Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Physiol ; 594(4): 915-36, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26627919

ABSTRACT

KEY POINTS: At the parallel fibre-Purkinje cell glutamatergic synapse, little or no Ca(2+) entry takes place through postsynaptic neurotransmitter receptors, although postsynaptic calcium increases are clearly involved in the synaptic plasticity. Postsynaptic voltage-gated Ca(2+) channels therefore constitute the sole rapid postsynaptic Ca(2+) signalling mechanism, making it essential to understand how they contribute to the synaptic signalling. Using a selective T-type calcium channel antagonist, we describe a T-type component of the EPSC that is activated by the AMPA receptor-mediated depolarization of the spine and thus will contribute to the local calcium dynamics. This component can amount up to 20% of the EPSC, and this fraction is maintained even at the high frequencies sometimes encountered in sensory processing. Modelling based on our biophysical characterization of T-type calcium channels in Purkinje cells suggests that the brief spine EPSCs cause the activated T-type channels to deactivate rather than inactivate, enabling repetitive activation. ABSTRACT: In the cerebellum, sensory information is conveyed to Purkinje cells (PC) via the granule cell/parallel fibre (PF) pathway. Plasticity at the PF-PC synapse is considered to be a mechanism of information storage in motor learning. The induction of synaptic plasticity in the cerebellum and elsewhere usually involves intracellular Ca(2+) signals. Unusually, postsynaptic Ca(2+) signalling in PF-PC spines does not involve ionotropic glutamatergic receptors because postsynaptic NMDA receptors are absent and the AMPA receptors are Ca(2+) -impermeable; postsynaptic voltage-gated Ca(2+) channels therefore constitute the sole rapid Ca(2+) signalling mechanism. Low-threshold activated T-type calcium channels are present at the synapse, although their contribution to PF-PC synaptic responses is unknown. Taking advantage of 3,5-dichloro-N-[1-(2,2-dimethyl-tetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide, a selective T-type channel antagonist, we show in the mouse that inhibition of these channels reduces PF-PC excitatory postsynaptic currents and excitatory postsynaptic potentials by 15-20%. This contribution was preserved during sparse input and repetitive activity. We characterized the biophysical properties of native T-type channels in young animals and modelled their activation during simulated dendritic excitatory postsynaptic potential waveforms. The comparison of modelled and observed synaptic responses suggests that T-type channels only activate in spines that are strongly depolarized by their synaptic input, a process requiring a high spine neck resistance. This brief and local activation ensures that T-type channels rapidly deactivate, thereby limiting inactivation during repetitive synaptic activity. T-type channels are therefore ideally situated to provide synaptic Ca(2+) entry at PF-PC spines.


Subject(s)
Calcium Channels, T-Type/metabolism , Excitatory Postsynaptic Potentials , Purkinje Cells/metabolism , Synapses/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling , Male , Mice , Mice, Inbred C57BL , Purkinje Cells/drug effects , Purkinje Cells/physiology , Synapses/physiology
2.
Sensors (Basel) ; 15(9): 24662-80, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26404317

ABSTRACT

Semiconductor nanocrystals (NCs) or quantum dots (QDs) are luminous point emitters increasingly being used to tag and track biomolecules in biological/biomedical imaging. However, their intracellular use as highlighters of single-molecule localization and nanobiosensors reporting ion microdomains changes has remained a major challenge. Here, we report the design, generation and validation of FRET-based nanobiosensors for detection of intracellular Ca(2+) and H⁺ transients. Our sensors combine a commercially available CANdot(®)565QD as an energy donor with, as an acceptor, our custom-synthesized red-emitting Ca(2+) or H⁺ probes. These 'Rubies' are based on an extended rhodamine as a fluorophore and a phenol or BAPTA (1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid) for H⁺ or Ca(2+) sensing, respectively, and additionally bear a linker arm for conjugation. QDs were stably functionalized using the same SH/maleimide crosslink chemistry for all desired reactants. Mixing ion sensor and cell-penetrating peptides (that facilitate cytoplasmic delivery) at the desired stoichiometric ratio produced controlled multi-conjugated assemblies. Multiple acceptors on the same central donor allow up-concentrating the ion sensor on the QD surface to concentrations higher than those that could be achieved in free solution, increasing FRET efficiency and improving the signal. We validate these nanosensors for the detection of intracellular Ca(2+) and pH transients using live-cell fluorescence imaging.


Subject(s)
Biosensing Techniques/instrumentation , Calcium/metabolism , Fluorescence Resonance Energy Transfer/instrumentation , Intracellular Space/metabolism , Molecular Imaging/methods , Protons , Animals , Biophysical Phenomena , Cell Line , Endocytosis , Endosomes/metabolism , Fluorescent Dyes/chemistry , Ions , Lysosomes/metabolism , Nanoparticles , Quantum Dots/chemistry , Rhodamines/chemistry , Titrimetry
3.
Elife ; 42015 Mar 31.
Article in English | MEDLINE | ID: mdl-25824291

ABSTRACT

The great demand for long-wavelength and high signal-to-noise Ca(2+) indicators has led us to develop CaRuby-Nano, a new functionalizable red calcium indicator with nanomolar affinity for use in cell biology and neuroscience research. In addition, we generated CaRuby-Nano dextran conjugates and an AM-ester variant for bulk loading of tissue. We tested the new indicator using in vitro and in vivo experiments demonstrating the high sensitivity of CaRuby-Nano as well as its power in dual color imaging experiments.


Subject(s)
Calcium/analysis , Fluorescent Dyes/chemistry , Indicators and Reagents/chemistry , Neurons/chemistry , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium/chemistry , Calcium Signaling , Color , Indicators and Reagents/chemical synthesis , Luminescent Measurements/methods , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Maleimides/chemistry , Membrane Potentials , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton/methods , Models, Chemical , Molecular Structure , Neurons/metabolism , Neurons/physiology , Reproducibility of Results
4.
Chem Sci ; 6(10): 5928-5937, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-29861916

ABSTRACT

Monitoring intracellular pH has drawn much attention due to its undeniably important function in cells. The widespread development of fluorescent imaging techniques makes pH sensitive fluorescent dyes valuable tools, especially red-emitting dyes which help to avoid the overcrowded green end of the spectral band. Herein, we present H-Rubies, a family of pH sensors based on a phenol moiety and a X-rhodamine fluorophore that display a bright red fluorescence upon acidification with pKa values spanning from 4 to 9. Slight structural modifications led to dramatic changes in their physicochemical properties and a relationship between their structures, their ability to form H-aggregates, and their apparent pKa was established. While molecular form H-Rubies can be used to monitor mitochondrial acidification of glioma cells, their functionalised forms were linked via click chemistry to dextrans or microbeads containing a near infrared Cy5 (Alexa-647) in order to provide ratiometric systems that were used to measure respectively the phagosomal and endosomal pH in macrophages (RAW 264.7 cells) using flow cytometry.

5.
Neuron ; 84(1): 137-151, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25220810

ABSTRACT

In cerebellar Purkinje cell dendrites, heterosynaptic calcium signaling induced by the proximal climbing fiber (CF) input controls plasticity at distal parallel fiber (PF) synapses. The substrate and regulation of this long-range dendritic calcium signaling are poorly understood. Using high-speed calcium imaging, we examine the role of active dendritic conductances. Under basal conditions, CF stimulation evokes T-type calcium signaling displaying sharp proximodistal decrement. Combined mGluR1 receptor activation and depolarization, two activity-dependent signals, unlock P/Q calcium spikes initiation and propagation, mediating efficient CF signaling at distal sites. These spikes are initiated in proximal smooth dendrites, independently from somatic sodium action potentials, and evoke high-frequency bursts of all-or-none fast-rising calcium transients in PF spines. Gradual calcium spike burst unlocking arises from increasing inactivation of mGluR1-modulated low-threshold A-type potassium channels located in distal dendrites. Evidence for graded activity-dependent CF calcium signaling at PF synapses refines current views on cerebellar supervised learning rules.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Dendrites/physiology , Kv Channel-Interacting Proteins/physiology , Purkinje Cells/physiology , Signal Transduction/physiology , Animals , Dendrites/ultrastructure , Ion Channel Gating/physiology , Mice , Organ Culture Techniques , Purkinje Cells/ultrastructure , Rats , Rats, Wistar
6.
Biochim Biophys Acta ; 1843(10): 2284-306, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24681159

ABSTRACT

Most chemical and, with only a few exceptions, all genetically encoded fluorimetric calcium (Ca(2+)) indicators (GECIs) emit green fluorescence. Many of these probes are compatible with red-emitting cell- or organelle markers. But the bulk of available fluorescent-protein constructs and transgenic animals incorporate green or yellow fluorescent protein (GFP and YFP respectively). This is, in part, not only heritage from the tendency to aggregate of early-generation red-emitting FPs, and due to their complicated photochemistry, but also resulting from the compatibility of green-fluorescent probes with standard instrumentation readily available in most laboratories and core imaging facilities. Photochemical constraints like limited water solubility and low quantum yield have contributed to the relative paucity of red-emitting Ca(2+) probes compared to their green counterparts, too. The increasing use of GFP and GFP-based functional reporters, together with recent developments in optogenetics, photostimulation and super-resolution microscopies, has intensified the quest for red-emitting Ca(2+) probes. In response to this demand more red-emitting chemical and FP-based Ca(2+)-sensitive indicators have been developed since 2009 than in the thirty years before. In this topical review, we survey the physicochemical properties of these red-emitting Ca(2+) probes and discuss their utility for biological Ca(2+) imaging. Using the spectral separability index Xijk (Oheim M., 2010. Methods in Molecular Biology 591: 3-16) we evaluate their performance for multi-color excitation/emission experiments, involving the identification of morphological landmarks with GFP/YFP and detecting Ca(2+)-dependent fluorescence in the red spectral band. We also establish a catalog of criteria for evaluating Ca(2+) indicators that ideally should be made available for each probe. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.


Subject(s)
Calcium/analysis , Fluorescent Dyes/chemistry , Molecular Imaging/methods , Optogenetics/methods , Photochemistry/methods , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Benzofurans/chemistry , Boron Compounds/chemistry , Calcium/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Humans , Imidazoles/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Rhodamines/chemistry , Spectrometry, Fluorescence , Thermodynamics , Red Fluorescent Protein
7.
Nano Lett ; 14(6): 2994-3001, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24754795

ABSTRACT

Small-molecule chemical calcium (Ca(2+)) indicators are invaluable tools for studying intracellular signaling pathways but have severe shortcomings for detecting local Ca(2+) entry. Nanobiosensors incorporating functionalized quantum dots (QDs) have emerged as promising alternatives but their intracellular use remains a major challenge. We designed cell-penetrating FRET-based Ca(2+) nanobiosensors for the detection of local Ca(2+) concentration transients, using commercially available CANdot565QD as a donor and CaRuby, a custom red-emitting Ca(2+) indicator, as an acceptor. With Ca(2+)-binding affinities covering the range of 3-20 µM, our CaRubies allow building sensors with a scalable affinity for detecting intracellular Ca(2+) transients at various concentrations. To facilitate their cytoplasmic delivery, QDs were further functionalized with a small cell-penetrating peptide (CPP) derived from hadrucalcin (HadUF1-11: H11), a ryanodine receptor-directed scorpion toxin identified within the venom of Hadrurus gertschi. Efficient internalization of QDs doubly functionalized with PEG5-CaRuby and H11 (in a molar ratio of 1:10:10, respectively) is demonstrated. In BHK cells expressing a N-methyl-d-aspartate receptor (NMDAR) construct, these nanobiosensors report rapid intracellular near-membrane Ca(2+) transients following agonist application when imaged by TIRF microscopy. Our work presents the elaboration of cell-penetrating FRET-based nanobiosensors and validates their function for detection of intracellular Ca(2+) transients.


Subject(s)
Biosensing Techniques/methods , Calcium Signaling/physiology , Calcium/metabolism , Cell-Penetrating Peptides/chemistry , Fluorescence Resonance Energy Transfer , Quantum Dots/chemistry , Animals , Cricetinae , HEK293 Cells , Humans , Scorpion Venoms/chemistry
8.
Proc Natl Acad Sci U S A ; 110(50): 20302-7, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277825

ABSTRACT

CaV3.1 T-type channels are abundant at the cerebellar synapse between parallel fibers and Purkinje cells where they contribute to synaptic depolarization. So far, no specific physiological function has been attributed to these channels neither as charge carriers nor more specifically as Ca(2+) carriers. Here we analyze their incidence on synaptic plasticity, motor behavior, and cerebellar motor learning, comparing WT animals and mice where T-type channel function has been abolished either by gene deletion or by acute pharmacological blockade. At the cellular level, we show that CaV3.1 channels are required for long-term potentiation at parallel fiber-Purkinje cell synapses. Moreover, basal simple spike discharge of the Purkinje cell in KO mice is modified. Acute or chronic T-type current blockade results in impaired motor performance in particular when a good body balance is required. Because motor behavior integrates reflexes and past memories of learned behavior, this suggests impaired learning. Indeed, subjecting the KO mice to a vestibulo-ocular reflex phase reversal test reveals impaired cerebellum-dependent motor learning. These data identify a role of low-voltage activated calcium channels in synaptic plasticity and establish a role for CaV3.1 channels in cerebellar learning.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Cerebellum/physiology , Learning/physiology , Long-Term Potentiation/drug effects , Purkinje Cells/metabolism , Synapses/metabolism , Animals , Benzamides , Calcium Channels, T-Type/genetics , Eye Movements/physiology , Mice , Mice, Knockout , Patch-Clamp Techniques , Piperidines , Rotarod Performance Test/adverse effects
9.
J Am Chem Soc ; 134(36): 14923-31, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22816677

ABSTRACT

We designed Calcium Rubies, a family of functionalizable BAPTA-based red-fluorescent calcium (Ca(2+)) indicators as new tools for biological Ca(2+) imaging. The specificity of this Ca(2+)-indicator family is its side arm, attached on the ethylene glycol bridge that allows coupling the indicator to various groups while leaving open the possibility of aromatic substitutions on the BAPTA core for tuning the Ca(2+)-binding affinity. Using this possibility we now synthesize and characterize three different CaRubies with affinities between 3 and 22 µM. Their long excitation and emission wavelengths (peaks at 586/604 nm) allow their use in otherwise challenging multicolor experiments, e.g., when combining Ca(2+) uncaging or optogenetic stimulation with Ca(2+) imaging in cells expressing fluorescent proteins. We illustrate this capacity by the detection of Ca(2+) transients evoked by blue light in cultured astrocytes expressing CatCh, a light-sensitive Ca(2+)-translocating channelrhodopsin linked to yellow fluorescent protein. Using time-correlated single-photon counting, we measured fluorescence lifetimes for all CaRubies and demonstrate a 10-fold increase in the average lifetime upon Ca(2+) chelation. Since only the fluorescence quantum yield but not the absorbance of the CaRubies is Ca(2+)-dependent, calibrated two-photon fluorescence excitation measurements of absolute Ca(2+) concentrations are feasible.


Subject(s)
Calcium/analysis , Egtazic Acid/analogs & derivatives , Fluorescent Dyes/chemistry , Indicators and Reagents/chemistry , Photons , Animals , Astrocytes/chemistry , Astrocytes/drug effects , Egtazic Acid/chemical synthesis , Egtazic Acid/chemistry , Egtazic Acid/pharmacokinetics , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacokinetics , Indicators and Reagents/chemical synthesis , Indicators and Reagents/pharmacokinetics , Mice , Mice, Inbred Strains , Molecular Structure
10.
ACS Nano ; 4(9): 5487-97, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20822159

ABSTRACT

The nanoscale size and unique optical properties of semiconductor quantum dots (QDs) have made them attractive as central photoluminescent scaffolds for a variety of biosensing platforms. In this report we functionalize QDs with dye-labeled peptides using two different linkage chemistries to yield Förster resonance energy transfer (FRET)-based sensors capable of monitoring either enzymatic activity or ionic presence. The first sensor targets the proteolytic activity of caspase 3, a key downstream effector of apoptosis. This QD conjugate utilized carbodiimide chemistry to covalently link dye-labeled peptide substrates to the terminal carboxyl groups on the QD's surface hydrophilic ligands in a quantitative manner. Caspase 3 cleaved the peptide substrate and disrupted QD donor-dye acceptor FRET providing signal transduction of enzymatic activity and allowing derivation of relevant Michaelis-Menten kinetic descriptors. The second sensor was designed to monitor Ca2+ ions that are ubiquitous in many biological processes. For this sensor, Cu+-catalyzed [3 + 2] azide-alkyne cycloaddition was exploited to attach a recently developed azide-functionalized CalciumRuby-Cl indicator dye to a cognate alkyne group present on the terminus of a modified peptide. The labeled peptide also expressed a polyhistidine sequence, which facilitated its subsequent metal-affinity coordination to the QD surface establishing the final FRET sensing construct. Adding exogenous Ca2+ to the sensor solution increased the dyes fluorescence, altering the donor-acceptor emission ratio and manifested a dissociation constant similar to that of the native dye. These results highlight the potential for combining peptides with QDs using different chemistries to create sensors for monitoring chemical compounds and biological processes.


Subject(s)
Biosensing Techniques/methods , Calcium/analysis , Caspase 3/metabolism , Peptides/chemistry , Quantum Dots , Amino Acid Sequence , Engineering , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Humans , Molecular Sequence Data
11.
Chemphyschem ; 11(3): 730-5, 2010 Feb 22.
Article in English | MEDLINE | ID: mdl-20135668

ABSTRACT

Ion sensors based on colloidal nanoparticles (NPs), either as actively ion-sensing NPs or as nanoscale carrier systems for organic ion-sensing fluorescent chelators typically require a charged surface in order to be colloidally stable. We demonstrate that this surface charge significantly impacts the ion binding and affects the read-out. Sensor read-out should be thus not determined by the bulk ion concentration, but by the local ion concentration in the nano-environment of the NP surface. We present a conclusive model corroborated by experimental data that reproduces the strong distance-dependence of the effect. The experimental data are based on the capability of tuning the distance of a pH-sensitive fluorophore to the surface of NPs in the nanometer (nm) range. This in turn allows for modification of the effective acid dissociation constant value (its logarithmic form, pK(a)) of analyte-sensitive fluorophores by tuning their distance to the underlying colloidal NPs.


Subject(s)
Colloids/chemistry , Nanoparticles/chemistry , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Ions , Surface Properties
12.
J Neurosci ; 29(31): 9668-82, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19657020

ABSTRACT

T-type voltage-gated calcium channels are expressed in the dendrites of many neurons, although their functional interactions with postsynaptic receptors and contributions to synaptic signaling are not well understood. We combine electrophysiological and ultrafast two-photon calcium imaging to demonstrate that mGluR1 activation potentiates cerebellar Purkinje cell Ca(v)3.1 T-type currents via a G-protein- and tyrosine-phosphatase-dependent pathway. Immunohistochemical and electron microscopic investigations on wild-type and Ca(v)3.1 gene knock-out animals show that Ca(v)3.1 T-type channels are preferentially expressed in Purkinje cell dendritic spines and colocalize with mGluR1s. We further demonstrate that parallel fiber stimulation induces fast subthreshold calcium signaling in dendritic spines and that the synaptic Ca(v)3.1-mediated calcium transients are potentiated by mGluR1 selectively during bursts of excitatory parallel fiber inputs. Our data identify a new fast calcium signaling pathway in Purkinje cell dendritic spines triggered by short burst of parallel fiber inputs and mediated by T-type calcium channels and mGluR1s.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium Signaling/physiology , Dendritic Spines/physiology , Purkinje Cells/physiology , Receptors, Metabotropic Glutamate/metabolism , Aging , Animals , Calcium Channels, T-Type/genetics , Cell Line , Dendritic Spines/ultrastructure , Humans , In Vitro Techniques , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Purkinje Cells/ultrastructure , Rats , Rats, Wistar , Synapses/physiology
13.
Langmuir ; 25(5): 3232-9, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19437725

ABSTRACT

Colloidal nanocrystal (NC) donors wrapped with a polymer coating including multiple organic acceptor molecules are promising scaffolds for fluorescence resonance energy transfer (FRET)-based nanobiosensors. Over other self-assembling donor-acceptor configurations, our preloaded polymers have the virtue of producing compact assemblies with a fixed donor/acceptor distance. This property, together with the possibility of stoichiometric polymer loading, allowed us to directly address how the FRET efficiency depended on the donor/acceptor. At the population level, nanoprobes based on commercial as well as custom CdSe/ZnS donors displayed the expected dose-dependent rise in transfer efficiency, saturating from about five ATTO dyes/NC. However, for a given acceptor concentration, both the intensity and lifetime of single-pair FRET data revealed a large dispersion of transfer efficiencies, highlighting an important heterogeneity among nominally identical FRET-based nanoprobes. Rigorous quality check during synthesis and shell assembly as well as postsynthesis sorting and purification are required to make hybrid semiconductor-organic nanoprobes a robust and viable alternative to organic or genetically encoded nanobiosensors.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Nanoparticles/chemistry , Nanotechnology/methods , Cyclohexanes/chemistry , Diffusion , Emulsions , Ethanol/chemistry , Hydrolysis , Kinetics , Magnetic Resonance Spectroscopy , Micelles , Microscopy, Electron, Transmission/methods , Models, Statistical , Oils , Temperature
14.
Cell Calcium ; 45(3): 275-83, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19167753

ABSTRACT

The limited choice and poor performance of red-emitting calcium (Ca(2+)) indicators have hampered microfluorometric measurements of the intracellular free Ca(2+) concentration in cells expressing yellow- or green-fluorescent protein constructs. A long-wavelength Ca(2+) indicator would also permit a better discrimination against cellular autofluorescence than the commonly used fluorescein-based probes. Here, we report an improved synthesis and characterization of Calcium Ruby, a red-emitting probe consisting of an extended rhodamine chromophore (578/602 nm peak excitation/emission) conjugated to BAPTA and having an additional NH(2) linker arm. The low-affinity variant (K(D,Ca) approximately 30 microM) with a chloride in meta position that was specifically designed for the detection of large and rapid Ca(2+) transients. While Calcium Ruby is a mitochondrial Ca(2+)probe, its conjugation, via the NH(2) tail, to a 10,000 MW dextran abolishes the sub-cellular compartmentalization and generates a cytosolic Ca(2+) probe with an affinity matched to microdomain Ca(2+) signals. As an example, we show depolarization-evoked Ca(2+) signals triggering the exocytosis of individual chromaffin granules. Calcium Ruby should be of use in a wide range of applications involving dual- or triple labeling schemes or targeted sub-cellular Ca(2+) measurements.


Subject(s)
Calcium/metabolism , Chromaffin Cells/metabolism , Cytoplasm/metabolism , Dextrans/metabolism , Green Fluorescent Proteins/metabolism , Mitochondria/metabolism , Rhodamines/metabolism , Adrenal Medulla/cytology , Animals , Calcium Signaling , Cattle , Dextrans/chemistry , Imaging, Three-Dimensional , Membrane Microdomains/metabolism , Rhodamines/chemistry , Spectrometry, Fluorescence , Staining and Labeling
15.
J Neurosci Methods ; 173(2): 259-70, 2008 Aug 30.
Article in English | MEDLINE | ID: mdl-18634822

ABSTRACT

Two-photon microscopy offers the promise of monitoring brain activity at multiple locations within intact tissue. However, serial sampling of voxels has been difficult to reconcile with millisecond timescales characteristic of neuronal activity. This is due to the conflicting constraints of scanning speed and signal amplitude. The recent use of acousto-optic deflector scanning to implement random-access multiphoton microscopy (RAMP) potentially allows to preserve long illumination dwell times while sampling multiple points-of-interest at high rates. However, the real-life abilities of RAMP microscopy regarding sensitivity and phototoxicity issues, which have so far impeded prolonged optical recordings at high frame rates, have not been assessed. Here, we describe the design, implementation and characterisation of an optimised RAMP microscope. We demonstrate the application of the microscope by monitoring calcium transients in Purkinje cells and cortical pyramidal cell dendrites and spines. We quantify the illumination constraints imposed by phototoxicity and show that stable continuous high-rate recordings can be obtained. During these recordings the fluorescence signal is large enough to detect spikes with a temporal resolution limited only by the calcium dye dynamics, improving upon previous techniques by at least an order of magnitude.


Subject(s)
Action Potentials/physiology , Brain/physiology , Microscopy, Fluorescence, Multiphoton/methods , Neurons/physiology , Neurophysiology/methods , Optics and Photonics/instrumentation , Animals , Brain/cytology , Calcium Signaling/physiology , Cerebellar Cortex/cytology , Cerebellar Cortex/physiology , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Fluorescent Dyes/standards , Image Cytometry/instrumentation , Image Cytometry/methods , Microscopy, Fluorescence, Multiphoton/instrumentation , Neurons/cytology , Neurophysiology/instrumentation , Organ Culture Techniques , Purkinje Cells/cytology , Purkinje Cells/physiology , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Rats , Rats, Wistar , Staining and Labeling/methods , Synaptic Transmission/physiology
16.
Nano Lett ; 7(9): 2613-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17691850

ABSTRACT

A fluorescence resonance energy transfer pair consisting of a colloidal quantum dot donor and multiple organic fluorophores as acceptors is reported and the photophysics of the system is characterized. Most nanoparticle-based biosensors reported so far use the detection of specific changes of the donor/acceptor distance under the influence of analyte binding. Our nanoparticle design on the other hand leads to sensors that detect spectral changes of the acceptor (under the influence of analyte binding) at fixed donor/acceptor distance by the introduction of the acceptor into the polymer coating. This approach allows for short acceptor-donor separation and thus for high-energy transfer efficiencies. Advantageously, the binding properties of the hydrophilic polymer coating further allows for addition of poly(ethylene glycol) shells for improved colloidal stability.


Subject(s)
Crystallization/methods , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Nanostructures/chemistry , Nanostructures/ultrastructure , Polyethylene Glycols/chemistry , Quantum Dots , Macromolecular Substances/chemistry , Materials Testing , Molecular Conformation , Nanotechnology/methods , Particle Size , Surface Properties
17.
Org Lett ; 9(14): 2629-32, 2007 Jul 05.
Article in English | MEDLINE | ID: mdl-17552530

ABSTRACT

Calcium Ruby m-Cl (X = H, Y = Cl) is a visible-light excited red-emitting calcium concentration ([Ca2+]) indicator dye (579/598 nm peak excitation/emission) with a side arm for conjugation via EDC or click chemistry. Its large molar extinction and high quantum yield rank it among the brightest long-wavelength Ca2+ indicators. Calcium Ruby is a promising alternative to existing dyes for imaging [Ca2+] in multicolor fluorescence applications or in the presence of yellow-green cellular autofluorescence.


Subject(s)
Calcium/chemistry , Indicators and Reagents/chemistry , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Egtazic Acid/analogs & derivatives , Egtazic Acid/chemical synthesis , Egtazic Acid/chemistry , Electrochemistry , Fluorescence , Indicators and Reagents/chemical synthesis , Spectrophotometry, Infrared , Trifluoroacetic Acid/chemistry
18.
J Biomed Biotechnol ; 2007(7): 68963, 2007.
Article in English | MEDLINE | ID: mdl-18273411

ABSTRACT

Semiconductor nanocrystals (NCs) are increasingly being used as photoluminescen markers in biological imaging. Their brightness, large Stokes shift, and high photostability compared to organic fluorophores permit the exploration of biological phenomena at the single-molecule scale with superior temporal resolution and spatial precision. NCs have predominantly been used as extracellular markers for tagging and tracking membrane proteins. Successful internalization and intracellular labelling with NCs have been demonstrated for both fixed immunolabelled and live cells. However, the precise localization and subcellular compartment labelled are less clear. Generally, live cell studies are limited by the requirement of fairly invasive protocols for loading NCs and the relatively large size of NCs compared to the cellular machinery, along with the subsequent sequestration of NCs in endosomal/lysosomal compartments. For long-period observation the potential cytotoxicity of cytoplasmically loaded NCs must be evaluated. This review focuses on the challenges of intracellular uses of NCs.

19.
Neurosci Res ; 56(3): 332-43, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16973229

ABSTRACT

Direct G protein inhibition of N-type calcium channels is recognized by characteristic biophysical modifications. In this study, we quantify and simulate the importance of G protein dissociation on the phenotype of G protein-regulated whole-cell currents. Based on the observation that the voltage-dependence of the time constant of recovery from G protein inhibition is correlated with the voltage-dependence of channel opening, we depict all G protein effects by a simple kinetic scheme. All landmark modifications in calcium currents, except inhibition, can be successfully described using three simple biophysical parameters (extent of block, extent of recovery, and time constant of recovery). Modifications of these parameters by auxiliary beta subunits are at the origin of differences in N-type channel regulation by G proteins. The simulation data illustrate that channel reluctance can occur as the result of an experimental bias linked to the variable extent of G protein dissociation when peak currents are measured at various membrane potentials. To produce alterations in channel kinetics, the two most important parameters are the extents of initial block and recovery. These data emphasize the contribution of the degree and kinetics of G protein dissociation in the modification of N-type currents.


Subject(s)
Calcium Channels, N-Type/physiology , GTP-Binding Proteins/chemistry , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Proteins/antagonists & inhibitors , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Microinjections/methods , Models, Neurological , Oocytes , Patch-Clamp Techniques/methods , Rabbits , Rats , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Xenopus
20.
Crit Rev Neurobiol ; 18(1-2): 169-78, 2006.
Article in English | MEDLINE | ID: mdl-17725519

ABSTRACT

Once the tools for controlling calcium gradients became available to electrophysiologists, they began the quest for understanding the role of Ca2+ in the control of neuronal activity. In the early 1970s Paul Feltz and I spent a rich period in K. Krnjevic's laboratory in Montreal, and I was already involved in a research, which showed that an increase in intracellular Ca2+ concentration can lead to hyperpolarization of motoneurones. At about the same time, a potassium conductance activated by intracellular calcium injection was identified in mammals and snails. Since then, most of my work has dealt with the study of Ca2+ entry in neurons. Here I review the progress that led fi rst to the biophysical characterization and, later, to the molecular identification of T-type calcium channels. With the advent of new optical methods, in particular two-photon microscopy, we may be on the brink of a step forward in our understanding of how T channels play a role in the integrative processes that take place in a large cortical neuron such as the Purkinje cell.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Nervous System/metabolism , Neurons/metabolism , Animals , Calcium Channels, T-Type/genetics , Humans , Microscopy/methods , Microscopy/trends , Neurosciences/methods , Neurosciences/trends , Purkinje Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...