Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(3): e2314699121, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38198527

ABSTRACT

Energy metabolism supports neuronal function. While it is well established that changes in energy metabolism underpin brain plasticity and function, less is known about how individual neurons modulate their metabolic states to meet varying energy demands. This is because most approaches used to examine metabolism in living organisms lack the resolution to visualize energy metabolism within individual circuits, cells, or subcellular regions. Here, we adapted a biosensor for glycolysis, HYlight, for use in Caenorhabditis elegans to image dynamic changes in glycolysis within individual neurons and in vivo. We determined that neurons cell-autonomously perform glycolysis and modulate glycolytic states upon energy stress. By examining glycolysis in specific neurons, we documented a neuronal energy landscape comprising three general observations: 1) glycolytic states in neurons are diverse across individual cell types; 2) for a given condition, glycolytic states within individual neurons are reproducible across animals; and 3) for varying conditions of energy stress, glycolytic states are plastic and adapt to energy demands. Through genetic analyses, we uncovered roles for regulatory enzymes and mitochondrial localization in the cellular and subcellular dynamic regulation of glycolysis. Our study demonstrates the use of a single-cell glycolytic biosensor to examine how energy metabolism is distributed across cells and coupled to dynamic states of neuronal function and uncovers unique relationships between neuronal identities and metabolic landscapes in vivo.


Subject(s)
Glycolysis , Neurons , Animals , Energy Metabolism , Caenorhabditis elegans , Neuronal Plasticity
2.
Proc Natl Acad Sci U S A ; 119(31): e2204407119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881794

ABSTRACT

Cellular metabolism is regulated over space and time to ensure that energy production is efficiently matched with consumption. Fluorescent biosensors are useful tools for studying metabolism as they enable real-time detection of metabolite abundance with single-cell resolution. For monitoring glycolysis, the intermediate fructose 1,6-bisphosphate (FBP) is a particularly informative signal as its concentration is strongly correlated with flux through the whole pathway. Using GFP insertion into the ligand-binding domain of the Bacillus subtilis transcriptional regulator CggR, we developed a fluorescent biosensor for FBP termed HYlight. We demonstrate that HYlight can reliably report the real-time dynamics of glycolysis in living cells and tissues, driven by various metabolic or pharmacological perturbations, alone or in combination with other physiologically relevant signals. Using this sensor, we uncovered previously unknown aspects of ß-cell glycolytic heterogeneity and dynamics.


Subject(s)
Biosensing Techniques , Fructose , Glycolysis , Single-Cell Analysis , Fluorescence , Fructose/analysis , Fructosediphosphates/analysis , Humans , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Single-Cell Analysis/methods
3.
Elife ; 82019 06 24.
Article in English | MEDLINE | ID: mdl-31232686

ABSTRACT

Exercise is a potent enhancer of learning and memory, yet we know little of the underlying mechanisms that likely include alterations in synaptic efficacy in the hippocampus. To address this issue, we exposed mice to a single episode of voluntary exercise, and permanently marked activated mature hippocampal dentate granule cells using conditional Fos-TRAP mice. Exercise-activated neurons (Fos-TRAPed) showed an input-selective increase in dendritic spines and excitatory postsynaptic currents at 3 days post-exercise, indicative of exercise-induced structural plasticity. Laser-capture microdissection and RNASeq of activated neurons revealed that the most highly induced transcript was Mtss1L, a little-studied I-BAR domain-containing gene, which we hypothesized could be involved in membrane curvature and dendritic spine formation. shRNA-mediated Mtss1L knockdown in vivo prevented the exercise-induced increases in spines and excitatory postsynaptic currents. Our results link short-term effects of exercise to activity-dependent expression of Mtss1L, which we propose as a novel effector of activity-dependent rearrangement of synapses.


Subject(s)
Hippocampus/physiology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Neuronal Plasticity , Neurons/physiology , Physical Conditioning, Animal , Action Potentials , Animals , Base Sequence , Gene Expression Profiling , Mice
4.
Curr Protoc Cytom ; 88(1): e54, 2019 04.
Article in English | MEDLINE | ID: mdl-30556645

ABSTRACT

Flow cytometry approaches combined with a genetically encoded targeted fluorescent biosensor are used to determine the subcellular compartmental availability of the oxidized form of nicotinamide adenine dinucleotide (NAD+ ). The availability of free NAD+ can affect the activities of NAD+ -consuming enzymes such as sirtuin, PARP/ARTD, and cyclic ADPR-hydrolase family members. Many methods for measuring the NAD+ available to these enzymes are limited because they cannot determine free NAD+ as it exists in various subcellular compartments distinctly from bound NAD+ or NADH. Here, an approach to express the sensor in mammalian cells, monitor NAD+ -dependent fluorescence intensity changes using flow cytometry approaches, and analyze data obtained is described. The benefit of flow cytometry approaches with the NAD+ sensor is the ability to monitor compartmentalized free NAD+ fluctuations simultaneously within many cells, which greatly facilitates analyses and calibration. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Biosensing Techniques/methods , Flow Cytometry/methods , Intracellular Space/metabolism , NAD/analysis , Acrylamides/pharmacology , Calibration , Digitonin/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescence , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mitochondria/drug effects , Mitochondria/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Piperidines/pharmacology , Small Molecule Libraries/pharmacology , Statistics as Topic
5.
Proc Natl Acad Sci U S A ; 115(42): 10654-10659, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30257945

ABSTRACT

Axon degeneration, a hallmark of chemotherapy-induced peripheral neuropathy (CIPN), is thought to be caused by a loss of the essential metabolite nicotinamide adenine dinucleotide (NAD+) via the prodegenerative protein SARM1. Some studies challenge this notion, however, and suggest that an aberrant increase in a direct precursor of NAD+, nicotinamide mononucleotide (NMN), rather than loss of NAD+, is responsible. In support of this idea, blocking NMN accumulation in neurons by expressing a bacterial NMN deamidase protected axons from degeneration. We hypothesized that protection could similarly be achieved by reducing NMN production pharmacologically. To achieve this, we took advantage of an alternative pathway for NAD+ generation that goes through the intermediate nicotinic acid mononucleotide (NAMN), rather than NMN. We discovered that nicotinic acid riboside (NAR), a precursor of NAMN, administered in combination with FK866, an inhibitor of the enzyme nicotinamide phosphoribosyltransferase that produces NMN, protected dorsal root ganglion (DRG) axons against vincristine-induced degeneration as well as NMN deamidase. Introducing a different bacterial enzyme that converts NAMN to NMN reversed this protection. Collectively, our data indicate that maintaining NAD+ is not sufficient to protect DRG neurons from vincristine-induced axon degeneration, and elevating NMN, by itself, is not sufficient to cause degeneration. Nonetheless, the combination of FK866 and NAR, which bypasses NMN formation, may provide a therapeutic strategy for neuroprotection.


Subject(s)
Acrylamides/pharmacology , NAD/metabolism , Nerve Degeneration/prevention & control , Neurons/drug effects , Niacinamide/analogs & derivatives , Nicotinamide Mononucleotide/analogs & derivatives , Piperidines/pharmacology , Vincristine/toxicity , Animals , Antineoplastic Agents, Phytogenic/toxicity , Drug Combinations , Francisella tularensis/enzymology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Neurons/metabolism , Neurons/pathology , Niacinamide/pharmacology , Nicotinamide Mononucleotide/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Pyridinium Compounds
6.
Methods Mol Biol ; 1813: 391-414, 2018.
Article in English | MEDLINE | ID: mdl-30097882

ABSTRACT

Free nicotinamide adenine dinucleotide (NAD+) serves as substrate for NAD+-consuming enzymes. As such, the local concentration of free NAD+ can influence enzymatic activities. Here we describe methods for using a fluorescent, genetically-encoded sensor to measure subcellular NAD+ concentrations. We also include a discussion of the limitations and potential applications for the current sensor. Presented in this chapter are (1) guidelines for calibrating instrumentation and experimental setups using a bead-based method, (2) instructions for incorporating required controls and properly performing ratiometric measurements in cells, and (3) descriptions of how to evaluate relative and quantitative fluctuations using appropriate statistical methods for ratio-of-ratio measurements.


Subject(s)
Biosensing Techniques/methods , Cell Nucleus/chemistry , Monitoring, Physiologic/methods , NAD/isolation & purification , Fluorescent Dyes/chemistry , NAD/chemistry
7.
Cell Rep ; 19(3): 505-520, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423315

ABSTRACT

The central circadian pacemaker, the suprachiasmatic nucleus (SCN), encodes day length information by mechanisms that are not well understood. Here, we report that genetic ablation of miR-132/212 alters entrainment to different day lengths and non-24 hr day-night cycles, as well as photoperiodic regulation of Period2 expression in the SCN. SCN neurons from miR-132/212-deficient mice have significantly reduced dendritic spine density, along with altered methyl CpG-binding protein (MeCP2) rhythms. In Syrian hamsters, a model seasonal rodent, day length regulates spine density on SCN neurons in a melatonin-independent manner, as well as expression of miR-132, miR-212, and their direct target, MeCP2. Genetic disruption of Mecp2 fully restores the level of dendritic spines of miR-132/212-deficient SCN neurons. Our results reveal that, by regulating the dendritic structure of SCN neurons through a MeCP2-dependent mechanism, miR-132/212 affects the capacity of the SCN to encode seasonal time.


Subject(s)
Adaptation, Physiological/genetics , Circadian Clocks/genetics , Dendrites/metabolism , MicroRNAs/metabolism , Seasons , Adaptation, Physiological/radiation effects , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Circadian Clocks/radiation effects , Dendrites/radiation effects , Dendritic Spines/metabolism , Dendritic Spines/radiation effects , Female , Gene Deletion , Gene Expression Regulation/radiation effects , Light , Male , Mesocricetus , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Neurons/metabolism , Photoperiod , Proteome/metabolism , Signal Transduction/radiation effects , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/radiation effects , TOR Serine-Threonine Kinases/metabolism , Time Factors
8.
Science ; 352(6292): 1474-7, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27313049

ABSTRACT

Nicotinamide adenine dinucleotide (NAD(+)) is an essential substrate for sirtuins and poly(adenosine diphosphate-ribose) polymerases (PARPs), which are NAD(+)-consuming enzymes localized in the nucleus, cytosol, and mitochondria. Fluctuations in NAD(+) concentrations within these subcellular compartments are thought to regulate the activity of NAD(+)-consuming enzymes; however, the challenge in measuring compartmentalized NAD(+) in cells has precluded direct evidence for this type of regulation. We describe the development of a genetically encoded fluorescent biosensor for directly monitoring free NAD(+) concentrations in subcellular compartments. We found that the concentrations of free NAD(+) in the nucleus, cytoplasm, and mitochondria approximate the Michaelis constants for sirtuins and PARPs in their respective compartments. Systematic depletion of enzymes that catalyze the final step of NAD(+) biosynthesis revealed cell-specific mechanisms for maintaining mitochondrial NAD(+) concentrations.


Subject(s)
Biosensing Techniques , Mitochondria/metabolism , NAD/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytosol/chemistry , Cytosol/metabolism , DNA Ligases/genetics , DNA Ligases/metabolism , HEK293 Cells , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitochondria/chemistry , NAD/analysis , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Point Mutation , Poly(ADP-ribose) Polymerases/metabolism , Sirtuins/metabolism
9.
ACS Chem Biol ; 11(6): 1532-6, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27074634

ABSTRACT

Translation regulation is a fundamental component of gene expression, allowing cells to respond rapidly to a variety of stimuli in the absence of new transcription. The lack of methods for profiling nascent proteomes in distinct cell populations in heterogeneous tissues has precluded an understanding of translational regulation in physiologically relevant contexts. Here, we describe a chemical genetic method that involves orthogonal enzyme-mediated incorporation of a clickable puromycin analogue into nascent polypeptides. Using this method, we show that we can label newly synthesized proteins in a cell-specific manner in cells grown in culture and in heterogeneous tissues. We also show that we can identify the nascent proteome in genetically targeted cell populations using affinity enrichment and tandem mass spectrometry. Our method has the potential to provide unprecedented insights into cell-specific translational regulation in heterogeneous tissues.


Subject(s)
Adenosine/analogs & derivatives , Proteome/chemistry , Puromycin/analogs & derivatives , Tyrosine/analogs & derivatives , Adenosine/chemistry , Adenosine/metabolism , Animals , Biotinylation , Click Chemistry , Fluorescent Dyes/chemistry , Glucagon-Secreting Cells/metabolism , HEK293 Cells , Humans , Insulin-Secreting Cells/metabolism , Mice , Penicillin Amidase/chemistry , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Proteome/genetics , Proteome/metabolism , Puromycin/chemistry , Tyrosine/chemistry , Tyrosine/metabolism
10.
eNeuro ; 3(1)2016.
Article in English | MEDLINE | ID: mdl-27011954

ABSTRACT

Despite representing only a small fraction of hippocampal granule cells, adult-generated newborn granule cells have been implicated in learning and memory (Aimone et al., 2011). Newborn granule cells undergo functional maturation and circuit integration over a period of weeks. However, it is difficult to assess the accompanying gene expression profiles in vivo with high spatial and temporal resolution using traditional methods. Here we used a novel method ["thiouracil (TU) tagging"] to map the profiles of nascent mRNAs in mouse immature newborn granule cells compared with mature granule cells. We targeted a nonmammalian uracil salvage enzyme, uracil phosphoribosyltransferase, to newborn neurons and mature granule cells using retroviral and lentiviral constructs, respectively. Subsequent injection of 4-TU tagged nascent RNAs for analysis by RNA sequencing. Several hundred genes were significantly enhanced in the retroviral dataset compared with the lentiviral dataset. We compared a selection of the enriched genes with steady-state levels of mRNAs using quantitative PCR. Ontology analysis revealed distinct patterns of nascent mRNA expression, with newly generated immature neurons showing enhanced expression for genes involved in synaptic function, and neural differentiation and development, as well as genes not previously associated with granule cell maturation. Surprisingly, the nascent mRNAs enriched in mature cells were related to energy homeostasis and metabolism, presumably indicative of the increased energy demands of synaptic transmission and their complex dendritic architecture. The high spatial and temporal resolution of our modified TU-tagging method provides a foundation for comparison with steady-state RNA analyses by traditional transcriptomic approaches in defining the functional roles of newborn neurons.


Subject(s)
Dentate Gyrus/metabolism , Gene Expression Profiling/methods , Neurogenesis , Neurons/metabolism , Thiouracil/metabolism , Animals , Base Sequence , Female , Genetic Vectors/administration & dosage , Lentivirus/genetics , Lentivirus/physiology , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Thiouracil/administration & dosage
12.
Nat Commun ; 6: 7718, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26212498

ABSTRACT

While microRNAs have emerged as an important component of gene regulatory networks, it remains unclear how microRNAs collaborate with transcription factors in the gene networks that determines neuronal cell fate. Here we show that in the developing spinal cord, the expression of miR-218 is directly upregulated by the Isl1-Lhx3 complex, which drives motor neuron fate. Inhibition of miR-218 suppresses the generation of motor neurons in both chick neural tube and mouse embryonic stem cells, suggesting that miR-218 plays a crucial role in motor neuron differentiation. Results from unbiased RISC-trap screens, in vivo reporter assays and overexpression studies indicated that miR-218 directly represses transcripts that promote developmental programs for interneurons. In addition, we found that miR-218 activity is required for Isl1-Lhx3 to effectively induce motor neurons and suppress interneuron fates. Together our results reveal an essential role of miR-218 as a downstream effector of the Isl1-Lhx3 complex in establishing motor neuron identity.


Subject(s)
Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/genetics , MicroRNAs/genetics , Motor Neurons/cytology , Neural Tube/embryology , Neurogenesis/genetics , Spinal Cord/embryology , Transcription Factors/genetics , Animals , Chick Embryo , Electroporation , HEK293 Cells , Humans , LIM-Homeodomain Proteins/metabolism , Mice , Mouse Embryonic Stem Cells , Neural Tube/cytology , Real-Time Polymerase Chain Reaction , Spinal Cord/cytology , Transcription Factors/metabolism , Up-Regulation
13.
Behav Brain Res ; 287: 15-26, 2015.
Article in English | MEDLINE | ID: mdl-25813747

ABSTRACT

The miR-132/212 family is thought to play an important role in neural function and plasticity, while its misregulation has been observed in various neurodegenerative disorders. In this study, we analyzed 6-month-old miR-132/212 knockout mice in a battery of cognitive and non-cognitive behavioral tests. No significant changes were observed in reflexes and basic sensorimotor functions as determined by the SHIRPA primary screen. Accordingly, miR-132/212 knockout mice did not differ from wild-type controls in general locomotor activity in an open-field test. Furthermore, no significant changes of anxiety were measured in an elevated plus maze task. However, the mutant mice showed retention phase defects in a novel object recognition test and in the T-water maze. Moreover, the learning and probe phases in the Barnes maze were clearly altered in knockout mice when compared to controls. Finally, changes in BDNF, CREB, and MeCP2 were identified in the miR-132/212-deficient mice, providing a potential mechanism for promoting memory loss. Taken together, these results further strengthen the role of miR-132/212 in memory formation and retention, and shed light on the potential consequences of its deregulation in neurodegenerative diseases.


Subject(s)
MicroRNAs/physiology , Retention, Psychology/physiology , Spatial Memory/physiology , Animals , Anxiety/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Maze Learning/physiology , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Motor Activity , Phosphorylation , Recognition, Psychology/physiology
15.
Neuron ; 81(6): 1255-1262, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24583023

ABSTRACT

Major nonprimate-primate differences in cortico-genesis include the dimensions, precursor lineages, and developmental timing of the germinal zones (GZs). microRNAs (miRNAs) of laser-dissected GZ compartments and cortical plate (CP) from embryonic E80 macaque visual cortex were deep sequenced. The CP and the GZ including ventricular zone (VZ) and outer and inner subcompartments of the outer subventricular zone (OSVZ) in area 17 displayed unique miRNA profiles. miRNAs present in primate, but absent in rodent, contributed disproportionately to the differential expression between GZ subregions. Prominent among the validated targets of these miRNAs were cell-cycle and neurogenesis regulators. Coevolution between the emergent miRNAs and their targets suggested that novel miRNAs became integrated into ancient gene circuitry to exert additional control over proliferation. We conclude that multiple cell-cycle regulatory events contribute to the emergence of primate-specific cortical features, including the OSVZ, generated enlarged supragranular layers, largely responsible for the increased primate cortex computational abilities.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation , Macaca/genetics , MicroRNAs/genetics , Neurogenesis/genetics , Neurons/cytology , Visual Cortex/cytology , Animals , Cell Cycle/physiology , Evolution, Molecular , Female , Neurogenesis/physiology , Neurons/metabolism , Visual Cortex/metabolism
16.
Proc Natl Acad Sci U S A ; 110(44): 17898-903, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127608

ABSTRACT

MicroRNA-134 (miR-134) serves as a widely accepted model for microRNA function in synaptic plasticity. In this model, synaptic activity stimulates miR-134 expression, which then regulates dendrite growth and spine formation. By using a ratiometric microRNA sensor, we found, unexpectedly, that miR-134 activity in cortical neurons was restricted to interneurons. Using an assay designed to trap microRNA-mRNA complexes, we determined that miR-134 interacted directly with the mRNA encoding the palmitoylation enzyme, DHHC9. This enzyme is known to palmitoylate H-Ras, a modification required for proper membrane trafficking. Treatment with bicuculline, a GABAA receptor antagonist, decreased DHHC9 expression in somatostatin-positive interneurons and membrane localization of an H-Ras reporter in a manner that depended on miR-134. Thus, although miR-134 has been proposed to affect all types of neurons, we showed that functionally active miR-134 is produced in only a selected population of neurons where it influences the expression of targets, such as DHHC9, that regulate membrane targeting of critical signaling molecules.


Subject(s)
Acyltransferases/metabolism , Interneurons/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , Somatostatin-Secreting Cells/metabolism , Synapses/metabolism , Animals , Bicuculline/pharmacology , Blotting, Western , Gene Expression Regulation, Enzymologic/drug effects , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Confocal , Mutagenesis , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
17.
Proc Natl Acad Sci U S A ; 109(50): 20473-8, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23184980

ABSTRACT

Identifying targets is critical for understanding the biological effects of microRNA (miRNA) expression. The challenge lies in characterizing the cohort of targets for a specific miRNA, especially when targets are being actively down-regulated in miRNA- RNA-induced silencing complex (RISC)-messengerRNA (mRNA) complexes. We have developed a robust and versatile strategy called RISCtrap to stabilize and purify targets from this transient interaction. Its utility was demonstrated by determining specific high-confidence target datasets for miR-124, miR-132, and miR-181 that contained known and previously unknown transcripts. Two previously unknown miR-132 targets identified with RISCtrap, adaptor protein CT10 regulator of kinase 1 (CRK1) and tight junction-associated protein 1 (TJAP1), were shown to be endogenously regulated by miR-132 in adult mouse forebrain. The datasets, moreover, differed in the number of targets and in the types and frequency of microRNA recognition element (MRE) motifs, thus revealing a previously underappreciated level of specificity in the target sets regulated by individual miRNAs.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Animals , Base Sequence , Binding Sites/genetics , HEK293 Cells , Humans , Macromolecular Substances , Mice , MicroRNAs/chemistry , Protein Subunits , Proto-Oncogene Proteins c-crk/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/chemistry , Sequence Deletion , Tight Junction Proteins/metabolism
18.
Mol Endocrinol ; 26(6): 989-99, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22539037

ABSTRACT

MicroRNA-375 (miR-375) is necessary for proper formation of pancreatic islets in vertebrates and is necessary for the development of ß-cells in mice, but regulation of miR-375 in these cells is poorly understood. Here, we show that miR-375 is transcriptionally repressed by the cAMP-protein kinase A (PKA) pathway and that this repression is mediated through a block in RNA polymerase II binding to the miR-375 promoter. cAMP analogs that are PKA selective repress miR-375, as do cAMP agonists and the glucagon-like peptide-1 receptor agonist, exendin-4. Repression of the miR-375 precursor occurs rapidly in rat insulinoma INS-1 832/13 cells, within 15 min after cAMP stimulation, although the mature microRNA declines more slowly due to the kinetics of RNA processing. Repression of miR-375 in isolated rat islets by exendin-4 also occurs slowly, after several hours of stimulation. Glucose is another reported antagonist of miR-375 expression, although we demonstrate here that glucose does not target the microRNA through the PKA pathway. As reported previously, miR-375 negatively regulates insulin secretion, and attenuation of miR-375 through the cAMP-PKA pathway may boost the insulin response in pancreatic ß-cells.


Subject(s)
Amino Acids, Cyclic/physiology , Down-Regulation , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Exenatide , Glucagon-Like Peptide-1 Receptor , Glucose/physiology , Insulin/metabolism , Insulin Secretion , Inverted Repeat Sequences , Male , MicroRNAs/genetics , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Signal Transduction , Transcription, Genetic , Venoms/pharmacology
19.
Proc Natl Acad Sci U S A ; 107(47): 20382-7, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059906

ABSTRACT

Newborn neurons in the dentate gyrus of the adult hippocampus rely upon cAMP response element binding protein (CREB) signaling for their differentiation into mature granule cells and their integration into the dentate network. Among its many targets, the transcription factor CREB activates expression of a gene locus that produces two microRNAs, miR-132 and miR-212. In cultured cortical and hippocampal neurons, miR-132 functions downstream from CREB to mediate activity-dependent dendritic growth and spine formation in response to a variety of signaling pathways. To investigate whether miR-132 and/or miR-212 contribute to the maturation of dendrites in newborn neurons in the adult hippocampus, we inserted LoxP sites surrounding the miR-212/132 locus and specifically targeted its deletion by stereotactically injecting a retrovirus expressing Cre recombinase. Deletion of the miR-212/132 locus caused a dramatic decrease in dendrite length, arborization, and spine density. The miR-212/132 locus may express up to four distinct microRNAs, miR-132 and -212 and their reverse strands miR-132* and -212*. Using ratiometric microRNA sensors, we determined that miR-132 is the predominantly active product in hippocampal neurons. We conclude that miR-132 is required for normal dendrite maturation in newborn neurons in the adult hippocampus and suggest that this microRNA also may participate in other examples of CREB-mediated signaling.


Subject(s)
Dendrites/genetics , Gene Expression Regulation/physiology , Hippocampus/growth & development , MicroRNAs/metabolism , Neurogenesis/physiology , Neurons/cytology , Signal Transduction/physiology , Animals , CREB-Binding Protein/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Flow Cytometry , Gene Knockout Techniques , Humans , Immunohistochemistry , Mice , Mice, Transgenic , MicroRNAs/genetics , Microscopy, Confocal
20.
Proc Natl Acad Sci U S A ; 107(45): 19231-6, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974915

ABSTRACT

Predictions of microRNA-mRNA interactions typically rely on bioinformatic algorithms, but these algorithms only suggest the possibility of microRNA binding and may miss important interactions as well as falsely predict others. We developed an affinity purification approach to empirically identify microRNAs associated with the 3'UTR of the mRNA encoding Hand2, a transcription factor essential for cardiac development. In addition to miR-1, a known regulator of Hand2 expression, we determined that the Hand2 3'UTR also associated with miR-133a, a microRNA cotranscribed with miR-1 in cardiac and muscle cells. Using a sequential binding assay, we showed that miR-1 and miR-133a could occupy the Hand2 3'UTR concurrently. miR-133a inhibited Hand2 expression in tissue culture models, and miR-133a double knockout mice had elevated levels of Hand2 mRNA and protein. We conclude that Hand2 is regulated by miR-133a in addition to miR-1. The affinity purification assay should be generally applicable for identifying other microRNA-mRNA interactions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/isolation & purification , Binding Sites , Heart , Humans , Mice , Mice, Knockout , MicroRNAs/isolation & purification , Myocardium/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...