Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 183(6): 1682-1698.e24, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33232692

ABSTRACT

In order to analyze how a signal transduction network converts cellular inputs into cellular outputs, ideally one would measure the dynamics of many signals within the network simultaneously. We found that, by fusing a fluorescent reporter to a pair of self-assembling peptides, it could be stably clustered within cells at random points, distant enough to be resolved by a microscope but close enough to spatially sample the relevant biology. Because such clusters, which we call signaling reporter islands (SiRIs), can be modularly designed, they permit a set of fluorescent reporters to be efficiently adapted for simultaneous measurement of multiple nodes of a signal transduction network within single cells. We created SiRIs for indicators of second messengers and kinases and used them, in hippocampal neurons in culture and intact brain slices, to discover relationships between the speed of calcium signaling, and the amplitude of PKA signaling, upon receiving a cAMP-driving stimulus.


Subject(s)
Fluorescent Dyes/metabolism , Genes, Reporter , Optical Imaging , Signal Transduction , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Green Fluorescent Proteins/metabolism , HeLa Cells , Hippocampus/metabolism , Humans , Mice , Neurons/metabolism , Peptides/metabolism , Proteins/metabolism , Pyramidal Cells/metabolism
2.
Nature ; 574(7778): 413-417, 2019 10.
Article in English | MEDLINE | ID: mdl-31597963

ABSTRACT

A longstanding goal in neuroscience has been to image membrane voltage across a population of individual neurons in an awake, behaving mammal. Here we describe a genetically encoded fluorescent voltage indicator, SomArchon, which exhibits millisecond response times and is compatible with optogenetic control, and which increases the sensitivity, signal-to-noise ratio, and number of neurons observable several-fold over previously published fully genetically encoded reagents1-8. Under conventional one-photon microscopy, SomArchon enables the routine population analysis of around 13 neurons at once, in multiple brain regions (cortex, hippocampus, and striatum) of head-fixed, awake, behaving mice. Using SomArchon, we detected both positive and negative responses of striatal neurons during movement, as previously reported by electrophysiology but not easily detected using modern calcium imaging techniques9-11, highlighting the power of voltage imaging to reveal bidirectional modulation. We also examined how spikes relate to the subthreshold theta oscillations of individual hippocampal neurons, with SomArchon showing that the spikes of individual neurons are more phase-locked to their own subthreshold theta oscillations than to local field potential theta oscillations. Thus, SomArchon reports both spikes and subthreshold voltage dynamics in awake, behaving mice.


Subject(s)
Environmental Biomarkers , Hippocampus/cytology , Neurons/physiology , Optical Imaging/methods , Wakefulness/physiology , Action Potentials/physiology , Animals , Environmental Biomarkers/genetics , Hippocampus/diagnostic imaging , Mice , Optogenetics
3.
J Biol Chem ; 294(11): 3806-3821, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30610117

ABSTRACT

Channelrhodopsins (ChRs) are light-gated ion channels in widespread use in neuroscience for mediating the genetically targetable optical control of neurons (optogenetics). ChRs pass multiple kinds of ions, and although nonspecific ChR-mediated conductance is not an issue in many neuroscience studies, conductance of calcium and protons, which can mediate diverse cellular signals, may be undesirable in some instances. Here, we turned our attention to the creation of ChRs that have high cation photocurrent but pass fewer calcium ions and protons. We developed an automated, time-resolved screening method capable of rapidly phenotyping channelrhodopsin-2 (ChR2) variants. We found substitution mutations throughout ChR2 that could boost current while altering ion selectivity and observed that the mutations that reduced calcium or proton conductance have additive effects. By combining four mutations, we obtained a ChR, ChromeQ, with improved photocurrent that possesses order-of-magnitude reductions in calcium and proton conductance and high fidelity in driving repetitive action potentials in neurons. The approach presented here offers a viable pathway toward customization of complex physiological properties of optogenetic tools. We propose that our screening method not only enables elucidation of new ChR variants that affect microbial opsin performance but may also reveal new principles of optogenetic protein engineering.


Subject(s)
Calcium/metabolism , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Electric Conductivity , Genetic Variation , Protons , Animals , Cloning, Molecular , Fluorescence , Genetic Variation/genetics , HEK293 Cells , Humans , Oxidation-Reduction , Phenotype , Photochemical Processes
5.
Nat Chem Biol ; 14(4): 352-360, 2018 04.
Article in English | MEDLINE | ID: mdl-29483642

ABSTRACT

We developed a new way to engineer complex proteins toward multidimensional specifications using a simple, yet scalable, directed evolution strategy. By robotically picking mammalian cells that were identified, under a microscope, as expressing proteins that simultaneously exhibit several specific properties, we can screen hundreds of thousands of proteins in a library in just a few hours, evaluating each along multiple performance axes. To demonstrate the power of this approach, we created a genetically encoded fluorescent voltage indicator, simultaneously optimizing its brightness and membrane localization using our microscopy-guided cell-picking strategy. We produced the high-performance opsin-based fluorescent voltage reporter Archon1 and demonstrated its utility by imaging spiking and millivolt-scale subthreshold and synaptic activity in acute mouse brain slices and in larval zebrafish in vivo. We also measured postsynaptic responses downstream of optogenetically controlled neurons in C. elegans.


Subject(s)
Directed Molecular Evolution/methods , Luminescent Proteins/chemistry , Protein Engineering/methods , Robotics , Zebrafish/embryology , Animals , Brain/diagnostic imaging , Caenorhabditis elegans , Cell Separation , Female , Flow Cytometry , Fluorescence , Gene Library , Genes, Reporter , HEK293 Cells , Hippocampus/cytology , Humans , Male , Mice , Microscopy, Fluorescence , Neurons/cytology , Optogenetics
6.
Nano Lett ; 17(10): 6131-6139, 2017 10 11.
Article in English | MEDLINE | ID: mdl-28933153

ABSTRACT

To decipher the molecular mechanisms of biological function, it is critical to map the molecular composition of individual cells or even more importantly tissue samples in the context of their biological environment in situ. Immunofluorescence (IF) provides specific labeling for molecular profiling. However, conventional IF methods have finite multiplexing capabilities due to spectral overlap of the fluorophores. Various sequential imaging methods have been developed to circumvent this spectral limit but are not widely adopted due to the common limitation of requiring multirounds of slow (typically over 2 h at room temperature to overnight at 4 °C in practice) immunostaining. We present here a practical and robust method, which we call DNA Exchange Imaging (DEI), for rapid in situ spectrally unlimited multiplexing. This technique overcomes speed restrictions by allowing for single-round immunostaining with DNA-barcoded antibodies, followed by rapid (less than 10 min) buffer exchange of fluorophore-bearing DNA imager strands. The programmability of DEI allows us to apply it to diverse microscopy platforms (with Exchange Confocal, Exchange-SIM, Exchange-STED, and Exchange-PAINT demonstrated here) at multiple desired resolution scales (from ∼300 nm down to sub-20 nm). We optimized and validated the use of DEI in complex biological samples, including primary neuron cultures and tissue sections. These results collectively suggest DNA exchange as a versatile, practical platform for rapid, highly multiplexed in situ imaging, potentially enabling new applications ranging from basic science, to drug discovery, and to clinical pathology.


Subject(s)
DNA/chemistry , Hippocampus/cytology , Immunoconjugates/chemistry , Microscopy, Confocal/methods , Neurons/ultrastructure , Optical Imaging/methods , Protein Interaction Mapping/methods , Animals , Brain/ultrastructure , Cells, Cultured , Fluorescent Dyes/chemistry , Hippocampus/ultrastructure , Mice , Microscopy, Fluorescence/methods , Neurons/cytology , Retina/cytology , Retina/ultrastructure , Staining and Labeling/methods , Synapsins/analysis , Synaptophysin/analysis
7.
bioRxiv ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39282466

ABSTRACT

Established methods for imaging the living mammalian brain have, to date, taken optical properties of the tissue as fixed; we here demonstrate that it is possible to modify the optical properties of the brain itself to significantly enhance at-depth imaging while preserving native physiology. Using a small amount of any of several biocompatible materials to raise the refractive index of solutions superfusing the brain prior to imaging, we could increase several-fold the signals from the deepest cells normally visible and, under both one-photon and two-photon imaging, visualize cells previously too dim to see. The enhancement was observed for both anatomical and functional fluorescent reporters across a broad range of emission wavelengths. Importantly, visual tuning properties of cortical neurons in awake mice, and electrophysiological properties of neurons assessed ex vivo, were not altered by this procedure.

8.
PLoS One ; 18(9): e0291506, 2023.
Article in English | MEDLINE | ID: mdl-37729182

ABSTRACT

Expansion microscopy (ExM), by physically enlarging specimens in an isotropic fashion, enables nanoimaging on standard light microscopes. Key to existing ExM protocols is the equipping of different kinds of molecules, with different kinds of anchoring moieties, so they can all be pulled apart from each other by polymer swelling. Here we present a multifunctional anchor, an acrylate epoxide, that enables proteins and RNAs to be equipped with anchors in a single experimental step. This reagent simplifies ExM protocols and reduces cost (by 2-10-fold for a typical multiplexed ExM experiment) compared to previous strategies for equipping RNAs with anchors. We show that this united ExM (uniExM) protocol can be used to preserve and visualize RNA transcripts, proteins in biologically relevant ultrastructures, and sets of RNA transcripts in patient-derived xenograft (PDX) cancer tissues and may support the visualization of other kinds of biomolecular species as well. uniExM may find many uses in the simple, multimodal nanoscale analysis of cells and tissues.


Subject(s)
Epoxy Compounds , Microscopy , Humans , Animals , Disease Models, Animal , Polymers , RNA
9.
Nat Biotechnol ; 41(5): 640-651, 2023 05.
Article in English | MEDLINE | ID: mdl-36593405

ABSTRACT

Observing cellular physiological histories is key to understanding normal and disease-related processes. Here we describe expression recording islands-a fully genetically encoded approach that enables both continual digital recording of biological information within cells and subsequent high-throughput readout in fixed cells. The information is stored in growing intracellular protein chains made of self-assembling subunits, human-designed filament-forming proteins bearing different epitope tags that each correspond to a different cellular state or function (for example, gene expression downstream of neural activity or pharmacological exposure), allowing the physiological history to be read out along the ordered subunits of protein chains with conventional optical microscopy. We use expression recording islands to record gene expression timecourse downstream of specific pharmacological and physiological stimuli in cultured neurons and in living mouse brain, with a time resolution of a fraction of a day, over periods of days to weeks.


Subject(s)
Microscopy , Neurons , Mice , Animals , Humans , Neurons/physiology
10.
Nat Biomed Eng ; 6(9): 1057-1073, 2022 09.
Article in English | MEDLINE | ID: mdl-36038771

ABSTRACT

Many crowded biomolecular structures in cells and tissues are inaccessible to labelling antibodies. To understand how proteins within these structures are arranged with nanoscale precision therefore requires that these structures be decrowded before labelling. Here we show that an iterative variant of expansion microscopy (the permeation of cells and tissues by a swellable hydrogel followed by isotropic hydrogel expansion, to allow for enhanced imaging resolution with ordinary microscopes) enables the imaging of nanostructures in expanded yet otherwise intact tissues at a resolution of about 20 nm. The method, which we named 'expansion revealing' and validated with DNA-probe-based super-resolution microscopy, involves gel-anchoring reagents and the embedding, expansion and re-embedding of the sample in homogeneous swellable hydrogels. Expansion revealing enabled us to use confocal microscopy to image the alignment of pre-synaptic calcium channels with post-synaptic scaffolding proteins in intact brain circuits, and to uncover periodic amyloid nanoclusters containing ion-channel proteins in brain tissue from a mouse model of Alzheimer's disease. Expansion revealing will enable the further discovery of previously unseen nanostructures within cells and tissues.


Subject(s)
Microscopy , Nanostructures , Animals , Brain/metabolism , Calcium Channels/metabolism , DNA/metabolism , Hydrogels , Mice , Microscopy/methods , Proteins/metabolism
11.
Protein Sci ; 31(3): 728-751, 2022 03.
Article in English | MEDLINE | ID: mdl-34913537

ABSTRACT

In vivo imaging of model organisms is heavily reliant on fluorescent proteins with high intracellular brightness. Here we describe a practical method for rapid optimization of fluorescent proteins via directed molecular evolution in cultured mammalian cells. Using this method, we were able to perform screening of large gene libraries containing up to 2 × 107 independent random genes of fluorescent proteins expressed in HEK cells, completing one iteration of directed evolution in a course of 8 days. We employed this approach to develop a set of green and near-infrared fluorescent proteins with enhanced intracellular brightness. The developed near-infrared fluorescent proteins demonstrated high performance for fluorescent labeling of neurons in culture and in vivo in model organisms such as Caenorhabditis elegans, Drosophila, zebrafish, and mice. Spectral properties of the optimized near-infrared fluorescent proteins enabled crosstalk-free multicolor imaging in combination with common green and red fluorescent proteins, as well as dual-color near-infrared fluorescence imaging. The described method has a great potential to be adopted by protein engineers due to its simplicity and practicality. We also believe that the new enhanced fluorescent proteins will find wide application for in vivo multicolor imaging of small model organisms.


Subject(s)
Directed Molecular Evolution , Zebrafish , Animals , Cell Line , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mammals/genetics , Mice , Neurons/metabolism , Optical Imaging , Zebrafish/genetics , Zebrafish/metabolism
12.
Neuron ; 107(1): 38-51.e8, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32353253

ABSTRACT

Optogenetics is among the most widely employed techniques to manipulate neuronal activity. However, a major drawback is the need for invasive implantation of optical fibers. To develop a minimally invasive optogenetic method that overcomes this challenge, we engineered a new step-function opsin with ultra-high light sensitivity (SOUL). We show that SOUL can activate neurons located in deep mouse brain regions via transcranial optical stimulation and elicit behavioral changes in SOUL knock-in mice. Moreover, SOUL can be used to modulate neuronal spiking and induce oscillations reversibly in macaque cortex via optical stimulation from outside the dura. By enabling external light delivery, our new opsin offers a minimally invasive tool for manipulating neuronal activity in rodent and primate models with fewer limitations on the depth and size of target brain regions and may further facilitate the development of minimally invasive optogenetic tools for the treatment of neurological disorders.


Subject(s)
Opsins , Optogenetics/methods , Animals , Brain/physiology , Macaca , Mice , Models, Animal , Neurons/physiology
13.
Nat Commun ; 10(1): 4377, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31558769

ABSTRACT

Synapses contain hundreds of distinct proteins whose heterogeneous expression levels are determinants of synaptic plasticity and signal transmission relevant to a range of diseases. Here, we use diffusible nucleic acid imaging probes to profile neuronal synapses using multiplexed confocal and super-resolution microscopy. Confocal imaging is performed using high-affinity locked nucleic acid imaging probes that stably yet reversibly bind to oligonucleotides conjugated to antibodies and peptides. Super-resolution PAINT imaging of the same targets is performed using low-affinity DNA imaging probes to resolve nanometer-scale synaptic protein organization across nine distinct protein targets. Our approach enables the quantitative analysis of thousands of synapses in neuronal culture to identify putative synaptic sub-types and co-localization patterns from one dozen proteins. Application to characterize synaptic reorganization following neuronal activity blockade reveals coordinated upregulation of the post-synaptic proteins PSD-95, SHANK3 and Homer-1b/c, as well as increased correlation between synaptic markers in the active and synaptic vesicle zones.


Subject(s)
Microscopy, Fluorescence/methods , Neurons/metabolism , Nucleic Acid Probes/metabolism , Oligonucleotides/metabolism , Animals , Animals, Newborn , Cells, Cultured , Diffusion , Disks Large Homolog 4 Protein/metabolism , Mice , Microfilament Proteins , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Neurons/cytology , Nucleic Acid Probes/chemistry , Oligonucleotides/chemistry , Rats, Sprague-Dawley , Synapses/metabolism , Synaptic Vesicles/metabolism
14.
Light Sci Appl ; 6: e16255, 2017 May 05.
Article in English | MEDLINE | ID: mdl-29152380

ABSTRACT

Three-photon wide-field depth-resolved excitation is used to overcome some of the limitations in conventional point-scanning two- and three-photon microscopy. Excitation of chromophores as diverse as channelrhodopsins and quantum dots is shown, and a penetration depth of more than 700 µm into fixed scattering brain tissue is achieved, approximately twice as deep as that achieved using two-photon wide-field excitation. Compatibility with live animal experiments is confirmed by imaging the cerebral vasculature of an anesthetized mouse; a complete focal stack was obtained without any evidence of photodamage. As an additional validation of the utility of wide-field three-photon excitation, functional excitation is demonstrated by performing three-photon optogenetic stimulation of cultured mouse hippocampal neurons expressing a channelrhodopsin; action potentials could reliably be excited without causing photodamage.

