Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Mol Cell ; 81(16): 3410-3421.e4, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34192510

ABSTRACT

Chromatosomes play a fundamental role in chromatin regulation, but a detailed understanding of their structure is lacking, partially due to their complex dynamics. Using single-molecule DNA unzipping with optical tweezers, we reveal that linker histone interactions with DNA are remarkably extended, with the C-terminal domain binding both DNA linkers as far as approximately ±140 bp from the dyad. In addition to a symmetrical compaction of the nucleosome core governed by globular domain contacts at the dyad, the C-terminal domain compacts the nucleosome's entry and exit. These interactions are dynamic, exhibit rapid binding and dissociation, are sensitive to phosphorylation of a specific residue, and are crucial to determining the symmetry of the chromatosome's core. Extensive unzipping of the linker DNA, which mimics its invasion by motor proteins, shifts H1 into an asymmetric, off-dyad configuration and triggers nucleosome decompaction, highlighting the plasticity of the chromatosome structure and its potential regulatory role.


Subject(s)
Chromatin/genetics , DNA/genetics , Histones/genetics , Nucleosomes/genetics , Biophysical Phenomena/genetics , DNA-Binding Proteins/genetics , Humans , Molecular Dynamics Simulation , Protein Binding/genetics , Protein Conformation , Single Molecule Imaging
2.
Trends Genet ; 37(4): 389-400, 2021 04.
Article in English | MEDLINE | ID: mdl-33092903

ABSTRACT

It is recognized that cell metabolism is tightly connected to other cellular processes such as regulation of gene expression. Metabolic pathways not only provide the precursor molecules necessary for gene expression, but they also provide ATP, the primary fuel driving gene expression. However, metabolic conditions are highly variable since nutrient uptake is not a uniform process. Thus, cells must continually calibrate gene expression to their changing metabolite and energy budgets. This review discusses recent advances in understanding the molecular and biophysical mechanisms that connect metabolism and gene regulation as cells navigate their growth, proliferation, and differentiation. Particular focus is given to these mechanisms in the context of organismal development.


Subject(s)
Energy Metabolism/genetics , Gene Expression Regulation/genetics , Metabolic Networks and Pathways/genetics , Adenosine Triphosphate/genetics , Animals , Biophysical Phenomena/genetics , Humans
3.
J Biol Chem ; 295(9): 2568-2569, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111719

ABSTRACT

Riboswitches alter gene expression in response to ligand binding, coupling sensing and regulatory functions to help bacteria respond to their environment. The structural determinants of ligand binding in the prequeuosine (7-aminomethyl-7-deazaguanine, preQ1) bacterial riboswitches have been studied, but the functional consequences of structural perturbations are less known. A new article combining biophysical and cell-based readouts of 15 mutants of the preQ1-II riboswitch from Lactobacillus rhamnosus demonstrates that ligand binding does not ensure successful gene regulation, providing new insights into these shapeshifting sequences.


Subject(s)
Bacteria/genetics , Lacticaseibacillus rhamnosus/genetics , Riboswitch/genetics , Bacteria/drug effects , Biophysical Phenomena/genetics , Gene Expression Regulation/drug effects , Lacticaseibacillus rhamnosus/drug effects , Ligands , Mutation/genetics , Nucleic Acid Conformation/drug effects , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Riboswitch/drug effects
4.
Proteins ; 88(1): 206-226, 2020 01.
Article in English | MEDLINE | ID: mdl-31344278

ABSTRACT

Computational design of binding sites in proteins remains difficult, in part due to limitations in our current ability to sample backbone conformations that enable precise and accurate geometric positioning of side chains during sequence design. Here we present a benchmark framework for comparison between flexible-backbone design methods applied to binding interactions. We quantify the ability of different flexible backbone design methods in the widely used protein design software Rosetta to recapitulate observed protein sequence profiles assumed to represent functional protein/protein and protein/small molecule binding interactions. The CoupledMoves method, which combines backbone flexibility and sequence exploration into a single acceptance step during the sampling trajectory, better recapitulates observed sequence profiles than the BackrubEnsemble and FastDesign methods, which separate backbone flexibility and sequence design into separate acceptance steps during the sampling trajectory. Flexible-backbone design with the CoupledMoves method is a powerful strategy for reducing sequence space to generate targeted libraries for experimental screening and selection.


