Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 364
Filter
1.
Front Neural Circuits ; 18: 1436915, 2024.
Article in English | MEDLINE | ID: mdl-39091993

ABSTRACT

We provide a brief (and unabashedly biased) overview of the pre-transcriptomic history of somatostatin interneuron taxonomy, followed by a chronological summary of the large-scale, NIH-supported effort over the last ten years to generate a comprehensive, single-cell RNA-seq-based taxonomy of cortical neurons. Focusing on somatostatin interneurons, we present the perspective of experimental neuroscientists trying to incorporate the new classification schemes into their own research while struggling to keep up with the ever-increasing number of proposed cell types, which seems to double every two years. We suggest that for experimental analysis, the most useful taxonomic level is the subdivision of somatostatin interneurons into ten or so "supertypes," which closely agrees with their more traditional classification by morphological, electrophysiological and neurochemical features. We argue that finer subdivisions ("t-types" or "clusters"), based on slight variations in gene expression profiles but lacking clear phenotypic differences, are less useful to researchers and may actually defeat the purpose of classifying neurons to begin with. We end by stressing the need for generating novel tools (mouse lines, viral vectors) for genetically targeting distinct supertypes for expression of fluorescent reporters, calcium sensors and excitatory or inhibitory opsins, allowing neuroscientists to chart the input and output synaptic connections of each proposed subtype, reveal the position they occupy in the cortical network and examine experimentally their roles in sensorimotor behaviors and cognitive brain functions.


Subject(s)
Interneurons , Somatostatin , Animals , Somatostatin/metabolism , Interneurons/classification , Interneurons/physiology , Interneurons/metabolism , Interneurons/cytology , Humans
2.
Nat Commun ; 15(1): 6164, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039043

ABSTRACT

Deciphering the striatal interneuron diversity is key to understanding the basal ganglia circuit and to untangling the complex neurological and psychiatric diseases affecting this brain structure. We performed snRNA-seq and spatial transcriptomics of postmortem human caudate nucleus and putamen samples to elucidate the diversity and abundance of interneuron populations and their inherent transcriptional structure in the human dorsal striatum. We propose a comprehensive taxonomy of striatal interneurons with eight main classes and fourteen subclasses, providing their full transcriptomic identity and spatial expression profile as well as additional quantitative FISH validation for specific populations. We have also delineated the correspondence of our taxonomy with previous standardized classifications and shown the main transcriptomic and class abundance differences between caudate nucleus and putamen. Notably, based on key functional genes such as ion channels and synaptic receptors, we found matching known mouse interneuron populations for the most abundant populations, the recently described PTHLH and TAC3 interneurons. Finally, we were able to integrate other published datasets with ours, supporting the generalizability of this harmonized taxonomy.


Subject(s)
Interneurons , Transcriptome , Humans , Interneurons/metabolism , Interneurons/classification , Interneurons/cytology , Male , Female , Corpus Striatum/cytology , Corpus Striatum/metabolism , Caudate Nucleus/metabolism , Caudate Nucleus/cytology , Putamen/metabolism , Putamen/cytology , Middle Aged , Animals , Aged , Mice , Gene Expression Profiling/methods , Adult
3.
Nature ; 632(8026): 858-868, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048816

ABSTRACT

Alzheimer's disease is the leading cause of dementia worldwide, but the cellular pathways that underlie its pathological progression across brain regions remain poorly understood1-3. Here we report a single-cell transcriptomic atlas of six different brain regions in the aged human brain, covering 1.3 million cells from 283 post-mortem human brain samples across 48 individuals with and without Alzheimer's disease. We identify 76 cell types, including region-specific subtypes of astrocytes and excitatory neurons and an inhibitory interneuron population unique to the thalamus and distinct from canonical inhibitory subclasses. We identify vulnerable populations of excitatory and inhibitory neurons that are depleted in specific brain regions in Alzheimer's disease, and provide evidence that the Reelin signalling pathway is involved in modulating the vulnerability of these neurons. We develop a scalable method for discovering gene modules, which we use to identify cell-type-specific and region-specific modules that are altered in Alzheimer's disease and to annotate transcriptomic differences associated with diverse pathological variables. We identify an astrocyte program that is associated with cognitive resilience to Alzheimer's disease pathology, tying choline metabolism and polyamine biosynthesis in astrocytes to preserved cognitive function late in life. Together, our study develops a regional atlas of the ageing human brain and provides insights into cellular vulnerability, response and resilience to Alzheimer's disease pathology.


Subject(s)
Alzheimer Disease , Brain , Gene Expression Profiling , Single-Cell Analysis , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Aging/metabolism , Aging/pathology , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Astrocytes/classification , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/pathology , Autopsy , Brain/anatomy & histology , Brain/cytology , Brain/metabolism , Brain/pathology , Case-Control Studies , Choline/metabolism , Cognition/physiology , Gene Regulatory Networks , Interneurons/classification , Interneurons/cytology , Interneurons/metabolism , Interneurons/pathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neural Inhibition , Neurons/classification , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Polyamines/metabolism , Reelin Protein , Signal Transduction , Thalamus/cytology , Thalamus/metabolism , Thalamus/pathology , Transcriptome
4.
Front Neural Circuits ; 18: 1427378, 2024.
Article in English | MEDLINE | ID: mdl-38933598

ABSTRACT

Various mammals have shown that sensory stimulation plays a crucial role in regulating the development of diverse structures, such as the olfactory bulb (OB), cerebral cortex, hippocampus, and retina. In the OB, the dendritic development of excitatory projection neurons like mitral/tufted cells is influenced by olfactory experiences. Odor stimulation is also essential for the dendritic development of inhibitory OB interneurons, such as granule and periglomerular cells, which are continuously produced in the ventricular-subventricular zone throughout life. Based on the morphological and molecular features, OB interneurons are classified into several subtypes. The role for each interneuron subtype in the control of olfactory behavior remains poorly understood due to lack of each specific marker. Among the several OB interneuron subtypes, a specific granule cell subtype, which expresses the oncofetal trophoblast glycoprotein (Tpbg or 5T4) gene, has been reported to be required for odor detection and discrimination behavior. This review will primarily focus on elucidating the contribution of different granule cell subtypes, including the Tpbg/5T4 subtype, to olfactory processing and behavior during the embryonic and adult stages.


Subject(s)
Interneurons , Olfactory Bulb , Animals , Interneurons/physiology , Interneurons/metabolism , Interneurons/classification , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Humans , Neurogenesis/physiology
5.
Nature ; 598(7879): 214-219, 2021 10.
Article in English | MEDLINE | ID: mdl-34616064

ABSTRACT

The cerebellar cortex is a well-studied brain structure with diverse roles in motor learning, coordination, cognition and autonomic regulation. However,  a complete inventory of cerebellar cell types is currently lacking. Here, using recent advances in high-throughput transcriptional profiling1-3, we molecularly define cell types across individual lobules of the adult mouse cerebellum. Purkinje neurons showed considerable regional specialization, with the greatest diversity occurring in the posterior lobules. For several types of cerebellar interneuron, the molecular variation within each type was more continuous, rather than discrete. In particular, for the unipolar brush cells-an interneuron population previously subdivided into discrete populations-the continuous variation in gene expression was associated with a graded continuum of electrophysiological properties. Notably, we found that molecular layer interneurons were composed of two molecularly and functionally distinct types. Both types show a continuum of morphological variation through the thickness of the molecular layer, but electrophysiological recordings revealed marked differences between the two types in spontaneous firing, excitability and electrical coupling. Together, these findings provide a comprehensive cellular atlas of the cerebellar cortex, and outline a methodological and conceptual framework for the integration of molecular, morphological and physiological ontologies for defining brain cell types.


Subject(s)
Cerebellar Cortex/cytology , Gene Expression Profiling , Transcriptome , Adult , Animals , Atlases as Topic , Electrophysiology , Female , Humans , Interneurons/classification , Interneurons/cytology , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroglia/classification , Neuroglia/cytology , Neuroglia/metabolism , Neurons/classification , Neurons/cytology , Neurons/metabolism
6.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800863

ABSTRACT

The ability to sense and move within an environment are complex functions necessary for the survival of nearly all species. The spinal cord is both the initial entry site for peripheral information and the final output site for motor response, placing spinal circuits as paramount in mediating sensory responses and coordinating movement. This is partly accomplished through the activation of complex spinal microcircuits that gate afferent signals to filter extraneous stimuli from various sensory modalities and determine which signals are transmitted to higher order structures in the CNS and to spinal motor pathways. A mechanistic understanding of how inhibitory interneurons are organized and employed within the spinal cord will provide potential access points for therapeutics targeting inhibitory deficits underlying various pathologies including sensory and movement disorders. Recent studies using transgenic manipulations, neurochemical profiling, and single-cell transcriptomics have identified distinct populations of inhibitory interneurons which express an array of genetic and/or neurochemical markers that constitute functional microcircuits. In this review, we provide an overview of identified neural components that make up inhibitory microcircuits within the dorsal and ventral spinal cord and highlight the importance of inhibitory control of sensorimotor pathways at the spinal level.


Subject(s)
Afferent Pathways/physiology , Interneurons/physiology , Movement/physiology , Neural Inhibition/physiology , Sensation/physiology , Sensory Gating/physiology , Spinal Cord/cytology , Animals , Anterior Horn Cells/chemistry , Anterior Horn Cells/classification , Anterior Horn Cells/physiology , Humans , Interneurons/chemistry , Interneurons/classification , Models, Neurological , Motor Neurons/physiology , Movement Disorders/physiopathology , Nerve Fibers/physiology , Nerve Tissue Proteins/analysis , Neuropeptides/analysis , Posterior Horn Cells/chemistry , Posterior Horn Cells/classification , Sensation Disorders/physiopathology , Sensory Receptor Cells/physiology , Spinal Cord/physiology , Synapses/physiology
7.
PLoS Comput Biol ; 16(7): e1008016, 2020 07.
Article in English | MEDLINE | ID: mdl-32716912

ABSTRACT

The mammalian sensory cortex is composed of multiple types of inhibitory and excitatory neurons, which form sophisticated microcircuits for processing and transmitting sensory information. Despite rapid progress in understanding the function of distinct neuronal populations, the parameters of connectivity that are required for the function of these microcircuits remain unknown. Recent studies found that two most common inhibitory interneurons, parvalbumin- (PV) and somatostatin-(SST) positive interneurons control sound-evoked responses, temporal adaptation and network dynamics in the auditory cortex (AC). These studies can inform our understanding of parameters for the connectivity of excitatory-inhibitory cortical circuits. Specifically, we asked whether a common microcircuit can account for the disparate effects found in studies by different groups. By starting with a cortical rate model, we find that a simple current-compensating mechanism accounts for the experimental findings from multiple groups. They key mechanisms are two-fold. First, PVs compensate for reduced SST activity when thalamic inputs are strong with less compensation when thalamic inputs are weak. Second, SSTs are generally disinhibited by reduced PV activity regardless of thalamic input strength. These roles are augmented by plastic synapses. These roles reproduce the differential effects of PVs and SSTs in stimulus-specific adaptation, forward suppression and tuning-curve adaptation, as well as the influence of PVs on feedforward functional connectivity in the circuit. This circuit exhibits a balance of inhibitory and excitatory currents that persists on stimulation. This approach brings together multiple findings from different laboratories and identifies a circuit that can be used in future studies of upstream and downstream sensory processing.


Subject(s)
Auditory Cortex/physiology , Computational Biology , Interneurons/physiology , Models, Neurological , Optogenetics , Algorithms , Animals , Computer Simulation , Humans , Interneurons/classification , Synapses/physiology , Thalamus/physiology , Time Factors
8.
Elife ; 92020 06 03.
Article in English | MEDLINE | ID: mdl-32490811

ABSTRACT

CCK-expressing interneurons (CCK+INs) are crucial for controlling hippocampal activity. We found two firing phenotypes of CCK+INs in rat hippocampal CA3 area; either possessing a previously undetected membrane potential-dependent firing or regular firing phenotype, due to different low-voltage-activated potassium currents. These different excitability properties destine the two types for distinct functions, because the former is essentially silenced during realistic 8-15 Hz oscillations. By contrast, the general intrinsic excitability, morphology and gene-profiles of the two types were surprisingly similar. Even the expression of Kv4.3 channels were comparable, despite evidences showing that Kv4.3-mediated currents underlie the distinct firing properties. Instead, the firing phenotypes were correlated with the presence of distinct isoforms of Kv4 auxiliary subunits (KChIP1 vs. KChIP4e and DPP6S). Our results reveal the underlying mechanisms of two previously unknown types of CCK+INs and demonstrate that alternative splicing of few genes, which may be viewed as a minor change in the cells' whole transcriptome, can determine cell-type identity.


Subject(s)
CA3 Region, Hippocampal/cytology , Cholecystokinin/metabolism , Interneurons , Shal Potassium Channels , Animals , Cells, Cultured , Interneurons/chemistry , Interneurons/classification , Interneurons/metabolism , Membrane Potentials/physiology , Phenotype , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar , Shal Potassium Channels/chemistry , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Transcriptome/genetics
9.
J Mol Neurosci ; 70(5): 796-805, 2020 May.
Article in English | MEDLINE | ID: mdl-32036579

ABSTRACT

Focal cortical dysplasia (FCD) is the main cause of medically intractable pediatric epilepsy. Previous studies have suggested that alteration of cortical interneurons and abnormal cytoarchitecture have been linked to initiation and development for seizure. However, whether each individual subpopulation of cortical interneurons is linked to distinct FCD subtypes remains largely unknown. Here, we retrospectively analyzed both control samples and epileptic specimens pathologically diagnosed with FCD types Ia, IIa, or IIb. We quantified three major interneuron (IN) subpopulations, including parvalbumin (PV)-, somatostatin (Sst)-, and vasoactive intestinal peptide (Vip)-positive INs across all the subgroups. Additionally, we calculated the ratio of the subpopulations of INs to the major INs (mINs) by defining the total number of the PV-, Sst-, and Vip-INs as mINs. Compared with the control, the density of the PV-INs in FCD type IIb was significantly lower, and the ratio of PV/mINs was lower in the superficial part of the cortex of the FCD type Ia and IIb groups. Interestingly, we found a significant increase in the ratio of Vip/mINs only in FCD type IIb. Overall, these results suggest that in addition to a reduction in PV-INs, the increase in Vip/mINs may be related to the initiation of epilepsy in FCD type IIb. Furthermore, the increase in Vip/mINs in FCD type IIb may, from the IN development perspective, indicate that FCD type IIb forms during earlier stages of pregnancy than FCD type Ia.


Subject(s)
Drug Resistant Epilepsy/pathology , Interneurons/metabolism , Malformations of Cortical Development/pathology , Adolescent , Adult , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Child , Child, Preschool , Drug Resistant Epilepsy/metabolism , Female , Humans , Infant , Interneurons/classification , Male , Malformations of Cortical Development/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
10.
J Neurosci ; 40(11): 2314-2331, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32005764

ABSTRACT

Distinct components of working memory are coordinated by different classes of inhibitory interneurons in the PFC, but the role of cholecystokinin (CCK)-positive interneurons remains enigmatic. In humans, this major population of interneurons shows histological abnormalities in schizophrenia, an illness in which deficient working memory is a core defining symptom and the best predictor of long-term functional outcome. Yet, CCK interneurons as a molecularly distinct class have proved intractable to examination by typical molecular methods due to widespread expression of CCK in the pyramidal neuron population. Using an intersectional approach in mice of both sexes, we have succeeded in labeling, interrogating, and manipulating CCK interneurons in the mPFC. Here, we describe the anatomical distribution, electrophysiological properties, and postsynaptic connectivity of CCK interneurons, and evaluate their role in cognition. We found that CCK interneurons comprise a larger proportion of the mPFC interneurons compared with parvalbumin interneurons, targeting a wide range of neuronal subtypes with a distinct connectivity pattern. Phase-specific optogenetic inhibition revealed that CCK, but not parvalbumin, interneurons play a critical role in the retrieval of working memory. These findings shine new light on the relationship between cortical CCK interneurons and cognition and offer a new set of tools to investigate interneuron dysfunction and cognitive impairments associated with schizophrenia.SIGNIFICANCE STATEMENT Cholecystokinin-expressing interneurons outnumber other interneuron populations in key brain areas involved in cognition and memory, including the mPFC. However, they have proved intractable to examination as experimental techniques have lacked the necessary selectivity. To the best of our knowledge, the present study is the first to report detailed properties of cortical cholecystokinin interneurons, revealing their anatomical organization, electrophysiological properties, postsynaptic connectivity, and behavioral function in working memory.


Subject(s)
Cholecystokinin/physiology , Interneurons/physiology , Memory, Short-Term/physiology , Mental Recall/physiology , Prefrontal Cortex/physiology , Animals , Appetitive Behavior/physiology , Discrimination Learning/physiology , Discrimination, Psychological/physiology , Female , Genes, Reporter , Interneurons/classification , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/analysis , Odorants , Optogenetics , Parvalbumins/analysis , Patch-Clamp Techniques , Reward , Schizophrenia/physiopathology , Smell/physiology , Synaptic Potentials/physiology
11.
J Cereb Blood Flow Metab ; 40(12): 2401-2415, 2020 12.
Article in English | MEDLINE | ID: mdl-31842665

ABSTRACT

Disturbances of cognitive functions occur rapidly during acute metabolic stress. However, the underlying mechanisms are not fully understood. Cortical gamma oscillations (30-100 Hz) emerging from precise synaptic transmission between excitatory principal neurons and inhibitory interneurons, such as fast-spiking GABAergic basket cells, are associated with higher brain functions, like sensory perception, selective attention and memory formation. We investigated the alterations of cholinergic gamma oscillations at the level of neuronal ensembles in the CA3 region of rat hippocampal slice cultures. We combined electrophysiology, calcium imaging (CamKII.GCaMP6f) and mild metabolic stress that was induced by rotenone, a lipophilic and highly selective inhibitor of complex I in the respiratory chain of mitochondria. The detected pyramidal cell ensembles showing repetitive patterns of activity were highly sensitive to mild metabolic stress. Whereas such synchronised multicellular activity diminished, the overall activity of individual pyramidal cells was unaffected. Additionally, mild metabolic stress had no effect on the rate of action potential generation in fast-spiking neural units. However, the partial disinhibition of slow-spiking neural units suggests that disturbances of ensemble formation likely result from alterations in synaptic inhibition. Our study bridges disturbances on the (multi-)cellular and network level to putative cognitive impairment on the system level.


Subject(s)
Cognitive Dysfunction/metabolism , Gamma Rhythm/physiology , Hippocampus/metabolism , Pyramidal Cells/drug effects , Stress, Physiological/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cognitive Dysfunction/physiopathology , Electrophysiology/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gamma Rhythm/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Interneurons/classification , Interneurons/drug effects , Interneurons/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Rats , Rats, Wistar , Rotenone/administration & dosage , Rotenone/pharmacology , Stress, Physiological/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Uncoupling Agents/administration & dosage , Uncoupling Agents/pharmacology
12.
J Neurosci ; 40(7): 1440-1452, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31826942

ABSTRACT

Neuronal diversity provides the spinal cord with the functional flexibility required to perform complex motor tasks. Spinal neurons arise during early embryonic development with the establishment of spatially and molecularly discrete progenitor domains that give rise to distinct, but highly heterogeneous, postmitotic interneuron (IN) populations. Our previous studies have shown that Sim1-expressing V3 INs, originating from the p3 progenitor domain, are anatomically and physiologically divergent. However, the developmental logic guiding V3 subpopulation diversity remains elusive. In specific cases of other IN classes, neurogenesis timing can play a role in determining the ultimate fates and unique characteristics of distinctive subpopulations. To examine whether neurogenesis timing contributes to V3 diversity, we systematically investigated the temporal neurogenesis profiles of V3 INs in the mouse spinal cord. Our work uncovered that V3 INs were organized into either early-born [embryonic day 9.5 (E9.5) to E10.5] or late-born (E11.5-E12.5) neurogenic waves. Early-born V3 INs displayed both ascending and descending commissural projections and clustered into subgroups across dorsoventral spinal laminae. In contrast, late-born V3 INs became fate-restricted to ventral laminae and displayed mostly descending and local commissural projections and uniform membrane properties. Furthermore, we found that the postmitotic transcription factor, Sim1, although expressed in all V3 INs, exclusively regulated the dorsal clustering and electrophysiological diversification of early-born, but not late-born, V3 INs, which indicates that neurogenesis timing may enable newborn V3 INs to interact with different postmitotic differentiation pathways. Thus, our work demonstrates neurogenesis timing as a developmental mechanism underlying the postmitotic differentiation of V3 INs into distinct subpopulation assemblies.SIGNIFICANCE STATEMENT Interneuron (IN) diversity empowers the spinal cord with the computation flexibility required to perform appropriate sensorimotor control. As such, uncovering the developmental logic guiding spinal IN diversity is fundamental to understanding the development of movement. In our current work, through a focus on the cardinal spinal V3 IN population, we investigated the role of neurogenesis timing on IN diversity. We uncovered that V3 INs are organized into early-born [embryonic day 9.5 (E9.5) to E10.5] or late-born (E11.5-E12.5) neurogenic waves, where late-born V3 INs display increasingly restricted subpopulation fates. Next, to better understand the consequences of V3 neurogenesis timing, we investigated the time-dependent functions of the Sim1 transcription factor, which is expressed in postmitotic V3 INs. Interestingly, Sim1 exclusively regulated the diversification of early-born, but not late-born, V3 INs. Thus, our current work indicates neurogenesis timing can modulate the functions of early postmitotic transcription factors and, thus, subpopulation fate specifications.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Interneurons/physiology , Neurogenesis , Repressor Proteins/physiology , Spinal Cord/cytology , Animals , Axonal Transport , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Cell Movement , Crosses, Genetic , Glutamic Acid/physiology , Interneurons/classification , Mice , Mice, Knockout , Neurotransmitter Agents/physiology , Patch-Clamp Techniques , Repressor Proteins/deficiency , Repressor Proteins/genetics , Spinal Cord/embryology , Spinal Cord/growth & development , Time Factors
13.
Mol Psychiatry ; 25(10): 2313-2329, 2020 10.
Article in English | MEDLINE | ID: mdl-31595033

ABSTRACT

Severe infections during pregnancy are one of the major risk factors for cognitive impairment in the offspring. It has been suggested that maternal inflammation leads to dysfunction of cortical GABAergic interneurons that in turn underlies cognitive impairment of the affected offspring. However, the evidence comes largely from studies of adult or mature brains and how the impairment of inhibitory circuits arises upon maternal inflammation is unknown. Here we show that maternal inflammation affects multiple steps of cortical GABAergic interneuron development, i.e., proliferation of precursor cells, migration and positioning of neuroblasts, as well as neuronal maturation. Importantly, the development of distinct subtypes of cortical GABAergic interneurons was discretely impaired as a result of maternal inflammation. This translated into a reduction in cell numbers, redistribution across cortical regions and layers, and changes in morphology and cellular properties. Furthermore, selective vulnerability of GABAergic interneuron subtypes was associated with the stage of brain development. Thus, we propose that maternally derived insults have developmental stage-dependent effects, which contribute to the complex etiology of cognitive impairment in the affected offspring.


Subject(s)
Cerebral Cortex , Inflammation , Interneurons , Mothers , Neurogenesis , Animals , Cell Movement , Cell Proliferation , Cerebral Cortex/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Female , GABAergic Neurons/pathology , Interneurons/classification , Interneurons/pathology , Male , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/pathology
14.
Acta Neurobiol Exp (Wars) ; 79(4): 328-337, 2019.
Article in English | MEDLINE | ID: mdl-31885390

ABSTRACT

For years, interneurons expressing vasoactive intestinal peptide (VIP) interneurons and their function within the neocortex have been shrouded in mystery. Their relatively small size and minimal representation in the cortex have made investigation difficult. Due to their service role performed in co­operation with glia and blood vessels to supply energy during neuronal activation in the brain, the contribution of VIP interneurons to local neuronal circuit function was not appreciated. VIP interneurons in the neocortex account for roughly 12% of all interneurons. They have been described as a subgroup of the third largest population of 5-hydroxytryptamine 3a (5HT3a) receptor­expressing interneurons, non­overlapping with interneuron populations expressing parvalbumin (PV) or somatostatin (SST). However, it was recently shown that only half of VIP interneurons display a 5HT3a receptor response and a subset of VIP interneurons in visual cortex co­express SST. Over the last several years, due to new technical advancements, many facts have emerged relating to VIP interneuron phylogenetic origin, operational mechanisms within local circuits and functional significance. Some of these discoveries have dramatically shifted the perception of VIP interneurons. This review focuses on the function of the VIP interneurons residing in layer 2/3 of the mouse neocortex.


Subject(s)
Interneurons/physiology , Neocortex/cytology , Vasoactive Intestinal Peptide/physiology , Action Potentials , Animals , Behavior, Animal/physiology , Connectome , Interneurons/chemistry , Interneurons/classification , Learning/physiology , Locomotion/physiology , Mice , Phylogeny , Pyramidal Cells/physiology , Receptors, Serotonin, 5-HT3/metabolism , Sensation/physiology , Somatostatin/analysis , Vision, Ocular/physiology , Visual Cortex/cytology
15.
Sci Data ; 6(1): 221, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641131

ABSTRACT

There is currently no unique catalog of cortical GABAergic interneuron types. In 2013, we asked 48 leading neuroscientists to classify 320 interneurons by inspecting images of their morphology. That study was the first to quantify the degree of agreement among neuroscientists in morphology-based interneuron classification, showing high agreement for the chandelier and Martinotti types, yet low agreement for most of the remaining types considered. Here we present the dataset containing the classification choices by the neuroscientists according to interneuron type as well as to five prominent morphological features. These data can be used as crisp or soft training labels for learning supervised machine learning interneuron classifiers, while further analyses can try to pinpoint anatomical characteristics that make an interneuron especially difficult or especially easy to classify.


Subject(s)
GABAergic Neurons/classification , Interneurons/classification , Animals , GABAergic Neurons/cytology , Humans , Interneurons/cytology
16.
Front Neural Circuits ; 13: 32, 2019.
Article in English | MEDLINE | ID: mdl-31164808

ABSTRACT

The striatum is predominantly composed of medium spiny projection neurons, with the remaining neurons consisting of several types of interneurons. Among the interneurons are a group of cells that express tyrosine hydroxylase (TH). Although the intrinsic electrical properties of these TH-expressing interneurons have been characterized, there is no agreement on the number of TH-expressing cell types and their electrical properties. Here, we have used transgenic mice in which YFP-tagged channelrhodopsin-2 (ChR2) was expressed in potential TH-expressing cells in a Cre-dependent manner. We found that the YFP+ neurons in the striatum were heterogeneous in their intrinsic electrical properties; unbiased clustering indicated that there are three main neuronal subtypes. One population of neurons had aspiny dendrites with high-frequency action potential (AP) firing and plateau potentials, resembling the TH interneurons (THIN) described previously. A second, very small population of labeled neurons resembled medium-sized spiny neurons (MSN). The third population of neurons had dendrites with an intermediate density of spines, showed substantial AP adaptation and generated prolonged spikes. This type of striatal neuron has not been previously identified in the adult mouse and we have named it the Frequency-Adapting Neuron with Spines (FANS). Because of their distinctive properties, FANS may play a unique role in striatal information processing.


Subject(s)
Corpus Striatum/cytology , Interneurons/classification , Interneurons/cytology , Animals , Mice , Mice, Transgenic
17.
BMC Bioinformatics ; 19(1): 511, 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30558530

ABSTRACT

BACKGROUND: The challenge of classifying cortical interneurons is yet to be solved. Data-driven classification into established morphological types may provide insight and practical value. RESULTS: We trained models using 217 high-quality morphologies of rat somatosensory neocortex interneurons reconstructed by a single laboratory and pre-classified into eight types. We quantified 103 axonal and dendritic morphometrics, including novel ones that capture features such as arbor orientation, extent in layer one, and dendritic polarity. We trained a one-versus-rest classifier for each type, combining well-known supervised classification algorithms with feature selection and over- and under-sampling. We accurately classified the nest basket, Martinotti, and basket cell types with the Martinotti model outperforming 39 out of 42 leading neuroscientists. We had moderate accuracy for the double bouquet, small and large basket types, and limited accuracy for the chandelier and bitufted types. We characterized the types with interpretable models or with up to ten morphometrics. CONCLUSION: Except for large basket, 50 high-quality reconstructions sufficed to learn an accurate model of a type. Improving these models may require quantifying complex arborization patterns and finding correlates of bouton-related features. Our study brings attention to practical aspects important for neuron classification and is readily reproducible, with all code and data available online.


Subject(s)
Algorithms , Dendrites/chemistry , Interneurons/classification , Interneurons/cytology , Neocortex/cytology , Animals , Cells, Cultured , Male , Rats , Rats, Wistar
18.
PLoS One ; 13(7): e0200567, 2018.
Article in English | MEDLINE | ID: mdl-30001424

ABSTRACT

Somatostatin-expressing (SOM+), inhibitory interneurons represent a heterogeneous group of cells and given their remarkable diversity, classification of SOM+ interneurons remains a challenging task. Electrophysiological, morphological and neurochemical classes of SOM+ interneurons have been proposed in the past but it remains unclear as to what extent these classes are congruent. We performed whole-cell patch-clamp recordings from 127 GFP-labeled SOM+ interneurons ('GIN') of the superficial cingulate cortex with subsequent biocytin-filling and immunocytochemical labeling. Principal component analysis followed by k-means clustering predicted two putative subtypes of SOM+ interneurons, which we designated as group I and group II GIN. A key finding of our study is the fact that these electrophysiologically and morphologically distinct groups of SOM+ interneurons can be correlated with two neurochemical subtypes of SOM+ interneurons described recently in our laboratory. In particular, all SOM+ interneurons expressing calbindin but no calretinin could be classified as group I GIN, whereas all but one neuropeptide Y- and calretinin-positive interneurons were found in group II.


Subject(s)
GABAergic Neurons , Gyrus Cinguli , Interneurons , Somatostatin/biosynthesis , Animals , Calbindins/biosynthesis , GABAergic Neurons/classification , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gyrus Cinguli/cytology , Gyrus Cinguli/metabolism , Interneurons/classification , Interneurons/cytology , Interneurons/metabolism , Mice , Neuropeptide Y/biosynthesis
19.
J Neurosci ; 38(34): 7378-7391, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30037831

ABSTRACT

Development of cortical interneurons continues until the end of human pregnancy. Premature birth deprives the newborns from the supply of maternal estrogen and a secure intrauterine environment. Indeed, preterm infants suffer from neurobehavioral disorders. This can result from both preterm birth and associated postnatal complications, which might disrupt recruitment and maturation of cortical interneurons. We hypothesized that interneuron subtypes, including parvalbumin-positive (PV+), somatostatin-positive (SST+), calretinin-positive (CalR+), and neuropeptide Y-positive (NPY+) interneurons, were recruited in the upper and lower cortical layers in a distinct manner with advancing gestational age. In addition, preterm birth would disrupt the heterogeneity of cortical interneurons, which might be reversed by estrogen treatment. These hypotheses were tested by analyzing autopsy samples from premature infants and evaluating the effect of estrogen supplementation in prematurely delivered rabbits. The PV+ and CalR+ neurons were abundant, whereas SST+ and NPY+ neurons were few in cortical layers of preterm human infants. Premature birth of infants reduced the density of PV+ or GAD67+ neurons and increased SST+ interneurons in the upper cortical layers. Importantly, 17 ß-estradiol treatment in preterm rabbits increased the number of PV+ neurons in the upper cortical layers relative to controls at postnatal day 14 (P14) and P21 and transiently reduced SST population at P14. Moreover, protein and mRNA levels of Arx, a key regulator of cortical interneuron maturation and migration, were higher in estrogen-treated rabbits relative to controls. Therefore, deficits in PV+ and excess of SST+ neurons in premature newborns are ameliorated by estrogen replacement, which can be attributed to elevated Arx levels. Estrogen replacement might enhance neurodevelopmental outcomes in extremely preterm infants.SIGNIFICANCE STATEMENT Premature birth often leads to neurodevelopmental delays and behavioral disorders, which may be ascribed to disturbances in the development and maturation of cortical interneurons. Here, we show that preterm birth in humans is associated with reduced population of parvalbumin-positive (PV+) neurons and an excess of somatostatin-expressing interneurons in the cerebral cortex. More importantly, 17 ß-estradiol treatment increased the number of PV+ neurons in preterm-born rabbits, which appears to be mediated by an elevation in the expression of Arx transcription factor. Hence the present study highlights prematurity-induced reduction in PV+ neurons in human infants and reversal in their population by estrogen replacement in preterm rabbits. Because preterm birth drops plasma estrogen level 100-fold, estrogen replacement in extremely preterm infants might improve their developmental outcome and minimize neurobehavioral disorders.


Subject(s)
Cerebral Cortex/pathology , Estradiol/pharmacology , Infant, Premature, Diseases/pathology , Interneurons/drug effects , Animals , Animals, Newborn , Calbindin 2/analysis , Cell Count , Female , Gestational Age , Glutamate Decarboxylase/analysis , Humans , Infant, Newborn , Infant, Premature , Interneurons/chemistry , Interneurons/classification , Interneurons/physiology , Male , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuropeptide Y/analysis , Parvalbumins/analysis , Rabbits , Somatostatin/analysis , Transcription Factors/biosynthesis , Transcription Factors/genetics
20.
J Neurosci ; 38(31): 6983-7003, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29954847

ABSTRACT

In cortical structures, principal cell activity is tightly regulated by different GABAergic interneurons (INs). Among these INs are vasoactive intestinal polypeptide-expressing (VIP+) INs, which innervate preferentially other INs, providing a structural basis for temporal disinhibition of principal cells. However, relatively little is known about VIP+ INs in the amygdaloid basolateral complex (BLA). In this study, we report that VIP+ INs have a variable density in the distinct subdivisions of the mouse BLA. Based on different anatomical, neurochemical, and electrophysiological criteria, VIP+ INs could be identified as IN-selective INs (IS-INs) and basket cells expressing CB1 cannabinoid receptors. Whole-cell recordings of VIP+ IS-INs revealed three different spiking patterns, none of which was associated with the expression of calretinin. Genetic targeting combined with optogenetics and in vitro recordings enabled us to identify several types of BLA INs innervated by VIP+ INs, including other IS-INs, basket and neurogliaform cells. Moreover, light stimulation of VIP+ basket cell axon terminals, characterized by CB1 sensitivity, evoked IPSPs in ∼20% of principal neurons. Finally, we show that VIP+ INs receive a dense innervation from both GABAergic inputs (although only 10% from other VIP+ INs) and distinct glutamatergic inputs, identified by their expression of different vesicular glutamate transporters.In conclusion, our study provides a wide-range analysis of single-cell properties of VIP+ INs in the mouse BLA and of their intrinsic and extrinsic connectivity. Our results reinforce the evidence that VIP+ INs are structurally and functionally heterogeneous and that this heterogeneity could mediate different roles in amygdala-dependent functions.SIGNIFICANCE STATEMENT We provide the first comprehensive analysis of the distribution of vasoactive intestinal polypeptide-expressing (VIP+) interneurons (INs) across the entire mouse amygdaloid basolateral complex (BLA), as well as of their morphological and physiological properties. VIP+ INs in the neocortex preferentially target other INs to form a disinhibitory network that facilitates principal cell firing. Our study is the first to demonstrate the presence of such a disinhibitory circuitry in the BLA. We observed structural and functional heterogeneity of these INs and characterized their input/output connectivity. We also identified several types of BLA INs that, when inhibited, may provide a temporal window for principal cell firing and facilitate associative plasticity, e.g., in fear learning.


Subject(s)
Basolateral Nuclear Complex/cytology , Interneurons/physiology , Vasoactive Intestinal Peptide/analysis , Action Potentials , Animals , Basolateral Nuclear Complex/physiology , Cell Count , Connectome , Crosses, Genetic , Genes, Reporter , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/radiation effects , Interneurons/chemistry , Interneurons/classification , Interneurons/radiation effects , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Optogenetics , Patch-Clamp Techniques , Presynaptic Terminals/ultrastructure , Receptor, Cannabinoid, CB1/analysis , Vesicular Glutamate Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL