Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.158
Filter
1.
J Comp Neurol ; 532(8): e25666, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39235159

ABSTRACT

We have investigated the hippocampal connectivity of the marmoset presubiculum (PreS) and reported that major connections of PreS in the rat were conserved in the marmoset. Moreover, our results indicated the presence of several additional projections that were almost absent in the rat brain, but abundant in the marmoset, such as direct projections from CA1 to PreS. However, little is known about the connectivity between the frontal brain regions and PreS or hippocampal formation. Therefore, we investigated the distribution of cells of the origins and terminals of the presubicular and hippocampal projections in the marmoset frontal brain regions using the retrograde and anterograde tracer cholera toxin B subunit. In cases of tracer injections into all layers of PreS, many neurons and terminals were labeled in the claustrum-endopiriform (Cl-En) complex almost entirely along the rostrocaudal axis. Even in cases where the injection site involved the superficial (not deep) layers of PreS, labeled neurons and terminals were distributed over a wide rostrocaudal range of the Cl-En complex, but their number and density were significantly lower than the whole-layer injection cases. In cases where the injection site was confined to the hippocampal formation, labeled cells and terminals were localized at a restricted portion of the Cl-En complex. Here, we demonstrate for what we believe to be the first time the strong, reciprocal connections of the Cl-En complex with PreS and projections from the Cl-En complex to the hippocampal regions (CA1 and the subiculum) in the marmoset. Our findings indicate that the Cl-En complex may exert a strong influence on the cortical and subcortical outputs from PreS and, in turn, the entire memory circuitry in the marmoset brain.


Subject(s)
Callithrix , Claustrum , Hippocampus , Neural Pathways , Animals , Callithrix/anatomy & histology , Hippocampus/anatomy & histology , Hippocampus/cytology , Neural Pathways/anatomy & histology , Neural Pathways/cytology , Male , Claustrum/anatomy & histology , Claustrum/physiology , Female , Neurons/cytology , Cholera Toxin/metabolism
2.
Neurosci Lett ; 836: 137894, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38997083

ABSTRACT

Reciprocal connections between the thalamus and the cortex are one of the most characteristic features of forebrain organization in mammals. To date, this circuit has been documented only in turtles. However, reptiles, including turtles, have an additional path from the dorsal thalamus to the telencephalon. This terminates in a pallial structure known as the dorsal ventricular ridge. Yet, no reciprocal connection from the dorsal ventricular ridge to thalamic nuclei has been uncovered. Since axons from the thalamus pass through the basal nuclei on route to the dorsal ventricular ridge, the basal nuclei might be a source of reciprocal connections. Accordingly, the location and distribution of neurons after retrograde tracer placement into the dorsal thalamus were examined. Retrogradely labeled neurons in the basal nuclei were indeed found. One possibility to explain this observation is that connections with the dorsal ventricular ridge are present during development but later pruned during embryogenesis.


Subject(s)
Neural Pathways , Turtles , Animals , Turtles/anatomy & histology , Neural Pathways/anatomy & histology , Neural Pathways/cytology , Thalamic Nuclei/anatomy & histology , Thalamic Nuclei/cytology , Neurons , Thalamus/anatomy & histology
3.
J Comp Neurol ; 532(6): e25629, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39031887

ABSTRACT

In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S, NPS) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray, then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known already about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts roles in threat response and circadian behavior.


Subject(s)
Parabrachial Nucleus , Animals , Parabrachial Nucleus/physiology , Parabrachial Nucleus/cytology , Mice , Efferent Pathways/cytology , Efferent Pathways/physiology , Mice, Transgenic , Neurons/metabolism , Male , Neuropeptides/metabolism , Neural Pathways/cytology
4.
J Comp Neurol ; 532(7): e25652, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38962882

ABSTRACT

Although the mammalian cerebral cortex is most often described as a hexalaminar structure, there are cortical areas (primary motor cortex) and species (elephants, cetaceans, and hippopotami), where a cytoarchitecturally indistinct, or absent, layer 4 is noted. Thalamocortical projections from the core, or first order, thalamic system terminate primarily in layers 4/inner 3. We explored the termination sites of core thalamocortical projections in cortical areas and in species where there is no cytoarchitecturally distinct layer 4 using the immunolocalization of vesicular glutamate transporter 2, a known marker of core thalamocortical axon terminals, in 31 mammal species spanning the eutherian radiation. Several variations from the canonical cortical column outline of layer 4 and core thalamocortical inputs were noted. In shrews/microchiropterans, layer 4 was present, but many core thalamocortical projections terminated in layer 1 in addition to layers 4 and inner 3. In primate primary visual cortex, the sublaminated layer 4 was associated with a specialized core thalamocortical projection pattern. In primate primary motor cortex, no cytoarchitecturally distinct layer 4 was evident and the core thalamocortical projections terminated throughout layer 3. In the African elephant, cetaceans, and river hippopotamus, no cytoarchitecturally distinct layer 4 was observed and core thalamocortical projections terminated primarily in inner layer 3 and less densely in outer layer 3. These findings are contextualized in terms of cortical processing, perception, and the evolutionary trajectory leading to an indistinct or absent cortical layer 4.


Subject(s)
Axons , Neocortex , Neural Pathways , Thalamus , Animals , Thalamus/cytology , Thalamus/anatomy & histology , Neocortex/cytology , Neocortex/anatomy & histology , Neural Pathways/cytology , Neural Pathways/anatomy & histology , Axons/physiology , Mammals/anatomy & histology , Vesicular Glutamate Transport Protein 2/metabolism , Species Specificity
5.
Nature ; 631(8020): 360-368, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926570

ABSTRACT

A deep understanding of how the brain controls behaviour requires mapping neural circuits down to the muscles that they control. Here, we apply automated tools to segment neurons and identify synapses in an electron microscopy dataset of an adult female Drosophila melanogaster ventral nerve cord (VNC)1, which functions like the vertebrate spinal cord to sense and control the body. We find that the fly VNC contains roughly 45 million synapses and 14,600 neuronal cell bodies. To interpret the output of the connectome, we mapped the muscle targets of leg and wing motor neurons using genetic driver lines2 and X-ray holographic nanotomography3. With this motor neuron atlas, we identified neural circuits that coordinate leg and wing movements during take-off. We provide the reconstruction of VNC circuits, the motor neuron atlas and tools for programmatic and interactive access as resources to support experimental and theoretical studies of how the nervous system controls behaviour.


Subject(s)
Connectome , Drosophila melanogaster , Motor Neurons , Nerve Tissue , Neural Pathways , Synapses , Animals , Female , Datasets as Topic , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Drosophila melanogaster/ultrastructure , Extremities/physiology , Extremities/innervation , Holography , Microscopy, Electron , Motor Neurons/cytology , Motor Neurons/physiology , Motor Neurons/ultrastructure , Movement , Muscles/innervation , Muscles/physiology , Nerve Tissue/anatomy & histology , Nerve Tissue/cytology , Nerve Tissue/physiology , Nerve Tissue/ultrastructure , Neural Pathways/cytology , Neural Pathways/physiology , Neural Pathways/ultrastructure , Synapses/physiology , Synapses/ultrastructure , Tomography, X-Ray , Wings, Animal/innervation , Wings, Animal/physiology
6.
Nature ; 631(8020): 369-377, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926579

ABSTRACT

Animal movement is controlled by motor neurons (MNs), which project out of the central nervous system to activate muscles1. MN activity is coordinated by complex premotor networks that facilitate the contribution of individual muscles to many different behaviours2-6. Here we use connectomics7 to analyse the wiring logic of premotor circuits controlling the Drosophila leg and wing. We find that both premotor networks cluster into modules that link MNs innervating muscles with related functions. Within most leg motor modules, the synaptic weights of each premotor neuron are proportional to the size of their target MNs, establishing a circuit basis for hierarchical MN recruitment. By contrast, wing premotor networks lack proportional synaptic connectivity, which may enable more flexible recruitment of wing steering muscles. Through comparison of the architecture of distinct motor control systems within the same animal, we identify common principles of premotor network organization and specializations that reflect the unique biomechanical constraints and evolutionary origins of leg and wing motor control.


Subject(s)
Connectome , Drosophila melanogaster , Extremities , Motor Neurons , Neural Pathways , Synapses , Wings, Animal , Animals , Female , Male , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Extremities/innervation , Extremities/physiology , Motor Neurons/physiology , Movement/physiology , Muscles/innervation , Muscles/physiology , Nerve Net/anatomy & histology , Nerve Net/cytology , Nerve Net/physiology , Neural Pathways/anatomy & histology , Neural Pathways/cytology , Neural Pathways/physiology , Synapses/physiology , Wings, Animal/innervation , Wings, Animal/physiology
7.
Sheng Li Xue Bao ; 76(2): 233-246, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38658373

ABSTRACT

The high-order cognitive and executive functions are necessary for an individual to survive. The densely bidirectional innervations between the medial prefrontal cortex (mPFC) and the mediodorsal thalamus (MD) play a vital role in regulating high-order functions. Pyramidal neurons in mPFC have been classified into several subclasses according to their morphological and electrophysiological properties, but the properties of the input-specific pyramidal neurons in mPFC remain poorly understood. The present study aimed to profile the morphological and electrophysiological properties of mPFC pyramidal neurons innervated by MD. In the past, the studies for characterizing the morphological and electrophysiological properties of neurons mainly relied on the electrophysiological recording of a large number of neurons and their morphologic reconstructions. But, it is a low efficient method for characterizing the circuit-specific neurons. The present study combined the advantages of traditional morphological and electrophysiological methods with machine learning to address the shortcomings of the past method, to establish a classification model for the morphological and electrophysiological properties of mPFC pyramidal neurons, and to achieve more accurate and efficient identification of the properties from a small size sample of neurons. We labeled MD-innervated pyramidal neurons of mPFC using the trans-synaptic neural circuitry tracing method and obtained their morphological properties using whole-cell patch-clamp recording and morphologic reconstructions. The results showed that the classification model established in the present study could predict the electrophysiological properties of MD-innervated pyramidal neurons based on their morphology. MD-innervated pyramidal neurons exhibit larger basal dendritic length but lower apical dendrite complexity compared to non-MD-innervated neurons in the mPFC. The morphological characteristics of the two subtypes (ET-1 and ET-2) of mPFC pyramidal neurons innervated by MD are different, with the apical dendrites of ET-1 neurons being longer and more complex than those of ET-2 neurons. These results suggest that the electrophysiological properties of MD- innervated pyramidal neurons within mPFC correlate with their morphological properties, indicating that the different roles of these two subclasses in local circuits within PFC, as well as in PFC-cortical/subcortical brain region circuits.


Subject(s)
Prefrontal Cortex , Pyramidal Cells , Pyramidal Cells/physiology , Pyramidal Cells/cytology , Prefrontal Cortex/physiology , Prefrontal Cortex/cytology , Animals , Rats , Mediodorsal Thalamic Nucleus/physiology , Mediodorsal Thalamic Nucleus/cytology , Male , Electrophysiological Phenomena , Neural Pathways/physiology , Neural Pathways/cytology , Machine Learning , Rats, Sprague-Dawley , Patch-Clamp Techniques
8.
Nature ; 624(7991): 355-365, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092919

ABSTRACT

Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.


Subject(s)
Brain , Epigenomics , Neural Pathways , Neurons , Animals , Mice , Amygdala , Brain/cytology , Brain/metabolism , Consensus Sequence , Datasets as Topic , Gene Expression Profiling , Hypothalamus/cytology , Mesencephalon/cytology , Neural Pathways/cytology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Regulatory Sequences, Nucleic Acid , Rhombencephalon/cytology , Single-Cell Analysis , Thalamus/cytology , Transcription Factors/metabolism
9.
Nature ; 624(7990): 130-137, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37993711

ABSTRACT

The termination of a meal is controlled by dedicated neural circuits in the caudal brainstem. A key challenge is to understand how these circuits transform the sensory signals generated during feeding into dynamic control of behaviour. The caudal nucleus of the solitary tract (cNTS) is the first site in the brain where many meal-related signals are sensed and integrated1-4, but how the cNTS processes ingestive feedback during behaviour is unknown. Here we describe how prolactin-releasing hormone (PRLH) and GCG neurons, two principal cNTS cell types that promote non-aversive satiety, are regulated during ingestion. PRLH neurons showed sustained activation by visceral feedback when nutrients were infused into the stomach, but these sustained responses were substantially reduced during oral consumption. Instead, PRLH neurons shifted to a phasic activity pattern that was time-locked to ingestion and linked to the taste of food. Optogenetic manipulations revealed that PRLH neurons control the duration of seconds-timescale feeding bursts, revealing a mechanism by which orosensory signals feed back to restrain the pace of ingestion. By contrast, GCG neurons were activated by mechanical feedback from the gut, tracked the amount of food consumed and promoted satiety that lasted for tens of minutes. These findings reveal that sequential negative feedback signals from the mouth and gut engage distinct circuits in the caudal brainstem, which in turn control elements of feeding behaviour operating on short and long timescales.


Subject(s)
Appetite Regulation , Brain Stem , Eating , Feedback, Physiological , Food , Satiation , Stomach , Appetite Regulation/physiology , Brain Stem/cytology , Brain Stem/physiology , Eating/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/metabolism , Prolactin-Releasing Hormone/metabolism , Satiation/physiology , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Stomach/physiology , Taste/physiology , Time Factors , Animals , Mice
10.
Science ; 378(6626): 1336-1343, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36548429

ABSTRACT

The primary motor cortex (M1) is involved in the control of voluntary movements and is extensively mapped in this capacity. Although the M1 is implicated in modulation of pain, the underlying circuitry and causal underpinnings remain elusive. We unexpectedly unraveled a connection from the M1 to the nucleus accumbens reward circuitry through a M1 layer 6-mediodorsal thalamus pathway, which specifically suppresses negative emotional valence and associated coping behaviors in neuropathic pain. By contrast, layer 5 M1 neurons connect with specific cell populations in zona incerta and periaqueductal gray to suppress sensory hypersensitivity without altering pain affect. Thus, the M1 employs distinct, layer-specific pathways to attune sensory and aversive-emotional components of neuropathic pain, which can be exploited for purposes of pain relief.


Subject(s)
Motor Cortex , Neural Pathways , Neuralgia , Motor Cortex/cytology , Motor Cortex/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Neuralgia/physiopathology , Neurons/physiology , Periaqueductal Gray/cytology , Periaqueductal Gray/physiology , Thalamus/cytology , Thalamus/physiology , Animals , Mice
11.
Nature ; 607(7919): 521-526, 2022 07.
Article in English | MEDLINE | ID: mdl-35794480

ABSTRACT

The direct and indirect pathways of the basal ganglia are classically thought to promote and suppress action, respectively1. However, the observed co-activation of striatal direct and indirect medium spiny neurons2 (dMSNs and iMSNs, respectively) has challenged this view. Here we study these circuits in mice performing an interval categorization task that requires a series of self-initiated and cued actions and, critically, a sustained period of dynamic action suppression. Although movement produced the co-activation of iMSNs and dMSNs in the sensorimotor, dorsolateral striatum (DLS), fibre photometry and photo-identified electrophysiological recordings revealed signatures of functional opponency between the two pathways during action suppression. Notably, optogenetic inhibition showed that DLS circuits were largely engaged to suppress-and not promote-action. Specifically, iMSNs on a given hemisphere were dynamically engaged to suppress tempting contralateral action. To understand how such regionally specific circuit function arose, we constructed a computational reinforcement learning model that reproduced key features of behaviour, neural activity and optogenetic inhibition. The model predicted that parallel striatal circuits outside the DLS learned the action-promoting functions, generating the temptation to act. Consistent with this, optogenetic inhibition experiments revealed that dMSNs in the associative, dorsomedial striatum, in contrast to those in the DLS, promote contralateral actions. These data highlight how opponent interactions between multiple circuit- and region-specific basal ganglia processes can lead to behavioural control, and establish a critical role for the sensorimotor indirect pathway in the proactive suppression of tempting actions.


Subject(s)
Corpus Striatum , Models, Neurological , Neural Inhibition , Neural Pathways , Neurons , Animals , Computer Simulation , Corpus Striatum/cytology , Corpus Striatum/physiology , Mice , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Optogenetics
12.
J Neurosci ; 42(6): 1068-1089, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34903572

ABSTRACT

The reuniens nucleus (RE) is situated at the most ventral position of the midline thalamus. In rats and mice RE is distinguished by bidirectional connections with the hippocampus and medial prefrontal cortex (mPFC) and a role in memory and cognition. In primates, many foundational questions pertaining to RE remain unresolved. We addressed these issues by investigating the composition of the rhesus monkey RE in both sexes by labeling for GABA, a marker of inhibitory neurons, and for the calcium-binding proteins parvalbumin (PV), calbindin (CB), and calretinin (CR), which label thalamic excitatory neurons that project to cortex. As in rats and mice, the macaque RE was mostly populated by CB and CR neurons, characteristic of matrix-dominant nuclei, and had bidirectional connections with hippocampus and mPFC area 25 (A25). Unlike rodents, we found GABAergic neurons in the monkey RE and a sparser but consistent population of core-associated thalamocortical PV neurons. RE had stronger connections with the basal amygdalar complex than in rats or mice. Amygdalar terminations were enriched with mitochondria and frequently formed successive synapses with the same postsynaptic structures, suggesting an active and robust pathway to RE. Significantly, hippocampal pathways formed multisynaptic complexes that uniquely involved excitatory projection neurons and dendrites of local inhibitory neurons in RE, extending this synaptic principle beyond sensory to high-order thalamic nuclei. Convergent pathways from hippocampus, A25, and amygdala in RE position it to flexibly coordinate activity for memory, cognition, and emotional context, which are disrupted in several psychiatric and neurologic diseases in humans.SIGNIFICANCE STATEMENT The primate RE is a central node for memory and cognition through connections with the hippocampus and mPFC. As in rats or mice, the primate RE is a matrix-dominant thalamic nucleus, suggesting signal traffic to the upper cortical layers. Unlike rats or mice, the primate RE contains inhibitory neurons, synaptic specializations with the hippocampal pathway, and robust connections with the amygdala, suggesting unique adaptations. Convergence of hippocampal, mPFC, and amygdalar pathways in RE may help unravel a circuit basis for binding diverse signals for conscious flexible behaviors and the synthesis of memory with affective significance in primates, whereas disruption of distinct circuit nodes may occur in psychiatric disorders in humans.


Subject(s)
Cognition/physiology , Emotions/physiology , Midline Thalamic Nuclei/physiology , Neural Pathways/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Axons/ultrastructure , Female , Hippocampus/cytology , Hippocampus/physiology , Macaca mulatta , Male , Midline Thalamic Nuclei/cytology , Neural Pathways/cytology
13.
J Neurosci ; 42(5): 749-761, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34887319

ABSTRACT

Neuronal remodeling after brain injury is essential for functional recovery. After unilateral cortical lesion, axons from the intact cortex ectopically project to the denervated midbrain, but the molecular mechanisms remain largely unknown. To address this issue, we examined gene expression profiles in denervated and intact mouse midbrains after hemispherectomy at early developmental stages using mice of either sex, when ectopic contralateral projection occurs robustly. The analysis showed that various axon growth-related genes were upregulated in the denervated midbrain, and most of these genes are reportedly expressed by glial cells. To identify the underlying molecules, the receptors for candidate upregulated molecules were knocked out in layer 5 projection neurons in the intact cortex, using the CRISPR/Cas9-mediated method, and axonal projection from the knocked-out cortical neurons was examined after hemispherectomy. We found that the ectopic projection was significantly reduced when integrin subunit ß three or neurotrophic receptor tyrosine kinase 2 (also known as TrkB) was knocked out. Overall, the present study suggests that denervated midbrain-derived glial factors contribute to lesion-induced remodeling of the cortico-mesencephalic projection via these receptors.SIGNIFICANCE STATEMENT After brain injury, compensatory neural circuits are established that contribute to functional recovery. However, little is known about the intrinsic mechanism that underlies the injury-induced remodeling. We found that after unilateral cortical ablation expression of axon-growth promoting factors is elevated in the denervated midbrain and is involved in the formation of ectopic axonal projection from the intact cortex. Evidence further demonstrated that these factors are expressed by astrocytes and microglia, which are activated in the denervated midbrain. Thus, our present study provides a new insight into the mechanism of lesion-induced axonal remodeling and further therapeutic strategies after brain injury.


Subject(s)
Brain Injuries/metabolism , Cerebral Cortex/metabolism , Hemispherectomy/trends , Mesencephalon/metabolism , Neuronal Plasticity/physiology , Animals , Brain Injuries/genetics , Brain Injuries/pathology , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Denervation/trends , Gene Knockout Techniques/methods , Mesencephalon/chemistry , Mesencephalon/cytology , Mice , Mice, Inbred ICR , Nerve Regeneration/physiology , Neural Pathways/cytology , Neural Pathways/metabolism , Organ Culture Techniques , Receptor, trkB/analysis , Receptor, trkB/genetics , Receptor, trkB/metabolism
14.
Cell Rep ; 37(8): 110031, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818557

ABSTRACT

Brain circuits are comprised of distinct interconnected neurons that are assembled by synaptic recognition molecules presented by defined pre- and post-synaptic neurons. This cell-cell recognition process is mediated by varying cellular adhesion molecules, including the latrophilin family of adhesion G-protein-coupled receptors. Focusing on parahippocampal circuitry, we find that latrophilin-2 (Lphn2; gene symbol ADGRL2) is specifically enriched in interconnected subregions of the medial entorhinal cortex (MEC), presubiculum (PrS), and parasubiculum (PaS). Retrograde viral tracing from the Lphn2-enriched region of the MEC reveals unique topographical patterning of inputs arising from the PrS and PaS that mirrors Lphn2 expression. Using a Lphn2 conditional knockout mouse model, we find that deletion of MEC Lphn2 expression selectively impairs retrograde viral labeling of inputs arising from the ipsilateral PrS. Combined with analysis of Lphn2 expression within the MEC, this study reveals Lphn2 to be selectively expressed by defined cell types and essential for MEC-PrS circuit connectivity.


Subject(s)
Entorhinal Cortex/physiology , Receptors, Peptide/genetics , Animals , Entorhinal Cortex/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Hippocampus/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/cytology , Neurons/physiology , Parahippocampal Gyrus/metabolism , Receptors, Peptide/metabolism
15.
Cell Rep ; 37(3): 109837, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686328

ABSTRACT

The selection of goal-directed behaviors is supported by neural circuits located within the frontal cortex. Frontal cortical afferents arise from multiple brain areas, yet the cell-type-specific targeting of these inputs is unclear. Here, we use monosynaptic retrograde rabies mapping to examine the distribution of afferent neurons targeting distinct classes of local inhibitory interneurons and excitatory projection neurons in mouse infralimbic frontal cortex. Interneurons expressing parvalbumin, somatostatin, or vasoactive intestinal peptide receive a large proportion of inputs from the hippocampus, while interneurons expressing neuron-derived neurotrophic factor receive a large proportion of inputs from thalamic regions. A similar dichotomy is present among the four different excitatory projection neurons. These results show a prominent bias among long-range hippocampal and thalamic afferent systems in their targeting to specific sets of frontal cortical neurons. Moreover, they suggest the presence of two distinct local microcircuits that control how different inputs govern frontal cortical information processing.


Subject(s)
Frontal Lobe/physiology , Hippocampus/physiology , Interneurons/physiology , Synapses/physiology , Thalamus/physiology , Animals , Behavior, Animal , Frontal Lobe/cytology , Frontal Lobe/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Interneurons/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neural Inhibition , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques , Parvalbumins/genetics , Parvalbumins/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Synapses/metabolism , Thalamus/cytology , Thalamus/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
16.
Cell Rep ; 37(3): 109855, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686344

ABSTRACT

The protein tau has been implicated in many brain disorders. In animal models, tau reduction suppresses epileptogenesis of diverse causes and ameliorates synaptic and behavioral abnormalities in various conditions associated with excessive excitation-inhibition (E/I) ratios. However, the underlying mechanisms are unknown. Global genetic ablation of tau in mice reduces the action potential (AP) firing and E/I ratio of pyramidal cells in acute cortical slices without affecting the excitability of these cells. Tau ablation reduces the excitatory inputs to inhibitory neurons, increases the excitability of these cells, and structurally alters their axon initial segments (AISs). In primary neuronal cultures subjected to prolonged overstimulation, tau ablation diminishes the homeostatic response of AISs in inhibitory neurons, promotes inhibition, and suppresses hypersynchrony. Together, these differential alterations in excitatory and inhibitory neurons help explain how tau reduction prevents network hypersynchrony and counteracts brain disorders causing abnormally increased E/I ratios.


Subject(s)
Excitatory Postsynaptic Potentials , Inhibitory Postsynaptic Potentials , Interneurons/metabolism , Neural Inhibition , Neural Pathways/metabolism , Pyramidal Cells/metabolism , Somatosensory Cortex/metabolism , tau Proteins/deficiency , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Cells, Cultured , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Female , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/cytology , Neuronal Plasticity , Somatosensory Cortex/cytology , Time Factors , tau Proteins/genetics
17.
J Neurosci ; 41(47): 9742-9755, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34649954

ABSTRACT

The subgenual (sgACC) and perigenual (pgACC) anterior cingulate are important afferents of the amygdala, with different cytoarchitecture, connectivity, and function. The sgACC is associated with arousal mechanisms linked to salient cues, whereas the pgACC is engaged in conflict decision-making, including in social contexts. After placing same-size, small volume tracer injections into sgACC and pgACC of the same hemisphere in male macaques, we examined anterogradely labeled fiber distribution to understand how these different functional systems communicate in the main amygdala nuclei at both mesocopic and cellular levels. The sgACC has broad-based termination patterns. In contrast, the pgACC has a more restricted pattern, which was always nested in sgACC terminals. Terminal overlap occurred in subregions of the accessory basal and basal nuclei, which we termed "hotspots." In triple-labeling confocal studies, the majority of randomly selected CaMKIIα-positive cells (putative amygdala glutamatergic neurons) in hotspots received dual contacts from the sgACC and pgACC. The ratio of dual contacts occurred over a surprisingly narrow range, suggesting a consistent, tight balance of afferent contacts on postsynaptic neurons. Large boutons, which are associated with greater synaptic strength, were ∼3 times more frequent on sgACC versus pgACC axon terminals in hotspots, consistent with a fast "driver" function. Together, the results reveal a nested interaction in which pgACC ("conflict/social monitoring") terminals converge with the broader sgACC ("salience") terminals at both the mesoscopic and cellular level. The presynaptic organization in hotspots suggests that shifts in arousal states can rapidly and flexibly influence decision-making functions in the amygdala.SIGNIFICANCE STATEMENT The subgenual (sgACC) and perigenual cingulate (pgACC) have distinct structural and functional characteristics and are important afferent modulators of the amygdala. The sgACC is critical for arousal, whereas the pgACC mediates conflict-monitoring, including in social contexts. Using dual tracer injections in the same monkey, we found that sgACC inputs broadly project in the main amygdala nuclei, whereas pgACC inputs were more restricted and nested in zones containing sgACC terminals (hotspots). The majority of CaMKIIα + (excitatory) amygdala neurons in hotspots received converging contacts, which were tightly balanced. pgACC and sgACC afferent streams are therefore highly interdependent in these specific amygdala subregions, permitting "internal arousal" states to rapidly shape responses of amygdala neurons involved in conflict and social monitoring networks.


Subject(s)
Amygdala/cytology , Gyrus Cinguli/cytology , Neural Pathways/cytology , Neurons, Afferent/cytology , Pyramidal Cells/cytology , Amygdala/physiology , Animals , Arousal/physiology , Gyrus Cinguli/physiology , Macaca fascicularis , Male , Neural Pathways/physiology , Neurons, Afferent/physiology , Pyramidal Cells/physiology
18.
J Neurosci ; 41(46): 9539-9560, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34642212

ABSTRACT

The lateral hypothalamic area (LHA) is a highly conserved brain region critical for maintaining physiological homeostasis and goal-directed behavior. LHA neurons that express melanin-concentrating hormone (MCH) are key regulators of arousal, energy balance, and motivated behavior. However, cellular and functional diversity among LHAMCH neurons is not well understood. Previous anatomic and molecular data suggest that LHAMCH neurons may be parsed into at least two distinct subpopulations, one of which is enriched in neurokinin-3 receptor (NK3R), the receptor for neurokinin B (NKB), encoded by the Tac2 gene. This tachykininergic ligand-receptor system has been implicated in reproduction, fear memory, and stress in other brain regions, but NKB interactions with LHAMCH neurons are poorly understood. We first identified how LHAMCH subpopulations may be distinguished anatomically and electrophysiologically. To dissect functional connectivity between NKB-expressing neurons and LHAMCH neurons, we used Cre-dependent retrograde and anterograde viral tracing in male Tac2-Cre mice and identified Tac2/EYFP+ neurons in the bed nucleus of the stria terminalis and central nucleus of the amygdala, the central extended amygdala, as major sources of NKB input onto LHAMCH neurons. In addition to innervating the LHA, these limbic forebrain NKB neurons also project to midbrain and brainstem targets. Finally, using a dual-virus approach, we found that optogenetic activation of these inputs in slices evokes GABA release onto a subset of LHAMCH neurons but lacked specificity for the NK3R+ subpopulation. Overall, these data define parallel tachykininergic/GABAergic limbic forebrain projections that are positioned to modulate multiple nodes of homeostatic and behavioral control.SIGNIFICANCE STATEMENT The LHA orchestrates fundamental behavioral states in the mammalian hypothalamus, including arousal, energy balance, memory, stress, and motivated behavior. The neuropeptide MCH defines one prominent population of LHA neurons, with multiple roles in the regulation of homeostatic behavior. Outstanding questions remain concerning the upstream inputs that control MCH neurons. We sought to define neurochemically distinct pathways in the mouse brain that may communicate with specific MCH neuron subpopulations using viral-based retrograde and anterograde neural pathway tracing and optogenetics in brain slices. Here, we identify a specific neuropeptide-defined forebrain circuit that makes functional synaptic connections with MCH neuron subpopulations. This work lays the foundation for further manipulating molecularly distinct neural circuits that modulate innate behavioral states.


Subject(s)
Central Amygdaloid Nucleus/cytology , Hypothalamic Area, Lateral/cytology , Neural Pathways/cytology , Neurons/cytology , Animals , Hypothalamic Hormones/metabolism , Male , Melanins/metabolism , Mice , Mice, Transgenic , Neural Pathways/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism
19.
Elife ; 102021 09 14.
Article in English | MEDLINE | ID: mdl-34519270

ABSTRACT

Synaptic connections in many brain circuits fluctuate, exhibiting substantial turnover and remodelling over hours to days. Surprisingly, experiments show that most of this flux in connectivity persists in the absence of learning or known plasticity signals. How can neural circuits retain learned information despite a large proportion of ongoing and potentially disruptive synaptic changes? We address this question from first principles by analysing how much compensatory plasticity would be required to optimally counteract ongoing fluctuations, regardless of whether fluctuations are random or systematic. Remarkably, we find that the answer is largely independent of plasticity mechanisms and circuit architectures: compensatory plasticity should be at most equal in magnitude to fluctuations, and often less, in direct agreement with previously unexplained experimental observations. Moreover, our analysis shows that a high proportion of learning-independent synaptic change is consistent with plasticity mechanisms that accurately compute error gradients.


Subject(s)
Behavior, Animal , Brain/physiology , Memory , Models, Neurological , Neural Pathways/physiology , Neuronal Plasticity , Neurons/physiology , Synaptic Transmission , Animals , Brain/cytology , Computer Simulation , Humans , Mice , Neural Pathways/cytology , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL