201
|
Abstract
Chronic alcohol consumption results in alcohol use disorder (AUD). Interestingly, however, sudden alcohol withdrawal (AW) after chronic alcohol exposure also leads to a devastating series of symptoms, referred to as alcohol withdrawal syndromes. One key feature of AW syndromes is to produce phenotypes that are opposite to AUD. For example, while the brain is characterized by a hypoactive state in the presence of alcohol, AW induces a hyperactive state, which is manifested as seizure expression. In this review, we discuss the idea that hippocampal neurogenesis and neural circuits play a key role in neuroadaptation and establishment of allostatic states in response to alcohol exposure and AW. The intrinsic properties of dentate granule cells (DGCs), and their contribution to the formation of a potent feedback inhibitory loop, endow the dentate gyrus with a "gate" function, which can limit the entry of excessive excitatory signals from the cortex into the hippocampus. We discuss the possibility that alcohol exposure and withdrawal disrupts structural development and circuitry integration of hippocampal newborn neurons, and that this altered neurogenesis impairs the gate function of the hippocampus. Failure of this gate function is expected to alter the ratio of excitatory to inhibitory (E/I) signals in the hippocampus and to induce seizure expression during AW. Recent functional studies have shown that specific activation and inhibition of hippocampal newborn DGCs are both necessary and sufficient for the expression of AW-associated seizures, further supporting the concept that neurogenesis-induced neuroadaptation is a critical target to understand and treat AUD and AW-associated seizures.
Collapse
Affiliation(s)
- Sreetama Basu
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Hoonkyo Suh
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
202
|
Mechanisms and plasticity of chemogenically induced interneuronal suppression of principal cells. Proc Natl Acad Sci U S A 2020; 118:2014157118. [PMID: 33372130 DOI: 10.1073/pnas.2014157118] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
How do firing patterns in a cortical circuit change when inhibitory neurons are excited? We virally expressed an excitatory designer receptor exclusively activated by a designer drug (Gq-DREADD) in all inhibitory interneuron types of the CA1 region of the hippocampus in the rat. While clozapine N-oxide (CNO) activation of interneurons suppressed firing of pyramidal cells, unexpectedly the majority of interneurons also decreased their activity. CNO-induced inhibition decreased over repeated sessions, which we attribute to long-term synaptic plasticity between interneurons and pyramidal cells. Individual interneurons did not display sustained firing but instead transiently enhanced their activity, interleaved with suppression of others. The power of the local fields in the theta band was unaffected, while power at higher frequencies was attenuated, likely reflecting reduced pyramidal neuron spiking. The incidence of sharp wave ripples decreased but the surviving ripples were associated with stronger population firing compared with the control condition. These findings demonstrate that DREADD activation of interneurons brings about both short-term and long-term circuit reorganization, which should be taken into account in the interpretation of chemogenic effects on behavior.
Collapse
|
203
|
VanDyke D, Wang W, Spangler JB. Innovative synthetic signaling technologies for immunotherapy. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020. [DOI: 10.1016/j.cobme.2020.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
204
|
Haaranen M, Schäfer A, Järvi V, Hyytiä P. Chemogenetic Stimulation and Silencing of the Insula, Amygdala, Nucleus Accumbens, and Their Connections Differentially Modulate Alcohol Drinking in Rats. Front Behav Neurosci 2020; 14:580849. [PMID: 33328918 PMCID: PMC7671963 DOI: 10.3389/fnbeh.2020.580849] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
The anterior insular cortex is hypothesized to represent interoceptive effects of drug reward in the service of goal-directed behavior. The insula is richly connected, but the insula circuitry in addiction remains poorly characterized. We examined the involvement of the anterior insula, amygdala, and nucleus accumbens, as well as the projections of the anterior insula to the central amygdala, basolateral amygdala (BLA), and nucleus accumbens core in voluntary alcohol drinking. We trained alcohol-preferring Alko Alcohol (AA) rats to drink alcohol during intermittent 2-h sessions. We then expressed excitatory or inhibitory designer receptors [designer receptors exclusively activated by designer drugs (DREADDs)] in the anterior insula, nucleus accumbens, or amygdala by means of adenovirus-mediated gene transfer and activated the DREADDs with clozapine-N-oxide (CNO) prior to the drinking sessions. Next, to examine the role of specific insula projections, we expressed FLEX-DREADDs in the efferent insula → nucleus accumbens core, insula → central amygdala, and insula → BLA projections by means of a retrograde AAV-Cre vector injected into the insula projection areas. In the anterior insula and amygdala, excitatory Gq-DREADDs significantly attenuated alcohol consumption. In contrast, in the nucleus accumbens, the Gq-DREADD stimulation increased alcohol drinking, and the inhibitory Gi-DREADDs suppressed it. The Gq-DREADDs expressed in the insula → nucleus accumbens core and insula → central amygdala projections increased alcohol intake, whereas inhibition of these projections had no effect. These data demonstrate that the anterior insula, along with the amygdala and nucleus accumbens, has a key role in controlling alcohol drinking by providing excitatory input to the central amygdala and nucleus accumbens to enhance alcohol reward.
Collapse
Affiliation(s)
- Mia Haaranen
- Department of Pharmacology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Annika Schäfer
- Department of Pharmacology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Vilja Järvi
- Department of Pharmacology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Petri Hyytiä
- Department of Pharmacology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
205
|
Obesity and Related Type 2 Diabetes: A Failure of the Autonomic Nervous System Controlling Gastrointestinal Function? GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The pandemic spread of obesity and type 2 diabetes is a serious health problem that cannot be contained with common therapies. At present, the most effective therapeutic tool is metabolic surgery, which substantially modifies the gastrointestinal anatomical structure. This review reflects the state of the art research in obesity and type 2 diabetes, describing the probable reason for their spread, how the various brain sectors are involved (with particular emphasis on the role of the vagal system controlling different digestive functions), and the possible mechanisms for the effectiveness of bariatric surgery. According to the writer’s interpretation, the identification of drugs that can modulate the activity of some receptor subunits of the vagal neurons and energy-controlling structures of the central nervous system (CNS), and/or specific physical treatment of cortical areas, could reproduce, non-surgically, the positive effects of metabolic surgery.
Collapse
|
206
|
Zhang J, Vardy E, Muise ES, Wang TM, Visconti R, Vadlamudi A, Pinto S, Peier AM. Utilizing Designed Receptors Exclusively Activated by Designer Drug Chemogenetic Tools to Identify Beneficial G Protein-Coupled Receptor Signaling for Fibrosis. J Pharmacol Exp Ther 2020; 375:357-366. [PMID: 32848074 DOI: 10.1124/jpet.120.000103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/04/2020] [Indexed: 12/21/2022] Open
Abstract
Fibrosis or accumulation of extracellular matrix is an evolutionarily conserved mechanism adopted by an organism as a response to chronic injury. Excessive fibrosis, however, leads to disruption of organ homeostasis and is a common feature of many chronic diseases. G protein-coupled receptors (GPCRs) are important cell signaling mediators and represent molecular targets for many Food and Drug Administration-approved drugs. To identify new targets for fibrosis, we used a synthetic GPCR system named designed receptors exclusively activated by designer drugs (DREADDs) to probe signaling pathways essential for fibrotic response. We found that upon expression in human lung fibroblasts, activation of Gq- and Gs-DREADDs abrogated the induction of TGFβ-induced fibrosis marker genes. Genome-wide transcriptome analysis identified dysregulation of multiple GPCRs in lung fibroblasts treated with TGFβ To investigate endogenous GPCR modulating TGFβ signaling, we selected 13 GPCRs that signal through Gq or Gs and activated them by using specific agonists. We examined the impact of each agonist and how activation of endogenous GPCR affects TGFβ signaling. Among the agonists examined, prostaglandin receptor agonists demonstrated the strongest inhibitory effect on fibrosis. Together, we have demonstrated that the DREADDs system is a valuable tool to identify beneficial GPCR signaling for fibrosis. This study in fibroblasts has served as a proof of concept and allowed us to further develop in vivo models for fibrosis GPCR discovery. SIGNIFICANCE STATEMENT: Fibrosis is the hallmark of many end-stage cardiometabolic diseases, and there is an unmet medical need to discover new antifibrotic therapies, reduce disease progression, and bring clinically meaningful efficacy to patients. Our work utilizes designed receptors exclusively activated by designer drug chemogenetic tools to identify beneficial GPCR signaling for fibrosis, providing new insights into GPCR drug discovery.
Collapse
Affiliation(s)
- Ji Zhang
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Eyal Vardy
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Eric S Muise
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Tzu-Ming Wang
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Richard Visconti
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Ashita Vadlamudi
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Shirly Pinto
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Andrea M Peier
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| |
Collapse
|
207
|
Enhanced Retrieval of Taste Associative Memory by Chemogenetic Activation of Locus Coeruleus Norepinephrine Neurons. J Neurosci 2020; 40:8367-8385. [PMID: 32994339 DOI: 10.1523/jneurosci.1720-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/28/2020] [Accepted: 09/21/2020] [Indexed: 01/08/2023] Open
Abstract
The ability of animals to retrieve memories stored in response to the environment is essential for behavioral adaptation. Norepinephrine (NE)-containing neurons in the brain play a key role in the modulation of synaptic plasticity underlying various processes of memory formation. However, the role of the central NE system in memory retrieval remains unclear. Here, we developed a novel chemogenetic activation strategy exploiting insect olfactory ionotropic receptors (IRs), termed "IR-mediated neuronal activation," and used it for selective stimulation of NE neurons in the locus coeruleus (LC). Drosophila melanogaster IR84a and IR8a subunits were expressed in LC NE neurons in transgenic mice. Application of phenylacetic acid (a specific ligand for the IR84a/IR8a complex) at appropriate doses induced excitatory responses of NE neurons expressing the receptors in both slice preparations and in vivo electrophysiological conditions, resulting in a marked increase of NE release in the LC nerve terminal regions (male and female). Ligand-induced activation of LC NE neurons enhanced the retrieval process of conditioned taste aversion without affecting taste sensitivity, general arousal state, and locomotor activity. This enhancing effect on taste memory retrieval was mediated, in part, through α1- and β-adrenergic receptors in the basolateral nucleus of the amygdala (BLA; male). Pharmacological inhibition of LC NE neurons confirmed the facilitative role of these neurons in memory retrieval via adrenergic receptors in the BLA (male). Our findings indicate that the LC NE system, through projections to the BLA, controls the retrieval process of taste associative memory.SIGNIFICANCE STATEMENT Norepinephrine (NE)-containing neurons in the brain play a key role in the modulation of synaptic plasticity underlying various processes of memory formation, but the role of the NE system in memory retrieval remains unclear. We developed a chemogenetic activation system based on insect olfactory ionotropic receptors and used it for selective stimulation of NE neurons in the locus coeruleus (LC) in transgenic mice. Ligand-induced activation of LC NE neurons enhanced the retrieval of conditioned taste aversion, which was mediated, in part, through adrenoceptors in the basolateral amygdala. Pharmacological blockade of LC activity confirmed the facilitative role of these neurons in memory retrieval. Our findings indicate that the LC-amygdala pathway plays an important role in the recall of taste associative memory.
Collapse
|
208
|
Yu X, Nagai J, Marti-Solano M, Soto JS, Coppola G, Babu MM, Khakh BS. Context-Specific Striatal Astrocyte Molecular Responses Are Phenotypically Exploitable. Neuron 2020; 108:1146-1162.e10. [PMID: 33086039 DOI: 10.1016/j.neuron.2020.09.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/13/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Astrocytes tile the central nervous system and are widely implicated in brain diseases, but the molecular mechanisms by which astrocytes contribute to brain disorders remain incompletely explored. By performing astrocyte gene expression analyses following 14 experimental perturbations of relevance to the striatum, we discovered that striatal astrocytes mount context-specific molecular responses at the level of genes, pathways, and upstream regulators. Through data mining, we also identified astrocyte pathways in Huntington's disease (HD) that were reciprocally altered with respect to the activation of striatal astrocyte G protein-coupled receptor (GPCR) signaling. Furthermore, selective striatal astrocyte stimulation of the Gi-GPCR pathway in vivo corrected several HD-associated astrocytic, synaptic, and behavioral phenotypes, with accompanying improvement of HD-associated astrocyte signaling pathways, including those related to synaptogenesis and neuroimmune functions. Overall, our data show that astrocytes are malleable, using context-specific responses that can be dissected molecularly and used for phenotypic benefit in brain disorders.
Collapse
Affiliation(s)
- Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Jun Nagai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Maria Marti-Solano
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Joselyn S Soto
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Giovanni Coppola
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
209
|
Echagarruga CT, Gheres KW, Norwood JN, Drew PJ. nNOS-expressing interneurons control basal and behaviorally evoked arterial dilation in somatosensory cortex of mice. eLife 2020; 9:e60533. [PMID: 33016877 PMCID: PMC7556878 DOI: 10.7554/elife.60533] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022] Open
Abstract
Cortical neural activity is coupled to local arterial diameter and blood flow. However, which neurons control the dynamics of cerebral arteries is not well understood. We dissected the cellular mechanisms controlling the basal diameter and evoked dilation in cortical arteries in awake, head-fixed mice. Locomotion drove robust arterial dilation, increases in gamma band power in the local field potential (LFP), and increases calcium signals in pyramidal and neuronal nitric oxide synthase (nNOS)-expressing neurons. Chemogenetic or pharmocological modulation of overall neural activity up or down caused corresponding increases or decreases in basal arterial diameter. Modulation of pyramidal neuron activity alone had little effect on basal or evoked arterial dilation, despite pronounced changes in the LFP. Modulation of the activity of nNOS-expressing neurons drove changes in the basal and evoked arterial diameter without corresponding changes in population neural activity.
Collapse
Affiliation(s)
| | - Kyle W Gheres
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Jordan N Norwood
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Patrick J Drew
- Bioengineering Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Departments of Engineering Science and Mechanics, Biomedical Engineering, and Neurosurgery, Pennsylvania State UniversityUniversity ParkUnited States
| |
Collapse
|
210
|
Schwabe MR, Taxier LR, Frick KM. It takes a neural village: Circuit-based approaches for estrogenic regulation of episodic memory. Front Neuroendocrinol 2020; 59:100860. [PMID: 32781195 PMCID: PMC7669700 DOI: 10.1016/j.yfrne.2020.100860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Cognitive behaviors, such as episodic memory formation, are complex processes involving coordinated activity in multiple brain regions. However, much of the research on hormonal regulation of cognition focuses on manipulation of one region at a time or provides a single snapshot of how a systemic treatment affects multiple brain regions without investigating how these regions might interact to mediate hormone effects. Here, we use estrogenic regulation of episodic memory as an example of how circuit-based approaches may be incorporated into future studies of hormones and cognition. We first review basic episodic memory circuitry, rapid mechanisms by which 17β-estradiol can alter circuit activity, and current knowledge about 17β-estradiol's effects on episodic memory. Next, we outline approaches that researchers can employ to consider circuit effects in their estrogen research and provide examples of how these methods have been used to examine hormonal regulation of memory and other behaviors.
Collapse
Affiliation(s)
- Miranda R Schwabe
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
211
|
Garcia-Marcos M, Parag-Sharma K, Marivin A, Maziarz M, Luebbers A, Nguyen LT. Optogenetic activation of heterotrimeric G-proteins by LOV2GIVe, a rationally engineered modular protein. eLife 2020; 9:60155. [PMID: 32936073 PMCID: PMC7515630 DOI: 10.7554/elife.60155] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
Heterotrimeric G-proteins are signal transducers involved in mediating the action of many natural extracellular stimuli and many therapeutic agents. Non-invasive approaches to manipulate the activity of G-proteins with high precision are crucial to understand their regulation in space and time. Here, we developed LOV2GIVe, an engineered modular protein that allows the activation of heterotrimeric G-proteins with blue light. This optogenetic construct relies on a versatile design that differs from tools previously developed for similar purposes, that is metazoan opsins, which are light-activated G-protein-coupled receptors (GPCRs). Instead, LOV2GIVe consists of the fusion of a G-protein activating peptide derived from a non-GPCR regulator of G-proteins to a small plant protein domain, such that light uncages the G-protein activating module. Targeting LOV2GIVe to cell membranes allowed for light-dependent activation of Gi proteins in different experimental systems. In summary, LOV2GIVe expands the armamentarium and versatility of tools available to manipulate heterotrimeric G-protein activity.
Collapse
Affiliation(s)
- Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Kshitij Parag-Sharma
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Arthur Marivin
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Marcin Maziarz
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Alex Luebbers
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Lien T Nguyen
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| |
Collapse
|
212
|
López-Ferreras L, Eerola K, Shevchouk OT, Richard JE, Nilsson FH, Jansson LE, Hayes MR, Skibicka KP. The supramammillary nucleus controls anxiety-like behavior; key role of GLP-1R. Psychoneuroendocrinology 2020; 119:104720. [PMID: 32563174 DOI: 10.1016/j.psyneuen.2020.104720] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 01/04/2023]
Abstract
Anxiety disorders are among the most prevalent categories of mental illnesses. The gut-brain axis, along with gastrointestinally-derived neuropeptides, like glucagon-like peptide-1 (GLP-1), are emerging as potential key regulators of emotionality, including anxiety behavior. However, the neuroanatomical substrates from which GLP-1 exerts its anxiogenic effect remain poorly characterized. Here we focus on a relatively new candidate nucleus, the supramammillary nucleus (SuM), located just caudal to the lateral hypothalamus and ventral to the ventral tegmental area. Our focus on the SuM is supported by previous data showing expression of GLP-1R mRNA throughout the SuM and activation of the SuM during anxiety-inducing behaviors in rodents. Data show that chemogenetic activation of neurons in the SuM results in an anxiolytic response in male and female rats. In contrast, selective activation of SuM GLP-1R, by microinjection of a GLP-1R agonist exendin-4 into the SuM resulted in potent anxiety-like behavior, measured in both open field and elevated plus maze tests in male and female rats. This anxiogenic effect of GLP-1R activation persisted after high-fat diet exposure. Importantly, reduction of GLP-1R expression in the SuM, by AAV-shRNA GLP-1R knockdown, resulted in a clear anxiolytic response; an effect only observed in female rats. Our data identify a new neural substrate for GLP-1 control of anxiety-like behavior and indicate that the SuM GLP-1R are sufficient for anxiogenesis in both sexes, but necessary only in females.
Collapse
Affiliation(s)
- L López-Ferreras
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden
| | - K Eerola
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Research Centre of Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Finland
| | - O T Shevchouk
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden
| | - J E Richard
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden
| | - F H Nilsson
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden
| | - L E Jansson
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden
| | - M R Hayes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - K P Skibicka
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
213
|
Ramirez V, Swain S, Murray K, Reardon C. Neural Immune Communication in the Control of Host-Bacterial Pathogen Interactions in the Gastrointestinal Tract. Infect Immun 2020; 88:e00928-19. [PMID: 32341116 PMCID: PMC7440759 DOI: 10.1128/iai.00928-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The orchestration of host immune responses to enteric bacterial pathogens is a complex process involving the integration of numerous signals, including from the nervous system. Despite the recent progress in understanding the contribution of neuroimmune interactions in the regulation of inflammation, the mechanisms and effects of this communication during enteric bacterial infection are only beginning to be characterized. As part of this neuroimmune communication, neurons specialized to detect painful or otherwise noxious stimuli can respond to bacterial pathogens. Highlighting the complexity of these systems, the immunological consequences of sensory neuron activation can be either host adaptive or maladaptive, depending on the pathogen and organ system. These are but one of many types of neuroimmune circuits, with the vagus nerve and sympathetic innervation of numerous organs now known to modulate immune cell function and therefore dictate immunological outcomes during health and disease. Here, we review the evidence for neuroimmune communication in response to bacterial pathogens, and then discuss the consequences to host morbidity and mortality during infection of the gastrointestinal tract.
Collapse
Affiliation(s)
- Valerie Ramirez
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Samantha Swain
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Kaitlin Murray
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Colin Reardon
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
214
|
Wang L, Zhu L, Meister J, Bone DBJ, Pydi SP, Rossi M, Wess J. Use of DREADD Technology to Identify Novel Targets for Antidiabetic Drugs. Annu Rev Pharmacol Toxicol 2020; 61:421-440. [PMID: 32746768 DOI: 10.1146/annurev-pharmtox-030220-121042] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
G protein-coupled receptors (GPCRs) form a superfamily of plasma membrane receptors that couple to four major families of heterotrimeric G proteins, Gs, Gi, Gq, and G12. GPCRs represent excellent targets for drug therapy. Since the individual GPCRs are expressed by many different cell types, the in vivo metabolic roles of a specific GPCR expressed by a distinct cell type are not well understood. The development of designer GPCRs known as DREADDs (designer receptors exclusively activated by a designer drug) that selectively couple to distinct classes of heterotrimeric G proteins has greatly facilitated studies in this area. This review focuses on the use of DREADD technology to explore the physiological and pathophysiological roles of distinct GPCR/G protein cascades in several metabolically important cell types. The novel insights gained from these studies should stimulate the development of GPCR-based treatments for major metabolic diseases such as type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Derek B J Bone
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Mario Rossi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| |
Collapse
|
215
|
Binning W, Hogan-Cann AE, Yae Sakae D, Maksoud M, Ostapchenko V, Al-Onaizi M, Matovic S, Lu WY, Prado MAM, Inoue W, Prado VF. Chronic hM3Dq signaling in microglia ameliorates neuroinflammation in male mice. Brain Behav Immun 2020; 88:791-801. [PMID: 32434046 DOI: 10.1016/j.bbi.2020.05.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
Microglia express muscarinic G protein-coupled receptors (GPCRs) that sense cholinergic activity and are activated by acetylcholine to potentially regulate microglial functions. Knowledge about how distinct types of muscarinic GPCR signaling regulate microglia function in vivo is still poor, partly due to the fact that some of these receptors are also present in astrocytes and neurons. We generated mice expressing the hM3Dq Designer Receptor Exclusively Activated by Designer Drugs (DREADD) selectively in microglia to investigate the role of muscarinic M3Gq-linked signaling. We show that activation of hM3Dq using clozapine N-oxide (CNO) elevated intracellular calcium levels and increased phagocytosis of FluoSpheres by microglia in vitro. Interestingly, whereas acute treatment with CNO increased synthesis of cytokine mRNA, chronic treatment attenuated LPS-induced cytokine mRNA changes in the brain. No effect of CNO on cytokine expression was observed in DREADD-negative mice. Interestingly, CNO activation of M3Dq in microglia was able to attenuate LPS-mediated decrease in social interactions. These results suggest that chronic activation of M3 muscarinic receptors (the hM3Dq progenitor) in microglia, and potentially other Gq-coupled GPCRs, can trigger an inflammatory-like response that preconditions microglia to decrease their response to further immunological challenges. Our results indicate that hM3Dq can be a useful tool to modulate neuroinflammation and study microglial immunological memory in vivo, which may be applicable for manipulations of neuroinflammation in neurodegenerative and psychiatric diseases.
Collapse
Affiliation(s)
- William Binning
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Aja E Hogan-Cann
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Diana Yae Sakae
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Matthew Maksoud
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Valeriy Ostapchenko
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Mohammed Al-Onaizi
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Sara Matovic
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Wei-Yang Lu
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Marco A M Prado
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Anatomy & Cell Biology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada.
| | - Wataru Inoue
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada.
| | - Vania F Prado
- Program in Neuroscience, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Department of Anatomy & Cell Biology, University of Western Ontario, London, Ontario N6A 5K8, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario N6A 5K8, Canada.
| |
Collapse
|
216
|
Marciante AB, Farmer GE, Cunningham JT. G q DREADD activation of CaMKIIa MnPO neurons stimulates nitric oxide activity. J Neurophysiol 2020; 124:591-609. [PMID: 32697679 PMCID: PMC7500373 DOI: 10.1152/jn.00239.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 11/22/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) modify cellular activity following administration of the exogenous ligand clozapine-N-oxide (CNO). However, some reports indicate CNO may have off-target effects. The current studies investigate the use of Gq DREADDs in CaMKIIa-expressing neurons in the median preoptic nucleus (MnPO). Male Sprague-Dawley rats (250 g) anesthetized with isoflurane were stereotaxically microinjected in the MnPO with the Gq DREADD (AAV5-CaMKIIa-HM3D-mCherry) or control virus (AAV5-CaMKIIa-mCherry). Following a 2-wk recovery, rats were used for either immunohistochemical Fos analysis or in vitro patch-clamp electrophysiology. In Gq DREADD-injected rats, CNO induced significant increases in Fos staining in the MnPO and in regions that receive direct or indirect projections from the MnPO. In electrophysiological studies, CNO depolarized and augmented firing frequency in both Gq DREADD-positive neurons (Gq DREADD) as well as unlabeled MnPO neurons in slices from Gq DREADD-injected rats (Gq DREADDx). Gq DREADDx neurons also displayed increases in spontaneous postsynaptic current (sPSC) frequency in response to CNO. Additionally, CaMKIIa-positive MnPO neurons, which also express nitric oxide synthase (NOS), were treated with Nω-nitro-l-arginine (l-NNA; competitive inhibitor of NOS) and hemoglobin (NO scavenger) to assess the role of NO in Gq DREADDx neuron recruitment. Both l-NNA and hemoglobin blocked CNO-induced effects in Gq DREADDx neurons without affecting Gq DREADD neurons. These findings indicate that Gq DREADD-mediated activation of CaMKIIa/NOS expressing neurons in the MnPO can influence the activity of neighboring neurons. Future studies utilizing the use of Gq DREADDs will need to consider the potential recruitment of additional cell populations.NEW & NOTEWORTHY Rats were injected in the median preoptic nucleus (MnPO) with either an adeno-associated virus (AAV) and excitatory (Gq) designer receptor exclusively activated by designer drugs (DREADD) construct or a control AAV. In the Gq DREADD-injected rats only, clozapine-N-oxide (CNO) increased Fos staining in the MnPO and its targets and increased neuron action potential frequency. In electrophysiology experiments with slices with DREADD cells, unlabeled cells were activated and this was likely due to nitric oxide release by the DREADD cells.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Fort Worth, Texas
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Fort Worth, Texas
| | | |
Collapse
|
217
|
Wang X, Zhang Y, Wang X, Dai J, Hua R, Zeng S, Li H. Anxiety-related cell-type-specific neural circuits in the anterior-dorsal bed nucleus of the stria terminalis. Sci Bull (Beijing) 2020; 65:1203-1216. [PMID: 36659150 DOI: 10.1016/j.scib.2020.03.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/15/2020] [Accepted: 02/01/2020] [Indexed: 01/21/2023]
Abstract
The bed nucleus of the stria terminalis (BNST) plays a critical role in regulating anxiety, yet the involved specific cell types and their connections functioning in anxiety-related behaviors remains elusive. Here we identified two cell subpopulations-corticotropin-releasing hormone-positive (CRH+) and protein kinase C-δ-positive (PKC-δ+) neurons-each displayed discrete emotionally valenced behaviors in the anterior-dorsal BNST (adBNST). Using whole-cell patch-clamp recordings and virus-assisted circuit tracing techniques, we delineated the local and long-range connectivity networks in a cell-type-specific manner. The results show that the CRH+ and PKC-δ+ neurons received inputs from similar brain regions and exhibited significant differences in the downstream projection density. In addition, in vivo calcium imaging as well as gain- and loss-of-function studies characterized the physiological response properties and the functional heterogeneities in modulating anxiety, further suggesting the similarity and individuality between the two adBNST cell types. These results provide novel insights into the circuit architecture of adBNST neurons underlying the functionally specific neural pathways that relate to anxiety disorders.
Collapse
Affiliation(s)
- Xinxin Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yongsheng Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xu Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiaqi Dai
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ruifang Hua
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haohong Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; Ministry of Education Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
218
|
Taylor MJ, Ullenbruch MR, Frucci EC, Rege J, Ansorge MS, Gomez-Sanchez CE, Begum S, Laufer E, Breault DT, Rainey WE. Chemogenetic activation of adrenocortical Gq signaling causes hyperaldosteronism and disrupts functional zonation. J Clin Invest 2020; 130:83-93. [PMID: 31738186 DOI: 10.1172/jci127429] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/18/2019] [Indexed: 02/04/2023] Open
Abstract
The mineralocorticoid aldosterone is produced in the adrenal zona glomerulosa (ZG) under the control of the renin-angiotensin II (AngII) system. Primary aldosteronism (PA) results from renin-independent production of aldosterone and is a common cause of hypertension. PA is caused by dysregulated localization of the enzyme aldosterone synthase (Cyp11b2), which is normally restricted to the ZG. Cyp11b2 transcription and aldosterone production are predominantly regulated by AngII activation of the Gq signaling pathway. Here, we report the generation of transgenic mice with Gq-coupled designer receptors exclusively activated by designer drugs (DREADDs) specifically in the adrenal cortex. We show that adrenal-wide ligand activation of Gq DREADD receptors triggered disorganization of adrenal functional zonation, with induction of Cyp11b2 in glucocorticoid-producing zona fasciculata cells. This result was consistent with increased renin-independent aldosterone production and hypertension. All parameters were reversible following termination of DREADD-mediated Gq signaling. These findings demonstrate that Gq signaling is sufficient for adrenocortical aldosterone production and implicate this pathway in the determination of zone-specific steroid production within the adrenal cortex. This transgenic mouse also provides an inducible and reversible model of hyperaldosteronism to investigate PA therapeutics and the mechanisms leading to the damaging effects of aldosterone on the cardiovascular system.
Collapse
Affiliation(s)
- Matthew J Taylor
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew R Ullenbruch
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Emily C Frucci
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mark S Ansorge
- The Sackler Institute for Developmental Psychobiology, Columbia University, New York, New York, USA
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Salma Begum
- Department of Obstetrics, Gynecology and Women's Health, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Edward Laufer
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - David T Breault
- Department of Pediatrics, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
219
|
Kuiper LB, Lucas KA, Mai V, Coolen LM. Enhancement of Drug Seeking Following Drug Taking in a Sexual Context Requires Anterior Cingulate Cortex Activity in Male Rats. Front Behav Neurosci 2020; 14:87. [PMID: 32670029 PMCID: PMC7330085 DOI: 10.3389/fnbeh.2020.00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/08/2020] [Indexed: 11/13/2022] Open
Abstract
Individual variance in vulnerability to develop addictions is influenced by social factors. Specifically, drug-taking in a sexual context appears to enhance further drug-seeking behavior in human users, as these users identify the effects of drugs to enhance sexual pleasure as a primary reason for continued drug use. Methamphetamine (Meth) is commonly used in this context. Similarly, male rats that self-administered Meth immediately followed by sexual behavior display enhanced drug-seeking behavior, including attenuation of extinction and increased reinstatement to seeking of Meth-associated cues. Hence, drug-taking in a sexual context enhances vulnerability for addiction. However, the neural mechanisms by which this occurs are unknown. Here the hypothesis was tested that medial prefrontal cortex is essential for this effect of Meth and sex when experienced concurrently. First it was shown that CaMKII neurons in the anterior cingulate area (ACA) were co-activated by both Meth and sex. Next, chemogenetic inactivation of ACA CaMKII cells using AAV5-CaMKIIa-hM4Di-mCherry was shown not to affect Meth-induced locomotor activity or sexual behavior. Subsequently, chemogenetic inactivation of ACA CaMKII neurons during Meth self-administration followed by sexual behavior was shown to prevent the effects of Meth and sex on enhanced reinstatement of Meth-seeking but did not affect enhanced drug-seeking during extinction tests. These results indicate that ACA CaMKII cell activation during exposure to Meth in a sexual context plays an essential role in the subsequent enhancement of drug-seeking during reinstatement tests.
Collapse
Affiliation(s)
- Lindsey B Kuiper
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kathryn A Lucas
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Vy Mai
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Lique M Coolen
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States.,Brain Health Research Institute, Kent State University, Kent, OH, United States.,Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
220
|
Neuronal regulation of the blood-brain barrier and neurovascular coupling. Nat Rev Neurosci 2020; 21:416-432. [PMID: 32636528 DOI: 10.1038/s41583-020-0322-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2020] [Indexed: 12/31/2022]
Abstract
To continuously process neural activity underlying sensation, movement and cognition, the CNS requires a homeostatic microenvironment that is not only enriched in nutrients to meet its high metabolic demands but that is also devoid of toxins that might harm the sensitive neural tissues. This highly regulated microenvironment is made possible by two unique features of CNS vasculature absent in the peripheral organs. First, the blood-blood barrier, which partitions the circulating blood from the CNS, acts as a gatekeeper to facilitate the selective trafficking of substances between the blood and the parenchyma. Second, neurovascular coupling ensures that, following local neural activation, regional blood flow is increased to quickly supply more nutrients and remove metabolic waste. Here, we review how neural and vascular activity act on one another with regard to these two properties.
Collapse
|
221
|
Sadahiro M, Demars MP, Burman P, Yevoo P, Zimmer A, Morishita H. Activation of Somatostatin Interneurons by Nicotinic Modulator Lypd6 Enhances Plasticity and Functional Recovery in the Adult Mouse Visual Cortex. J Neurosci 2020; 40:5214-5227. [PMID: 32467358 PMCID: PMC7329312 DOI: 10.1523/jneurosci.1373-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 01/27/2023] Open
Abstract
The limitation of plasticity in the adult brain impedes functional recovery later in life from brain injury or disease. This pressing clinical issue may be resolved by enhancing plasticity in the adult brain. One strategy for triggering robust plasticity in adulthood is to reproduce one of the hallmark physiological events of experience-dependent plasticity observed during the juvenile critical period: to rapidly reduce the activity of parvalbumin (PV)-expressing interneurons and disinhibit local excitatory neurons. This may be achieved through the enhancement of local inhibitory inputs, particularly those of somatostatin (SST)-expressing interneurons. However, to date the means for manipulating SST interneurons for enhancing cortical plasticity in the adult brain are not known. We show that SST interneuron-selective overexpression of Lypd6, an endogenous nicotinic signaling modulator, enhances ocular dominance plasticity in the adult primary visual cortex (V1). Lypd6 overexpression mediates a rapid experience-dependent increase in the visually evoked activity of SST interneurons as well as a simultaneous reduction in PV interneuron activity and disinhibition of excitatory neurons. Recapitulating this transient activation of SST interneurons using chemogenetics similarly enhanced V1 plasticity. Notably, we show that SST-selective Lypd6 overexpression restores visual acuity in amblyopic mice that underwent early long-term monocular deprivation. Our data in both male and female mice reveal selective modulation of SST interneurons and a putative downstream circuit mechanism as an effective method for enhancing experience-dependent cortical plasticity as well as functional recovery in adulthood.SIGNIFICANCE STATEMENT The decline of cortical plasticity after closure of juvenile critical period consolidates neural circuits and behavior, but this limits functional recovery from brain diseases and dysfunctions in later life. Here we show that activation of cortical somatostatin (SST) interneurons by Lypd6, an endogenous modulator of nicotinic acetylcholine receptors, enhances experience-dependent plasticity and recovery from amblyopia in adulthood. This manipulation triggers rapid reduction of PV interneuron activity and disinhibition of excitatory neurons, which are known hallmarks of cortical plasticity during juvenile critical periods. Our study demonstrates modulation of SST interneurons by Lypd6 to achieve robust levels of cortical plasticity in the adult brain and may provide promising targets for restoring brain function in the event of brain trauma or disease.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/physiology
- Animals
- Dominance, Ocular/genetics
- Evoked Potentials, Visual/genetics
- Evoked Potentials, Visual/physiology
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/physiology
- Immunohistochemistry
- Interneurons/physiology
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Neuronal Plasticity/genetics
- Neuronal Plasticity/physiology
- Phosphatidylinositols/pharmacology
- Receptors, Nicotinic/genetics
- Recovery of Function/genetics
- Somatostatin/physiology
- Vision, Monocular/genetics
- Vision, Monocular/physiology
- Visual Acuity/genetics
- Visual Cortex/physiology
Collapse
Affiliation(s)
- Masato Sadahiro
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Michael P Demars
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Poromendro Burman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Priscilla Yevoo
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
222
|
Sorrentino A, Michel T. Redox à la carte: Novel chemogenetic models of heart failure. Br J Pharmacol 2020; 177:3162-3167. [PMID: 32368791 PMCID: PMC7312266 DOI: 10.1111/bph.15093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022] Open
Abstract
Many current animal models of heart failure are hampered by intrinsic methodological complexities, while other models yield only a subtle cardiac phenotype even after prolonged in vivo treatments. A new 'chemogenetic' animal model of heart failure reproduces a critical characteristic shared by many disease states that lead to heart failure in humans: an increase in redox stress in the heart. This 'chemogenetic' approach exploits a recombinant yeast enzyme that can be dynamically and specifically activated in vivo to generate the ROS hydrogen peroxide (H2 O2 ) in cardiac myocytes. Redox stress can be rapidly, selectively and reversibly manipulated by chemogenetic generation of ROS in cardiac myocytes, yielding a new model of dilated cardiomyopathy. Treatment of animals with the angiotensin receptor antagonist valsartan promotes recovery of ventricular function and resolution of adverse cardiac remodelling. This mini-review discusses in vivo chemogenetic approaches to manipulate and analyse oxidative stress in the heart.
Collapse
Affiliation(s)
- Andrea Sorrentino
- Cardiovascular Division, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Thomas Michel
- Cardiovascular Division, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
223
|
Chen LY, Liang J, Fei F, Ruan YP, Cheng HM, Wang Y, Chen Z, Xu CL. Pharmaco-genetic inhibition of pyramidal neurons retards hippocampal kindling-induced epileptogenesis. CNS Neurosci Ther 2020; 26:1111-1120. [PMID: 32596972 PMCID: PMC7564188 DOI: 10.1111/cns.13434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
AIMS Pharmaco-genetics emerges as a new promising approach for epileptic seizures. Whether it can modulate epileptogenesis is still unknown. METHODS Here, parvalbumin neurons and pyramidal neurons of the seizure focus were transfected with engineered excitatory Gq-coupled human muscarinic receptor hM3Dq and engineered inhibitory Gi-coupled human muscarinic receptor hM4Di, respectively. And their therapeutic value in mouse hippocampal kindling-induced epileptogenesis was tested. RESULTS Pharmaco-genetic activating parvalbumin neurons limitedly retarded the progression of behavioral seizure stage and afterdischarge duration (ADD) during epileptogenesis induced by kindling. Activating parvalbumin neurons delayed seizure development only in the early stage, but accelerated it in late stages. On the contrary, pharmaco-genetic inhibiting pyramidal neurons robustly retarded the progression of seizure stages and ADDs, which greatly delayed seizure development in both early and late stages. Although both pharmaco-genetic therapeutics efficiently alleviated the severity of acute kindling-induced seizures, pharmaco-genetic inhibiting pyramidal neurons were able to reverse the enhanced synaptic plasticity during epileptogenesis, compared with that of pharmaco-genetic activating parvalbumin neurons. CONCLUSION Our results demonstrated that pharmaco-genetic inhibiting pyramidal neurons retard hippocampal kindling-induced epileptogenesis and reverse the enhanced synaptic plasticity during epileptogenesis, compared with that of pharmaco-genetic activating parvalbumin neurons. It suggests that pharmaco-genetics targeting pyramidal neurons may be a promising treatment for epileptogenesis.
Collapse
Affiliation(s)
- Li-Ying Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiao Liang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Department of Neurology, Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Fei
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ye-Ping Ruan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - He-Ming Cheng
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Department of Neurology, Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ceng-Lin Xu
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Department of Neurology, Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
224
|
Saika F, Matsuzaki S, Kobayashi D, Ideguchi Y, Nakamura TY, Kishioka S, Kiguchi N. Chemogenetic Regulation of CX3CR1-Expressing Microglia Using Gi-DREADD Exerts Sex-Dependent Anti-Allodynic Effects in Mouse Models of Neuropathic Pain. Front Pharmacol 2020; 11:925. [PMID: 32636748 PMCID: PMC7318895 DOI: 10.3389/fphar.2020.00925] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/08/2020] [Indexed: 12/25/2022] Open
Abstract
Despite growing evidence suggesting that spinal microglia play an important role in the molecular mechanism underlying experimental neuropathic pain (NP) in male rodents, evidence regarding the sex-dependent role of these microglia in NP is insufficient. In this study, we evaluated the effects of microglial regulation on NP using Gi-designer receptors exclusively activated by designer drugs (Gi-DREADD) driven by the microglia-specific Cx3cr1 promoter. For the Cre-dependent expression of human Gi-coupled M4 muscarinic receptors (hM4Di) in CX3C chemokine receptor 1-expressing (CX3CR1+) cells, R26-LSL-hM4Di-DREADD mice were crossed with CX3CR1-Cre mice. Mouse models of NP were generated by partial sciatic nerve ligation (PSL) and treatment with anti-cancer agent paclitaxel (PTX) or oxaliplatin (OXA), and mechanical allodynia was evaluated using the von Frey test. Immunohistochemistry revealed that hM4Di was specifically expressed on Iba1+ microglia, but not on astrocytes or neurons in the spinal dorsal horn of CX3CR1-hM4Di mice. PSL-induced mechanical allodynia was significantly attenuated by systemic (intraperitoneal, i.p.) administration of 10 mg/kg of clozapine N-oxide (CNO), a hM4Di-selective ligand, in male CX3CR1-hM4Di mice. The mechanical threshold in naive CX3CR1-hM4Di mice was not altered by i.p. administration of CNO. Consistently, local (intrathecal, i.t.) administration of CNO (20 nmol) significantly relieved PSL-induced mechanical allodynia in male CX3CR1-hM4Di mice. However, neither i.p. nor i.t. administration of CNO affected PSL-induced mechanical allodynia in female CX3CR1-hM4Di mice. Both i.p. and i.t. administration of CNO relieved PTX-induced mechanical allodynia in male CX3CR1-hM4Di mice, and a limited effect of i.p. CNO was observed in female CX3CR1-hM4Di mice. Unlike PTX-induced allodynia, OXA-induced mechanical allodynia was slightly improved, but not significantly relieved, by i.p. administration of CNO in both male and female CX3CR1-hM4Di mice. These results suggest that spinal microglia can be regulated by Gi-DREADD and support the notion that CX3CR1+ spinal microglia play sex-dependent roles in nerve injury-induced NP; however, their roles may vary among different models of NP.
Collapse
Affiliation(s)
- Fumihiro Saika
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Shinsuke Matsuzaki
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Daichi Kobayashi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan.,Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuya Ideguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Tomoe Y Nakamura
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan.,Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care, Wakayama, Japan
| | - Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
225
|
In Vivo Attenuation of M-Current Suppression Impairs Consolidation of Object Recognition Memory. J Neurosci 2020; 40:5847-5856. [PMID: 32554550 DOI: 10.1523/jneurosci.0348-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/08/2020] [Accepted: 06/06/2020] [Indexed: 11/21/2022] Open
Abstract
The M-current is a low voltage-activated potassium current generated by neuronal Kv7 channels. A prominent role of the M-current is to a create transient increase of neuronal excitability in response to neurotransmitters through the suppression of this current. Accordingly, M-current suppression is assumed to be involved in higher brain functions including learning and memory. However, there is little evidence supporting such a role to date. To address this gap, we examined behavioral tasks to assess learning and memory in homozygous Kv7.2 knock-in mice, Kv7.2(S559A), which show reduced M-current suppression while maintaining a normal basal M-current activity in neurons. We found that Kv7.2(S559A) mice had normal object location memory and contextual fear memory, but impaired long-term object recognition memory. Furthermore, short-term memory for object recognition was intact in Kv7.2(S559A) mice. The deficit in long-term object recognition memory was restored by the administration of a selective Kv7 channel inhibitor, XE991, when delivered during the memory consolidation phase. Lastly, c-Fos induction 2 h after training in Kv7.2(S559A) mice was normal in the hippocampus, which corresponds to intact object location memory, but was reduced in the perirhinal cortex, which corresponds to impaired long-term object recognition memory. Together, these results support the overall conclusion that M-current suppression is important for memory consolidation of specific types of memories.SIGNIFICANCE STATEMENT Dynamic regulation of neuronal excitation is a fundamental mechanism for information processing in the brain, which is mediated by changes in synaptic transmissions or by changes in ion channel activity. Some neurotransmitters can facilitate action potential firing by suppression of a low voltage-activated potassium current, M-current. We demonstrate that M-current suppression is critical for establishment of long-term object recognition memory, but is not required for establishment of hippocampus-dependent location memory or contextual memory. This study suggests that M-current suppression is important for stable encoding of specific types of memories.
Collapse
|
226
|
Shchepinova MM, Hanyaloglu AC, Frost GS, Tate EW. Chemical biology of noncanonical G protein-coupled receptor signaling: Toward advanced therapeutics. Curr Opin Chem Biol 2020; 56:98-110. [PMID: 32446179 DOI: 10.1016/j.cbpa.2020.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling membrane proteins, are the target of more than 30% of the drugs on the market. Recently, it has become clear that GPCR functions are far more multidimensional than previously thought, with multiple noncanonical aspects coming to light, including biased, oligomeric, and compartmentalized signaling. These additional layers of functional selectivity greatly expand opportunities for advanced therapeutic interventions, but the development of new chemical biology tools is absolutely required to improve our understanding of noncanonical GPCR regulation and pave the way for future drugs. In this opinion, we highlight the most notable examples of chemical and chemogenetic tools addressing new paradigms in GPCR signaling, discuss their promises and limitations, and explore future directions.
Collapse
Affiliation(s)
- Maria M Shchepinova
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Dept. Surgery and Cancer, Imperial College, London, UK
| | - Gary S Frost
- Department of Medicine, Faculty of Medicine, Nutrition and Dietetic Research Group, Imperial College, London, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| |
Collapse
|
227
|
Anterior insula stimulation suppresses appetitive behavior while inducing forebrain activation in alcohol-preferring rats. Transl Psychiatry 2020; 10:150. [PMID: 32424183 PMCID: PMC7235223 DOI: 10.1038/s41398-020-0833-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
The anterior insular cortex plays a key role in the representation of interoceptive effects of drug and natural rewards and their integration with attention, executive function, and emotions, making it a potential target region for intervention to control appetitive behaviors. Here, we investigated the effects of chemogenetic stimulation or inhibition of the anterior insula on alcohol and sucrose consumption. Excitatory or inhibitory designer receptors (DREADDs) were expressed in the anterior insula of alcohol-preferring rats by means of adenovirus-mediated gene transfer. Rats had access to either alcohol or sucrose solution during intermittent sessions. To characterize the brain network recruited by chemogenetic insula stimulation we measured brain-wide activation patterns using pharmacological magnetic resonance imaging (phMRI) and c-Fos immunohistochemistry. Anterior insula stimulation by the excitatory Gq-DREADDs significantly attenuated both alcohol and sucrose consumption, whereas the inhibitory Gi-DREADDs had no effects. In contrast, anterior insula stimulation failed to alter locomotor activity or deprivation-induced water drinking. phMRI and c-Fos immunohistochemistry revealed downstream activation of the posterior insula and medial prefrontal cortex, as well as of the mediodorsal thalamus and amygdala. Our results show the critical role of the anterior insula in regulating reward-directed behavior and delineate an insula-centered functional network associated with the effects of insula stimulation. From a translational perspective, our data demonstrate the therapeutic potential of circuit-based interventions and suggest that potentiation of insula excitability with neuromodulatory methods, such as repetitive transcranial magnetic stimulation (rTMS), could be useful in the treatment of alcohol use disorders.
Collapse
|
228
|
Singh G, Inoue A, Gutkind JS, Russell RB, Raimondi F. PRECOG: PREdicting COupling probabilities of G-protein coupled receptors. Nucleic Acids Res 2020; 47:W395-W401. [PMID: 31143927 PMCID: PMC6602504 DOI: 10.1093/nar/gkz392] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/13/2019] [Accepted: 05/01/2019] [Indexed: 01/08/2023] Open
Abstract
G-protein coupled receptors (GPCRs) control multiple physiological states by transducing a multitude of extracellular stimuli into the cell via coupling to intra-cellular heterotrimeric G-proteins. Deciphering which G-proteins couple to each of the hundreds of GPCRs present in a typical eukaryotic organism is therefore critical to understand signalling. Here, we present PRECOG (precog.russelllab.org): a web-server for predicting GPCR coupling, which allows users to: (i) predict coupling probabilities for GPCRs to individual G-proteins instead of subfamilies; (ii) visually inspect the protein sequence and structural features that are responsible for a particular coupling; (iii) suggest mutations to rationally design artificial GPCRs with new coupling properties based on predetermined coupling features.
Collapse
Affiliation(s)
- Gurdeep Singh
- CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert B Russell
- CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Francesco Raimondi
- CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| |
Collapse
|
229
|
Guida F, De Gregorio D, Palazzo E, Ricciardi F, Boccella S, Belardo C, Iannotta M, Infantino R, Formato F, Marabese I, Luongo L, de Novellis V, Maione S. Behavioral, Biochemical and Electrophysiological Changes in Spared Nerve Injury Model of Neuropathic Pain. Int J Mol Sci 2020; 21:ijms21093396. [PMID: 32403385 PMCID: PMC7246983 DOI: 10.3390/ijms21093396] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023] Open
Abstract
Neuropathic pain is a pathological condition induced by a lesion or disease affecting the somatosensory system, with symptoms like allodynia and hyperalgesia. It has a multifaceted pathogenesis as it implicates several molecular signaling pathways involving peripheral and central nervous systems. Affective and cognitive dysfunctions have been reported as comorbidities of neuropathic pain states, supporting the notion that pain and mood disorders share some common pathogenetic mechanisms. The understanding of these pathophysiological mechanisms requires the development of animal models mimicking, as far as possible, clinical neuropathic pain symptoms. Among them, the Spared Nerve Injury (SNI) model has been largely characterized in terms of behavioral and functional alterations. This model is associated with changes in neuronal firing activity at spinal and supraspinal levels, and induces late neuropsychiatric disorders (such as anxious-like and depressive-like behaviors, and cognitive impairments) comparable to an advanced phase of neuropathy. The goal of this review is to summarize current findings in preclinical research, employing the SNI model as a tool for identifying pathophysiological mechanisms of neuropathic pain and testing pharmacological agent.
Collapse
Affiliation(s)
- Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
- Correspondence: (F.G.); (S.M.)
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montréal, QC H3A1A1, Canada;
| | - Enza Palazzo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Flavia Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Monica Iannotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Federica Formato
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Vito de Novellis
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Naples, 80138 Naples, Italy; (E.P.); (F.R.); (S.B.); (C.B.); (M.I.); (R.I.); (F.F.); (I.M.); (L.L.); (V.d.N.)
- Correspondence: (F.G.); (S.M.)
| |
Collapse
|
230
|
Wang Y, Wang Y, Xu C, Wang S, Tan N, Chen C, Chen L, Wu X, Fei F, Cheng H, Lin W, Qi Y, Chen B, Liang J, Zhao J, Xu Z, Guo Y, Zhang S, Li X, Zhou Y, Duan S, Chen Z. Direct Septum-Hippocampus Cholinergic Circuit Attenuates Seizure Through Driving Somatostatin Inhibition. Biol Psychiatry 2020; 87:843-856. [PMID: 31987494 DOI: 10.1016/j.biopsych.2019.11.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/24/2019] [Accepted: 11/12/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Previous studies indicated the involvement of cholinergic neurons in seizure; however, the specific role of the medial septum (MS)-hippocampus cholinergic circuit in temporal lobe epilepsy (TLE) has not yet been completely elucidated. METHODS In the current study, we used magnetic resonance imaging and diffusion tensor imaging to characterize the pathological change of the MS-hippocampus circuit in 42 patients with TLE compared with 22 healthy volunteers. Using optogenetics and chemogenetics, combined with in vivo or in vitro electrophysiology and retrograde rabies virus tracing, we revealed a direct MS-hippocampus cholinergic circuit that potently attenuates seizure through driving somatostatin inhibition in animal TLE models. RESULTS We found that patients with TLE with hippocampal sclerosis showed a decrease of neuronal fiber connectivity of the MS-hippocampus compared with healthy people. In the mouse TLE model, MS cholinergic neurons ceased firing during hippocampal seizures. Optogenetic and chemogenetic activation of MS cholinergic neurons (but not glutamatergic or GABAergic [gamma-aminobutyric acidergic] neurons) significantly attenuated hippocampal seizures, while specific inhibition promoted hippocampal seizures. Electrophysiology combined with modified rabies virus tracing studies showed that direct (but not indirect) MS-hippocampal cholinergic projections mediated the antiseizure effect by preferentially targeting hippocampal GABAergic neurons. Furthermore, chemogenetic inhibition of hippocampal somatostatin-positive (rather than parvalbumin-positive) subtype of GABAergic neurons reversed the antiseizure effect of the MS-hippocampus cholinergic circuit, which was mimicked by activating somatostatin-positive neurons. CONCLUSIONS These findings underscore the notable antiseizure role of the direct cholinergic MS-hippocampus circuit in TLE through driving the downstream somatostatin effector. This may provide a better understanding of the changes of the seizure circuit and the precise spatiotemporal control of epilepsy.
Collapse
Affiliation(s)
- Ying Wang
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Cenglin Xu
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuang Wang
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Tan
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cong Chen
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liying Chen
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaohua Wu
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Fei
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Heming Cheng
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wenkai Lin
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yingbei Qi
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bin Chen
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiao Liang
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junli Zhao
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhenghao Xu
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Guo
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shihong Zhang
- Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoming Li
- Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China; Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yudong Zhou
- Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China; Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shumin Duan
- Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China; Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Institute of Neuroscience, Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China; Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
231
|
Abstract
The search for more effective treatments for depression is a long-standing primary objective in both psychiatry and translational neuroscience. From initial models centered on neurochemical deficits, such as the monoamine hypothesis, research toward this goal has shifted toward a focus on network and circuit models to explain how key nodes in the limbic system and beyond interact to produce persistent shifts in affective states. To build these models, researchers have turned to two complementary approaches: neuroimaging studies in human patients (and their healthy counterparts) and neurophysiology studies in animal models, facilitated in large part by optogenetic and chemogenetic techniques. As the authors discuss, functional neuroimaging studies in humans have included largely task-oriented experiments, which have identified brain regions differentially activated during processing of affective stimuli, and resting-state functional MRI experiments, which have identified brain-wide networks altered in depressive states. Future work in this area will build on a multisite approach, assembling large data sets across diverse populations, and will also leverage the statistical power afforded by longitudinal imaging studies in patient samples. Translational studies in rodents have used optogenetic and chemogenetic tools to identify not just nodes but also connections within the networks of the limbic system that are both critical and permissive for the expression of motivated behavior and affective phenotypes. Future studies in this area will exploit mesoscale imaging and multisite electrophysiology recordings to construct network models with cell-type specificity and high statistical power, identifying candidate circuit and molecular pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Timothy Spellman
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, New York
| | - Conor Liston
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, New York
| |
Collapse
|
232
|
Cheloha RW, Fischer FA, Woodham AW, Daley E, Suminski N, Gardella TJ, Ploegh HL. Improved GPCR ligands from nanobody tethering. Nat Commun 2020; 11:2087. [PMID: 32350260 PMCID: PMC7190724 DOI: 10.1038/s41467-020-15884-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Antibodies conjugated to bioactive compounds allow targeted delivery of therapeutics to cell types of choice based on that antibody's specificity. Here we develop a new type of conjugate that consists of a nanobody and a peptidic ligand for a G protein-coupled receptor (GPCR), fused via their C-termini. We address activation of parathyroid hormone receptor-1 (PTHR1) and improve the signaling activity and specificity of otherwise poorly active N-terminal peptide fragments of PTH by conjugating them to nanobodies (VHHs) that recognize PTHR1. These C-to-C conjugates show biological activity superior to that of the parent fragment peptide in vitro. In an exploratory experiment in mice, a VHH-PTH peptide conjugate showed biological activity, whereas the corresponding free peptide did not. The lead conjugate also possesses selectivity for PTHR1 superior to that of PTH(1-34). This design approach, dubbed "conjugation of ligands and antibodies for membrane proteins" (CLAMP), can yield ligands with high potency and specificity.
Collapse
Affiliation(s)
- Ross W Cheloha
- Boston Children's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
| | - Fabian A Fischer
- Boston Children's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
| | - Andrew W Woodham
- Boston Children's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
| | - Eileen Daley
- Massachusetts General Hospital and Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Naomi Suminski
- Boston Children's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
| | - Thomas J Gardella
- Massachusetts General Hospital and Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA.
| | - Hidde L Ploegh
- Boston Children's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
233
|
Weiss AR, Liguore WA, Domire JS, Button D, McBride JL. Intra-striatal AAV2.retro administration leads to extensive retrograde transport in the rhesus macaque brain: implications for disease modeling and therapeutic development. Sci Rep 2020; 10:6970. [PMID: 32332773 PMCID: PMC7181773 DOI: 10.1038/s41598-020-63559-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/01/2020] [Indexed: 11/09/2022] Open
Abstract
Recently, AAV2.retro, a new capsid variant capable of efficient retrograde transport in brain, was generated in mice using a directed evolution approach. However, it remains unclear to what degree transport will be recapitulated in the substantially larger and more complex nonhuman primate (NHP) brain. Here, we compared the biodistribution of AAV2.retro with its parent serotype, AAV2, in adult macaques following delivery into the caudate and putamen, brain regions which comprise the striatum. While AAV2 transduction was primarily limited to the injected brain regions, AAV2.retro transduced cells in the striatum and in dozens of cortical and subcortical regions with known striatal afferents. We then evaluated the capability of AAV2.retro to deliver disease-related gene cargo to biologically-relevant NHP brain circuits by packaging a fragment of human mutant HTT, the causative gene mutation in Huntington’s disease. Following intra-striatal delivery, pathological mHTT-positive protein aggregates were distributed widely among cognitive, motor, and limbic cortico-basal ganglia circuits. Together, these studies demonstrate strong retrograde transport of AAV2.retro in NHP brain, highlight its utility in developing novel NHP models of brain disease and suggest its potential for querying circuit function and delivering therapeutic genes in the brain, particularly where treating dysfunctional circuits, versus single brain regions, is warranted.
Collapse
Affiliation(s)
- Alison R Weiss
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - William A Liguore
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - Jacqueline S Domire
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - Dana Button
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA. .,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, USA. .,Department of Neurology, Oregon Health and Science University, Portland, USA.
| |
Collapse
|
234
|
Pan-Vazquez A, Wefelmeyer W, Gonzalez Sabater V, Neves G, Burrone J. Activity-Dependent Plasticity of Axo-axonic Synapses at the Axon Initial Segment. Neuron 2020; 106:265-276.e6. [PMID: 32109363 PMCID: PMC7181187 DOI: 10.1016/j.neuron.2020.01.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 12/06/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022]
Abstract
The activity-dependent rules that govern the wiring of GABAergic interneurons are not well understood. Chandelier cells (ChCs) are a type of GABAergic interneuron that control pyramidal cell output through axo-axonic synapses that target the axon initial segment. In vivo imaging of ChCs during development uncovered a narrow window (P12-P18) over which axons arborized and formed connections. We found that increases in the activity of either pyramidal cells or individual ChCs during this temporal window result in a reversible decrease in axo-axonic connections. Voltage imaging of GABAergic transmission at the axon initial segment (AIS) showed that axo-axonic synapses were depolarizing during this period. Identical manipulations of network activity in older mice (P40-P46), when ChC synapses are inhibitory, resulted instead in an increase in axo-axonic synapses. We propose that the direction of ChC synaptic plasticity follows homeostatic rules that depend on the polarity of axo-axonic synapses.
Collapse
Affiliation(s)
- Alejandro Pan-Vazquez
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Winnie Wefelmeyer
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Victoria Gonzalez Sabater
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Guilherme Neves
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| | - Juan Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
| |
Collapse
|
235
|
Sharif B, Ase AR, Ribeiro-da-Silva A, Séguéla P. Differential Coding of Itch and Pain by a Subpopulation of Primary Afferent Neurons. Neuron 2020; 106:940-951.e4. [PMID: 32298640 DOI: 10.1016/j.neuron.2020.03.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/21/2019] [Accepted: 03/20/2020] [Indexed: 12/21/2022]
Abstract
Itch and pain are distinct unpleasant sensations that can be triggered from the same receptive fields in the skin, raising the question of how pruriception and nociception are coded and discriminated. Here, we tested the multimodal capacity of peripheral first-order neurons, focusing on the genetically defined subpopulation of mouse C-fibers that express the chloroquine receptor MrgprA3. Using optogenetics, chemogenetics, and pharmacology, we assessed the behavioral effects of their selective stimulation in a wide variety of conditions. We show that metabotropic Gq-linked stimulation of these C-afferents, through activation of native MrgprA3 receptors or DREADDs, evokes stereotypical pruriceptive rather than nocifensive behaviors. In contrast, fast ionotropic stimulation of these same neurons through light-gated cation channels or native ATP-gated P2X3 channels predominantly evokes nocifensive rather than pruriceptive responses. We conclude that C-afferents display intrinsic multimodality, and we provide evidence that optogenetic and chemogenetic interventions on the same neuronal populations can drive distinct behavioral outputs.
Collapse
Affiliation(s)
- Behrang Sharif
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada; Alan Edwards Centre for Research on Pain, Montreal, QC H3A 0G1, Canada; Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Ariel R Ase
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada; Alan Edwards Centre for Research on Pain, Montreal, QC H3A 0G1, Canada
| | - Alfredo Ribeiro-da-Silva
- Alan Edwards Centre for Research on Pain, Montreal, QC H3A 0G1, Canada; Department of Pharmacology & Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Philippe Séguéla
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada; Alan Edwards Centre for Research on Pain, Montreal, QC H3A 0G1, Canada.
| |
Collapse
|
236
|
Chen P, Lou S, Huang ZH, Wang Z, Shan QH, Wang Y, Yang Y, Li X, Gong H, Jin Y, Zhang Z, Zhou JN. Prefrontal Cortex Corticotropin-Releasing Factor Neurons Control Behavioral Style Selection under Challenging Situations. Neuron 2020; 106:301-315.e7. [DOI: 10.1016/j.neuron.2020.01.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 10/31/2019] [Accepted: 01/24/2020] [Indexed: 02/07/2023]
|
237
|
Chisholm A, Iannuzzi J, Rizzo D, Gonzalez N, Fortin É, Bumbu A, Batallán Burrowes AA, Chapman CA, Shalev U. The role of the paraventricular nucleus of the thalamus in the augmentation of heroin seeking induced by chronic food restriction. Addict Biol 2020; 25:e12708. [PMID: 30623532 DOI: 10.1111/adb.12708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 01/12/2023]
Abstract
Drug addiction is a chronic disorder that is characterized by compulsive drug seeking and involves cycling between periods of compulsive drug use, abstinence, and relapse. In both human addicts and animal models of addiction, chronic food restriction has been shown to increase rates of relapse. Previously, our laboratory has demonstrated a robust increase in drug seeking following a period of withdrawal in chronically food-restricted rats compared with sated rats. To date, the neural mechanisms that mediate the effect of chronic food restriction on drug seeking have not been elucidated. However, the paraventricular nucleus of the thalamus (PVT) appears to be a promising target to investigate. The objective of the current study was to examine the role of the PVT in the augmentation of heroin seeking induced by chronic food restriction. Male Long-Evans rats were trained to self-administer heroin for 10 days. Rats were then removed from the training chambers and experienced a 14-day withdrawal period with either unrestricted (sated) or mildly restricted (FDR) access to food. On day 14, rats underwent a 1-hour heroin-seeking test under extinction conditions, during which neural activity in the PVT was either inhibited or increased using pharmacological or chemogenetic approaches. Unexpectedly, inhibition of the PVT did not alter heroin seeking in food-restricted or sated rats, while enhancing neural activity in the PVT-attenuated heroin seeking in food-restricted rats. These results indicate that PVT activity can modulate heroin seeking induced by chronic food restriction.
Collapse
Affiliation(s)
- Alexandra Chisholm
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Jessica Iannuzzi
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Damaris Rizzo
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Natasha Gonzalez
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Émilie Fortin
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Alexandra Bumbu
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Ariel A. Batallán Burrowes
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - C. Andrew Chapman
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| | - Uri Shalev
- Department of Psychology, Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie ComportementaleConcordia University Montreal Canada
| |
Collapse
|
238
|
Peeters LM, Missault S, Keliris AJ, Keliris GA. Combining designer receptors exclusively activated by designer drugs and neuroimaging in experimental models: A powerful approach towards neurotheranostic applications. Br J Pharmacol 2020; 177:992-1002. [PMID: 31658365 PMCID: PMC7042113 DOI: 10.1111/bph.14885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 11/30/2022] Open
Abstract
The combination of chemogenetics targeting specific brain cell populations with in vivo imaging techniques provides scientists with a powerful new tool to study functional neural networks at the whole-brain scale. A number of recent studies indicate the potential of this approach to increase our understanding of brain function in health and disease. In this review, we discuss the employment of a specific chemogenetic tool, designer receptors exclusively activated by designer drugs, in conjunction with non-invasive neuroimaging techniques such as PET and MRI. We highlight the utility of using this multiscale approach in longitudinal studies and its ability to identify novel brain circuits relevant to behaviour that can be monitored in parallel. In addition, some identified shortcomings in this technique and more recent efforts to overcome them are also presented. Finally, we discuss the translational potential of designer receptors exclusively activated by designer drugs in neuroimaging and the promise it holds for future neurotheranostic applications.
Collapse
|
239
|
Choi JE, Kim J, Kim J. Capturing activated neurons and synapses. Neurosci Res 2020; 152:25-34. [DOI: 10.1016/j.neures.2019.12.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/12/2022]
|
240
|
A disinhibitory nigra-parafascicular pathway amplifies seizure in temporal lobe epilepsy. Nat Commun 2020; 11:923. [PMID: 32066723 PMCID: PMC7026152 DOI: 10.1038/s41467-020-14648-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 01/26/2020] [Indexed: 01/11/2023] Open
Abstract
The precise circuit of the substantia nigra pars reticulata (SNr) involved in temporal lobe epilepsy (TLE) is still unclear. Here we found that optogenetic or chemogenetic activation of SNr parvalbumin+ (PV) GABAergic neurons amplifies seizure activities in kindling- and kainic acid-induced TLE models, whereas selective inhibition of these neurons alleviates seizure activities. The severity of seizures is bidirectionally regulated by optogenetic manipulation of SNr PV fibers projecting to the parafascicular nucleus (PF). Electrophysiology combined with rabies virus-assisted circuit mapping shows that SNr PV neurons directly project to and functionally inhibit posterior PF GABAergic neurons. Activity of these neurons also regulates seizure activity. Collectively, our results reveal that a long-range SNr-PF disinhibitory circuit participates in regulating seizure in TLE and inactivation of this circuit can alleviate severity of epileptic seizures. These findings provide a better understanding of pathological changes from a circuit perspective and suggest a possibility to precisely control epilepsy. The neural circuits through which the substantia nigra pars reticulata (SNr) exerts its role in epilepsy control are not known. Here the authors reveal that a long-range SNr-parafascicular nucleus disinhibitory circuit participates in regulating seizures in temporal lobe epilepsy and inhibition of this circuit can alleviate severity of epileptic seizures.
Collapse
|
241
|
Delaney SL, Gendreau JL, D'Souza M, Feng AY, Ho AL. Optogenetic Modulation for the Treatment of Traumatic Brain Injury. Stem Cells Dev 2020; 29:187-197. [DOI: 10.1089/scd.2019.0187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
| | | | | | - Austin Y. Feng
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, Georgia
| | - Allen L. Ho
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, Georgia
| |
Collapse
|
242
|
Cho C, Lee S, Kim A, Yarishkin O, Ryoo K, Lee Y, Jung H, Yang E, Lee DY, Lee B, Kim H, Oh U, Im H, Hwang EM, Park J. TMEM16A expression in cholinergic neurons of the medial habenula mediates anxiety-related behaviors. EMBO Rep 2020; 21:e48097. [PMID: 31782602 PMCID: PMC7001509 DOI: 10.15252/embr.201948097] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 11/09/2022] Open
Abstract
TMEM16A, a Ca2+ -activated Cl- channel, is known to modulate the excitability of various types of cells; however, its function in central neurons is largely unknown. Here, we show the specific expression of TMEM16A in the medial habenula (mHb) via RNAscope in situ hybridization, immunohistochemistry, and electrophysiology. When TMEM16A is ablated in the mHb cholinergic neurons (TMEM16A cKO mice), the slope of after-hyperpolarization of spontaneous action potentials decreases and the firing frequency is reduced. Reduced mHb activity also decreases the activity of the interpeduncular nucleus (IPN). Moreover, TMEM16A cKO mice display anxiogenic behaviors and deficits in social interaction without despair-like phenotypes or cognitive dysfunctions. Finally, chemogenetic inhibition of mHb cholinergic neurons using the DREADD (Designer Receptors Exclusively Activated by Designer Drugs) approach reveals similar behavioral phenotypes to those of TMEM16A cKO mice. We conclude that TMEM16A plays a key role in anxiety-related behaviors regulated by mHb cholinergic neurons and could be a potential therapeutic target against anxiety-related disorders.
Collapse
Affiliation(s)
- Chang‐Hoon Cho
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| | - Sangjoon Lee
- Convergence Research Center for Diagnosis, Treatment and Care System of DementiaKISTSeoulKorea
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulKorea
- Center for NeuroscienceBrain Science InstituteKorea Institute of Science and Technology (KIST)SeoulKorea
| | - Ajung Kim
- Center for Functional ConnectomicsKISTSeoulKorea
- KHU‐KIST Department of Converging Science and TechnologyGraduate SchoolKyung Hee UniversitySeoulKorea
| | | | - Kanghyun Ryoo
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| | - Young‐Sun Lee
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| | - Hyun‐Gug Jung
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
- Center for Functional ConnectomicsKISTSeoulKorea
| | - Esther Yang
- Department of AnatomyCollege of MedicineKorea UniversitySeoulKorea
| | - Da Yong Lee
- Center for Functional ConnectomicsKISTSeoulKorea
| | - Byeongjun Lee
- Sensory Research CenterCRIBrain Science InstituteKorea Institute of Science and TechnologySeoulKorea
| | - Hyun Kim
- Department of AnatomyCollege of MedicineKorea UniversitySeoulKorea
| | - Uhtaek Oh
- Sensory Research CenterCRIBrain Science InstituteKorea Institute of Science and TechnologySeoulKorea
| | - Heh‐In Im
- Convergence Research Center for Diagnosis, Treatment and Care System of DementiaKISTSeoulKorea
- Center for NeuroscienceBrain Science InstituteKorea Institute of Science and Technology (KIST)SeoulKorea
- Division of Bio‐Medical Science & TechnologyKIST SchoolKorea University of Science and TechnologySeoulKorea
| | - Eun Mi Hwang
- Center for Functional ConnectomicsKISTSeoulKorea
- KHU‐KIST Department of Converging Science and TechnologyGraduate SchoolKyung Hee UniversitySeoulKorea
- Division of Bio‐Medical Science & TechnologyKIST SchoolKorea University of Science and TechnologySeoulKorea
| | - Jae‐Yong Park
- School of Biosystems and Biomedical SciencesCollege of Health SciencesKorea UniversitySeoulKorea
| |
Collapse
|
243
|
Dong Z, Chen W, Chen C, Wang H, Cui W, Tan Z, Robinson H, Gao N, Luo B, Zhang L, Zhao K, Xiong WC, Mei L. CUL3 Deficiency Causes Social Deficits and Anxiety-like Behaviors by Impairing Excitation-Inhibition Balance through the Promotion of Cap-Dependent Translation. Neuron 2020; 105:475-490.e6. [PMID: 31780330 PMCID: PMC7007399 DOI: 10.1016/j.neuron.2019.10.035] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/11/2019] [Accepted: 10/27/2019] [Indexed: 01/30/2023]
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders with symptoms including social deficits, anxiety, and communication difficulties. However, ASD pathogenic mechanisms are poorly understood. Mutations of CUL3, which encodes Cullin 3 (CUL3), a component of an E3 ligase complex, are thought of as risk factors for ASD and schizophrenia (SCZ). CUL3 is abundant in the brain, yet little is known of its function. Here, we show that CUL3 is critical for neurodevelopment. CUL3-deficient mice exhibited social deficits and anxiety-like behaviors with enhanced glutamatergic transmission and neuronal excitability. Proteomic analysis revealed eIF4G1, a protein for Cap-dependent translation, as a potential target of CUL3. ASD-associated cellular and behavioral deficits could be rescued by pharmacological inhibition of the eIF4G1 function and chemogenetic inhibition of neuronal activity. Thus, CUL3 is critical to neural development, neurotransmission, and excitation-inhibition (E-I) balance. Our study provides novel insight into the pathophysiological mechanisms of ASD and SCZ.
Collapse
Affiliation(s)
- Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wenbing Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Chao Chen
- The Laboratory of Vector Biology and Control, College of Engineering, Beijing Normal University (Zhuhai), Zhuhai 519085, China
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wanpeng Cui
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zhibing Tan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Heath Robinson
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Nannan Gao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Lei Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Kai Zhao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
244
|
Guarino S, Conrad SE, Papini MR. Frustrative nonreward: Chemogenetic inactivation of the central amygdala abolishes the effect of reward downshift without affecting alcohol intake. Neurobiol Learn Mem 2020; 169:107173. [PMID: 32001338 DOI: 10.1016/j.nlm.2020.107173] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 11/16/2022]
Abstract
The role of the central amygdala (CeA) in the adjustment to a 32-to-2% sucrose downshift in the consummatory successive negative contrast (cSNC) task and in a free-choice 10% alcohol-water preference task (PT) was studied using chemogenetic inactivation. cSNC is a model of frustrative nonreward that enhances alcohol consumption. In Experiment 1, sessions 1-10 involved 5-min access to 32% sucrose and sessions 11-12 involved access to 2% sucrose. Vehicle or clozapine N-oxide (CNO; 1 or 3 mg/kg, ip), used later to activate the inhibitory designer receptor, was administered 30 min before sessions 11-12. There was no evidence that CNO affected consummatory behavior after the sucrose downshift. In Experiment 2, all animals received an infusion of the inhibitory designer receptor hM4D(Gi) into the CeA. After recovery, animals received access to either 32% or 2% sucrose on sessions 1-10, followed by 2% sucrose on sessions 11-12. Immediately after each 5-min sucrose session, animals received a 2-bottle, 1-h PT with 10% alcohol and water. CNO (3 mg/kg, ip) or vehicle was administered 30 min before sessions 11-12. CeA inactivation prior to sucrose downshift eliminated the cSNC effect, which was observed in vehicle controls. However, there was no evidence that CeA inactivation affected preference for 10% alcohol over water. These results support the hypothesis that CeA activity is critical for cSNC effect, an outcome consistent with the view that the amygdala plays a central role in frustrative nonreward.
Collapse
Affiliation(s)
- Sara Guarino
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Shannon E Conrad
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Mauricio R Papini
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA.
| |
Collapse
|
245
|
Sorrentino A, Eroglu E, Michel T. In vivo applications of chemogenetics in redox (patho)biology. OXIDATIVE STRESS 2020:97-112. [DOI: 10.1016/b978-0-12-818606-0.00007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
246
|
Synaptic control of spinal GRPR + neurons by local and long-range inhibitory inputs. Proc Natl Acad Sci U S A 2019; 116:27011-27017. [PMID: 31806757 DOI: 10.1073/pnas.1905658116] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal gastrin-releasing peptide receptor-expressing (GRPR+) neurons play an essential role in itch signal processing. However, the circuit mechanisms underlying the modulation of spinal GRPR+ neurons by direct local and long-range inhibitory inputs remain elusive. Using viral tracing and electrophysiological approaches, we dissected the neural circuits underlying the inhibitory control of spinal GRPR+ neurons. We found that spinal galanin+ GABAergic neurons form inhibitory synapses with GRPR+ neurons in the spinal cord and play an important role in gating the GRPR+ neuron-dependent itch signaling pathway. Spinal GRPR+ neurons also receive inhibitory inputs from local neurons expressing neuronal nitric oxide synthase (nNOS). Moreover, spinal GRPR+ neurons are gated by strong inhibitory inputs from the rostral ventromedial medulla. Thus, both local and long-range inhibitory inputs could play important roles in gating itch processing in the spinal cord by directly modulating the activity of spinal GRPR+ neurons.
Collapse
|
247
|
Abstract
The peripheral nervous system (PNS) is highly complicated and heterogenous. Conventional neuromodulatory approaches have revealed numerous essential biological functions of the PNS and provided excellent tools to treat a large variety of human diseases. Yet growing evidence indicated the importance of cell-type-specific neuromodulation in the PNS in not only biological research using animal models but also potential human therapies. Optogenetics is a recently developed neuromodulatory approach combining optics and genetics that can effectively stimulate or silence neuronal activity with high spatial and temporal precision. Here, I review research regarding optogenetic manipulations for cell-type-specific control of the PNS, highlighting the advantages and challenges of current optogenetic tools, and discuss their potential future applications.
Collapse
Affiliation(s)
- Rui B Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
248
|
Todd TP, Fournier DI, Bucci DJ. Retrosplenial cortex and its role in cue-specific learning and memory. Neurosci Biobehav Rev 2019; 107:713-728. [PMID: 31055014 PMCID: PMC6906080 DOI: 10.1016/j.neubiorev.2019.04.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 10/26/2022]
Abstract
The retrosplenial cortex (RSC) contributes to spatial navigation, as well as contextual learning and memory. However, a growing body of research suggests that the RSC also contributes to learning and memory for discrete cues, such as auditory or visual stimuli. In this review, we summarize and assess the Pavlovian and instrumental conditioning experiments that have examined the role of the RSC in cue-specific learning and memory. We use the term cue-specific to refer to these putatively non-spatial conditioning paradigms that involve discrete cues. Although these paradigms emphasize behavior related to cue presentations, we note that cue-specific learning and memory always takes place against a background of contextual stimuli. We review multiple ways by which contexts can influence responding to discrete cues and suggest that RSC contributions to cue-specific learning and memory are intimately tied to contextual learning and memory. Indeed, although the RSC is involved in several forms of cue-specific learning and memory, we suggest that many of these can be linked to processing of contextual stimuli.
Collapse
Affiliation(s)
- Travis P Todd
- Dartmouth College, Department of Psychological and Brain Sciences, 6207 Moore Hall, NH, 03755, USA.
| | - Danielle I Fournier
- Dartmouth College, Department of Psychological and Brain Sciences, 6207 Moore Hall, NH, 03755, USA
| | - David J Bucci
- Dartmouth College, Department of Psychological and Brain Sciences, 6207 Moore Hall, NH, 03755, USA
| |
Collapse
|
249
|
Milton AL. Fear not: recent advances in understanding the neural basis of fear memories and implications for treatment development. F1000Res 2019; 8:F1000 Faculty Rev-1948. [PMID: 31824654 PMCID: PMC6880271 DOI: 10.12688/f1000research.20053.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2019] [Indexed: 01/01/2023] Open
Abstract
Fear is a highly adaptive emotion that has evolved to promote survival and reproductive fitness. However, maladaptive expression of fear can lead to debilitating stressor-related and anxiety disorders such as post-traumatic stress disorder. Although the neural basis of fear has been extensively researched for several decades, recent technological advances in pharmacogenetics and optogenetics have allowed greater resolution in understanding the neural circuits that underlie fear. Alongside conceptual advances in the understanding of fear memory, this increased knowledge has clarified mechanisms for some currently available therapies for post-traumatic stress disorder and has identified new potential treatment targets.
Collapse
Affiliation(s)
- Amy L. Milton
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| |
Collapse
|
250
|
Chemogenetic Activation of Excitatory Neurons Alters Hippocampal Neurotransmission in a Dose-Dependent Manner. eNeuro 2019; 6:ENEURO.0124-19.2019. [PMID: 31645362 PMCID: PMC6860986 DOI: 10.1523/eneuro.0124-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 11/21/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tools are extensively used to manipulate neuronal activity in a cell type-specific manner. Whole-cell patch-clamp recordings indicate membrane depolarization, coupled with increased neuronal firing rate, following administration of the DREADD ligand, clozapine-N-oxide (CNO) to activate the Gq-coupled DREADD, hM3Dq. Although hM3Dq has been used to enhance neuronal firing in order to manipulate diverse behaviors, often within 30 min to 1 h after CNO administration, the physiological effects on excitatory neurotransmission remain poorly understood. We investigated the influence of CNO-mediated hM3Dq DREADD activation on distinct aspects of hippocampal excitatory neurotransmission at the Schaffer collateral-CA1 synapse in hippocampal slices derived from mice expressing hM3Dq in Ca2+/calmodulin-dependent protein kinase α (CamKIIα)-positive excitatory neurons. Our results indicate a clear dose-dependent effect on field EPSP (fEPSP) slope, with no change noted at the lower dose of CNO (1 µM) and a significant, long-term decline in fEPSP slope observed at higher doses (5-20 µM). Further, we noted a robust θ burst stimulus (TBS) induced long-term potentiation (LTP) in the presence of the lower CNO (1 µM) dose, which was significantly attenuated at the higher CNO (20 µM) dose. Whole-cell patch-clamp recording revealed both complex dose-dependent regulation of excitability, and spontaneous and evoked activity of CA1 pyramidal neurons in response to hM3Dq activation across CNO concentrations. Our data indicate that CNO-mediated activation of the hM3Dq DREADD results in dose-dependent regulation of excitatory hippocampal neurotransmission and highlight the importance of careful interpretation of behavioral experiments involving chemogenetic manipulation.
Collapse
|