Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
EMBO Rep ; 22(12): e53200, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34633730

ABSTRACT

Astrocytes display extraordinary morphological complexity that is essential to support brain circuit development and function. Formin proteins are key regulators of the cytoskeleton; however, their role in astrocyte morphogenesis across diverse brain regions and neural circuits is unknown. Here, we show that loss of the formin protein Daam2 in astrocytes increases morphological complexity in the cortex and olfactory bulb, but elicits opposing effects on astrocytic calcium dynamics. These differential physiological effects result in increased excitatory synaptic activity in the cortex and increased inhibitory synaptic activity in the olfactory bulb, leading to altered olfactory behaviors. Proteomic profiling and immunoprecipitation experiments identify Slc4a4 as a binding partner of Daam2 in the cortex, and combined deletion of Daam2 and Slc4a4 restores the morphological alterations seen in Daam2 mutants. Our results reveal new mechanisms regulating astrocyte morphology and show that congruent changes in astrocyte morphology can differentially influence circuit function.


Subject(s)
Astrocytes , Microfilament Proteins/genetics , rho GTP-Binding Proteins/genetics , Formins , Morphogenesis , Olfactory Bulb/metabolism , Proteomics , Sodium-Bicarbonate Symporters
2.
Nature ; 538(7624): 253-256, 2016 Oct 13.
Article in English | MEDLINE | ID: mdl-27698417

ABSTRACT

Atypical food intake is a primary cause of obesity and other eating and metabolic disorders. Insight into the neural control of feeding has previously focused mainly on signalling mechanisms associated with the hypothalamus, the major centre in the brain that regulates body weight homeostasis. However, roles of non-canonical central nervous system signalling mechanisms in regulating feeding behaviour have been largely uncharacterized. Acetylcholine has long been proposed to influence feeding owing in part to the functional similarity between acetylcholine and nicotine, a known appetite suppressant. Nicotine is an exogenous agonist for acetylcholine receptors, suggesting that endogenous cholinergic signalling may play a part in normal physiological regulation of feeding. However, it remains unclear how cholinergic neurons in the brain regulate food intake. Here we report that cholinergic neurons of the mouse basal forebrain potently influence food intake and body weight. Impairment of cholinergic signalling increases food intake and results in severe obesity, whereas enhanced cholinergic signalling decreases food consumption. We found that cholinergic circuits modulate appetite suppression on downstream targets in the hypothalamus. Together our data reveal the cholinergic basal forebrain as a major modulatory centre underlying feeding behaviour.


Subject(s)
Appetite Regulation/physiology , Basal Forebrain/cytology , Basal Forebrain/physiology , Cholinergic Neurons/metabolism , Feeding Behavior/physiology , Satiety Response/physiology , Acetylcholine/metabolism , Animals , Body Weight/physiology , Cell Death , Choline O-Acetyltransferase/deficiency , Cholinergic Agonists , Cholinergic Neurons/pathology , Eating/physiology , Eating/psychology , Feeding Behavior/psychology , Female , Homeostasis , Hyperphagia/enzymology , Hyperphagia/genetics , Hyperphagia/pathology , Hypothalamus/cytology , Hypothalamus/physiology , Male , Mice , Mice, Knockout , Models, Neurological , Nicotine/metabolism , Obesity/enzymology , Obesity/genetics , Obesity/pathology , Receptors, Cholinergic/metabolism
3.
J Neurosci Res ; 98(8): 1605-1618, 2020 08.
Article in English | MEDLINE | ID: mdl-32426930

ABSTRACT

Astrocytes are the most abundant glial cell in the central nervous system. They modulate synaptic function through a variety of mechanisms, and yet remain relatively understudied with respect to overall neuronal circuit function. Exploiting the tractability of the mouse olfactory system, we manipulated astrocyte activity and examined how astrocytes modulate olfactory bulb responses. Toward this, we genetically targeted both astrocytes and neurons for in vivo widefield imaging of Ca2+ responses to odor stimuli. We found that astrocytes exhibited odor response maps that overlap with excitatory neuronal activity. By manipulating Ca2+ activity in astrocytes using chemical genetics we found that odor-evoked neuronal activity was reciprocally affected, suggesting that astrocyte activation inhibits neuronal odor responses. Subsequently, behavioral experiments revealed that astrocyte manipulations affect both odor detection threshold and discrimination, suggesting that astrocytes play an active role in olfactory sensory processing circuits. Together, these studies show that astrocyte calcium signaling contributes to olfactory behavior through modulation of sensory circuits.


Subject(s)
Astrocytes/physiology , Calcium Signaling/physiology , Olfactory Bulb/physiology , Animals , Astrocytes/metabolism , Calcium/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Odorants , Olfactory Bulb/metabolism , Olfactory Pathways/physiology
4.
J Neurosci ; 36(34): 8856-71, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27559168

ABSTRACT

UNLABELLED: Elucidating patterns of functional synaptic connectivity and deciphering mechanisms of how plasticity influences such connectivity is essential toward understanding brain function. In the mouse olfactory bulb (OB), principal neurons (mitral/tufted cells) make reciprocal connections with local inhibitory interneurons, including granule cells (GCs) and external plexiform layer (EPL) interneurons. Our current understanding of the functional connectivity between these cell types, as well as their experience-dependent plasticity, remains incomplete. By combining acousto-optic deflector-based scanning microscopy and genetically targeted expression of Channelrhodopsin-2, we mapped connections in a cell-type-specific manner between mitral cells (MCs) and GCs or between MCs and EPL interneurons. We found that EPL interneurons form broad patterns of connectivity with MCs, whereas GCs make more restricted connections with MCs. Using an olfactory associative learning paradigm, we found that these circuits displayed differential features of experience-dependent plasticity. Whereas reciprocal connectivity between MCs and EPL interneurons was nonplastic, the connections between GCs and MCs were dynamic and adaptive. Interestingly, experience-dependent plasticity of GCs occurred only in certain stages of neuronal maturation. We show that different interneuron subtypes form distinct connectivity maps and modes of experience-dependent plasticity in the OB, which may reflect their unique functional roles in information processing. SIGNIFICANCE STATEMENT: Deducing how specific interneuron subtypes contribute to normal circuit function requires understanding the dynamics of their connections. In the olfactory bulb (OB), diverse interneuron subtypes vastly outnumber principal excitatory cells. By combining acousto-optic deflector-based scanning microscopy, electrophysiology, and genetically targeted expression of Channelrhodopsin-2, we mapped the functional connectivity between mitral cells (MCs) and OB interneurons in a cell-type-specific manner. We found that, whereas external plexiform layer (EPL) interneurons show broadly distributed patterns of stable connectivity with MCs, adult-born granule cells show dynamic and plastic patterns of synaptic connectivity with task learning. Together, these findings reveal the diverse roles for interneuons within sensory circuits toward information learning and processing.


Subject(s)
Association Learning/physiology , Brain Mapping , Interneurons/physiology , Nerve Net/physiology , Neuronal Plasticity/physiology , Olfactory Bulb/cytology , Analysis of Variance , Animals , Channelrhodopsins , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/physiology , Interneurons/classification , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Light , Mice , Mice, Transgenic , Microscopy, Confocal , Neural Inhibition/genetics , Neural Inhibition/physiology , Neuronal Plasticity/genetics , Odorants , Optogenetics , Patch-Clamp Techniques , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Glia ; 63(10): 1840-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25946682

ABSTRACT

Wnt signaling plays an essential role in developmental and regenerative myelination of the CNS, therefore it is critical to understand how the factors associated with the various regulatory layers of this complex pathway contribute to these processes. Recently, Apcdd1 was identified as a negative regulator of proximal Wnt signaling, however its role in oligodendrocyte (OL) differentiation and reymelination in the CNS remain undefined. Analysis of Apcdd1 expression revealed dynamic expression during OL development, where its expression is upregulated during differentiation. Functional studies using ex vivo and in vitro OL systems revealed that Apcdd1 promotes OL differentiation, suppresses Wnt signaling, and associates with ß-catenin. Application of these findings to white matter injury (WMI) models revealed that Apcdd1 similarly promotes OL differentiation after gliotoxic injury in vivo and acute hypoxia ex vivo. Examination of Apcdd1 expression in white matter lesions from neonatal WMI and adult multiple sclerosis revealed its expression in subsets of oligodendrocyte (OL) precursors. These studies describe, for the first time, the role of Apcdd1 in OLs after WMI and reveal that negative regulators of the proximal Wnt pathway can influence regenerative myelination, suggesting a new therapeutic strategy for modulating Wnt signaling and stimulating repair after WMI.


Subject(s)
Cell Differentiation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Oligodendroglia/physiology , White Muscle Disease/pathology , Age Factors , Animals , Disease Models, Animal , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hypoxia/complications , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Lysophosphatidylcholines/toxicity , Membrane Proteins/genetics , Mice , Organ Culture Techniques , Spinal Cord/pathology , Stem Cells/metabolism , Stem Cells/physiology , White Muscle Disease/chemically induced , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
6.
eNeuro ; 11(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38383587

ABSTRACT

Obesity results from excessive caloric input associated with overeating and presents a major public health challenge. The hypothalamus has received significant attention for its role in governing feeding behavior and body weight homeostasis. However, extrahypothalamic brain circuits also regulate appetite and consumption by altering sensory perception, motivation, and reward. We recently discovered a population of basal forebrain cholinergic (BFc) neurons that regulate appetite suppression. Through viral tracing methods in the mouse model, we found that BFc neurons densely innervate the basolateral amygdala (BLA), a limbic structure involved in motivated behaviors. Using channelrhodopsin-assisted circuit mapping, we identified cholinergic responses in BLA neurons following BFc circuit manipulations. Furthermore, in vivo acetylcholine sensor and genetically encoded calcium indicator imaging within the BLA (using GACh3 and GCaMP, respectively) revealed selective response patterns of activity during feeding. Finally, through optogenetic manipulations in vivo, we found that increased cholinergic signaling from the BFc to the BLA suppresses appetite and food intake. Together, these data support a model in which cholinergic signaling from the BFc to the BLA directly influences appetite and feeding behavior.


Subject(s)
Basal Forebrain , Basolateral Nuclear Complex , Mice , Animals , Basolateral Nuclear Complex/physiology , Basal Forebrain/physiology , Cholinergic Neurons/physiology , Cholinergic Agents , Eating/physiology
7.
Stem Cells ; 30(10): 2140-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996827

ABSTRACT

Transsynaptic circuit tracing using genetically modified rabies virus (RV) is an emerging technology for identifying synaptic connections between neurons. Complementing this methodology, it is now possible to assay the basic molecular and cellular properties of neuronal lineages derived from embryonic stem cells (ESCs) in vitro, and these properties are under intense investigation toward devising cell replacement therapies. Here, we report the generation of a novel mouse ESC (mESC) line that harbors the genetic elements to allow RV-mediated transsynaptic circuit tracing in ESC-derived neurons and their synaptic networks. To facilitate transsynaptic tracing, we have engineered a new reporter allele by introducing cDNA encoding tdTomato, the Rabies-G glycoprotein, and the avian TVA receptor into the ROSA26 locus by gene targeting. We demonstrate high-efficiency differentiation of these novel mESCs into functional neurons, show their capacity to synaptically connect with primary neuronal cultures as evidenced by immunohistochemistry and electrophysiological recordings, and show their ability to act as source cells for presynaptic tracing of neuronal networks in vitro and in vivo. Together, our data highlight the potential for using genetically engineered stem cells to investigate fundamental mechanisms of synapse and circuit formation with unambiguous identification of presynaptic inputs onto neuronal populations of interest.


Subject(s)
Embryonic Stem Cells/metabolism , Neurons/metabolism , Proteins/metabolism , Synapses/physiology , Alleles , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Avian Proteins/genetics , Avian Proteins/metabolism , Biomarkers/metabolism , Birds , Cell Differentiation , Cell Engineering , Cell Line , Embryo, Mammalian , Embryonic Stem Cells/cytology , Founder Effect , Glycoproteins/genetics , Glycoproteins/metabolism , Mice , Nerve Net , Neurons/cytology , Primary Cell Culture , Proteins/genetics , RNA, Untranslated , Rabies virus/genetics , Receptors, Virus/genetics , Receptors, Virus/metabolism , Synapses/ultrastructure , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
8.
Nat Commun ; 12(1): 5230, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34471129

ABSTRACT

The role of transcription factors during astrocyte development and their subsequent effects on neuronal development has been well studied. Less is known about astrocytes contributions towards circuits and behavior in the adult brain. Astrocytes play important roles in synaptic development and modulation, however their contributions towards neuronal sensory function and maintenance of neuronal circuit architecture remain unclear. Here, we show that loss of the transcription factor Sox9 results in both anatomical and functional changes in adult mouse olfactory bulb (OB) astrocytes, affecting sensory processing. Indeed, astrocyte-specific deletion of Sox9 in the OB results in decreased odor detection thresholds and discrimination and it is associated with aberrant neuronal sensory response maps. At functional level, loss of astrocytic Sox9 impairs the electrophysiological properties of mitral and tufted neurons. RNA-sequencing analysis reveals widespread changes in the gene expression profiles of OB astrocytes. In particular, we observe reduced GLT-1 expression and consequential alterations in glutamate transport. Our findings reveal that astrocytes are required for physiological sensory processing and we identify astrocytic Sox9 as an essential transcriptional regulator of mature astrocyte function in the mouse OB.


Subject(s)
Astrocytes/metabolism , Olfactory Bulb/physiology , SOX9 Transcription Factor/metabolism , Sensation/physiology , Animals , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neurons , SOX9 Transcription Factor/genetics , Transcription Factors
9.
Elife ; 82019 05 10.
Article in English | MEDLINE | ID: mdl-31074744

ABSTRACT

Appetite is driven by nutritional state, environmental cues, mood, and reward pathways. Environmental cues strongly influence feeding behavior, as they can dramatically induce or diminish the drive to consume food despite homeostatic state. Here, we have uncovered an excitatory neuronal population in the basal forebrain that is activated by food-odor related stimuli, and potently drives hypophagia. Notably, we found that the basal forebrain directly integrates environmental sensory cues to govern feeding behavior, and that basal forebrain signaling, mediated through projections to the lateral hypothalamus, promotes selective avoidance of food and food-related stimuli. Together, these findings reveal a novel role for the excitatory basal forebrain in regulating appetite suppression through food avoidance mechanisms, highlighting a key function for this structure as a potent integrator of sensory information towards governing consummatory behaviors.


Subject(s)
Appetite Regulation , Basal Forebrain/physiology , Feeding Behavior , Nerve Net/physiology , Odorants , Olfactory Perception , Animals , Food , Mice
10.
Nat Commun ; 10(1): 3369, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358754

ABSTRACT

Inhibitory interneurons are integral to sensory processing, yet revealing their cell type-specific roles in sensory circuits remains an ongoing focus. To Investigate the mouse olfactory system, we selectively remove GABAergic transmission from a subset of olfactory bulb interneurons, EPL interneurons (EPL-INs), and assay odor responses from their downstream synaptic partners - tufted cells and mitral cells. Using a combination of in vivo electrophysiological and imaging analyses, we find that inactivating this single node of inhibition leads to differential effects in magnitude, reliability, tuning width, and temporal dynamics between the two principal neurons. Furthermore, tufted and not mitral cell responses to odor mixtures become more linearly predictable without EPL-IN inhibition. Our data suggest that olfactory bulb interneurons, through exerting distinct inhibitory functions onto their different synaptic partners, play a significant role in the processing of odor information.


Subject(s)
Interneurons/physiology , Neural Inhibition/physiology , Neurons/physiology , Olfactory Bulb/physiology , Olfactory Pathways/physiology , Animals , Interneurons/cytology , Interneurons/metabolism , Mice, Knockout , Mice, Transgenic , Neural Inhibition/genetics , Neurons/cytology , Neurons/metabolism , Odorants , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Smell , Synaptic Transmission/genetics , Synaptic Transmission/physiology
11.
Nat Neurosci ; 20(2): 189-199, 2017 02.
Article in English | MEDLINE | ID: mdl-28024159

ABSTRACT

Sensory maps are created by networks of neuronal responses that vary with their anatomical position, such that representations of the external world are systematically and topographically organized in the brain. Current understanding from studying excitatory maps is that maps are sculpted and refined throughout development and/or through sensory experience. Investigating the mouse olfactory bulb, where ongoing neurogenesis continually supplies new inhibitory granule cells into existing circuitry, we isolated the development of sensory maps formed by inhibitory networks. Using in vivo calcium imaging of odor responses, we compared functional responses of both maturing and established granule cells. We found that, in contrast to the refinement observed for excitatory maps, inhibitory sensory maps became broader with maturation. However, like excitatory maps, inhibitory sensory maps are sensitive to experience. These data describe the development of an inhibitory sensory map as a network, highlighting the differences from previously described excitatory maps.


Subject(s)
Nerve Net/growth & development , Neurogenesis/physiology , Neurons/physiology , Olfactory Bulb/growth & development , Smell/physiology , Animals , Female , Male , Mice, Transgenic , Odorants/analysis
12.
J Vis Exp ; (68): e50004, 2012 Oct 29.
Article in English | MEDLINE | ID: mdl-23128465

ABSTRACT

Elucidating patterns of neuronal connectivity has been a challenge for both clinical and basic neuroscience. Electrophysiology has been the gold standard for analyzing patterns of synaptic connectivity, but paired electrophysiological recordings can be both cumbersome and experimentally limiting. The development of optogenetics has introduced an elegant method to stimulate neurons and circuits, both in vitro(1) and in vivo(2,3). By exploiting cell-type specific promoter activity to drive opsin expression in discrete neuronal populations, one can precisely stimulate genetically defined neuronal subtypes in distinct circuits(4-6). Well described methods to stimulate neurons, including electrical stimulation and/or pharmacological manipulations, are often cell-type indiscriminate, invasive, and can damage surrounding tissues. These limitations could alter normal synaptic function and/or circuit behavior. In addition, due to the nature of the manipulation, the current methods are often acute and terminal. Optogenetics affords the ability to stimulate neurons in a relatively innocuous manner, and in genetically targeted neurons. The majority of studies involving in vivo optogenetics currently use a optical fiber guided through an implanted cannula(6,7); however, limitations of this method include damaged brain tissue with repeated insertion of an optical fiber, and potential breakage of the fiber inside the cannula. Given the burgeoning field of optogenetics, a more reliable method of chronic stimulation is necessary to facilitate long-term studies with minimal collateral tissue damage. Here we provide our modified protocol as a video article to complement the method effectively and elegantly described in Sparta et al.(8) for the fabrication of a fiber optic implant and its permanent fixation onto the cranium of anesthetized mice, as well as the assembly of the fiber optic coupler connecting the implant to a light source. The implant, connected with optical fibers to a solid-state laser, allows for an efficient method to chronically photostimulate functional neuronal circuitry with less tissue damage(9) using small, detachable, tethers. Permanent fixation of the fiber optic implants provides consistent, long-term in vivo optogenetic studies of neuronal circuits in awake, behaving mice(10) with minimal tissue damage.


Subject(s)
Brain/physiology , Fiber Optic Technology/methods , Prostheses and Implants , Animals , Brain/surgery , Fiber Optic Technology/instrumentation , Mice
SELECTION OF CITATIONS
SEARCH DETAIL