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










Publication year range
1.
Small Methods ; 7(10): e2300218, 2023 10.
Article in English | MEDLINE | ID: mdl-37421204

ABSTRACT

Imaging of living synapses has relied for over two decades on the overexpression of synaptic proteins fused to fluorescent reporters. This strategy alters the stoichiometry of synaptic components and ultimately affects synapse physiology. To overcome these limitations, here a nanobody is presented that binds the calcium sensor synaptotagmin-1 (NbSyt1). This nanobody functions as an intrabody (iNbSyt1) in living neurons and is minimally invasive, leaving synaptic transmission almost unaffected, as suggested by the crystal structure of the NbSyt1 bound to Synaptotagmin-1 and by the physiological data. Its single-domain nature enables the generation of protein-based fluorescent reporters, as showcased here by measuring spatially localized presynaptic Ca2+ with a NbSyt1- jGCaMP8 chimera. Moreover, the small size of NbSyt1 makes it ideal for various super-resolution imaging methods. Overall, NbSyt1 is a versatile binder that will enable imaging in cellular and molecular neuroscience with unprecedented precision across multiple spatiotemporal scales.


Subject(s)
Microscopy , Synapses , Synapses/metabolism , Synaptic Transmission/physiology , Neurons , Calcium/metabolism
2.
Cereb Cortex ; 33(6): 2838-2856, 2023 03 10.
Article in English | MEDLINE | ID: mdl-35788286

ABSTRACT

Focal cortical epilepsies are frequently refractory to available anticonvulsant drug therapies. One key factor contributing to this state is the limited availability of animal models that allow to reliably study focal cortical seizures and how they recruit surrounding brain areas in vivo. In this study, we selectively expressed the inhibitory chemogenetic receptor, hM4D, in GABAergic neurons in focal cortical areas using viral gene transfer. GABAergic silencing using Clozapine-N-Oxide (CNO) demonstrated reliable induction of local epileptiform events in the electroencephalogram signal of awake freely moving mice. Anesthetized mice experiments showed consistent induction of focal epileptiform-events in both the barrel cortex (BC) and the medial prefrontal cortex (mPFC), accompanied by high-frequency oscillations, a known characteristic of human seizures. Epileptiform-events showed propagation indication with favored propagation pathways: from the BC on 1 hemisphere to its counterpart and from the BC to the mPFC, but not vice-versa. Lastly, sensory whisker-pad stimulation evoked BC epileptiform events post-CNO, highlighting the potential use of this model in studying sensory-evoked seizures. Combined, our results show that targeted chemogenetic inhibition of GABAergic neurons using hM4D can serve as a novel, versatile, and reliable model of focal cortical epileptic activity suitable for systematically studying cortical ictogenesis in different cortical areas.


Subject(s)
Clozapine , Epilepsies, Partial , GABAergic Neurons , Neurons , Gene Expression Regulation, Viral , Clozapine/analogs & derivatives , Electroencephalography , Seizures , Animals
3.
Brain Sci ; 12(6)2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35741613

ABSTRACT

The Locus coeruleus (LC) modulates various neuronal circuits throughout the brain. Its unique architectural organization encompasses a net of axonal innervation that spans the entire brain, while its somatic core is highly compact. Recent research revealed an unexpected cellular input specificity within the nucleus that can give rise to various network states that either broadcast norepinephrine signals throughout the brain or pointedly modulate specific brain areas. Such adaptive input-output functions likely surpass our existing network models that build upon a given synaptic wiring configuration between neurons. As the distances between noradrenergic neurons in the core of the LC are unusually small, neighboring neurons could theoretically impact each other via volume transmission of NE. We therefore set out to investigate if such interaction could be mediated through noradrenergic alpha2-receptors in a spiking neuron model of the LC. We validated our model of LC neurons through comparison with experimental patch-clamp data and identified key variables that impact alpha2-mediated inhibition of neighboring LC neurons. Our simulation confirmed a reliable autoinhibition of LC neurons after episodes of high neuronal activity that continue even after neuronal activity subsided. Additionally, dendro-somatic synapses inhibited spontaneous spiking in the somatic compartment of connected neurons in our model. We determined the exact position of hundreds of LC neurons in the mouse brain stem via a tissue clearing approach and, based on this, further determined that 25 percent of noradrenergic neurons have a neighboring LC neuron within less than a 25-micrometer radius. By modeling NE diffusion, we estimated that more than 15 percent of the alpha2-adrenergic receptors fraction can bind NE within such a diffusion radius. Our spiking neuron model of LC neurons predicts that repeated or long-lasting episodes of high neuronal activity induce partitioning of the gross LC network and reduce the spike rate in neighboring neurons at distances smaller than 25 µm. As these volume-mediating neighboring effects are challenging to test with the current methodology, our findings can guide future experimental approaches to test this phenomenon and its physiological consequences.

4.
Front Cell Dev Biol ; 8: 579513, 2020.
Article in English | MEDLINE | ID: mdl-33363141

ABSTRACT

Correct brain wiring depends on reliable synapse formation. Nevertheless, signaling codes promoting synaptogenesis are not fully understood. Here, we report a spinogenic mechanism that operates during neuronal development and is based on the interaction of tumor necrosis factor receptor-associated factor 6 (TRAF6) with the synaptic cell adhesion molecule neuroplastin. The interaction between these proteins was predicted in silico and verified by co-immunoprecipitation in extracts from rat brain and co-transfected HEK cells. Binding assays show physical interaction between neuroplastin's C-terminus and the TRAF-C domain of TRAF6 with a K d value of 88 µM. As the two proteins co-localize in primordial dendritic protrusions, we used young cultures of rat and mouse as well as neuroplastin-deficient mouse neurons and showed with mutagenesis, knock-down, and pharmacological blockade that TRAF6 is required by neuroplastin to promote early spinogenesis during in vitro days 6-9, but not later. Time-framed TRAF6 blockade during days 6-9 reduced mEPSC amplitude, number of postsynaptic sites, synapse density and neuronal activity as neurons mature. Our data unravel a new molecular liaison that may emerge during a specific window of the neuronal development to determine excitatory synapse density in the rodent brain.

5.
Neuron ; 95(3): 504-529, 2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28772120

ABSTRACT

Reversible silencing of neuronal activity is a powerful approach for isolating the roles of specific neuronal populations in circuit dynamics and behavior. In contrast with neuronal excitation, for which the majority of studies have used a limited number of optogenetic and chemogenetic tools, the number of genetically encoded tools used for inhibition of neuronal activity has vastly expanded. Silencing strategies vary widely in their mechanism of action and in their spatial and temporal scales. Although such manipulations are commonly applied, the design and interpretation of neuronal silencing experiments present unique challenges, both technically and conceptually. Here, we review the most commonly used tools for silencing neuronal activity and provide an in-depth analysis of their mechanism of action and utility for particular experimental applications. We further discuss the considerations that need to be given to experimental design, analysis, and interpretation of collected data. Finally, we discuss future directions for the development of new silencing approaches in neuroscience.


Subject(s)
Action Potentials/physiology , Brain/physiology , Light , Neurons/physiology , Neurosciences , Optogenetics , Animals , Humans , Optogenetics/methods , Rhodopsin/genetics
6.
PLoS One ; 12(7): e0179232, 2017.
Article in English | MEDLINE | ID: mdl-28749956

ABSTRACT

Sodium pumping rhodopsins (NaRs) are a unique member of the microbial-type I rhodopsin family which actively transport Na+ and H+ depending on ionic condition. In this study, we surveyed 12 different NaRs from various sources of eubacteria for their electrophysiological as well as spectroscopic properties. In mammalian cells several of these NaRs exhibited a Na+ based pump photocurrent and four interesting candidates were chosen for further characterization. Voltage dependent photocurrent amplitudes revealed a membrane potential-sensitive turnover rate, indicating the presence of an electrically-charged intermediate(s) in the photocycle reaction. The NaR from Salinarimonas rosea DSM21201 exhibited a red-shifted absorption spectrum, and slower kinetics compared to the first described sodium pump, KR2. Although the ratio of Na+ to H+ ion transport varied among the NaRs we tested, the NaRs from Flagellimonas sp_DIK and Nonlabens sp_YIK_SED-11 showed significantly higher Na+ selectivity when compared to KR2. All four further investigated NaRs showed a functional expression in dissociated hippocampal neuron culture and hyperpolarizing activity upon light-stimulation. Additionally, all four NaRs allowed optical inhibition of electrically-evoked neuronal spiking. Although efficiency of silencing was 3-5 times lower than silencing with the enhanced version of the proton pump AR3 from Halorubrum sodomense, our data outlines a new approach for hyperpolarization of excitable cells without affecting the intracellular and extracellular proton environment.


Subject(s)
Rhodopsin/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cells, Cultured , Gene Silencing , Humans , Mice , Optogenetics , Rats, Sprague-Dawley , Spectrum Analysis
7.
Proc Natl Acad Sci U S A ; 114(26): E5167-E5176, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28611221

ABSTRACT

Key mitochondrial functions such as ATP production, Ca2+ uptake and release, and substrate accumulation depend on the proton electrochemical gradient (ΔµH+) across the inner membrane. Although several drugs can modulate ΔµH+, their effects are hardly reversible, and lack cellular specificity and spatial resolution. Although channelrhodopsins are widely used to modulate the plasma membrane potential of excitable cells, mitochondria have thus far eluded optogenetic control. Here we describe a toolkit of optometabolic constructs based on selective targeting of channelrhodopsins with distinct functional properties to the inner mitochondrial membrane of intact cells. We show that our strategy enables a light-dependent control of the mitochondrial membrane potential (Δψm) and coupled mitochondrial functions such as ATP synthesis by oxidative phosphorylation, Ca2+ dynamics, and respiratory metabolism. By directly modulating Δψm, the mitochondria-targeted opsins were used to control complex physiological processes such as spontaneous beats in cardiac myocytes and glucose-dependent ATP increase in pancreatic ß-cells. Furthermore, our optometabolic tools allow modulation of mitochondrial functions in single cells and defined cell regions.


Subject(s)
Calcium Signaling/physiology , Channelrhodopsins/metabolism , Insulin-Secreting Cells/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Optogenetics , Animals , HEK293 Cells , HeLa Cells , Humans , Insulin-Secreting Cells/cytology , Oxygen Consumption/physiology , Rats , Rats, Sprague-Dawley
8.
Nat Neurosci ; 20(6): 836-844, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28288126

ABSTRACT

Fear-related disorders are thought to reflect strong and persistent fear memories. The basolateral amygdala (BLA) and the medial prefrontal cortex (mPFC) form strong reciprocal synaptic connections that play a key role in acquisition and extinction of fear memories. While synaptic contacts of BLA cells onto mPFC neurons are likely to play a crucial role in this process, the BLA connects with several additional nuclei within the fear circuit that could relay fear-associated information to the mPFC, and the contribution of direct monosynaptic BLA-mPFC inputs is not yet clear. Here we establish an optogenetic stimulation protocol that induces synaptic depression in BLA-mPFC synapses. In behaving mice, optogenetic high-frequency stimulation of BLA inputs to mPFC interfered with retention of cued associations, attenuated previously acquired cue-associated responses in mPFC neurons and facilitated extinction. Our findings demonstrate the contribution of BLA inputs to mPFC in forming and maintaining cued fear associations.


Subject(s)
Amygdala/physiology , Conditioning, Psychological/physiology , Fear/physiology , Neural Pathways/physiology , Optogenetics/methods , Prefrontal Cortex/physiology , Animals , Extinction, Psychological/physiology , Long-Term Synaptic Depression/physiology , Male , Memory/physiology , Mice , Mice, Transgenic , Neurons/physiology
9.
Methods Mol Biol ; 1408: 141-65, 2016.
Article in English | MEDLINE | ID: mdl-26965121

ABSTRACT

After the discovery of Channelrhodopsin, a light-gated ion channel, only a few people saw the diverse range of applications for such a protein. Now, more than 10 years later Channelrhodopsins have become widely accepted as the ultimate tool to control the membrane potential of excitable cells via illumination. The demand for more application-specific Channelrhodopsin variants started a race between protein engineers to design improved variants. Even though many engineered variants have undisputable advantages compared to wild-type variants, many users are alienated by the tremendous amount of new variants and their perplexing names. Here, we review new variants whose efficacy has already been proven in neurophysiological experiments, or variants which are likely to extend the optogenetic toolbox. Variants are described based on their mechanistic and operational properties in terms of expression, kinetics, ion selectivity, and wavelength responsivity.


Subject(s)
Chlorophyta/genetics , Plant Proteins/genetics , Protein Engineering/methods , Rhodopsin/genetics , Animals , Chlorophyta/metabolism , Gene Expression , Humans , Light , Models, Molecular , Optogenetics/methods , Plant Proteins/metabolism , Rhodopsin/metabolism
10.
Nat Neurosci ; 19(4): 554-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950004

ABSTRACT

We investigated the efficacy of optogenetic inhibition at presynaptic terminals using halorhodopsin, archaerhodopsin and chloride-conducting channelrhodopsins. Precisely timed activation of both archaerhodopsin and halorhodpsin at presynaptic terminals attenuated evoked release. However, sustained archaerhodopsin activation was paradoxically associated with increased spontaneous release. Activation of chloride-conducting channelrhodopsins triggered neurotransmitter release upon light onset. Thus, the biophysical properties of presynaptic terminals dictate unique boundary conditions for optogenetic manipulation.


Subject(s)
Biophysical Phenomena/physiology , Neural Inhibition/physiology , Optogenetics/methods , Presynaptic Terminals/metabolism , Synaptic Transmission/physiology , Animals , Cells, Cultured , Female , HEK293 Cells , Halorhodopsins/metabolism , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
11.
Nature ; 525(7570): 519-22, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26375004

ABSTRACT

It is commonly assumed, but has rarely been demonstrated, that sex differences in behaviour arise from sexual dimorphism in the underlying neural circuits. Parental care is a complex stereotypic behaviour towards offspring that is shared by numerous species. Mice display profound sex differences in offspring-directed behaviours. At their first encounter, virgin females behave maternally towards alien pups while males will usually ignore the pups or attack them. Here we show that tyrosine hydroxylase (TH)-expressing neurons in the anteroventral periventricular nucleus (AVPV) of the mouse hypothalamus are more numerous in mothers than in virgin females and males, and govern parental behaviours in a sex-specific manner. In females, ablating the AVPV TH(+) neurons impairs maternal behaviour whereas optogenetic stimulation or increased TH expression in these cells enhance maternal care. In males, however, this same neuronal cluster has no effect on parental care but rather suppresses inter-male aggression. Furthermore, optogenetic activation or increased TH expression in the AVPV TH(+) neurons of female mice increases circulating oxytocin, whereas their ablation reduces oxytocin levels. Finally, we show that AVPV TH(+) neurons relay a monosynaptic input to oxytocin-expressing neurons in the paraventricular nucleus. Our findings uncover a previously unknown role for this neuronal population in the control of maternal care and oxytocin secretion, and provide evidence for a causal relationship between sexual dimorphism in the adult brain and sex differences in parental behaviour.


Subject(s)
Hypothalamus/cytology , Hypothalamus/physiology , Maternal Behavior/physiology , Oxytocin/metabolism , Sex Characteristics , Aggression , Animals , Anterior Hypothalamic Nucleus/cytology , Anterior Hypothalamic Nucleus/enzymology , Anterior Hypothalamic Nucleus/physiology , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/metabolism , Female , Hypothalamus/enzymology , Male , Mice , Oxytocin/blood , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiology , Postpartum Period , Synapses/metabolism , Tyrosine 3-Monooxygenase/metabolism
12.
IEEE Rev Biomed Eng ; 7: 3-30, 2014.
Article in English | MEDLINE | ID: mdl-24802525

ABSTRACT

The brain is a large network of interconnected neurons where each cell functions as a nonlinear processing element. Unraveling the mysteries of information processing in the complex networks of the brain requires versatile neurostimulation and imaging techniques. Optogenetics is a new stimulation method which allows the activity of neurons to be modulated by light. For this purpose, the cell-types of interest are genetically targeted to produce light-sensitive proteins. Once these proteins are expressed, neural activity can be controlled by exposing the cells to light of appropriate wavelengths. Optogenetics provides a unique combination of features, including multimodal control over neural function and genetic targeting of specific cell-types. Together, these versatile features combine to a powerful experimental approach, suitable for the study of the circuitry of psychiatric and neurological disorders. The advent of optogenetics was followed by extensive research aimed to produce new lines of light-sensitive proteins and to develop new technologies: for example, to control the distribution of light inside the brain tissue or to combine optogenetics with other modalities including electrophysiology, electrocorticography, nonlinear microscopy, and functional magnetic resonance imaging. In this paper, the authors review some of the recent advances in the field of optogenetics and related technologies and provide their vision for the future of the field.


Subject(s)
Biomedical Research , Brain/physiology , Electroencephalography , Magnetic Resonance Imaging , Optogenetics , Animals , Caenorhabditis elegans , Humans , Microtechnology , Nervous System Diseases , Neurons/physiology , Rats
13.
Front Mol Neurosci ; 6: 2, 2013.
Article in English | MEDLINE | ID: mdl-23459413

ABSTRACT

Genetically encoded calcium indicators (GECIs) are powerful tools for systems neuroscience. Here we describe red, single-wavelength GECIs, "RCaMPs," engineered from circular permutation of the thermostable red fluorescent protein mRuby. High-resolution crystal structures of mRuby, the red sensor RCaMP, and the recently published red GECI R-GECO1 give insight into the chromophore environments of the Ca(2+)-bound state of the sensors and the engineered protein domain interfaces of the different indicators. We characterized the biophysical properties and performance of RCaMP sensors in vitro and in vivo in Caenorhabditis elegans, Drosophila larvae, and larval zebrafish. Further, we demonstrate 2-color calcium imaging both within the same cell (registering mitochondrial and somatic [Ca(2+)]) and between two populations of cells: neurons and astrocytes. Finally, we perform integrated optogenetics experiments, wherein neural activation via channelrhodopsin-2 (ChR2) or a red-shifted variant, and activity imaging via RCaMP or GCaMP, are conducted simultaneously, with the ChR2/RCaMP pair providing independently addressable spectral channels. Using this paradigm, we measure calcium responses of naturalistic and ChR2-evoked muscle contractions in vivo in crawling C. elegans. We systematically compare the RCaMP sensors to R-GECO1, in terms of action potential-evoked fluorescence increases in neurons, photobleaching, and photoswitching. R-GECO1 displays higher Ca(2+) affinity and larger dynamic range than RCaMP, but exhibits significant photoactivation with blue and green light, suggesting that integrated channelrhodopsin-based optogenetics using R-GECO1 may be subject to artifact. Finally, we create and test blue, cyan, and yellow variants engineered from GCaMP by rational design. This engineered set of chromatic variants facilitates new experiments in functional imaging and optogenetics.

14.
PLoS One ; 7(10): e46827, 2012.
Article in English | MEDLINE | ID: mdl-23056472

ABSTRACT

The C. elegans nervous system is particularly well suited for optogenetic analyses of circuit function: Essentially all connections have been mapped, and light can be directed at the neuron of interest in the freely moving, transparent animals, while behavior is observed. Thus, different nodes of a neuronal network can be probed for their role in controlling a particular behavior, using different optogenetic tools for photo-activation or -inhibition, which respond to different colors of light. As neurons may act in concert or in opposing ways to affect a behavior, one would further like to excite these neurons concomitantly, yet independent of each other. In addition to the blue-light activated Channelrhodopsin-2 (ChR2), spectrally red-shifted ChR variants have been explored recently. Here, we establish the green-light activated ChR chimera C1V1 (from Chlamydomonas and Volvox ChR1's) for use in C. elegans. We surveyed a number of red-shifted ChRs, and found that C1V1-ET/ET (E122T; E162T) works most reliable in C. elegans, with 540-580 nm excitation, which leaves ChR2 silent. However, as C1V1-ET/ET is very light sensitive, it still becomes activated when ChR2 is stimulated, even at 400 nm. Thus, we generated a highly efficient blue ChR2, the H134R; T159C double mutant (ChR2-HR/TC). Both proteins can be used in the same animal, in different neurons, to independently control each cell type with light, enabling a further level of complexity in circuit analyses.


Subject(s)
Caenorhabditis elegans/metabolism , Caenorhabditis elegans/radiation effects , Neurons/cytology , Neurons/radiation effects , Recombinant Fusion Proteins/metabolism , Rhodopsin/metabolism , Action Potentials/radiation effects , Animals , Behavior, Animal/physiology , Behavior, Animal/radiation effects , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Color , GABAergic Neurons/cytology , GABAergic Neurons/radiation effects , Motor Neurons/cytology , Motor Neurons/radiation effects , Muscle Contraction/radiation effects , Muscles/cytology , Muscles/physiology , Muscles/radiation effects , Neurons/metabolism , Spectrum Analysis
15.
J Biol Chem ; 287(38): 31804-12, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22843694

ABSTRACT

Channelrhodopsin-2 is a light-gated ion channel and a major tool of optogenetics. It is used to control neuronal activity via blue light. Here we describe the construction of color-tuned high efficiency channelrhodopsins (ChRs), based on chimeras of Chlamydomonas channelrhodopsin-1 and Volvox channelrhodopsin-1. These variants show superb expression and plasma membrane integration, resulting in 3-fold larger photocurrents in HEK cells compared with channelrhodopsin-2. Further molecular engineering gave rise to chimeric variants with absorption maxima ranging from 526 to 545 nm, dovetailing well with maxima of channelrhodopsin-2 derivatives ranging from 461 to 492 nm. Additional kinetic fine-tuning led to derivatives in which the lifetimes of the open state range from 19 ms to 5 s. Finally, combining green- with blue-absorbing variants allowed independent activation of two distinct neural cell populations at 560 and 405 nm. This novel panel of channelrhodopsin variants may serve as an important toolkit element for dual-color cell stimulation in neural circuits.


Subject(s)
Chlamydomonas/metabolism , Optogenetics/methods , Rhodopsin/chemistry , Volvox/metabolism , Animals , Calcium/chemistry , Calcium/metabolism , Color , Electrophysiology/methods , Genetic Engineering/methods , HEK293 Cells , Hippocampus/metabolism , Humans , Ions , Kinetics , Light , Models, Neurological , Oocytes/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Xenopus
16.
Cell ; 147(7): 1446-57, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22196724

ABSTRACT

The capture and utilization of light is an exquisitely evolved process. The single-component microbial opsins, although more limited than multicomponent cascades in processing, display unparalleled compactness and speed. Recent advances in understanding microbial opsins have been driven by molecular engineering for optogenetics and by comparative genomics. Here we provide a Primer on these light-activated ion channels and pumps, describe a group of opsins bridging prior categories, and explore the convergence of molecular engineering and genomic discovery for the utilization and understanding of these remarkable molecular machines.


Subject(s)
Opsins/genetics , Opsins/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Humans , Models, Molecular , Molecular Sequence Data , Opsins/chemistry , Phylogeny , Protein Engineering
17.
Nature ; 477(7363): 171-8, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21796121

ABSTRACT

Severe behavioural deficits in psychiatric diseases such as autism and schizophrenia have been hypothesized to arise from elevations in the cellular balance of excitation and inhibition (E/I balance) within neural microcircuitry. This hypothesis could unify diverse streams of pathophysiological and genetic evidence, but has not been susceptible to direct testing. Here we design and use several novel optogenetic tools to causally investigate the cellular E/I balance hypothesis in freely moving mammals, and explore the associated circuit physiology. Elevation, but not reduction, of cellular E/I balance within the mouse medial prefrontal cortex was found to elicit a profound impairment in cellular information processing, associated with specific behavioural impairments and increased high-frequency power in the 30-80 Hz range, which have both been observed in clinical conditions in humans. Consistent with the E/I balance hypothesis, compensatory elevation of inhibitory cell excitability partially rescued social deficits caused by E/I balance elevation. These results provide support for the elevated cellular E/I balance hypothesis of severe neuropsychiatric disease-related symptoms.


Subject(s)
Models, Neurological , Neural Inhibition/physiology , Neurons/metabolism , Prefrontal Cortex/physiology , Prefrontal Cortex/physiopathology , Social Behavior , Animals , Autistic Disorder/physiopathology , Disease Models, Animal , HEK293 Cells , Hippocampus/cytology , Humans , Learning , Mental Disorders/physiopathology , Mice , Motor Activity , Opsins/metabolism , Schizophrenia/physiopathology
18.
Channels (Austin) ; 4(3): 241-7, 2010.
Article in English | MEDLINE | ID: mdl-20714225

ABSTRACT

Channelrhodopsin-2 (ChR2) is a light-gated ion channel that is successfully used in neurosciences to depolarize cells with blue light. In this regard control of membrane voltage with light opens new perspectives for the characterization of ion channels and the search for inhibitors or modulators. Here, we report a control of membrane potential with ChR2 and the potassium channel mTrek for the purpose of screening for ion channel specific drugs. To verify principle we have chosen the voltage gated calcium channel Ca(V)3.2 as potential drug target. For this purpose we transfected the ChR2 gene into a HEK293T-cell line that permanently expresses Ca(V)3.2 and the K-channel mTrek. The resting potential was adjusted with low concentration of extracellular potassium ions whereas transient depolarization was achieved by activation of ChR2 with short pulses of blue light. Calcium ion influx through Ca(V)3.2 was monitored by observing fura-2 fluorescence. This approach allowed a repetitive activation of Ca(V)3.2. The Ca(2+) influx was specifically blocked by the inhibitor mibefradil. Since this assay is genetically-encoded, it may be employed for a variety of voltage-gated calcium channels and should be applicable to multi-well reader formats for high-throughput screening.


Subject(s)
Calcium Channels, T-Type/metabolism , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Ion Channels/drug effects , Light , Animals , Calcium Channel Blockers/pharmacology , Cell Line , Channelrhodopsins , Humans , Ion Channel Gating , Membrane Potentials , Mibefradil/pharmacology , Mice , Potassium Channels, Tandem Pore Domain/metabolism , Transfection
19.
Biophys J ; 98(5): 753-61, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20197028

ABSTRACT

Channelrhodopsins are light-gated ion channels that mediate vision in phototactic green algae like Chlamydomonas. In neurosciences, channelrhodopsins are widely used to light-trigger action potentials in transfected cells. All known channelrhodopsins preferentially conduct H(+). Previous studies have indicated the existence of an early and a late conducting state within the channelrhodopsin photocycle. Here, we show that for channelrhodopsin-2 expressed in Xenopus oocytes and HEK cells, the two open states have different ion selectivities that cause changes in the channelrhodopsin-2 reversal voltage during a light pulse. An enzyme kinetic algorithm was applied to convert the reversal voltages in various ionic conditions to conductance ratios for H(+) and divalent cations (Ca(2+) and/or Mg(2+)), as compared to monovalent cations (Na(+) and/or K(+)). Compared to monovalent cation conductance, the H(+) conductance, alpha, is approximately 3 x 10(6) and the divalent cation conductance, beta, is approximately 0.01 in the early conducting state. In the stationary mixture of the early and late states, alpha is larger and beta smaller, both by a factor of approximately 2. The results suggest that the ionic basis of light perception in Chlamydomonas is relatively nonspecific in the beginning of a light pulse but becomes more selective for protons during longer light exposures.


Subject(s)
Ion Channel Gating/radiation effects , Light , Protons , Rhodopsin/metabolism , Animals , Cations, Divalent/pharmacology , Cell Line , Humans , Ion Channel Gating/drug effects , Models, Biological , Oocytes/drug effects , Oocytes/metabolism , Oocytes/radiation effects , Xenopus/metabolism
20.
Nat Neurosci ; 11(6): 631-3, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18432196

ABSTRACT

The introduction of two microbial opsin-based tools, channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), to neuroscience has generated interest in fast, multimodal, cell type-specific neural circuit control. Here we describe a cation-conducting channelrhodopsin (VChR1) from Volvox carteri that can drive spiking at 589 nm, with excitation maximum red-shifted approximately 70 nm compared with ChR2. These results demonstrate fast photostimulation with yellow light, thereby defining a functionally distinct third category of microbial rhodopsin proteins.


Subject(s)
Carrier Proteins/physiology , Color , Neurons/physiology , Photic Stimulation/methods , Volvox/chemistry , Animals , Animals, Newborn , Carrier Proteins/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Electric Stimulation , Halorhodopsins/physiology , Hippocampus/cytology , Humans , Ion Channels , Light , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Transfection , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...