Subject(s)
Computational Biology , Protein Conformation , Protein Interaction Mapping , Proteins/ultrastructure , Algorithms , Amino Acid Sequence/genetics , Binding Sites/genetics , Biophysical Phenomena/genetics , Humans , Models, Molecular , Protein Binding/genetics , Protein Engineering/trends , Proteins/chemistry , Software
5.
Int J Mol Sci ; 21(15)2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32752103

ABSTRACT

The flight muscle of Manduca sexta (DLM1) is an emerging model system for biophysical studies of muscle contraction. Unlike the well-studied indirect flight muscle of Lethocerus and Drosophila, the DLM1 of Manduca is a synchronous muscle, as are the vertebrate cardiac and skeletal muscles. Very little has been published regarding the ultrastructure and protein composition of this muscle. Previous studies have demonstrated that DLM1 express two projectin isoform, two kettin isoforms, and two large Salimus (Sls) isoforms. Such large Sls isoforms have not been observed in the asynchronous flight muscles of Lethocerus and Drosophila. The spatial localization of these proteins was unknown. Here, immuno-localization was used to show that the N-termini of projectin and Salimus are inserted into the Z-band. Projectin spans across the I-band, and the C-terminus is attached to the thick filament in the A-band. The C-terminus of Sls was also located in the A-band. Using confocal microscopy and experimental force-length curves, thin filament lengths were estimated as ~1.5 µm and thick filament lengths were measured as ~2.5 µm. This structural information may help provide an interpretive framework for future studies using this muscle system.


Subject(s)
Connectin/genetics , Manduca/physiology , Muscle Contraction/physiology , Muscle Proteins/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/ultrastructure , Amino Acid Sequence/genetics , Animals , Biophysical Phenomena/genetics , Drosophila/genetics , Flight, Animal/physiology , Manduca/genetics , Muscle Contraction/genetics , Muscle, Skeletal/physiology , Muscle, Skeletal/ultrastructure , Myofibrils/genetics , Myofibrils/physiology , Myofibrils/ultrastructure , Sarcomeres/genetics , Sarcomeres/physiology , Sarcomeres/ultrastructure
6.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1799-1808, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28732675

ABSTRACT

Cells sense and respond to the biophysical properties of their surrounding environment by interacting with the extracellular matrix (ECM). Therefore, the optimization of these cell-matrix interactions is critical in tissue engineering. The vascular system is adapted to specific functions in diverse tissues and organs. Appropriate arterial-venous differentiation is vital for the establishment of functional vasculature in angiogenesis. Here, we have developed a polydimethylsiloxane (PDMS)-based substrate capable of simulating the physiologically relevant stiffness of both venous (7kPa) and arterial (128kPa) tissues. This substrate was utilized to investigate the effects of changes in substrate stiffness on the differentiation of endothelial progenitor cells (EPCs). As EPCs derived from mouse bone marrow were cultured on substrates of increasing stiffness, the mRNA and protein levels of the specific arterial endothelial cell marker ephrinB2 were found to increase, while the expression of the venous marker EphB4 decreased. Further experiments were performed to identify the mechanotransduction pathway involved in this process. The results indicated that substrate stiffness regulates the arterial and venous differentiation of EPCs via the Ras/Mek pathway. This work shows that modification of substrate stiffness may represent a method for regulating arterial-venous differentiation for the fulfilment of diverse functions of the vasculature.


Subject(s)
Cell Differentiation/genetics , Endothelial Progenitor Cells/metabolism , Ephrin-B2/genetics , Extracellular Matrix/metabolism , Receptor, EphB4/genetics , Animals , Arteries/growth & development , Arteries/metabolism , Biophysical Phenomena/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Dimethylpolysiloxanes/chemistry , Dimethylpolysiloxanes/metabolism , Extracellular Matrix/genetics , Gene Expression Regulation , Mechanotransduction, Cellular/genetics , Mice , RNA, Messenger/genetics , Substrate Specificity , Tissue Engineering , Vascular Stiffness/genetics , Vascular Stiffness/physiology , Veins/growth & development , Veins/metabolism
7.
Trends Biochem Sci ; 39(9): 381-99, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25129887

ABSTRACT

Transcription factors (TFs) influence cell fate by interpreting the regulatory DNA within a genome. TFs recognize DNA in a specific manner; the mechanisms underlying this specificity have been identified for many TFs based on 3D structures of protein-DNA complexes. More recently, structural views have been complemented with data from high-throughput in vitro and in vivo explorations of the DNA-binding preferences of many TFs. Together, these approaches have greatly expanded our understanding of TF-DNA interactions. However, the mechanisms by which TFs select in vivo binding sites and alter gene expression remain unclear. Recent work has highlighted the many variables that influence TF-DNA binding, while demonstrating that a biophysical understanding of these many factors will be central to understanding TF function.


Subject(s)
Biophysical Phenomena/genetics , DNA/genetics , Genome/genetics , Transcription Factors/metabolism , Animals , Binding Sites , Computational Biology , DNA/metabolism , Humans , Protein Binding
8.
Proc Natl Acad Sci U S A ; 111(22): 8043-8, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843124

ABSTRACT

DNA base flipping is a fundamental theme in DNA biophysics. The dynamics for a B-DNA base to spontaneously flip out of the double helix has significant implications in various DNA-protein interactions but are still poorly understood. The spontaneous base-flipping rate obtained previously via the imino proton exchange assay is most likely the rate of base wobbling instead of flipping. Using the diffusion-decelerated fluorescence correlation spectroscopy together with molecular dynamics simulations, we show that a base of a single mismatched base pair (T-G, T-T, or T-C) in a double-stranded DNA can spontaneously flip out of the DNA duplex. The extrahelical lifetimes are on the order of 10 ms, whereas the intrahelical lifetimes range from 0.3 to 20 s depending on the stability of the base pairs. These findings provide detailed understanding on the dynamics of DNA base flipping and lay down foundation to fully understand how exactly the repair proteins search and locate the target mismatched base among a vast excess of matched DNA bases.


Subject(s)
Base Pair Mismatch/genetics , Base Pairing/genetics , Biophysical Phenomena/genetics , DNA, B-Form/chemistry , DNA/chemistry , Molecular Dynamics Simulation , Thermodynamics , DNA/genetics , DNA, B-Form/genetics , Fluorescence , Nucleic Acid Conformation , Photochemistry/methods
9.
J Neurosci ; 35(35): 12198-206, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26338330

ABSTRACT

The mutation F184C in Kv1.1 leads to development of episodic ataxia type I (EA1). Although the mutation has been said to alter activation kinetics and to lower expression, we show here that the underlying molecular mechanisms may be more complex. Although F184 is positioned in the "peripheral" S1 helix, it occupies a central position in the 3D fold. We show in cut-open oocyte voltage-clamp recordings of gating and ionic currents of the Shaker Kv channel expressed in Xenopus oocytes that F184 not only interacts directly with the gating charges of the S4, but also creates a functional link to the selectivity filter of the neighboring subunit. This link leads to impaired fast and slow inactivation. The effect on fast inactivation is of an allosteric nature considering that fast inactivation is caused by a linked cytosolic ball peptide. The extensive effects of F184C provide a new mechanism underlying EA. SIGNIFICANCE STATEMENT: Episodic ataxia (EA) is an inherited disease that leads to occasional loss of motor control in combination with variable other symptoms such as vertigo or migraine. EA type I (EA1), studied here, is caused by mutations in a voltage-gated potassium channel that contributes to the generation of electrical signals in the brain. The mechanism by which mutations in voltage-gated potassium channels lead to EA is still unknown and there is no consistent pharmacological treatment. By studying in detail one disease-causing mutation in Kv1.1, we describe a novel molecular mechanism distinct from mechanisms described previously. This mechanism contributes to the understanding of potassium channel function in general and might lead to a better understanding of how EA develops.


Subject(s)
Ion Channel Gating/genetics , Kv1.1 Potassium Channel/genetics , Membrane Potentials/genetics , Point Mutation/genetics , Animals , Ataxia/genetics , Biophysical Phenomena/genetics , Crystallography, X-Ray , Electric Stimulation , Humans , Models, Molecular , Myokymia/genetics , Oocytes , Patch-Clamp Techniques , Time Factors , Xenopus laevis
10.
J Neurosci ; 35(42): 14397-405, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26490875

ABSTRACT

Inwardly rectifying potassium channels enforce tight control of resting membrane potential in excitable cells. The Kir3.2 channel, a member of the Kir3 subfamily of G-protein-activated potassium channels (GIRKs), plays several roles in the nervous system, including key responsibility in the GABAB pathway of inhibition, in pain perception pathways via opioid receptors, and is also involved in alcoholism. PKC phosphorylation acts on the channel to reduce activity, yet the mechanism is incompletely understood. Using the heterologous Xenopus oocyte system combined with molecular dynamics simulations, we show that PKC modulation of channel activity is dependent on Ser-196 in Kir3.2 such that, when this site is phosphorylated, the channel is less sensitive to PKC inhibition. This reduced inhibition is dependent on an interaction between phospho-Ser (SEP)-196 and Arg-201, reducing Arg-201 interaction with the sodium-binding site Asp-228. Neutralization of either SEP-196 or Arg-201 leads to a channel with reduced activity and increased sensitivity to PKC inhibition. This study clarifies the role of Ser-196 as an allosteric modulator of PKC inhibition and suggests that the SEP-196/Arg-201 interaction is critical for maintaining maximal channel activity. SIGNIFICANCE STATEMENT: The inwardly rectifying potassium 3.2 (Kir3.2) channel is found principally in neurons that regulate diverse brain functions, including pain perception, alcoholism, and substance addiction. Activation or inhibition of this channel leads to changes in neuronal firing and chemical message transmission. The Kir3.2 channel is subject to regulation by intracellular signals including sodium, G-proteins, ethanol, the phospholipid phosphatidylinositol bis-phosphate, and phosphorylation by protein kinases. Here, we take advantage of the recently published structure of Kir3.2 to provide an in-depth molecular view of how phosphorylation of a specific residue previously thought to be the target of PKC promotes channel gating and acts as an allosteric modulator of PKC-mediated inhibition.


Subject(s)
Biophysical Phenomena/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/genetics , Membrane Potentials/physiology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Electric Stimulation , Enzyme Inhibitors/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Membrane Potentials/drug effects , Microinjections , Models, Molecular , Oocytes , Patch-Clamp Techniques , Phorbol 12,13-Dibutyrate/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation , Point Mutation/genetics , Protein Kinase C/metabolism , Serine/genetics , Xenopus laevis
11.
J Neurosci ; 35(19): 7626-42, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972186

ABSTRACT

The superficial dorsal horn of the spinal cord contains numerous inhibitory interneurons, which regulate the transmission of information perceived as touch, pain, or itch. Despite the importance of these cells, our understanding of their roles in the neuronal circuitry is limited by the difficulty in identifying functional populations. One group that has been identified and characterized consists of cells in the mouse that express green fluorescent protein (GFP) under control of the prion protein (PrP) promoter. Previous reports suggested that PrP-GFP cells belonged to a single morphological class (central cells), received inputs exclusively from unmyelinated primary afferents, and had axons that remained in lamina II. However, we recently reported that the PrP-GFP cells expressed neuronal nitric oxide synthase (nNOS) and/or galanin, and it has been shown that nNOS-expressing cells are more diverse in their morphology and synaptic connections. We therefore used a combined electrophysiological, pharmacological, and anatomical approach to reexamine the PrP-GFP cells. We provide evidence that they are morphologically diverse (corresponding to "unclassified" cells) and receive synaptic input from a variety of primary afferents, with convergence onto individual cells. We also show that their axons project into adjacent laminae and that they target putative projection neurons in lamina I. This indicates that the neuronal circuitry involving PrP-GFP cells is more complex than previously recognized, and suggests that they are likely to have several distinct roles in regulating the flow of somatosensory information through the dorsal horn.


Subject(s)
Afferent Pathways/physiology , Green Fluorescent Proteins/metabolism , Interneurons/metabolism , Prions/metabolism , Spinal Cord/cytology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Capsaicin/pharmacology , Female , Green Fluorescent Proteins/genetics , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/pharmacology , Nitric Oxide Synthase Type I/metabolism , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Prions/genetics , Receptors, Neurokinin-1/metabolism , Sensory System Agents/pharmacology
12.
J Neurosci ; 35(38): 13133-47, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26400943

ABSTRACT

Night blindness can result from impaired photoreceptor function and a subset of cases have been linked to dysfunction of Cav1.4 calcium channels and in turn compromised synaptic transmission. Here, we show that active zone proteins RIM1/2 are important regulators of Cav1.4 channel function in mouse rod photoreceptors and thus synaptic activity. The conditional double knock-out (cdko) of RIM1 and RIM2 from rods starting a few weeks after birth did not change Cav1.4 protein expression at rod ribbon synapses nor was the morphology of the ribbon altered. Heterologous overexpression of RIM2 with Cav1.4 had no significant influence on current density when examined with BaCl2 as the charge carrier. Nonetheless, whole-cell voltage-clamp recordings from cdko rods revealed a profound reduction in Ca(2+) currents. Concomitantly, we observed a 4-fold reduction in spontaneous miniature release events from the cdko rod terminals and an almost complete absence of evoked responses when monitoring changes in membrane incorporation after strong step depolarizations. Under control conditions, 49 and 83 vesicles were released with 0.2 and 1 s depolarizations, respectively, which is close to the maximal number of vesicles estimated to be docked at the base of the ribbon active zone, but without RIM1/2, only a few vesicles were stimulated for release after a 1 s stimulation. In conclusion, our study shows that RIM1/2 potently enhance the influx of Ca(2+) into rod terminals through Cav1.4 channels, which is vitally important for the release of vesicles from the rod ribbon. Significance statement: Active zone scaffolding proteins are thought to bring multiple components involved in Ca(2+)-dependent exocytosis into functional interactions. We show that removal of scaffolding proteins RIM1/2 from rod photoreceptor ribbon synapses causes a dramatic loss of Ca(2+) influx through Cav1.4 channels and a correlated reduction in evoked release, yet the channels remain localized to synaptic ribbons in a normal fashion. Our findings strongly argue that RIM1/2 facilitate Ca(2+) entry and in turn Ca(2+) evoked release by modulating Cav1.4 channel openings; however, RIM1/2 are not needed for the retention of Cav1.4 at the synapse. In summary, a key function of RIM1/2 at rod ribbons is to enhance Cav1.4 channel activity, possibly through direct or indirect modulation of the channel.


Subject(s)
Biophysical Phenomena/genetics , Calcium Channels/metabolism , Calcium/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Retinal Rod Photoreceptor Cells/physiology , rab3 GTP-Binding Proteins/metabolism , Animals , Aspartic Acid/pharmacology , Barium Compounds/pharmacology , Biophysical Phenomena/drug effects , Calcium Channels/genetics , Calcium Channels, L-Type , Chlorides/pharmacology , Excitatory Amino Acid Agents/pharmacology , GTP-Binding Proteins/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Retina/cytology , Retinal Rod Photoreceptor Cells/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , rab3 GTP-Binding Proteins/genetics
13.
J Neurophysiol ; 116(2): 686-97, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27193322

ABSTRACT

Horizontal cells form the first laterally interacting network of inhibitory interneurons in the retina. Dopamine released onto horizontal cells under photic and circadian control modulates horizontal cell function. Using isolated, identified horizontal cells from a connexin-57-iCre × ROSA26-tdTomato transgenic mouse line, we investigated dopaminergic modulation of calcium channel currents (ICa) with whole cell patch-clamp techniques. Dopamine (10 µM) blocked 27% of steady-state ICa, an action blunted to 9% in the presence of the L-type Ca channel blocker verapamil (50 µM). The dopamine type 1 receptor (D1R) agonist SKF38393 (20 µM) inhibited ICa by 24%. The D1R antagonist SCH23390 (20 µM) reduced dopamine and SKF38393 inhibition. Dopamine slowed ICa activation, blocking ICa by 38% early in a voltage step. Enhanced early inhibition of ICa was eliminated by applying voltage prepulses to +120 mV for 100 ms, increasing ICa by 31% and 11% for early and steady-state currents, respectively. Voltage-dependent facilitation of ICa and block of dopamine inhibition after preincubation with a Gßγ-blocking peptide suggested involvement of Gßγ proteins in the D1R-mediated modulation. When the G protein activator guanosine 5'-O-(3-thiotriphosphate) (GTPγS) was added intracellularly, ICa was smaller and showed the same slowed kinetics seen during D1R activation. With GTPγS in the pipette, additional block of ICa by dopamine was only 6%. Strong depolarizing voltage prepulses restored the GTPγS-reduced early ICa amplitude by 36% and steady-state ICa amplitude by 3%. These results suggest that dopaminergic inhibition of ICa via D1Rs is primarily mediated through the action of Gßγ proteins in horizontal cells.


Subject(s)
Calcium Channels/physiology , Membrane Potentials/physiology , Receptors, Dopamine D1/metabolism , Retinal Horizontal Cells/physiology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Connexins/genetics , Connexins/metabolism , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plant Lectins/genetics , Plant Lectins/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Retina/cytology , Retinal Horizontal Cells/drug effects , Spiperone/pharmacology , omega-Conotoxin GVIA/pharmacology
14.
Hippocampus ; 26(5): 589-600, 2016 May.
Article in English | MEDLINE | ID: mdl-26482652

ABSTRACT

It is well known that the neuronal effects of vascular endothelial growth factor (VEGF) include modulating learning and memory, plasticity of mature neurons, and synaptic transmission in addition to neurogenesis. However, there is conflicting evidence particularly of its role in the regulation of excitatory synaptic activity. In this study, application of the patch-clamp technique revealed that lower doses (10 and 50 ng/mL) of VEGF enhanced excitatory neurotransmission in hippocampal slices of mice through both presynaptic and postsynaptic mechanisms. However, the effects were reversed by higher doses of VEGF (>100 ng/mL), which inhibited excitatory neurotransmission via a presynaptic mechanism. These competing, concentration-dependent effects of VEGF suggested that different pathways were involved. The involvement of the Notch1 receptor was tested in the modulation of VEGF on synaptic activity by using heterozygous Notch1(+/-) mice. Notch1 knockdown did not influence the inhibitory effect of high VEGF doses (200 ng/mL) but reduced the enhancement effects of low concentration of VEGF (50 ng/mL) at the postsynaptic level, which might be due to the decreased level of VEGF receptor. The results indicate that the Notch1 receptor plays a role in VEGF-induced modulation of synaptic activity, which provides new insights into a complex VEGF/Notch signaling cross-talk. These findings set the groundwork for understanding new mechanisms of Notch signaling and the neurotrophic effects of VEGF, which is beneficial to develop new therapeutic targets to the VEGF/Notch axis and improve current treatments for neural diseases.


Subject(s)
Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Hippocampus/cytology , Neurons/drug effects , Receptor, Notch1/deficiency , Vascular Endothelial Growth Factor A/pharmacology , Animals , Animals, Newborn , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Dose-Response Relationship, Drug , Electric Stimulation , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/physiology , Receptor, Notch1/genetics , Statistics, Nonparametric , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Muscle Nerve ; 53(2): 260-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26012778

ABSTRACT

INTRODUCTION: Reports of muscle testing are frequently limited to maximal force alone. The experiments reported here show that force generation and relaxation rates can be obtained from the same experiments and provide a more complete functional characterization. METHODS: Partial in situ testing was performed on the tibialis anterior of young wild-type (WT) mice, young mdx mice, and old mdx mice. Force, force generation rate, and relaxation rates were measured during a fatigue test, 2 frequency-force tests, and a passive tension test. RESULTS: We measured increased force but decreased force generation rate in WT compared with mdx muscles, and increased force but decreased relaxation rate of old compared with young mdx muscles. Young mdx muscles were the most sensitive to increases in passive tension. CONCLUSIONS: These measurements offer an improved understanding of muscle capability and are readily acquired by further analysis of the same tests used to obtain force measurements.


Subject(s)
Muscle Contraction/physiology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Age Factors , Animals , Biophysical Phenomena/genetics , Electric Stimulation , Fatigue/etiology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics
16.
Neurobiol Dis ; 73: 106-17, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25281316

ABSTRACT

Dominant loss-of-function mutations in voltage-gated sodium channel NaV1.1 cause Dravet Syndrome, an intractable childhood-onset epilepsy. NaV1.1(+/-) Dravet Syndrome mice in C57BL/6 genetic background exhibit severe seizures, cognitive and social impairments, and premature death. Here we show that Dravet Syndrome mice in pure 129/SvJ genetic background have many fewer seizures and much less premature death than in pure C57BL/6 background. These mice also have a higher threshold for thermally induced seizures, fewer myoclonic seizures, and no cognitive impairment, similar to patients with Genetic Epilepsy with Febrile Seizures Plus. Consistent with this mild phenotype, mutation of NaV1.1 channels has much less physiological effect on neuronal excitability in 129/SvJ mice. In hippocampal slices, the excitability of CA1 Stratum Oriens interneurons is selectively impaired, while the excitability of CA1 pyramidal cells is unaffected. NaV1.1 haploinsufficiency results in increased rheobase and threshold for action potential firing and impaired ability to sustain high-frequency firing. Moreover, deletion of NaV1.1 markedly reduces the amplification and integration of synaptic events, further contributing to reduced excitability of interneurons. Excitability is less impaired in inhibitory neurons of Dravet Syndrome mice in 129/SvJ genetic background. Because specific deletion of NaV1.1 in forebrain GABAergic interneuons is sufficient to cause the symptoms of Dravet Syndrome in mice, our results support the conclusion that the milder phenotype in 129/SvJ mice is caused by lesser impairment of sodium channel function and electrical excitability in their forebrain interneurons. This mild impairment of excitability of interneurons leads to a milder disease phenotype in 129/SvJ mice, similar to Genetic Epilepsy with Febrile Seizures Plus in humans.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neural Inhibition/genetics , Action Potentials/genetics , Animals , Animals, Newborn , Biophysical Phenomena/genetics , Conditioning, Psychological/physiology , Disease Models, Animal , Epilepsies, Myoclonic/etiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fear/psychology , Hippocampus/cytology , Hyperthermia, Induced/adverse effects , In Vitro Techniques , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/physiology , Sodium Channel Blockers/pharmacology
17.
Neurosignals ; 23(1): 20-33, 2015.
Article in English | MEDLINE | ID: mdl-26673921

ABSTRACT

BACKGROUND/AIMS: Kir2.1 (KCNJ2) channels are expressed in neurons, skeletal muscle and cardiac tissue and maintain the resting membrane potential. The activity of those channels is regulated by diverse signalling molecules. The present study explored whether Kir2.1 channels are sensitive to the transporter and channels regulating kinases SPAK (SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase 1), which are in turn regulated by WNK (with-no-K[Lys]) kinases. METHODS: cRNA encoding Kir2.1 was injected into Xenopus laevis oocytes with or without additional injection of cRNA encoding wild-type SPAK, constitutively active T233E SPAK, WNK insensitive T233A SPAK, catalytically inactive D212A SPAK, wild-type OSR1, constitutively active T185E OSR1, WNK insensitive T185A OSR1 and catalytically inactive D164A OSR1. Inwardly rectifying K+ channel activity was quantified utilizing dual electrode voltage clamp and Kir2.1 channel protein abundance in the cell membrane was measured utilizing chemiluminescence of Kir2.1 containing an extracellular HA-tag epitope. RESULTS: Kir2.1 activity was significantly enhanced by wild-type SPAK and T233E SPAK, but not by T233A SPAK and D212A SPAK, as well as by wild-type OSR1 and T185E OSR1, but not by T185A OSR1 and D164A OSR1. As shown for SPAK, the kinases enhanced Kir2.1 protein abundance in the cell membrane. The difference of current and conductance between oocytes expressing Kir2.1 together with SPAK or OSR1 and oocytes expressing Kir2.1 alone was dissipated following a 24 hours inhibition of channel insertion into the cell membrane by brefeldin A (5 µM). CONCLUSIONS: SPAK and OSR1 are both stimulators of Kir2.1 activity. They are presumably effective by enhancing channel insertion into the cell membrane.


Subject(s)
Potassium Channels, Inwardly Rectifying/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Brefeldin A/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Microinjections , Mutation/genetics , Oocytes , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Protein Serine-Threonine Kinases/genetics , Protein Synthesis Inhibitors/pharmacology , Xenopus laevis
18.
Mol Cell Neurosci ; 61: 110-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24914823

ABSTRACT

The polygenic origin of generalized absence epilepsy results in dysfunction of ion channels that allows the switch from physiological asynchronous to pathophysiological highly synchronous network activity. Evidence from rat and mouse models of absence epilepsy indicates that altered Ca(2+) channel activity contributes to cellular and network alterations that lead to seizure activity. Under physiological circumstances, high voltage-activated (HVA) Ca(2+) channels are important in determining the thalamic firing profile. Here, we investigated a possible contribution of HVA channels to the epileptic phenotype using a rodent genetic model of absence epilepsy. In this study, HVA Ca(2+) currents were recorded from neurons of three different thalamic nuclei that are involved in both sensory signal transmission and rhythmic-synchronized activity during epileptic spike-and-wave discharges (SWD), namely the dorsal part of the lateral geniculate nucleus (dLGN), the ventrobasal thalamic complex (VB) and the reticular thalamic nucleus (NRT) of epileptic Wistar Albino Glaxo rats from Rijswijk (WAG/Rij) and non-epileptic August Copenhagen Irish (ACI) rats. HVA Ca(2+) current densities in dLGN neurons were significantly increased in epileptic rats compared with non-epileptic controls while other thalamic regions revealed no differences between the strains. Application of specific channel blockers revealed that the increased current was carried by L-type Ca(2+) channels. Electrophysiological evidence of increased L-type current correlated with up-regulated mRNA and protein expression of a particular L-type channel, namely Cav1.3, in dLGN of epileptic rats. No significant changes were found for other HVA Ca(2+) channels. Moreover, pharmacological inactivation of L-type Ca(2+) channels results in altered firing profiles of thalamocortical relay (TC) neurons from non-epileptic rather than from epileptic rats. While HVA Ca(2+) channels influence tonic and burst firing in ACI and WAG/Rij differently, it is discussed that increased Cav1.3 expression may indirectly contribute to increased robustness of burst firing and thereby the epileptic phenotype of absence epilepsy.


Subject(s)
Calcium Channels/metabolism , Epilepsy/pathology , Membrane Potentials/physiology , Thalamic Nuclei/metabolism , Up-Regulation/physiology , Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/analogs & derivatives , Albuterol/pharmacology , Animals , Animals, Newborn , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Biophysical Phenomena/physiology , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Disease Models, Animal , Electric Stimulation , Epilepsy/genetics , Epilepsy/physiopathology , Immunosuppressive Agents/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutation Rate , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar , Salmeterol Xinafoate , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Thalamic Nuclei/pathology , Up-Regulation/genetics
19.
Neurodegener Dis ; 15(2): 93-108, 2015.
Article in English | MEDLINE | ID: mdl-25871323

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a late-onset fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the gene coding for the protein huntingtin and is characterised by progressive motor, psychiatric and cognitive decline. We previously demonstrated that normal synaptic function in HD could be restored by application of dopamine receptor agonists, suggesting that changes in the release or bioavailability of dopamine may be a contributing factor to the disease process. OBJECTIVE: In the present study, we examined the properties of midbrain dopaminergic neurones and dopamine release in presymptomatic and symptomatic transgenic HD mice. METHODS AND RESULTS: Using intracellular sharp recordings and immunohistochemistry, we found that neuronal excitability was increased due to a loss of slow afterhyperpolarisation and that these changes were related to an apparent functional loss and abnormal distribution of SK3 channels (KCa2.3 encoded by the KCNN3 gene), a class of small-conductance calcium-activated potassium channels. Electrochemical detection of dopamine showed that this observation was associated with an enhanced dopamine release in presymptomatic transgenic mice and a drastic reduction in symptomatic animals. These changes occurred in the context of a progressive expansion in the CAG repeat number and nuclear localisation of mutant protein within the substantia nigra pars compacta. CONCLUSIONS: Dopaminergic neuronal dysfunction is a key early event in HD disease progression. The initial increase in dopamine release appears to be related to a loss of SK3 channel function, a protein containing a polyglutamine tract. Implications for polyglutamine-mediated sequestration of SK3 channels, dopamine-associated DNA damage and CAG expansion are discussed in the context of HD.


Subject(s)
Brain/pathology , Dopaminergic Neurons/physiology , Huntington Disease/pathology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Biophysical Phenomena/genetics , Disease Models, Animal , Dopamine/metabolism , Electric Stimulation , Female , Gene Expression Regulation/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , In Vitro Techniques , Male , Membrane Potentials/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Trinucleotide Repeat Expansion/genetics , Tyrosine 3-Monooxygenase/metabolism
20.
Learn Mem ; 21(5): 298-304, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741110

ABSTRACT

The proper regulation of translation is required for the expression of long-lasting synaptic plasticity. A major site of translational control involves the phosphorylation of eukaryotic initiation factor 2 α (eIF2α) by PKR-like endoplasmic reticulum (ER) kinase (PERK). To determine the role of PERK in hippocampal synaptic plasticity, we used the Cre-lox expression system to selectively disrupt PERK expression in the adult mouse forebrain. Here, we demonstrate that in hippocampal area CA1, metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD) is associated with increased eIF2α phosphorylation, whereas stimulation of early- and late-phase long-term potentiation (E-LTP and L-LTP, respectively) is associated with decreased eIF2α phosphorylation. Interesting, although PERK-deficient mice exhibit exaggerated mGluR-LTD, both E-LTP and L-LTP remained intact. We also found that mGluR-LTD is associated with a PERK-dependent increase in eIF2α phosphorylation. Our findings are consistent with the notion that eIF2α phosphorylation is a key site for the bidirectional control of persistent forms of synaptic LTP and LTD and suggest a distinct role for PERK in mGluR-LTD.


Subject(s)
CA1 Region, Hippocampal/physiology , Long-Term Synaptic Depression/physiology , Receptors, Metabotropic Glutamate/metabolism , eIF-2 Kinase/metabolism , Analysis of Variance , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , DNA-Binding Proteins/metabolism , Electric Stimulation , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Transcription Factors/metabolism , eIF-2 Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL