Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Neurosci Res ; 100(3): 880-896, 2022 03.
Article in English | MEDLINE | ID: mdl-35043465

ABSTRACT

Recently, we have identified CaMKIIα and CaMKIIß mutations in patients with neurodevelopmental disorders by whole exome sequencing study. Most CaMKII mutants have increased phosphorylation of Thr286/287, which induces autonomous activity of CaMKII, using cell culture experiments. In this study, we explored the pathological mechanism of motor dysfunction observed exclusively in a patient with Pro213Leu mutation in CaMKIIß using a mouse model of the human disease. The homozygous CaMKIIß Pro213Leu knockin mice showed age-dependent motor dysfunction and growth failure from 2 weeks after birth. In the cerebellum, the mutation did not alter the mRNA transcript level, but the CaMKIIß protein level was dramatically decreased. Furthermore, in contrast to previous result from cell culture, Thr287 phosphorylation of CaMKIIß was also reduced. CaMKIIß Pro213Leu knockin mice showed similar motor dysfunction as CaMKIIß knockout mice, newly providing evidence for a loss of function rather than a gain of function. Our disease model mouse showed similar phenotypes of the patient, except for epileptic seizures. We clearly demonstrated that the pathological mechanism is a reduction of mutant CaMKIIß in the brain, and the physiological aspects of mutation were greatly different between in vivo and cell culture.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cerebellum , Animals , Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebellum/metabolism , Humans , Mice , Mutation/genetics , Phosphorylation
2.
Int J Mol Sci ; 23(19)2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36233218

ABSTRACT

Specific antibodies are necessary for cellular and tissue expression, biochemical, and functional analyses of protein complexes. However, generating a specific antibody is often time-consuming and effort-intensive. The epitope tagging of an endogenous protein at an appropriate position can overcome this problem. Here, we investigated epitope tag position using AlphaFold2 protein structure prediction and developed Flag/DYKDDDDK tag knock-in CaMKIIα and CaMKIIß mice by combining CRISPR-Cas9 genome editing with electroporation (i-GONAD). With i-GONAD, it is possible to insert a small fragment of up to 200 bp into the genome of the target gene, enabling efficient and convenient tagging of a small epitope. Experiments with commercially available anti-Flag antibodies could readily detect endogenous CaMKIIα and ß proteins by Western blotting, immunoprecipitation, and immunohistochemistry. Our data demonstrated that the generation of Flag/DYKDDDDK tag knock-in mice by i-GONAD is a useful and convenient choice, especially if specific antibodies are unavailable.


Subject(s)
Electroporation , Gene Editing , Animals , Antibodies/metabolism , Blotting, Western , CRISPR-Cas Systems/genetics , Epitopes/genetics , Epitopes/metabolism , Gonads/metabolism , Mice
3.
Am J Hum Genet ; 102(2): 321-329, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29394991

ABSTRACT

Early-onset epileptic encephalopathies, including West syndrome (WS), are a group of neurological disorders characterized by developmental impairments and intractable seizures from early infancy. We have now identified biallelic CNPY3 variants in three individuals with WS; these include compound-heterozygous missense and frameshift variants in a family with two affected siblings (individuals 1 and 2) and a homozygous splicing variant in a consanguineous family (individual 3). All three individuals showed hippocampal malrotation. In individuals 1 and 2, electroencephalography (EEG) revealed characteristic fast waves and diffuse sharp- and slow-wave complexes. The fast waves were clinically associated with seizures. CNPY3 encodes a co-chaperone in the endoplasmic reticulum and regulates the subcellular distribution and responses of multiple Toll-like receptors. The amount of CNPY3 in lymphoblastoid cells derived from individuals 1 and 2 was severely lower than that in control cells. Cnpy3-knockout mice exhibited spastic or dystonic features under resting conditions and hyperactivity and anxiolytic behavior during the open field test. Also, their resting EEG showed enhanced activity in the fast beta frequency band (20-35 Hz), which could mimic the fast waves in individuals 1 and 2. These data suggest that CNPY3 and Cnpy3 perform essential roles in brain function in addition to known Toll-like receptor-dependent immune responses.


Subject(s)
Molecular Chaperones/genetics , Mutation , Seizures/genetics , Spasms, Infantile/genetics , Adolescent , Amino Acid Sequence , Animals , Child , Consanguinity , Electroencephalography , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Family , Female , Gene Expression , Heterozygote , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Infant , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Seizures/diagnostic imaging , Seizures/physiopathology , Sequence Alignment , Sequence Homology, Amino Acid , Siblings , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/physiopathology
4.
J Hum Genet ; 66(12): 1189-1192, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34168248

ABSTRACT

Brain malformations have heterogeneous genetic backgrounds. Tubulinopathies are a wide range of brain malformations caused by variants in tubulin and microtubules-associated genes. Recently biallelic variants in TTC5, also known as stress responsive activator of p300, have been reported in 11 patients from seven families with developmental delay, intellectual disability, and brain malformations. Here, we report compound heterozygous frameshift variants in TTC5 in a Japanese boy who showed severe psychomotor developmental delay and pseudobulbar palsy with growth failure. Brain magnetic resonance imaging showed a simplified gyral pattern and undetectable anterior limb of the internal capsule, suggesting tubulinopathies. Immunoblotting using lymphoblastoid cells derived from the patient showed undetectable TTC5 protein. Ttc5 silencing by RNA interference in Neuro2a cells reduced Tubulin ß3 protein level and caused abnormal cell cycle. Our report suggests a possible link between TTC5-related brain malformation and tubulinopathies.


Subject(s)
Brain/abnormalities , Frameshift Mutation , Genetic Predisposition to Disease , Nervous System Malformations/diagnosis , Nervous System Malformations/genetics , Phenotype , Transcription Factors/genetics , Tubulin/genetics , Brain/diagnostic imaging , Genetic Association Studies , Humans , Japan , Magnetic Resonance Imaging , Male , Tubulin/metabolism
5.
Circ Res ; 117(5): 401-12, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26078285

ABSTRACT

RATIONALE: Monitoring and controlling cardiac myocyte activity with optogenetic tools offer exciting possibilities for fundamental and translational cardiovascular research. Genetically encoded voltage indicators may be particularly attractive for minimal invasive and repeated assessments of cardiac excitation from the cellular to the whole heart level. OBJECTIVE: To test the hypothesis that cardiac myocyte-targeted voltage-sensitive fluorescence protein 2.3 (VSFP2.3) can be exploited as optogenetic tool for the monitoring of electric activity in isolated cardiac myocytes and the whole heart as well as function and maturity in induced pluripotent stem cell-derived cardiac myocytes. METHODS AND RESULTS: We first generated mice with cardiac myocyte-restricted expression of VSFP2.3 and demonstrated distinct localization of VSFP2.3 at the t-tubulus/junctional sarcoplasmic reticulum microdomain without any signs for associated pathologies (assessed by echocardiography, RNA-sequencing, and patch clamping). Optically recorded VSFP2.3 signals correlated well with membrane voltage measured simultaneously by patch clamping. The use of VSFP2.3 for human action potential recordings was confirmed by simulation of immature and mature action potentials in murine VSFP2.3 cardiac myocytes. Optical cardiograms could be monitored in whole hearts ex vivo and minimally invasively in vivo via fiber optics at physiological heart rate (10 Hz) and under pacing-induced arrhythmia. Finally, we reprogrammed tail-tip fibroblasts from transgenic mice and used the VSFP2.3 sensor for benchmarking functional and structural maturation in induced pluripotent stem cell-derived cardiac myocytes. CONCLUSIONS: We introduce a novel transgenic voltage-sensor model as a new method in cardiovascular research and provide proof of concept for its use in optogenetic sensing of physiological and pathological excitation in mature and immature cardiac myocytes in vitro and in vivo.


Subject(s)
Membrane Potentials/physiology , Myocytes, Cardiac/physiology , Optogenetics/methods , Animals , Humans , Mice , Mice, Transgenic , Voltage-Sensitive Dye Imaging/methods
6.
J Neurosci ; 34(50): 16611-20, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505314

ABSTRACT

Complex cognitive processes require neuronal activity to be coordinated across multiple scales, ranging from local microcircuits to cortex-wide networks. However, multiscale cortical dynamics are not well understood because few experimental approaches have provided sufficient support for hypotheses involving multiscale interactions. To address these limitations, we used, in experiments involving mice, genetically encoded voltage indicator imaging, which measures cortex-wide electrical activity at high spatiotemporal resolution. Here we show that, as mice recovered from anesthesia, scale-invariant spatiotemporal patterns of neuronal activity gradually emerge. We show for the first time that this scale-invariant activity spans four orders of magnitude in awake mice. In contrast, we found that the cortical dynamics of anesthetized mice were not scale invariant. Our results bridge empirical evidence from disparate scales and support theoretical predictions that the awake cortex operates in a dynamical regime known as criticality. The criticality hypothesis predicts that small-scale cortical dynamics are governed by the same principles as those governing larger-scale dynamics. Importantly, these scale-invariant principles also optimize certain aspects of information processing. Our results suggest that during the emergence from anesthesia, criticality arises as information processing demands increase. We expect that, as measurement tools advance toward larger scales and greater resolution, the multiscale framework offered by criticality will continue to provide quantitative predictions and insight on how neurons, microcircuits, and large-scale networks are dynamically coordinated in the brain.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Voltage-Sensitive Dye Imaging/methods , Wakefulness/physiology , Animals , Cerebral Cortex/chemistry , Electroencephalography/methods , Female , Male , Mice
7.
Pflugers Arch ; 465(3): 361-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23271451

ABSTRACT

Optical imaging has a long history in physiology and in neurophysiology in particular. Over the past 15 years or so, new methodologies have emerged that combine genetic engineering with light-based imaging methods. This merger has resulted in a tool box of genetically encoded optical indicators that enable nondestructive and long-lasting monitoring of neuronal activities at the cellular, circuit, and system level. This review describes the historical roots and fundamental concepts underlying these new approaches, evaluates current progress in this field, and concludes with a forward-looking perspective on current work and potential future developments in this field.


Subject(s)
Neuroimaging/methods , Optical Imaging/methods , Optogenetics/methods , Animals , History, 20th Century , Humans , Ion Channels/genetics , Ion Channels/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials , Neuroimaging/trends , Neurons/physiology , Optical Imaging/history , Optogenetics/trends , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
Nat Methods ; 7(8): 643-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20622860

ABSTRACT

Cortical information processing relies on synaptic interactions between diverse classes of neurons with distinct electrophysiological and connection properties. Uncovering the operational principles of these elaborate circuits requires the probing of electrical activity from selected populations of defined neurons. Here we show that genetically encoded voltage-sensitive fluorescent proteins (VSFPs) provide an optical voltage report from targeted neurons in culture, acute brain slices and living mice. By expressing VSFPs in pyramidal cells of mouse somatosensory cortex, we also demonstrate that these probes can report cortical electrical responses to single sensory stimuli in vivo. These protein-based voltage probes will facilitate the analysis of cortical circuits in genetically defined cell populations and are hence a valuable addition to the optogenetic toolbox.


Subject(s)
Brain/physiology , Diagnostic Imaging/methods , Luminescent Proteins , Membrane Potentials , Animals , Electrophysiological Phenomena , Methods , Mice , Pyramidal Cells , Somatosensory Cortex
9.
J Neurosci Methods ; 383: 109730, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36280087

ABSTRACT

BACKGROUND: Identification of biallelic CNPY3 mutations in patients with epileptic encephalopathy and abnormal electroencephalography findings of Cnpy3 knock-out mice have indicated that the loss of CNPY3 function causes neurological disorders such as epilepsy. However, the basic property of CNPY3 in the brain remains unclear. NEW METHOD: We generated C-terminal 2xHA-tag knock-in Cnpy3 mice by i-GONAD in vivo genome editing system to investigate the expression and function of Cnpy3 in the mouse brain. RESULTS: 2xHA-tagged Cnpy3 was confirmed by immunoblot analysis using anti-HA and CNPY3 antibodies, although HA tagging caused the decreased Cnpy3 protein level. Immunohistochemical analysis of Cnpy32xHA knock-in mice showed that Cnpy3-2xHA was predominantly expressed in the neuron. In addition, Cnpy3 and Cnpy3-2xHA were both localized in the endoplasmic reticulum and synaptosome and showed age-dependent expression changes in the brain. COMPARISON WITH EXISTING METHODS: Conventional Cnpy3 antibodies could not allow us to investigate the distribution of Cnpy3 expression in the brain, while HA-tagging revealed the expression of CNPY3 in neuronal cells. CONCLUSIONS: Taken together, we demonstrated that Cnpy32xHA knock-in mice would be useful to further elucidate the property of Cnpy3 in brain function and neurological disorders.


Subject(s)
Epilepsy , Neurons , Animals , Mice , Neurons/physiology , Brain/metabolism , Electroencephalography , Mice, Knockout , Antibodies
10.
Biophys J ; 103(4): 669-76, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22947928

ABSTRACT

Membrane proteins that respond to changes in transmembrane voltage are critical in regulating the function of living cells. The voltage-sensing domains (VSDs) of voltage-gated ion channels are extensively studied to elucidate voltage-sensing mechanisms, and yet many aspects of their structure-function relationship remain elusive. Here, we transplanted homologous amino acid motifs from the tetrameric voltage-activated potassium channel Kv3.1 to the monomeric VSD of Ciona intestinalis voltage-sensitive phosphatase (Ci-VSP) to explore which portions of Kv3.1 subunits depend on the tetrameric structure of Kv channels and which properties of Kv3.1 can be transferred to the monomeric Ci-VSP scaffold. By attaching fluorescent proteins to these chimeric VSDs, we obtained an optical readout to establish membrane trafficking and kinetics of voltage-dependent structural rearrangements. We found that motifs extending from 10 to roughly 100 amino acids can be readily transplanted from Kv3.1 into Ci-VSP to form engineered VSDs that efficiently incorporate into the plasma membrane and sense voltage. Some of the functional features of these engineered VSDs are reminiscent of Kv3.1 channels, indicating that these properties do not require interactions between Kv subunits or between the voltage sensing and the pore domains of Kv channels.


Subject(s)
Ciona intestinalis/enzymology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Engineering/methods , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Molecular Sequence Data , PC12 Cells , Phosphoric Monoester Hydrolases/chemistry , Porosity , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shaw Potassium Channels/chemistry
11.
J Neurophysiol ; 108(8): 2323-37, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22815406

ABSTRACT

Population signals from neuronal ensembles in cortex during behavior are commonly measured with EEG, local field potential (LFP), and voltage-sensitive dyes. A genetically encoded voltage indicator would be useful for detection of such signals in specific cell types. Here we describe how this goal can be achieved with Butterfly, a voltage-sensitive fluorescent protein (VSFP) with a subthreshold detection range and enhancements designed for voltage imaging from single neurons to brain in vivo. VSFP-Butterfly showed reliable membrane targeting, maximum response gain around standard neuronal resting membrane potential, fast kinetics for single-cell synaptic responses, and a high signal-to-noise ratio. Butterfly reports excitatory postsynaptic potentials (EPSPs) in cortical neurons, whisker-evoked responses in barrel cortex, 25-Hz gamma oscillations in hippocampal slices, and 2- to 12-Hz slow waves during brain state modulation in vivo. Our findings demonstrate that cell class-specific voltage imaging is practical with VSFP-Butterfly, and expand the genetic toolbox for the detection of neuronal population dynamics.


Subject(s)
Luminescent Proteins/genetics , Neurons/physiology , Voltage-Sensitive Dye Imaging/methods , Animals , Brain Waves , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Excitatory Postsynaptic Potentials , Fluorescence Resonance Energy Transfer , Hippocampus/cytology , Hippocampus/physiology , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Neurons/classification , Optogenetics , PC12 Cells , Phosphoric Monoester Hydrolases/chemistry , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 106(27): 11388-93, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19549872

ABSTRACT

Metabotropic glutamate receptor (mGluR) activation has been extensively studied under steady-state conditions. However, at central synapses, mGluRs are exposed to brief submillisecond glutamate transients and may not reach steady-state. The lack of information on the kinetics of mGluR activation impairs accurate predictions of their operation during synaptic transmission. Here, we report experiments designed to investigate mGluR kinetics in real-time. We inserted either CFP or YFP into the second intracellular loop of mGluR1beta. When these constructs were coexpressed in PC12 cells, glutamate application induced a conformational change that could be monitored, using fluorescence resonance energy transfer (FRET), with an EC(50) of 7.5 microM. The FRET response was mimicked by the agonist DHPG, abolished by the competitive antagonist MCPG, and partially inhibited by mGluR1-selective allosteric modulators. These results suggest that the FRET response reports active conformations of mGluR1 dimers. The solution exchange at the cell membrane was optimized for voltage-clamped cells by recording the current induced by co-application of 30 mM potassium. When glutamate was applied at increasing concentrations up to 2 mM, the activation time course decreased to a minimum of approximately 10 ms, whereas the deactivation time course remained constant (approximately 50 ms). During long-lasting applications, no desensitization was observed. In contrast, we observed a robust sensitization of the FRET response that developed over approximately 400 ms. Activation, deactivation, and sensitization time courses and amplitudes were used to derive a kinetic scheme and rate constants, from which we inferred the EC(50) and frequency dependence of mGluR1 activation under non-steady-state conditions, as occurs during synaptic transmission.


Subject(s)
Fluorescence Resonance Energy Transfer , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/metabolism , Animals , Kinetics , Models, Biological , PC12 Cells , Protein Conformation , Protein Multimerization , Rats
13.
Exp Physiol ; 96(1): 13-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20851856

ABSTRACT

Over the last decade, researchers in our laboratory have engineered and developed several series of genetically encoded voltage-sensitive fluorescent proteins (VSFPs) by molecular fusion of a voltage-sensing domain operand with different fluorescent reporter proteins. These genetically encoded VSFPs have been shown to provide a reliable optical report of membrane potential from targeted neurons and muscle cells in culture or in living animals. However, these various reporters also exhibit discrepancies in both their voltage-sensing and targeting properties that are essentially related to the intrinsic characteristics of the fluorescent reporter proteins. It is therefore important carefully to select the sensor that is most appropriate for the particular question being investigated experimentally. Here we examine the current state of this subfield of optogenetics, address current limitations and challenges, and discuss what is likely to be feasible in the near future.


Subject(s)
Genetic Engineering/methods , Luminescent Proteins/chemistry , Membrane Potentials/physiology , Muscle Cells/physiology , Neurons/physiology , Optics and Photonics/methods , Voltage-Sensitive Dye Imaging/methods , Animals , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Muscle Cells/cytology , Muscle Cells/metabolism , Neurons/cytology , Neurons/metabolism
14.
Nat Commun ; 12(1): 2107, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833240

ABSTRACT

Vacuolar H+-ATPases (V-ATPases) transport protons across cellular membranes to acidify various organelles. ATP6V0A1 encodes the a1-subunit of the V0 domain of V-ATPases, which is strongly expressed in neurons. However, its role in brain development is unknown. Here we report four individuals with developmental and epileptic encephalopathy with ATP6V0A1 variants: two individuals with a de novo missense variant (R741Q) and the other two individuals with biallelic variants comprising one almost complete loss-of-function variant and one missense variant (A512P and N534D). Lysosomal acidification is significantly impaired in cell lines expressing three missense ATP6V0A1 mutants. Homozygous mutant mice harboring human R741Q (Atp6v0a1R741Q) and A512P (Atp6v0a1A512P) variants show embryonic lethality and early postnatal mortality, respectively, suggesting that R741Q affects V-ATPase function more severely. Lysosomal dysfunction resulting in cell death, accumulated autophagosomes and lysosomes, reduced mTORC1 signaling and synaptic connectivity, and lowered neurotransmitter contents of synaptic vesicles are observed in the brains of Atp6v0a1A512P/A512P mice. These findings demonstrate the essential roles of ATP6V0A1/Atp6v0a1 in neuronal development in terms of integrity and connectivity of neurons in both humans and mice.


Subject(s)
Brain Diseases/genetics , Brain/growth & development , Neurons/physiology , Neurotransmitter Agents/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Animals , Autophagosomes/pathology , Brain Mapping/methods , Cathepsin D/metabolism , Cell Line , HEK293 Cells , Humans , Loss of Function Mutation/genetics , Lysosomes/pathology , Magnetic Resonance Imaging/methods , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mutation, Missense/genetics , Neurons/cytology , Synaptic Vesicles/pathology
15.
Biophys J ; 96(10): 3959-76, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19450468

ABSTRACT

Fluorescent protein voltage sensors are recombinant proteins that are designed as genetically encoded cellular probes of membrane potential using mechanisms of voltage-dependent modulation of fluorescence. Several such proteins, including VSFP2.3 and VSFP3.1, were recently reported with reliable function in mammalian cells. They were designed as molecular fusions of the voltage sensor of Ciona intestinalis voltage sensor containing phosphatase with a fluorescence reporter domain. Expression of these proteins in cell membranes is accompanied by additional dynamic membrane capacitance, or "sensing capacitance", with feedback effect on the native electro-responsiveness of targeted cells. We used recordings of sensing currents and fluorescence responses of VSFP2.3 and of VSFP3.1 to derive kinetic models of the voltage-dependent signaling of these proteins. Using computational neuron simulations, we quantitatively investigated the perturbing effects of sensing capacitance on the input/output relationship in two central neuron models, a cerebellar Purkinje and a layer 5 pyramidal neuron. Probe-induced sensing capacitance manifested as time shifts of action potentials and increased synaptic input thresholds for somatic action potential initiation with linear dependence on the membrane density of the probe. Whereas the fluorescence signal/noise grows with the square root of the surface density of the probe, the growth of sensing capacitance is linear. We analyzed the trade-off between minimization of sensing capacitance and signal/noise of the optical read-out depending on kinetic properties and cellular distribution of the probe. The simulation results suggest ways to reduce capacitive effects at a given level of signal/noise. Yet, the simulations indicate that significant improvement of existing probes will still be required to report action potentials in individual neurons in mammalian brain tissue in single trials.


Subject(s)
Electric Conductivity , Luminescent Proteins/pharmacology , Neurons/drug effects , Neurons/physiology , Cell Membrane/metabolism , Computer Simulation , Electric Capacitance , Fluorescence , Gene Expression Regulation , Luminescent Proteins/metabolism , Models, Biological , Neurons/cytology , Purkinje Cells/drug effects , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiology
16.
Biophys J ; 96(2): L19-21, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19167283

ABSTRACT

The N-terminus of Ciona intestinalis (Ci-VSP) is a voltage-sensing domain (VSD) controlling the activity of a phosphatase domain on the C terminus. By replacing the phosphatase domain with a tandem of fluorescent proteins, CFP and YFP, a family of fluorescence resonance energy transfer-based, genetically encoded voltage-sensing fluorescent protein (VSFP) was created. VSFP2.3, one of the latest versions of this family, showed large changes in YFP emission upon changes in membrane potential with CFP excitation when expressed in Xenopus laevis oocytes. The time course of the fluorescence has two components: the fast component correlates with the time course of sensing current produced by the charge movement, while the slow component is at least one order-of-magnitude slower than the sensing current. This suggests that the tandem of fluorescent proteins reports a secondary conformational transition of the VSD which resembles the relaxation of the VSD of Ci-VSP described in detail for the Ci-VSP. This observation indicates that the relaxation of the VSD of VSFP2.3 is a global conformational change that encompasses the entire S4 segment.


Subject(s)
Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Membrane Potentials/physiology , Animals , Ciona intestinalis , Fluorescence , Fluorometry/methods , Ion Transport , Oocytes/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Xenopus laevis
17.
Ann Clin Transl Neurol ; 5(3): 280-296, 2018 03.
Article in English | MEDLINE | ID: mdl-29560374

ABSTRACT

Objective: α (CAMK2A) and ß (CAMK2B) isoforms of Calcium/calmodulin-dependent protein kinase II (CaMKII) play a pivotal role in neuronal plasticity and in learning and memory processes in the brain. Here, we explore the possible involvement of α- and ß-CaMKII variants in neurodevelopmental disorders. Methods: Whole-exome sequencing was performed for 976 individuals with intellectual disability, developmental delay, and epilepsy. The effect of CAMK2A and CAMK2B variants on CaMKII structure and firing of neurons was evaluated by computational structural analysis, immunoblotting, and electrophysiological analysis. Results: We identified a total of five de novo CAMK2A and CAMK2B variants in three and two individuals, respectively. Seizures were common to three individuals with CAMK2A variants. Using a minigene splicing assay, we demonstrated that a splice site variant caused skipping of exon 11 leading to an in-frame deletion of the regulatory segment of CaMKII α. By structural analysis, four missense variants are predicted to impair the interaction between the kinase domain and the regulatory segment responsible for the autoinhibition of its kinase activity. The Thr286/Thr287 phosphorylation as a result of release from autoinhibition was increased in three mutants when the mutants were stably expressed in Neuro-2a neuroblastoma cells. Expression of a CaMKII α mutant in primary hippocampal neurons significantly increased A-type K+ currents, which facilitated spike repolarization of single action potentials. Interpretation: Our data highlight the importance of CaMKII α and CaMKII ß and their autoinhibitory regulation in human brain function, and suggest the enhancement of A-type K+ currents as a possible pathophysiological basis.

18.
J Neurosci ; 25(17): 4252-9, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15858051

ABSTRACT

The synapses formed by the olfactory nerve (ON) convey sensory information to olfactory glomeruli, the first stage of central odor processing. Morphological and behavioral studies suggest that glomerular odor processing is plastic in neonate rodents. However, long-term synaptic plasticity, a cellular correlate of functional and structural plasticity, has not yet been demonstrated in this system. Here, we report that ON-->mitral cell (MC) synapses of 5- to 8-d-old mice express long-term depression (LTD) after brief low-frequency ON stimulation. Pharmacological techniques and imaging of presynaptic calcium signals demonstrate that ON-MC LTD is expressed presynaptically and requires the activation of metabotropic glutamate receptors but does not require fast synaptic transmission. LTD at the ON--> MC synapse is potentially relevant for the establishment, maintenance, and experience-dependent refinement of odor maps in the olfactory bulb.


Subject(s)
Long-Term Synaptic Depression/physiology , Neurons/cytology , Olfactory Bulb/cytology , Synapses/physiology , Animals , Animals, Newborn , Calcium/metabolism , Diagnostic Imaging/methods , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Evoked Potentials/drug effects , Evoked Potentials/physiology , Evoked Potentials/radiation effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Glycine/analogs & derivatives , Glycine/pharmacology , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/radiation effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred ICR , N-Methylaspartate/pharmacology , Patch-Clamp Techniques/methods , Quinoxalines/pharmacology , Sodium Channel Blockers/pharmacology , Sulpiride/pharmacology , Synapses/drug effects , Synapses/radiation effects , Tetrodotoxin/pharmacology , Time Factors , Valine/analogs & derivatives , Valine/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
19.
Sci Adv ; 2(8): e1501723, 2016 08.
Article in English | MEDLINE | ID: mdl-27540587

ABSTRACT

Corticotropin-releasing hormone (CRH), which is synthesized in the paraventricular nucleus (PVN) of the hypothalamus, plays an important role in the endocrine stress response. The excitability of CRH neurons is regulated by γ-aminobutyric acid (GABA)-containing neurons projecting to the PVN. We investigated the role of GABA in the regulation of CRH release. The release of CRH was impaired, accumulating in the cell bodies of CRH neurons in heterozygous GAD67-GFP (green fluorescent protein) knock-in mice (GAD67(+/GFP)), which exhibited decreased GABA content. The GABAA receptor (GABAAR) and the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1), but not the K(+)-Cl(-) cotransporter (KCC2), were expressed in the terminals of the CRH neurons at the median eminence (ME). In contrast, CRH neuronal somata were enriched with KCC2 but not with NKCC1. Thus, intracellular Cl(-) concentrations ([Cl(-)]i) may be increased at the terminals of CRH neurons compared with concentrations in the cell body. Moreover, GABAergic terminals projecting from the arcuate nucleus were present in close proximity to CRH-positive nerve terminals. Furthermore, a GABAAR agonist increased the intracellular calcium (Ca(2+)) levels in the CRH neuron terminals but decreased the Ca(2+) levels in their somata. In addition, the increases in Ca(2+) concentrations were prevented by an NKCC1 inhibitor. We propose a novel mechanism by which the excitatory action of GABA maintains a steady-state CRH release from axon terminals in the ME.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Receptors, GABA-A/metabolism , Solute Carrier Family 12, Member 2/metabolism , Symporters/genetics , gamma-Aminobutyric Acid/metabolism , Animals , Axons/metabolism , Calcium Signaling , GABAergic Neurons/metabolism , Gene Expression Regulation , Gene Knock-In Techniques , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Hypothalamus/metabolism , Median Eminence/metabolism , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, GABA-A/genetics , Solute Carrier Family 12, Member 2/genetics , Symporters/metabolism , K Cl- Cotransporters
20.
Neurophotonics ; 2(2)2015.
Article in English | MEDLINE | ID: mdl-26082930

ABSTRACT

The invention of membrane voltage protein indicators widens the reach of optical voltage imaging in cell physiology, most notably neurophysiology, by enabling membrane voltage recordings from genetically defined cell types in chronic and life-long preparations. While the last years have seen a dramatic improvement in the technical performance of these indicators, concomitant innovations in optogenetics, optical axon tracing, and high-speed digital microscopy are beginning to fulfill the age-old vision of an all-optical analysis of neuronal circuits, reaching beyond the limits of traditional electrode-based recordings. We will present our personal account of the development of protein voltage indicators from the pioneering days to the present state, including their applications in neurophysiology that has inspired our own work for more than a decade.

SELECTION OF CITATIONS
SEARCH DETAIL