgenic binding affinities have been found in these effluents, suggesting that the impairment of reproduction is through an endocrine-related mode of action. To date, a great deal of attention has been paid to phytoestrogens and resin acids that are present in mill process streams as a result of pulping trees. Estrogen and estrogen mimics interact directly with the estrogen receptor and have near immediate effects on gene transcription by turning on the expression of a unique set of genes. Using differential display (DD) RT-PCR, we examined changes in gene expression induced by exposure to paper mill effluents. Largemouth bass were exposed to 0, 10, 20, 40, and 80% paper mill effluent concentrations in large flow-through tanks for varied periods of time including 7, 28 or 56 days. Plasma hormone levels in males and females and plasma vitellogenin (Vtg) in females decreased with dose and time. Measurements of changes in gene expression using DD RT-PCR suggest that the gene expression patterns of male fish do not change much with exposure, except for the induction of a few genes including CYP 1A, a protein that is induced through the action of the Ah receptor in response to dioxin and similar polyaromatic hydrocarbons. However, in the case of females, exposure to these effluents resulted in an up-regulation of CYP 1A that was accompanied by a generalized down-regulation of genes normally expressed during the reproductive season. These antiestrogenic changes are in agreement with previous studies in bass exposed to these effluents, and could result in decreased reproductive success in affected populations.


Subject(s)
Bass/genetics , Industrial Waste/adverse effects , Paper , Water Pollutants, Chemical/toxicity , Amino Acid Sequence , Animals , Bass/growth & development , Body Constitution , Cytochrome P-450 CYP1A1/analysis , Female , Gene Expression Profiling , Liver/chemistry , Male , Molecular Sequence Data , Organ Size/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Vitellogenins/analysis
12.
Mol Brain ; 7: 46, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24929669

ABSTRACT

BACKGROUND: Huntington's Disease (HD) is a progressive neurodegenerative disorder with a single causal mutation in the Huntingtin (HTT) gene. MicroRNAs (miRNAs) have recently been implicated as epigenetic regulators of neurological disorders, however, their role in HD pathogenesis is not well defined. Here we study transgenic HD monkeys (HD monkeys) to examine miRNA dysregulation in a primate model of the disease. RESULTS: In this report, 11 miRNAs were found to be significantly associated (P value < 0.05) with HD in the frontal cortex of the HD monkeys. We further focused on one of those candidates, miR-128a, due to the corresponding disruption in humans and mice with HD as well as its intriguing lists of gene targets. miR-128a was downregulated in our HD monkey model by the time of birth. We then confirmed that miR-128a was also downregulated in the brains of pre-symptomatic and post-symptomatic HD patients. Additionally, our studies confirmed a panel of canonical HD signaling genes regulated by miR-128a, including HTT and Huntingtin Interaction Protein 1 (HIP1). CONCLUSION: Our studies found that miR-128a may play a critical role in HD and could be a viable candidate as a therapeutic or biomarker of the disease.


Subject(s)
Gene Expression Regulation , Haplorhini/genetics , Huntington Disease/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Animals, Genetically Modified , Caspase 3/metabolism , Enzyme Activation , Frontal Lobe/enzymology , Frontal Lobe/pathology , Gene Expression Profiling , Genotyping Techniques , Glial Fibrillary Acidic Protein/metabolism , HEK293 Cells , Humans , Mice , MicroRNAs/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics
13.
Stem Cells Dev ; 22(8): 1198-205, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23190281

ABSTRACT

A transgenic primate model for Huntington's Disease (HD) first reported by our group that (HD monkeys) carry the mutant Huntingtin (HTT) gene with expanded polyglutamine (CAG) repeats and, develop chorea, dystonia, and other involuntary motor deficiencies similar to HD [ 1 ]. More recently, we have found that longitudinal magnetic resonance imaging of the HD monkey brain revealed significant atrophy in regions associated with cognitive deficits symptomatic in HD patients, providing the first animal model which replicates clinical phenotypes of diagnosed humans. Here we report germline transmission of the pathogenic mutant HTT in HD monkey by the production of embryos and subsequent derivation of HD monkey embryonic stem cells (rHD-ESCs) using HD monkey sperm. rHD-ESCs inherit mutant HTT and green fluorescent protein (GFP) genes through the gametes of HD monkey. rHD-ESCs express mutant HTT and form intranuclear inclusion, a classical cellular feature of HD. Notably, mosaicism of the pathogenic polyQ region in the sperm as well as derived ESCs were also observed, consistent with intraindividual and intergenerational reports of mosaic CAG repeats [ 2 , 3 ]and CAG expansion in HD patients [ 4-7 ]. The confirmation of transgene inheritability and development of pathogenic HD phenotype in derived rHD-ESCs reported in this study is a milestone in the pursuit of a transgenic primate model with inherited mutant HTT for development of novel disease biomarkers and therapeutics.


Subject(s)
Disease Models, Animal , Germ Cells/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Animals , Animals, Genetically Modified , Base Sequence , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Germ Cells/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Immunohistochemistry , Macaca mulatta , Male , Mice , Mice, SCID , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/metabolism , Peptides/genetics , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Trinucleotide Repeat Expansion/genetics
14.
Front Genet ; 3: 82, 2012.
Article in English | MEDLINE | ID: mdl-22629284

ABSTRACT

The microRNA (miRNA) class of non-coding RNAs exhibit a diverse range of regulatory roles in neuronal functions that are conserved from lower vertebrates to primates. Disruption of miRNA expression has compellingly been linked to pathogenesis in neuropsychiatric and neurodegenerative disorders, such as schizophrenia, Alzheimer's disease, and autism. The list of transcript targets governed by a single miRNA provide a molecular paradigm applicable for therapeutic intervention. Indeed, reports have shown that specific manipulation of a miRNA in cell or animal models can significantly alter phenotypes linked with neurological disease. Here, we review how a diverse range of biological systems, including Drosophila, rodents, and primates such as monkeys and humans, can be integrated into the translation of miRNAs as novel clinical targets.

15.
Mol Neurodegener ; 7: 53, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23046583

ABSTRACT

BACKGROUND: Mutations in the gene encoding the RNA-binding protein fused in sarcoma (FUS) can cause familial and sporadic amyotrophic lateral sclerosis (ALS) and rarely frontotemproal dementia (FTD). FUS accumulates in neuronal cytoplasmic inclusions (NCIs) in ALS patients with FUS mutations. FUS is also a major pathologic marker for a group of less common forms of frontotemporal lobar degeneration (FTLD), which includes atypical FTLD with ubiquitinated inclusions (aFTLD-U), neuronal intermediate filament inclusion disease (NIFID) and basophilic inclusion body disease (BIBD). These diseases are now called FUS proteinopathies, because they share this disease marker. It is unknown how FUS mutations cause disease and the role of FUS in FTD-FUS cases, which do not have FUS mutations. In this paper we report the development of somatic brain transgenic (SBT) mice using recombinant adeno-associated virus (rAAV) to investigate how FUS mutations lead to neurodegeneration. RESULTS: We compared SBT mice expressing wild-type human FUS (FUSWT), and two ALS-linked mutations: FUSR521C and FUSΔ14, which lacks the nuclear localization signal. Both FUS mutants accumulated in the cytoplasm relative to FUSWT. The degree of this shift correlated with the severity of the FUS mutation as reflected by disease onset in humans. Mice expressing the most aggressive mutation, FUSΔ14, recapitulated many aspects of FUS proteinopathies, including insoluble FUS, basophilic and eosiniphilic NCIs, and other pathologic markers, including ubiquitin, p62/SQSTM1, α-internexin, and the poly-adenylate(A)-binding protein 1 (PABP-1). However, TDP-43 did not localize to inclusions. CONCLUSIONS: Our data supports the hypothesis that ALS or FTD-linked FUS mutations cause neurodegeneration by increasing cyotplasmic FUS. Accumulation of FUS in the cytoplasm may retain RNA targets and recruit additional RNA-binding proteins, such as PABP-1, into stress-granule like aggregates that coalesce into permanent inclusions that could negatively affect RNA metabolism. Identification of mutations in other genes that cause ALS/FTD, such as C9ORF72, sentaxin, and angiogenin, lends support to the idea that defective RNA metabolism is a critical pathogenic pathway. The SBT FUS mice described here will provide a valuable platform for dissecting the pathogenic mechanism of FUS mutations, define the relationship between FTD and ALS-FUS, and help identify therapeutic targets that are desperately needed for these devastating neurodegenerative disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , RNA-Binding Protein FUS/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Immunohistochemistry , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice , Mice, Transgenic , Mutation , Nerve Degeneration/metabolism , Neurons/metabolism , Neurons/pathology , RNA-Binding Protein FUS/metabolism
16.
Endocrinology ; 151(1): 341-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19906818

ABSTRACT

The steroidogenic acute regulatory (StAR) protein mediates the rate-limiting step of mitochondrial transport of cholesterol for steroid biosynthesis. To investigate the regulation of this protein in lower vertebrates, we cloned the StAR coding region from large-mouth bass for analysis. Induction of the mRNA corresponded with increasing levels of plasma sex steroids in vivo. Cultures of largemouth bass ovarian follicles were exposed to dibutyryl cAMP (dbcAMP), a potent signaling molecule for steroidogenesis. StAR mRNA expression was significantly up-regulated by dbcAMP signaling, suggesting that the 5' regulatory region of the gene is functionally conserved. To further analyze its transcriptional regulation, a 2.9-kb portion of the promoter was cloned and transfected into Y-1 cells, a steroidogenic mouse adrenocortical cell line. The promoter activity was induced in a dose-responsive manner upon stimulation with dbcAMP; however, deletion of 1 kb from the 5' end of the promoter segment significantly diminished the transcriptional activation. A putative retinoic acid-related receptor-alpha/rev-erb alpha element was identified between the -1.86- and -2.9-kb region and mutated to assess its potential role in dbcAMP regulation of the promoter. Mutation of the rev-erb alpha element significantly impeded dbcAMP-induced activation. Chromatin immunoprecipitation and EMSA results revealed rev-erb alpha and retinoic acid-related receptor-alpha enrichment at the site under basal and dbcAMP-induced conditions, respectively. These results implicate important roles for these proteins previously uncharacterized for the StAR promoter. Altogether these data suggest novel regulatory mechanisms for dbcAMP up-regulation of StAR transcription in the distal part of the largemouth bass promoter.


Subject(s)
Bass/genetics , Gene Expression Regulation , Orphan Nuclear Receptors/physiology , Phosphoproteins/genetics , Animals , Bass/metabolism , Cells, Cultured , Cloning, Molecular , Female , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/physiology , Orphan Nuclear Receptors/metabolism , Phosphoproteins/metabolism , Promoter Regions, Genetic , Signal Transduction/genetics , Transcription, Genetic , Transfection
17.
PLoS One ; 5(10)2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20957177

ABSTRACT

Natural antisense transcripts represent a class of regulatory RNA molecules, which are characterized by their complementary sequence to another RNA transcript. Extensive sequencing efforts suggest that natural antisense transcripts are prevalent throughout the mammalian genome; however, their biological significance has not been well defined. We performed a loss-of-function RNA interference (RNAi) screen, which targeted 797 evolutionary conserved antisense transcripts, and found evidence for a regulatory role for a number of natural antisense transcripts. Specifically, we found that natural antisense transcripts for CCPG1 and RAPGEF3 may functionally disrupt signaling pathways and corresponding biological phenotypes, such as cell viability, either independently or in parallel with the corresponding sense transcript. Our results show that the large-scale siRNA screen can be applied to evaluate natural antisense transcript modulation of fundamental cellular events.


Subject(s)
RNA Interference , RNA, Antisense/genetics , RNA, Messenger/genetics , Cell Line , Cell Proliferation , Gene Knockdown Techniques , Humans , Oligonucleotide Array Sequence Analysis
18.
Neuromolecular Med ; 11(3): 162-72, 2009.
Article in English | MEDLINE | ID: mdl-19536656

ABSTRACT

In recent years, there has been a shift in the conventional paradigms for transcriptional and translational regulation as extensive sequencing efforts have yielded new insights into the landscape of the human genome and transcriptome. Hundreds of non-coding regulatory RNA molecules called microRNAs (miRNAs) have been identified in the mammalian central nervous system (CNS) and are reported to mediate pivotal roles in many aspects of neuronal functions. Disruption of miRNA-based post-transcriptional regulation has been implicated in a range of CNS disorders as one miRNA is predicted to impact the expression of numerous downstream mRNA targets. The intricate molecular networks mediated by an miRNA form a robust mechanism for rapid and potent responses to cellular events throughout the development of the human brain. Recent studies have identified a molecular and ultimately pathogenic role for a subset of miRNAs in Alzheimer's disease and schizophrenia, including the characterization of their downstream CNS mRNA targets, such as beta-secretase (BACE1) and calmodulin-dependent protein kinase II (CaMKII). Here, we present an overview of the current progress in miRNA research related to CNS disorders and also highlight the utility of LNA (locked nucleic acid)-modified oligonucleotides in the detection and modulation of miRNA activity.


Subject(s)
Central Nervous System Diseases/genetics , MicroRNAs , Animals , Central Nervous System/anatomy & histology , Central Nervous System/physiology , Central Nervous System Diseases/pathology , Central Nervous System Diseases/physiopathology , Disease Models, Animal , Gene Expression Regulation , Gene Silencing , Genetic Variation , Genome, Human , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Structure , Nucleic Acids/chemistry , Nucleic Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL