Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Cell ; 183(7): 1986-2002.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33333022

ABSTRACT

Serotonin plays a central role in cognition and is the target of most pharmaceuticals for psychiatric disorders. Existing drugs have limited efficacy; creation of improved versions will require better understanding of serotonergic circuitry, which has been hampered by our inability to monitor serotonin release and transport with high spatial and temporal resolution. We developed and applied a binding-pocket redesign strategy, guided by machine learning, to create a high-performance, soluble, fluorescent serotonin sensor (iSeroSnFR), enabling optical detection of millisecond-scale serotonin transients. We demonstrate that iSeroSnFR can be used to detect serotonin release in freely behaving mice during fear conditioning, social interaction, and sleep/wake transitions. We also developed a robust assay of serotonin transporter function and modulation by drugs. We expect that both machine-learning-guided binding-pocket redesign and iSeroSnFR will have broad utility for the development of other sensors and in vitro and in vivo serotonin detection, respectively.


Subject(s)
Directed Molecular Evolution , Machine Learning , Serotonin/metabolism , Algorithms , Amino Acid Sequence , Amygdala/physiology , Animals , Behavior, Animal , Binding Sites , Brain/metabolism , HEK293 Cells , Humans , Kinetics , Linear Models , Mice , Mice, Inbred C57BL , Photons , Protein Binding , Serotonin Plasma Membrane Transport Proteins/metabolism , Sleep/physiology , Wakefulness/physiology
2.
Cell ; 173(5): 1265-1279.e19, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775595

ABSTRACT

Chronic social isolation causes severe psychological effects in humans, but their neural bases remain poorly understood. 2 weeks (but not 24 hr) of social isolation stress (SIS) caused multiple behavioral changes in mice and induced brain-wide upregulation of the neuropeptide tachykinin 2 (Tac2)/neurokinin B (NkB). Systemic administration of an Nk3R antagonist prevented virtually all of the behavioral effects of chronic SIS. Conversely, enhancing NkB expression and release phenocopied SIS in group-housed mice, promoting aggression and converting stimulus-locked defensive behaviors to persistent responses. Multiplexed analysis of Tac2/NkB function in multiple brain areas revealed dissociable, region-specific requirements for both the peptide and its receptor in different SIS-induced behavioral changes. Thus, Tac2 coordinates a pleiotropic brain state caused by SIS via a distributed mode of action. These data reveal the profound effects of prolonged social isolation on brain chemistry and function and suggest potential new therapeutic applications for Nk3R antagonists.


Subject(s)
Brain/metabolism , Neurokinin B/metabolism , Protein Precursors/metabolism , Social Isolation , Stress, Psychological , Tachykinins/metabolism , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Brain/pathology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurokinin B/genetics , Neurons/cytology , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/antagonists & inhibitors , Protein Precursors/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Tachykinin/antagonists & inhibitors , Receptors, Tachykinin/metabolism , Tachykinins/antagonists & inhibitors , Tachykinins/genetics , Up-Regulation/drug effects
3.
Cell ; 167(6): 1469-1480.e12, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912057

ABSTRACT

The intestinal microbiota influence neurodevelopment, modulate behavior, and contribute to neurological disorders. However, a functional link between gut bacteria and neurodegenerative diseases remains unexplored. Synucleinopathies are characterized by aggregation of the protein α-synuclein (αSyn), often resulting in motor dysfunction as exemplified by Parkinson's disease (PD). Using mice that overexpress αSyn, we report herein that gut microbiota are required for motor deficits, microglia activation, and αSyn pathology. Antibiotic treatment ameliorates, while microbial re-colonization promotes, pathophysiology in adult animals, suggesting that postnatal signaling between the gut and the brain modulates disease. Indeed, oral administration of specific microbial metabolites to germ-free mice promotes neuroinflammation and motor symptoms. Remarkably, colonization of αSyn-overexpressing mice with microbiota from PD-affected patients enhances physical impairments compared to microbiota transplants from healthy human donors. These findings reveal that gut bacteria regulate movement disorders in mice and suggest that alterations in the human microbiome represent a risk factor for PD.


Subject(s)
Parkinson Disease/microbiology , Parkinson Disease/pathology , Animals , Brain/pathology , Dysbiosis/pathology , Fatty Acids/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice , Microglia/pathology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , alpha-Synuclein/metabolism
4.
Annu Rev Neurosci ; 45: 447-469, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35440143

ABSTRACT

Recombinant adeno-associated viruses (AAVs) are commonly used gene delivery vehicles for neuroscience research. They have two engineerable features: the capsid (outer protein shell) and cargo (encapsulated genome). These features can be modified to enhance cell type or tissue tropism and control transgene expression, respectively. Several engineered AAV capsids with unique tropisms have been identified, including variants with enhanced central nervous system transduction, cell type specificity, and retrograde transport in neurons. Pairing these AAVs with modern gene regulatory elements and state-of-the-art reporter, sensor, and effector cargo enables highly specific transgene expression for anatomical and functional analyses of brain cells and circuits. Here, we discuss recent advances that provide a comprehensive (capsid and cargo) AAV toolkit for genetic access to molecularly defined brain cell types.


Subject(s)
Dependovirus , Genetic Vectors , Brain , Capsid/metabolism , Dependovirus/genetics , Gene Transfer Techniques
5.
Cell ; 158(4): 945-958, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25088144

ABSTRACT

Understanding the structure-function relationships at cellular, circuit, and organ-wide scale requires 3D anatomical and phenotypical maps, currently unavailable for many organs across species. At the root of this knowledge gap is the absence of a method that enables whole-organ imaging. Herein, we present techniques for tissue clearing in which whole organs and bodies are rendered macromolecule-permeable and optically transparent, thereby exposing their cellular structure with intact connectivity. We describe PACT (passive clarity technique), a protocol for passive tissue clearing and immunostaining of intact organs; RIMS (refractive index matching solution), a mounting media for imaging thick tissue; and PARS (perfusion-assisted agent release in situ), a method for whole-body clearing and immunolabeling. We show that in rodents PACT, RIMS, and PARS are compatible with endogenous-fluorescence, immunohistochemistry, RNA single-molecule FISH, long-term storage, and microscopy with cellular and subcellular resolution. These methods are applicable for high-resolution, high-content mapping and phenotyping of normal and pathological elements within intact organs and bodies.


Subject(s)
Cells/classification , Imaging, Three-Dimensional/methods , Single-Cell Analysis , Whole Body Imaging , Animals , Brain/cytology , Cells/metabolism , Fluorescence , Mice , Microscopy, Confocal/methods , Microscopy, Electron, Scanning , Phenotype
6.
Nature ; 595(7867): 409-414, 2021 07.
Article in English | MEDLINE | ID: mdl-34194038

ABSTRACT

Social interactions among animals mediate essential behaviours, including mating, nurturing, and defence1,2. The gut microbiota contribute to social activity in mice3,4, but the gut-brain connections that regulate this complex behaviour and its underlying neural basis are unclear5,6. Here we show that the microbiome modulates neuronal activity in specific brain regions of male mice to regulate canonical stress responses and social behaviours. Social deviation in germ-free and antibiotic-treated mice is associated with elevated levels of the stress hormone corticosterone, which is primarily produced by activation of the hypothalamus-pituitary-adrenal (HPA) axis. Adrenalectomy, antagonism of glucocorticoid receptors, or pharmacological inhibition of corticosterone synthesis effectively corrects social deficits following microbiome depletion. Genetic ablation of glucocorticoid receptors in specific brain regions or chemogenetic inactivation of neurons in the paraventricular nucleus of the hypothalamus that produce corticotrophin-releasing hormone (CRH) reverse social impairments in antibiotic-treated mice. Conversely, specific activation of CRH-expressing neurons in the paraventricular nucleus induces social deficits in mice with a normal microbiome. Via microbiome profiling and in vivo selection, we identify a bacterial species, Enterococcus faecalis, that promotes social activity and reduces corticosterone levels in mice following social stress. These studies suggest that specific gut bacteria can restrain the activation of the HPA axis, and show that the microbiome can affect social behaviours through discrete neuronal circuits that mediate stress responses in the brain.


Subject(s)
Brain/cytology , Brain/physiology , Gastrointestinal Microbiome/physiology , Neurons/metabolism , Social Behavior , Stress, Psychological , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Enterococcus faecalis/metabolism , Germ-Free Life , Glucocorticoids/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Glucocorticoid/metabolism , Signal Transduction
7.
Annu Rev Neurosci ; 41: 323-348, 2018 07 08.
Article in English | MEDLINE | ID: mdl-29709207

ABSTRACT

Recombinant viruses allow for targeted transgene expression in specific cell populations throughout the nervous system. The adeno-associated virus (AAV) is among the most commonly used viruses for neuroscience research. Recombinant AAVs (rAAVs) are highly versatile and can package most cargo composed of desired genes within the capsid's ∼5-kb carrying capacity. Numerous regulatory elements and intersectional strategies have been validated in rAAVs to enable cell type-specific expression. rAAVs can be delivered to specific neuronal populations or globally throughout the animal. The AAV capsids have natural cell type or tissue tropism and trafficking that can be modified for increased specificity. Here, we describe recently engineered AAV capsids and associated cargo that have extended the utility of AAVs in targeting molecularly defined neurons throughout the nervous system, which will further facilitate neuronal circuit interrogation and discovery.


Subject(s)
Central Nervous System/physiology , Genetic Engineering , Peripheral Nervous System/physiology , Animals , Dependovirus/genetics , Humans
8.
Cell ; 147(3): 678-89, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22019004

ABSTRACT

Prevailing theory suggests that long-term memories are encoded via a two-phase process requiring early involvement of the hippocampus followed by the neocortex. Contextual fear memories in rodents rely on the hippocampus immediately following training but are unaffected by hippocampal lesions or pharmacological inhibition weeks later. With fast optogenetic methods, we examine the real-time contribution of hippocampal CA1 excitatory neurons to remote memory and find that contextual fear memory recall, even weeks after training, can be reversibly abolished by temporally precise optogenetic inhibition of CA1. When this inhibition is extended to match the typical time course of pharmacological inhibition, remote hippocampus dependence converts to hippocampus independence, suggesting that long-term memory retrieval normally depends on the hippocampus but can adaptively shift to alternate structures. Further revealing the plasticity of mechanisms required for memory recall, we confirm the remote-timescale importance of the anterior cingulate cortex (ACC) and implicate CA1 in ACC recruitment for remote recall.


Subject(s)
Hippocampus/physiology , Memory, Long-Term , Animals , Fear , Gyrus Cinguli/metabolism , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/physiology
9.
Nature ; 580(7803): 381-385, 2020 04.
Article in English | MEDLINE | ID: mdl-32296178

ABSTRACT

The spread of protein aggregates during disease progression is a common theme underlying many neurodegenerative diseases. The microtubule-associated protein tau has a central role in the pathogenesis of several forms of dementia known as tauopathies-including Alzheimer's disease, frontotemporal dementia and chronic traumatic encephalopathy1. Progression of these diseases is characterized by the sequential spread and deposition of protein aggregates in a predictable pattern that correlates with clinical severity2. This observation and complementary experimental studies3,4 have suggested that tau can spread in a prion-like manner, by passing to naive cells in which it templates misfolding and aggregation. However, although the propagation of tau has been extensively studied, the underlying cellular mechanisms remain poorly understood. Here we show that the low-density lipoprotein receptor-related protein 1 (LRP1) controls the endocytosis of tau and its subsequent spread. Knockdown of LRP1 significantly reduced tau uptake in H4 neuroglioma cells and in induced pluripotent stem cell-derived neurons. The interaction between tau and LRP1 is mediated by lysine residues in the microtubule-binding repeat region of tau. Furthermore, downregulation of LRP1 in an in vivo mouse model of tau spread was found to effectively reduce the propagation of tau between neurons. Our results identify LRP1 as a key regulator of tau spread in the brain, and therefore a potential target for the treatment of diseases that involve tau spread and aggregation.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/metabolism , tau Proteins/metabolism , Animals , Cell Line , Endocytosis , Female , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Male , Mice , Neurons/metabolism
10.
Nature ; 586(7829): 417-423, 2020 10.
Article in English | MEDLINE | ID: mdl-32999463

ABSTRACT

Microglia, the brain's resident macrophages, help to regulate brain function by removing dying neurons, pruning non-functional synapses, and producing ligands that support neuronal survival1. Here we show that microglia are also critical modulators of neuronal activity and associated behavioural responses in mice. Microglia respond to neuronal activation by suppressing neuronal activity, and ablation of microglia amplifies and synchronizes the activity of neurons, leading to seizures. Suppression of neuronal activation by microglia occurs in a highly region-specific fashion and depends on the ability of microglia to sense and catabolize extracellular ATP, which is released upon neuronal activation by neurons and astrocytes. ATP triggers the recruitment of microglial protrusions and is converted by the microglial ATP/ADP hydrolysing ectoenzyme CD39 into AMP; AMP is then converted into adenosine by CD73, which is expressed on microglia as well as other brain cells. Microglial sensing of ATP, the ensuing microglia-dependent production of adenosine, and the adenosine-mediated suppression of neuronal responses via the adenosine receptor A1R are essential for the regulation of neuronal activity and animal behaviour. Our findings suggest that this microglia-driven negative feedback mechanism operates similarly to inhibitory neurons and is essential for protecting the brain from excessive activation in health and disease.


Subject(s)
Feedback, Physiological , Microglia/physiology , Neural Inhibition , Neurons/physiology , 5'-Nucleotidase/metabolism , Action Potentials , Adenosine/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Calcium/metabolism , Corpus Striatum/cytology , Corpus Striatum/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Neural Inhibition/genetics , Receptor, Adenosine A1/metabolism , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Time Factors
11.
Nat Rev Neurosci ; 21(5): 298, 2020 05.
Article in English | MEDLINE | ID: mdl-32152524

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Nat Rev Neurosci ; 21(2): 61-79, 2020 02.
Article in English | MEDLINE | ID: mdl-31896771

ABSTRACT

State-of-the-art tissue-clearing methods provide subcellular-level optical access to intact tissues from individual organs and even to some entire mammals. When combined with light-sheet microscopy and automated approaches to image analysis, existing tissue-clearing methods can speed up and may reduce the cost of conventional histology by several orders of magnitude. In addition, tissue-clearing chemistry allows whole-organ antibody labelling, which can be applied even to thick human tissues. By combining the most powerful labelling, clearing, imaging and data-analysis tools, scientists are extracting structural and functional cellular and subcellular information on complex mammalian bodies and large human specimens at an accelerated pace. The rapid generation of terabyte-scale imaging data furthermore creates a high demand for efficient computational approaches that tackle challenges in large-scale data analysis and management. In this Review, we discuss how tissue-clearing methods could provide an unbiased, system-level view of mammalian bodies and human specimens and discuss future opportunities for the use of these methods in human neuroscience.


Subject(s)
Histological Techniques/methods , Microscopy/methods , Nervous System/cytology , Animals , Histological Techniques/instrumentation , Humans , Imaging, Three-Dimensional/methods , Mammals , Microscopy/instrumentation , Neurosciences
13.
Cell ; 141(1): 154-165, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20303157

ABSTRACT

Optogenetic technologies employ light to control biological processes within targeted cells in vivo with high temporal precision. Here, we show that application of molecular trafficking principles can expand the optogenetic repertoire along several long-sought dimensions. Subcellular and transcellular trafficking strategies now permit (1) optical regulation at the far-red/infrared border and extension of optogenetic control across the entire visible spectrum, (2) increased potency of optical inhibition without increased light power requirement (nanoampere-scale chloride-mediated photocurrents that maintain the light sensitivity and reversible, step-like kinetic stability of earlier tools), and (3) generalizable strategies for targeting cells based not only on genetic identity, but also on morphology and tissue topology, to allow versatile targeting when promoters are not known or in genetically intractable organisms. Together, these results illustrate use of cell-biological principles to enable expansion of the versatile fast optogenetic technologies suitable for intact-systems biology and behavior.


Subject(s)
Genetic Techniques , Light , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Humans , Neurons/metabolism , Opsonin Proteins/genetics , Opsonin Proteins/metabolism , Rats , Systems Biology/methods
14.
Mol Ther ; 32(6): 1687-1700, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38582966

ABSTRACT

Deep-learning-based methods for protein structure prediction have achieved unprecedented accuracy, yet their utility in the engineering of protein-based binders remains constrained due to a gap between the ability to predict the structures of candidate proteins and the ability toprioritize proteins by their potential to bind to a target. To bridge this gap, we introduce Automated Pairwise Peptide-Receptor Analysis for Screening Engineered proteins (APPRAISE), a method for predicting the target-binding propensity of engineered proteins. After generating structural models of engineered proteins competing for binding to a target using an established structure prediction tool such as AlphaFold-Multimer or ESMFold, APPRAISE performs a rapid (under 1 CPU second per model) scoring analysis that takes into account biophysical and geometrical constraints. As proof-of-concept cases, we demonstrate that APPRAISE can accurately classify receptor-dependent vs. receptor-independent adeno-associated viral vectors and diverse classes of engineered proteins such as miniproteins targeting the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike, nanobodies targeting a G-protein-coupled receptor, and peptides that specifically bind to transferrin receptor or programmed death-ligand 1 (PD-L1). APPRAISE is accessible through a web-based notebook interface using Google Colaboratory (https://tiny.cc/APPRAISE). With its accuracy, interpretability, and generalizability, APPRAISE promises to expand the utility of protein structure prediction and accelerate protein engineering for biomedical applications.


Subject(s)
Protein Binding , Protein Engineering , SARS-CoV-2 , Protein Engineering/methods , Humans , SARS-CoV-2/metabolism , SARS-CoV-2/genetics , Models, Molecular , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Protein Conformation , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Deep Learning , COVID-19/virology , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/chemistry , Dependovirus/genetics , Genetic Vectors/chemistry , Genetic Vectors/genetics , Genetic Vectors/metabolism
15.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34001602

ABSTRACT

The goal of cancer-drug delivery is to achieve high levels of therapeutics within tumors with minimal systemic exposure that could cause toxicity. Producing biologics directly in situ where they diffuse and act locally is an attractive alternative to direct administration of recombinant therapeutics, as secretion by the tumor itself provides high local concentrations that act in a paracrine fashion continuously over an extended duration (paracrine delivery). We have engineered a SHielded, REtargeted ADenovirus (SHREAD) gene therapy platform that targets specific cells based on chosen surface markers and converts them into biofactories secreting therapeutics. In a proof of concept, a clinically approved antibody is delivered to orthotopic tumors in a model system in which precise biodistribution can be determined using tissue clearing with passive CLARITY technique (PACT) with high-resolution three-dimensional imaging and feature quantification within the tumors made transparent. We demonstrate high levels of tumor cell-specific transduction and significant and durable antibody production. PACT gives a localized quantification of the secreted therapeutic and allows us to directly observe enhanced pore formation in the tumor and destruction of the intact vasculature. In situ production of the antibody led to an 1,800-fold enhanced tumor-to-serum antibody concentration ratio compared to direct administration. Our detailed biochemical and microscopic analyses thus show that paracrine delivery with SHREAD could enable the use of highly potent therapeutic combinations, including those with systemic toxicity, to reach adequate therapeutic windows.


Subject(s)
Antibodies/pharmacology , Drug Delivery Systems , Genetic Therapy , Neoplasms/drug therapy , Adenoviridae/genetics , Animals , Antibodies/genetics , Antibodies/immunology , Antigens, Surface/genetics , Antineoplastic Agents/pharmacology , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Imaging, Three-Dimensional , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Paracrine Communication/drug effects
16.
BMC Biol ; 21(1): 232, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37957716

ABSTRACT

BACKGROUND: Copy number variations, and particularly duplications of genomic regions, have been strongly associated with various neurodegenerative conditions including autism spectrum disorder (ASD). These genetic variations have been found to have a significant impact on brain development and function, which can lead to the emergence of neurological and behavioral symptoms. Developing strategies to target these genomic duplications has been challenging, as the presence of endogenous copies of the duplicate genes often complicates the editing strategies. RESULTS: Using the ASD and anxiety mouse model Flailer, which contains a partial genomic duplication working as a dominant negative for MyoVa, we demonstrate the use of DN-CRISPRs to remove a 700 bp genomic region in vitro and in vivo. Importantly, DN-CRISPRs have not been used to remove genomic regions using sgRNA with an offset greater than 300 bp. We found that editing the flailer gene in primary cortical neurons reverts synaptic transport and transmission defects. Moreover, long-term depression (LTD), disrupted in Flailer animals, is recovered after gene editing. Delivery of DN-CRISPRs in vivo shows that local delivery to the ventral hippocampus can rescue some of the mutant behaviors, while intracerebroventricular delivery, completely recovers the Flailer animal phenotype associated to anxiety and ASD. CONCLUSIONS: Our results demonstrate the potential of DN-CRISPR to efficiently remove larger genomic duplications, working as a new gene therapy approach for treating neurodegenerative diseases.


Subject(s)
Autism Spectrum Disorder , Mice , Animals , Autism Spectrum Disorder/genetics , DNA Copy Number Variations , RNA, Guide, CRISPR-Cas Systems , Synaptic Transmission/genetics , Genomics
17.
Nat Methods ; 17(5): 541-550, 2020 05.
Article in English | MEDLINE | ID: mdl-32313222

ABSTRACT

Recombinant adeno-associated viruses (rAAVs) are efficient gene delivery vectors via intravenous delivery; however, natural serotypes display a finite set of tropisms. To expand their utility, we evolved AAV capsids to efficiently transduce specific cell types in adult mouse brains. Building upon our Cre-recombination-based AAV targeted evolution (CREATE) platform, we developed Multiplexed-CREATE (M-CREATE) to identify variants of interest in a given selection landscape through multiple positive and negative selection criteria. M-CREATE incorporates next-generation sequencing, synthetic library generation and a dedicated analysis pipeline. We have identified capsid variants that can transduce the central nervous system broadly, exhibit bias toward vascular cells and astrocytes, target neurons with greater specificity or cross the blood-brain barrier across diverse murine strains. Collectively, the M-CREATE methodology accelerates the discovery of capsids for use in neuroscience and gene-therapy applications.


Subject(s)
Brain/virology , Capsid Proteins/metabolism , Dependovirus/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Genetic Vectors/genetics , Integrases/metabolism , Animals , Blood-Brain Barrier/metabolism , Capsid Proteins/genetics , Female , Genetic Therapy , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Viral Tropism
18.
Nat Methods ; 17(4): 422-429, 2020 04.
Article in English | MEDLINE | ID: mdl-32203389

ABSTRACT

Brain circuits comprise vast numbers of interconnected neurons with diverse molecular, anatomical and physiological properties. To allow targeting of individual neurons for structural and functional studies, we created light-inducible site-specific DNA recombinases based on Cre, Dre and Flp (RecVs). RecVs can induce genomic modifications by one-photon or two-photon light induction in vivo. They can produce targeted, sparse and strong labeling of individual neurons by modifying multiple loci within mouse and zebrafish genomes. In combination with other genetic strategies, they allow intersectional targeting of different neuronal classes. In the mouse cortex they enable sparse labeling and whole-brain morphological reconstructions of individual neurons. Furthermore, these enzymes allow single-cell two-photon targeted genetic modifications and can be used in combination with functional optical indicators with minimal interference. In summary, RecVs enable spatiotemporally precise optogenomic modifications that can facilitate detailed single-cell analysis of neural circuits by linking genetic identity, morphology, connectivity and function.


Subject(s)
Genomics/methods , Optogenetics , Recombinases/metabolism , Animals , Brain/cytology , Gene Expression Regulation , Genetic Engineering , Mice , Neurons/metabolism , Recombinases/genetics , Zebrafish
19.
J Neurosci ; 41(12): 2645-2655, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33563725

ABSTRACT

The ability to recognize motivationally salient events and adaptively respond to them is critical for survival. Here, we tested whether dopamine (DA) neurons in the dorsal raphe nucleus (DRN) contribute to this process in both male and female mice. Population recordings of DRNDA neurons during associative learning tasks showed that their activity dynamically tracks the motivational salience, developing excitation to both reward-paired and shock-paired cues. The DRNDA response to reward-predicting cues was diminished after satiety, suggesting modulation by internal states. DRNDA activity was also greater for unexpected outcomes than for expected outcomes. Two-photon imaging of DRNDA neurons demonstrated that the majority of individual neurons developed activation to reward-predicting cues and reward but not to shock-predicting cues, which was surprising and qualitatively distinct from the population results. Performing the same fear learning procedures in freely-moving and head-fixed groups revealed that head-fixation itself abolished the neural response to aversive cues, indicating its modulation by behavioral context. Overall, these results suggest that DRNDA neurons encode motivational salience, dependent on internal and external factors.SIGNIFICANCE STATEMENT Dopamine (DA) contributes to motivational control, composed of at least two functional cell types, one signaling for motivational value and another for motivational salience. Here, we demonstrate that DA neurons in the dorsal raphe nucleus (DRN) encode the motivational salience in associative learning tasks. Neural responses were dynamic and modulated by the animal's internal state. The majority of single-cells developed responses to reward or paired cues, but not to shock-predicting cues. Additional experiments with freely-moving and head-fixed mice showed that head-fixation abolished the development of cue responses during fear learning. This work provides further characterization on the functional roles of overlooked DRNDA populations and an example that neural responses can be altered by head-fixation, which is commonly used in neuroscience.


Subject(s)
Dopaminergic Neurons/physiology , Dorsal Raphe Nucleus/physiology , Habituation, Psychophysiologic/physiology , Learning/physiology , Motivation/physiology , Neurons/physiology , Animals , Dopaminergic Neurons/chemistry , Dorsal Raphe Nucleus/chemistry , Dorsal Raphe Nucleus/cytology , Female , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton/methods , Neurons/chemistry , Photometry/methods , Signal Transduction/physiology
20.
Nat Methods ; 16(11): 1176-1184, 2019 11.
Article in English | MEDLINE | ID: mdl-31611694

ABSTRACT

We engineered light-gated channelrhodopsins (ChRs) whose current strength and light sensitivity enable minimally invasive neuronal circuit interrogation. Current ChR tools applied to the mammalian brain require intracranial surgery for transgene delivery and implantation of fiber-optic cables to produce light-dependent activation of a small volume of tissue. To facilitate expansive optogenetics without the need for invasive implants, our engineering approach leverages the substantial literature of ChR variants to train statistical models for the design of high-performance ChRs. With Gaussian process models trained on a limited experimental set of 102 functionally characterized ChRs, we designed high-photocurrent ChRs with high light sensitivity. Three of these, ChRger1-3, enable optogenetic activation of the nervous system via systemic transgene delivery. ChRger2 enables light-induced neuronal excitation without fiber-optic implantation; that is, this opsin enables transcranial optogenetics.


Subject(s)
Channelrhodopsins/genetics , Machine Learning , Optogenetics , Protein Engineering/methods , Animals , Channelrhodopsins/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL