Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Methods ; 19(10): 1208-1220, 2022 10.
Article in English | MEDLINE | ID: mdl-35618955

ABSTRACT

Circular RNAs (circRNAs) are formed in all domains of life and via different mechanisms. There has been an explosion in the number of circRNA papers in recent years; however, as a relatively young field, circRNA biology has an urgent need for common experimental standards for isolating, analyzing, expressing and depleting circRNAs. Here we propose a set of guidelines for circRNA studies based on the authors' experience. This Perspective will specifically address the major class of circRNAs in Eukarya that are generated by a spliceosome-catalyzed back-splicing event. We hope that the implementation of best practice principles for circRNA research will help move the field forward and allow a better functional understanding of this fascinating group of RNAs.


Subject(s)
RNA, Circular , RNA , RNA/genetics , RNA/metabolism , RNA Splicing
2.
EMBO Mol Med ; 12(9): e11942, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32715657

ABSTRACT

Circular RNAs (circRNAs) are brain-abundant RNAs of mostly unknown functions. To seek their roles in Parkinson's disease (PD), we generated an RNA sequencing resource of several brain region tissues from dozens of PD and control donors. In the healthy substantia nigra (SN), circRNAs accumulate in an age-dependent manner, but in the PD SN this correlation is lost and the total number of circRNAs reduced. In contrast, the levels of circRNAs are increased in the other studied brain regions of PD patients. We also found circSLC8A1 to increase in the SN of PD individuals. CircSLC8A1 carries 7 binding sites for miR-128 and is strongly bound to the microRNA effector protein Ago2. Indeed, RNA targets of miR-128 are also increased in PD individuals, suggesting that circSLC8A1 regulates miR-128 function and/or activity. CircSLC8A1 levels also increased in cultured cells exposed to the oxidative stress-inducing agent paraquat but were decreased in cells treated with the neuroprotective antioxidant regulator drug Simvastatin. Together, our work links circSLC8A1 to oxidative stress-related Parkinsonism and suggests further exploration of its molecular function in PD.


Subject(s)
MicroRNAs , Parkinson Disease , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidative Stress , Parkinson Disease/genetics , RNA, Circular , Substantia Nigra/metabolism
3.
Aging Cell ; 19(3): e13115, 2020 03.
Article in English | MEDLINE | ID: mdl-32080970

ABSTRACT

Recent reports highlight regulatory functions of long noncoding RNAs (lncRNAs) in neurodegeneration and aging, but biomedical implications remain limited. Here, we report an rRNA-depletion-based long RNA-Sequencing Resource of 65 substantia nigra, amygdala, and medial temporal gyrus samples from Parkinson's disease (PD) and matched control brains. Using a lncRNA-focused analysis approach to identify functionally important transcripts, we discovered and prioritized many lncRNAs dysregulated in PD. Those included pronounced elevation of the P53-induced noncoding transcript LINC-PINT in the substantia nigra of PD patients, as well as in additional models of oxidative stress and PD. Intriguingly, we found that LINC-PINT is a primarily neuronal transcript which showed conspicuous increases in maturing primary culture neurons. LINC-PINT also accumulated in several brain regions of Alzheimer's and Huntington's disease patients and decreased with healthy brain aging, suggesting a general role in aging and neurodegeneration for this lncRNA. RNAi-mediated depletion of LINC-PINT exacerbated the death of cultured N2A and SH-SY5Y cells exposed to oxidative stress, highlighting a previously undiscovered neuroprotective role for this tumor-inducible lncRNA in the brains of patients with neurodegenerative disorders.


Subject(s)
Neuroprotection/genetics , Parkinson Disease/metabolism , RNA, Long Noncoding/metabolism , Substantia Nigra/metabolism , Aged , Aged, 80 and over , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neuroblastoma/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Peroxides/pharmacology , RNA Interference , RNA, Long Noncoding/genetics , RNA-Seq
5.
FASEB J ; 33(10): 11223-11234, 2019 10.
Article in English | MEDLINE | ID: mdl-31311324

ABSTRACT

Recent reports attribute numerous regulatory functions to the nuclear paraspeckle-forming long noncoding RNA, nuclear enriched assembly transcript 1 (NEAT1), but the implications of its involvement in Parkinson's disease (PD) remain controversial. To address this issue, we assessed NEAT1 expression levels and cell type patterns in the substantia nigra (SN) from 53 donors with and without PD, as well as in interference tissue culture tests followed by multiple in-house and web-available models of PD. PCR quantification identified elevated levels of NEAT1 expression in the PD SN compared with control brains, an elevation that was reproducible across a multitude of disease models. In situ RNA hybridization supported neuron-specific formation of NEAT1-based paraspeckles at the SN and demonstrated coincreases of NEAT1 and paraspeckles in cultured cells under paraquat (PQ)-induced oxidative stress. Furthermore, neuroprotective agents, including fenofibrate and simvastatin, induced NEAT1 up-regulation, whereas RNA interference-mediated depletion of NEAT1 exacerbated death of PQ-exposed cells in a leucine-rich repeat kinase 2-mediated manner. Our findings highlight a novel protective role for NEAT1 in PD and suggest a previously unknown mechanism for the neuroprotective traits of widely used preventive therapeutics.-Simchovitz, A., Hanan, M., Niederhoffer, N., Madrer, N., Yayon, N., Bennett, E. R., Greenberg, D. S., Kadener, S., Soreq, H. NEAT1 is overexpressed in Parkinson's disease substantia nigra and confers drug-inducible neuroprotection from oxidative stress.


Subject(s)
Neuroprotection/physiology , Oxidative Stress/physiology , Parkinson Disease/metabolism , RNA, Long Noncoding/metabolism , Substantia Nigra/metabolism , Brain/metabolism , Cell Line , HEK293 Cells , Humans , Neurons/metabolism , RNA Interference/physiology
6.
EMBO J ; 38(17): e101064, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31359456

ABSTRACT

Alzheimer's disease is the most prevalent type of dementia and is caused by the deposition of extracellular amyloid-beta and abnormal tau phosphorylation. Neuroinflammation has emerged as an additional pathological component. Microglia, representing the brain's major innate immune cells, play an important role during Alzheimer's. Once activated, microglia show changes in their morphology, characterized by a retraction of cell processes. Systemic inflammation is known to increase the risk for cognitive decline in human neurogenerative diseases including Alzheimer's. Here, we assess for the first time microglial changes upon a peripheral immune challenge in the context of aging and Alzheimer's in vivo, using 2-photon laser scanning microscopy. Microglia were monitored at 2 and 10 days post-challenge by lipopolysaccharide. Microglia exhibited a reduction in the number of branches and the area covered at 2 days, a phenomenon that resolved at 10 days. Systemic inflammation reduced microglial clearance of amyloid-beta in APP/PS1 mice. NLRP3 inflammasome knockout blocked many of the observed microglial changes upon lipopolysaccharide, including alterations in microglial morphology and amyloid pathology. NLRP3 inhibition may thus represent a novel therapeutic target that may protect the brain from toxic peripheral inflammation during systemic infection.


Subject(s)
Aging/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Aging/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/immunology , Animals , Disease Models, Animal , Gene Knockout Techniques , Humans , Inflammation/chemically induced , Inflammation/diagnostic imaging , Lipopolysaccharides/adverse effects , Mice , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Microscopy, Confocal , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
7.
Nucleic Acids Res ; 45(11): e95, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28335028

ABSTRACT

Cells regulate biological responses in part through changes in transcription start sites (TSS) or cleavage and polyadenylation sites (PAS). To fully understand gene regulatory networks, it is therefore critical to accurately annotate cell type-specific TSS and PAS. Here we present a simple and straightforward approach for genome-wide annotation of 5΄- and 3΄-RNA ends. Our approach reliably discerns bona fide PAS from false PAS that arise due to internal poly(A) tracts, a common problem with current PAS annotation methods. We applied our methodology to study the impact of temperature on the Drosophila melanogaster head transcriptome. We found hundreds of previously unidentified TSS and PAS which revealed two interesting phenomena: first, genes with multiple PASs tend to harbor a motif near the most proximal PAS, which likely represents a new cleavage and polyadenylation signal. Second, motif analysis of promoters of genes affected by temperature suggested that boundary element association factor of 32 kDa (BEAF-32) and DREF mediates a transcriptional program at warm temperatures, a result we validated in a fly line where beaf-32 is downregulated. These results demonstrate the utility of a high-throughput platform for complete experimental and computational analysis of mRNA-ends to improve gene annotation.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , 3' Flanking Region , 5' Flanking Region , Animals , Base Sequence , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Exonucleases/chemistry , Genes, Insect , Molecular Sequence Annotation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease H/chemistry , Transcriptome
8.
Mol Cell ; 66(1): 9-21.e7, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28344080

ABSTRACT

Circular RNAs (circRNAs) are abundant and evolutionarily conserved RNAs of largely unknown function. Here, we show that a subset of circRNAs is translated in vivo. By performing ribosome footprinting from fly heads, we demonstrate that a group of circRNAs is associated with translating ribosomes. Many of these ribo-circRNAs use the start codon of the hosting mRNA, are bound by membrane-associated ribosomes, and have evolutionarily conserved termination codons. In addition, we found that a circRNA generated from the muscleblind locus encodes a protein, which we detected in fly head extracts by mass spectrometry. Next, by performing in vivo and in vitro translation assays, we show that UTRs of ribo-circRNAs (cUTRs) allow cap-independent translation. Moreover, we found that starvation and FOXO likely regulate the translation of a circMbl isoform. Altogether, our study provides strong evidence for translation of circRNAs, revealing the existence of an unexplored layer of gene activity.


Subject(s)
Drosophila Proteins/biosynthesis , Drosophila melanogaster/metabolism , Nuclear Proteins/biosynthesis , Protein Biosynthesis , RNA/metabolism , Ribosomes/metabolism , Animals , Cell Line , Codon, Initiator , Codon, Terminator , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Forkhead Transcription Factors/metabolism , Genotype , Head , Mass Spectrometry , Mice , Mutation , Nuclear Proteins/genetics , Nucleic Acid Conformation , Nutritional Status , Phenotype , RNA/chemistry , RNA/genetics , RNA Caps/chemistry , RNA Caps/genetics , RNA, Circular , Rats , Ribosomes/chemistry , Ribosomes/genetics , Starvation/genetics , Starvation/metabolism , Structure-Activity Relationship , Transfection
9.
RNA Biol ; 14(8): 1028-1034, 2017 08 03.
Article in English | MEDLINE | ID: mdl-27892769

ABSTRACT

Circular RNAs (circRNAs) are highly abundant and evolutionarily conserved non-coding RNAs produced by circularization of specific exons. Since their re-discovery as potential regulators of gene expression, thousands of circRNAs were detected in different tissues and cell types across most organisms. Accumulating data suggest key roles for them in the central nervous system. Neuronal-expressed RNAs are diverted to yield highly enriched CircRNAs in human, mouse, pig and flies, with many of them enriched in neuronal tissues. CircRNA levels are dynamically modulated in neurons, both during differentiation and following bursts of electrical activity, and accumulate with age, and many of them are enriched in synapses. Together, available data suggest that circRNAs have important roles in synaptic plasticity and neuronal function. This review covers current advances in the field and lays out hypotheses regarding functions of circRNAs in the brain as well as their putative involvement in initiation and progression of neurodegenerative processes.


Subject(s)
Brain/metabolism , MicroRNAs/genetics , Neurons/metabolism , RNA-Binding Proteins/genetics , RNA/genetics , Synapses/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Exons , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression Regulation , Humans , Mice , MicroRNAs/metabolism , Neuronal Plasticity , Neurons/cytology , RNA/metabolism , RNA, Circular , RNA-Binding Proteins/metabolism , Swine/genetics , Swine/metabolism
10.
EMBO J ; 34(11): 1538-53, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-25916830

ABSTRACT

Nutrient sensing pathways adjust metabolism and physiological functions in response to food intake. For example, sugar feeding promotes lipogenesis by activating glycolytic and lipogenic genes through the Mondo/ChREBP-Mlx transcription factor complex. Concomitantly, other metabolic routes are inhibited, but the mechanisms of transcriptional repression upon sugar sensing have remained elusive. Here, we characterize cabut (cbt), a transcription factor responsible for the repressive branch of the sugar sensing transcriptional network in Drosophila. We demonstrate that cbt is rapidly induced upon sugar feeding through direct regulation by Mondo-Mlx. We found that CBT represses several metabolic targets in response to sugar feeding, including both isoforms of phosphoenolpyruvate carboxykinase (pepck). Deregulation of pepck1 (CG17725) in mlx mutants underlies imbalance of glycerol and glucose metabolism as well as developmental lethality. Furthermore, we demonstrate that cbt provides a regulatory link between nutrient sensing and the circadian clock. Specifically, we show that a subset of genes regulated by the circadian clock are also targets of CBT. Moreover, perturbation of CBT levels leads to deregulation of the circadian transcriptome and circadian behavioral patterns.


Subject(s)
Circadian Clocks/physiology , Drosophila Proteins/metabolism , Energy Metabolism/physiology , Feeding Behavior/physiology , Glucose/metabolism , Transcription Factors/metabolism , Transcriptome/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Glucose/genetics , Glycerol/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Transcription Factors/genetics
11.
Mol Cell ; 58(5): 870-85, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25921068

ABSTRACT

Circular RNAs (circRNAs) are an endogenous class of animal RNAs. Despite their abundance, their function and expression in the nervous system are unknown. Therefore, we sequenced RNA from different brain regions, primary neurons, isolated synapses, as well as during neuronal differentiation. Using these and other available data, we discovered and analyzed thousands of neuronal human and mouse circRNAs. circRNAs were extraordinarily enriched in the mammalian brain, well conserved in sequence, often expressed as circRNAs in both human and mouse, and sometimes even detected in Drosophila brains. circRNAs were overall upregulated during neuronal differentiation, highly enriched in synapses, and often differentially expressed compared to their mRNA isoforms. circRNA expression correlated negatively with expression of the RNA-editing enzyme ADAR1. Knockdown of ADAR1 induced elevated circRNA expression. Together, we provide a circRNA brain expression atlas and evidence for important circRNA functions and values as biomarkers.


Subject(s)
Brain/metabolism , RNA/metabolism , Animals , Base Sequence , Cell Line , Drosophila melanogaster , Humans , Mice , Molecular Sequence Data , Neurogenesis , Organ Specificity , RNA/genetics , RNA, Circular , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Synapses/metabolism
12.
Mol Cell ; 56(1): 55-66, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25242144

ABSTRACT

Circular RNAs (circRNAs) are widely expressed noncoding RNAs. However, their biogenesis and possible functions are poorly understood. Here, by studying circRNAs that we identified in neuronal tissues, we provide evidence that animal circRNAs are generated cotranscriptionally and that their production rate is mainly determined by intronic sequences. We demonstrate that circularization and splicing compete against each other. These mechanisms are tissue specific and conserved in animals. Interestingly, we observed that the second exon of the splicing factor muscleblind (MBL/MBNL1) is circularized in flies and humans. This circRNA (circMbl) and its flanking introns contain conserved muscleblind binding sites, which are strongly and specifically bound by MBL. Modulation of MBL levels strongly affects circMbl biosynthesis, and this effect is dependent on the MBL binding sites. Together, our data suggest that circRNAs can function in gene regulation by competing with linear splicing. Furthermore, we identified muscleblind as a factor involved in circRNA biogenesis.


Subject(s)
Drosophila/genetics , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA/biosynthesis , Animals , Cells, Cultured , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , HEK293 Cells , Humans , Models, Genetic , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , RNA, Circular , Transcription, Genetic
13.
Brain ; 131(Pt 1): 109-19, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18056160

ABSTRACT

Alzheimer's disease has long been known to involve cholinergic deficits, but the linkage between cholinergic gene expression and the Alzheimer's disease amyloid pathology has remained incompletely understood. One known link involves synaptic acetylcholinesterase (AChE-S), shown to accelerate amyloid fibrils formation. Here, we report that the 'Readthrough' AChE-R splice variant, which differs from AChE-S in its 26 C-terminal residues, inversely exerts neuroprotective effects from amyloid beta (Abeta) induced toxicity. In vitro, highly purified AChE-R dose-dependently suppressed the formation of insoluble Abeta oligomers and fibrils and abolished Abeta toxicity to cultured cells, competing with the prevalent AChE-S protein which facilitates these processes. In vivo, double transgenic APPsw/AChE-R mice showed lower plaque burden, fewer reactive astrocytes and less dendritic damage than single APPsw mice, inverse to reported acceleration of these features in double APPsw/AChE-S mice. In hippocampi from Alzheimer's disease patients (n = 10), dentate gyrus neurons showed significantly elevated AChE-R mRNA and reduced AChE-S mRNA. However, immunoblot analyses revealed drastic reductions in the levels of intact AChE-R protein, suggesting that its selective loss in the Alzheimer's disease brain exacerbates the Abeta-induced damages and revealing a previously unforeseen linkage between cholinergic and amyloidogenic events.


Subject(s)
Acetylcholinesterase/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Acetylcholinesterase/genetics , Acetylcholinesterase/physiology , Adult , Aged , Aged, 80 and over , Alternative Splicing , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/drug effects , Animals , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Dendrites/pathology , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Enzymologic , Hippocampus/enzymology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...