16.
J Neurophysiol ; 95(6): 3955-60, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16554503

ABSTRACT

Daily biological rhythms in both prokaryotes and eukaryotes are controlled by circadian clocks. In Drosophila, there is a good basic understanding of both the molecular and anatomical components of the clock. In this study we directly measure, for the first time, electrophysiological properties and anatomy of individual filled large lateral PDF-positive clock neurons, a cell group believed to be involved in synchronization of the clock in constant conditions. We find that the large PDF-positive neurons are morphologically homogeneous and that their resting membrane potential is modulated both by the clock and by light inputs. Expression of a leak channel, dORK-deltaC, which has been shown to disrupt circadian locomotor rhythms, hyperpolarizes these neurons, and blocks firing. These data imply that the firing properties of large PDF neurons are both regulated by and critical for clock function.


Subject(s)
Biological Clocks/physiology , Brain/cytology , Brain/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Neurons/cytology , Neurons/physiology , Neuropeptides/metabolism , Action Potentials/physiology , Animals , Female
17.
J Neurophysiol ; 91(5): 2353-65, 2004 May.
Article in English | MEDLINE | ID: mdl-14695352

ABSTRACT

We have used dye fills and electrophysiological recordings to identify and characterize a cluster of motor neurons in the third instar larval ventral ganglion. This cluster of neurons is similar in position to the well-studied embryonic RP neurons. Dye fills of larval dorsomedial neurons demonstrate that individual neurons within the cluster can be reproducibly identified by observing their muscle targets and bouton morphology. The terminal targets of these five neurons are body wall muscles 6/7, 1, 14, and 30 and the intersegmental nerve (ISN) terminal muscles (1, 2, 3, 4, 9, 10, 19, 20). All cells except the ISN neuron, which has a type Is ending, display type Ib boutons. Two of these neurons appear to be identical to the embryonic RP3 and aCC cells, which define the most proximal and distal innervations within a hemisegment. The targets of the other neurons in the larval dorsomedial cluster do not correspond to embryonic targets of the neurons in the RP cluster, suggesting rewiring of this circuit during early larval stages. Electrophysiological studies of the five neurons in current clamp revealed that type Is neurons have a longer delay in the appearance of the first spike compared with type Ib neurons. Genetic, biophysical, and pharmacological studies in current and voltage clamp show this delay is controlled by the kinetics and voltage sensitivity of inactivation of a current whose properties suggest that it may be the Shal I(A) current. The combination of genetic identification and whole cell recording allows us to directly explore the cellular substrates of neural and locomotor behavior in an intact system.


Subject(s)
Central Nervous System/cytology , Central Nervous System/physiology , Drosophila/physiology , Motor Neurons/physiology , Motor Neurons/ultrastructure , 4-Aminopyridine/pharmacology , Animals , Axons/physiology , Axons/ultrastructure , Central Nervous System/ultrastructure , Dendrites/physiology , Dendrites/ultrastructure , Electrophysiology , Female , Fluorescent Dyes , Green Fluorescent Proteins , Larva , Locomotion/physiology , Luminescent Proteins , Male , Membrane Potentials/physiology , Muscles/innervation , Muscles/physiology , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/physiology
18.
J Neurobiol ; 52(1): 24-42, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12115891

ABSTRACT

The ability of calcium/calmodulin-dependent protein kinase II (CaMKII) to become calcium independent after autophosphorylation makes this enzyme a temporal marker of neuronal activity. Here we show that the calcium-independent form of CaMKII has unique effects on larval viability, locomotion, and neuronal excitability in Drosophila. Expression of constitutively active T287D, but not calcium-dependent T287A, mutant CaMKII in Drosophila neurons resulted in decreased viability, behavioral defects, and failure of action potential propagation. The actions of T287D may be mediated, at least in part, by increased potassium conductances. Expression of T287D CaMKII also stimulated an increase in the number of boutons at the larval neuromuscular junction, but did not affect the mechanics of release. This study defines a role for autophosphorylation of CaMKII in the regulation of multiple neuronal functions including the intrinsic properties of neurons.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drosophila/enzymology , Motor Neurons/enzymology , Saccharomyces cerevisiae Proteins , Anesthetics, Local/pharmacology , Animals , Animals, Genetically Modified , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calmodulin/metabolism , DNA-Binding Proteins , Drosophila/growth & development , Evoked Potentials/drug effects , Evoked Potentials/physiology , Female , Fungal Proteins/genetics , Gene Expression Regulation, Enzymologic , Larva/enzymology , Male , Motor Activity/physiology , Muscles/enzymology , Neuromuscular Junction/enzymology , Phenotype , Phosphorylation , Potassium/metabolism , Presynaptic Terminals/enzymology , Sodium Channels/genetics , Tetrodotoxin/pharmacology , Transcription Factors/genetics , Transgenes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL