1
|
Miyake T, Tanaka K, Inoue Y, Nagai Y, Nishimura R, Seta T, Nakagawa S, Inoue KI, Hasegawa E, Minamimoto T, Doi M. Size-reduced DREADD derivatives for AAV-assisted multimodal chemogenetic control of neuronal activity and behavior. CELL REPORTS METHODS 2024; 4:100881. [PMID: 39437713 DOI: 10.1016/j.crmeth.2024.100881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are engineered G-protein-coupled receptors that afford reversible manipulation of neuronal activity in vivo. Here, we introduce size-reduced DREADD derivatives miniDq and miniDi, which inherit the basic receptor properties from the Gq-coupled excitatory receptor hM3Dq and the Gi-coupled inhibitory receptor hM4Di, respectively, while being approximately 30% smaller in size. Taking advantage of the compact size of the receptors, we generated an adeno-associated virus (AAV) vector carrying both miniDq and the other DREADD family receptor (κ-opioid receptor-based inhibitory DREADD [KORD]) within the maximum AAV capacity (4.7 kb), allowing us to modulate neuronal activity and animal behavior in both excitatory and inhibitory directions using a single viral vector. We confirmed that expressing miniDq, but not miniDi, allowed activation of striatum activity in the cynomolgus monkey (Macaca fascicularis). The compact DREADDs may thus widen the opportunity for multiplexed interrogation and/or intervention in neuronal regulation in mice and non-human primates.
Collapse
Affiliation(s)
- Takahito Miyake
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan.
| | - Kaho Tanaka
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Yutsuki Inoue
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Yuji Nagai
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Reo Nishimura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Takehito Seta
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Shumpei Nakagawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama 484-8506, Japan
| | - Emi Hasegawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Takafumi Minamimoto
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
2
|
Neřoldová M, Stuchlík A. Chemogenetic Tools and their Use in Studies of Neuropsychiatric Disorders. Physiol Res 2024; 73:S449-S470. [PMID: 38957949 PMCID: PMC11412350 DOI: 10.33549/physiolres.935401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Chemogenetics is a newly developed set of tools that allow for selective manipulation of cell activity. They consist of a receptor mutated irresponsive to endogenous ligands and a synthetic ligand that does not interact with the wild-type receptors. Many different types of these receptors and their respective ligands for inhibiting or excitating neuronal subpopulations were designed in the past few decades. It has been mainly the G-protein coupled receptors (GPCRs) selectively responding to clozapine-N-oxide (CNO), namely Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), that have been employed in research. Chemogenetics offers great possibilities since the activity of the receptors is reversible, inducible on demand by the ligand, and non-invasive. Also, specific groups or types of neurons can be selectively manipulated thanks to the delivery by viral vectors. The effect of the chemogenetic receptors on neurons lasts longer, and even chronic activation can be achieved. That can be useful for behavioral testing. The great advantage of chemogenetic tools is especially apparent in research on brain diseases since they can manipulate whole neuronal circuits and connections between different brain areas. Many psychiatric or other brain diseases revolve around the dysfunction of specific brain networks. Therefore, chemogenetics presents a powerful tool for investigating the underlying mechanisms causing the disease and revealing the link between the circuit dysfunction and the behavioral or cognitive symptoms observed in patients. It could also contribute to the development of more effective treatments.
Collapse
Affiliation(s)
- M Neřoldová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic. E-mail:
| | | |
Collapse
|
3
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Wu C, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024; 57:1243-1259.e8. [PMID: 38744291 PMCID: PMC11168877 DOI: 10.1016/j.immuni.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.
Collapse
Affiliation(s)
- Tyler E Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Derek B Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew B Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Darshan N Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucille M Rich
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, CA, USA
| | - Jason S Debley
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
4
|
Clark PJ, Brodnik ZD, España RA. Chemogenetic Signaling in Space and Time: Considerations for Designing Neuroscience Experiments Using DREADDs. Neuroscientist 2024; 30:328-346. [PMID: 36408535 DOI: 10.1177/10738584221134587] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
The use of designer receptors exclusively activated by designer drugs (DREADDs) has led to significant advances in our understanding of the neural circuits that govern behavior. By allowing selective control over cellular activity and signaling, DREADDs have become an integral tool for defining the pathways and cellular phenotypes that regulate sleep, pain, motor activity, goal-directed behaviors, and a variety of other processes. In this review, we provide a brief overview of DREADDs and discuss notable discoveries in the neurosciences with an emphasis on circuit mechanisms. We then highlight methodological approaches to achieve pathway specific activation of DREADDs. Finally, we discuss spatial and temporal constraints of DREADDs signaling and how these features can be incorporated into experimental designs to precisely dissect circuits of interest.
Collapse
Affiliation(s)
- Philip J Clark
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Zachary D Brodnik
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Rodrigo A España
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
5
|
Gonzalez-Ramos A, Puigsasllosas-Pastor C, Arcas-Marquez A, Tornero D. Updated Toolbox for Assessing Neuronal Network Reconstruction after Cell Therapy. Bioengineering (Basel) 2024; 11:487. [PMID: 38790353 PMCID: PMC11118929 DOI: 10.3390/bioengineering11050487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Cell therapy has proven to be a promising treatment for a range of neurological disorders, including Parkinson Disease, drug-resistant epilepsy, and stroke, by restoring function after brain damage. Nevertheless, evaluating the true effectiveness of these therapeutic interventions requires a deep understanding of the functional integration of grafted cells into existing neural networks. This review explores a powerful arsenal of molecular techniques revolutionizing our ability to unveil functional integration of grafted cells within the host brain. From precise manipulation of neuronal activity to pinpoint the functional contribution of transplanted cells by using opto- and chemo-genetics, to real-time monitoring of neuronal dynamics shedding light on functional connectivity within the reconstructed circuits by using genetically encoded (calcium) indicators in vivo. Finally, structural reconstruction and mapping communication pathways between grafted and host neurons can be achieved by monosynaptic tracing with viral vectors. The cutting-edge toolbox presented here holds immense promise for elucidating the impact of cell therapy on neural circuitry and guiding the development of more effective treatments for neurological disorders.
Collapse
Affiliation(s)
- Ana Gonzalez-Ramos
- Stanley Center for Psychiatric Research at Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Claudia Puigsasllosas-Pastor
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ainhoa Arcas-Marquez
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Daniel Tornero
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
6
|
Iguchi Y, Fukabori R, Kato S, Takahashi K, Eifuku S, Maejima Y, Shimomura K, Mizuma H, Mawatari A, Doi H, Cui Y, Onoe H, Hikishima K, Osanai M, Nishijo T, Momiyama T, Benton R, Kobayashi K. Chemogenetic activation of mammalian brain neurons expressing insect Ionotropic Receptors by systemic ligand precursor administration. Commun Biol 2024; 7:547. [PMID: 38714803 PMCID: PMC11076466 DOI: 10.1038/s42003-024-06223-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/22/2024] [Indexed: 05/10/2024] Open
Abstract
Chemogenetic approaches employing ligand-gated ion channels are advantageous regarding manipulation of target neuronal population functions independently of endogenous second messenger pathways. Among them, Ionotropic Receptor (IR)-mediated neuronal activation (IRNA) allows stimulation of mammalian neurons that heterologously express members of the insect chemosensory IR repertoire in response to their cognate ligands. In the original protocol, phenylacetic acid, a ligand of the IR84a/IR8a complex, was locally injected into a brain region due to its low permeability of the blood-brain barrier. To circumvent this invasive injection, we sought to develop a strategy of peripheral administration with a precursor of phenylacetic acid, phenylacetic acid methyl ester, which is efficiently transferred into the brain and converted to the mature ligand by endogenous esterase activities. This strategy was validated by electrophysiological, biochemical, brain-imaging, and behavioral analyses, demonstrating high utility of systemic IRNA technology in the remote activation of target neurons in the brain.
Collapse
Affiliation(s)
- Yoshio Iguchi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Ryoji Fukabori
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kazumi Takahashi
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Satoshi Eifuku
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Hiroshi Mizuma
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Aya Mawatari
- Laboratory for Labeling Chemistry, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Hisashi Doi
- Laboratory for Labeling Chemistry, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Research, Institute for Drug Discovery Science, Collaborative Creation Research Center, Organization for Research Promotion, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, 599-8531, Japan
| | - Yilong Cui
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Hirotaka Onoe
- Human Brain Research Center, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-Cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Keigo Hikishima
- Medical Devices Research Group, Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-2-1 Namiki, Tsukuba, 305-8564, Japan
| | - Makoto Osanai
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, 565-0871, Japan
| | - Takuma Nishijo
- Department of Pharmacology, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Tokyo, 105-8461, Japan
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya-cho, Kasugai, 480-0392, Japan
| | - Toshihiko Momiyama
- Department of Pharmacology, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Tokyo, 105-8461, Japan
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
| |
Collapse
|
7
|
Cea Salazar VI, Perez MD, Robison AJ, Trainor BC. Impacts of sex differences on optogenetic, chemogenetic, and calcium-imaging tools. Curr Opin Neurobiol 2024; 84:102817. [PMID: 38042130 PMCID: PMC11374099 DOI: 10.1016/j.conb.2023.102817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 12/04/2023]
Abstract
Technical innovation in neuroscience introduced powerful tools for measuring and manipulating neuronal activity via optical, chemogenetic, and calcium-imaging tools. These tools were initially tested primarily in male animals but are now increasingly being used in females as well. In this review, we consider how these tools may work differently in males and females. For example, we review sex differences in the metabolism of chemogenetic ligands and their downstream signaling effects. Optical tools more directly alter depolarization or hyperpolarization of neurons, but biological sex and gonadal hormones modulate synaptic inputs and intrinsic excitability. We review studies demonstrating that optogenetic manipulations are sometimes consistent across the rodent estrous cycle but within certain circuits; manipulations can vary across the ovarian cycle. Finally, calcium-imaging methods utilize genetically encoded calcium indicators to measure neuronal activity. Testosterone and estradiol can directly modulate calcium influx, and we consider these implications for interpreting the results of calcium-imaging studies. Together, our findings suggest that these neuroscientific tools may sometimes work differently in males and females and that users should be aware of these differences when applying these methods.
Collapse
Affiliation(s)
| | - Melvin D Perez
- Department of Physiology, University of California, Davis, CA 95616, USA
| | - A J Robison
- Department of Psychology, University of California, Davis, CA 95616, USA
| | - Brian C Trainor
- Neuroscience Graduate Group, University of California, Davis, CA 95616, USA; Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
8
|
Aomine Y, Oyama Y, Sakurai K, Macpherson T, Ozawa T, Hikida T. Clozapine N-oxide, compound 21, and JHU37160 do not influence effortful reward-seeking behavior in mice. Psychopharmacology (Berl) 2024; 241:89-96. [PMID: 37792024 PMCID: PMC10774210 DOI: 10.1007/s00213-023-06465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023]
Abstract
RATIONALE Clozapine N-oxide (CNO) has been developed as a ligand to selectively activate designer receptors exclusively activated by designer drugs (DREADDs). However, previous studies have revealed that peripherally injected CNO is reverse-metabolized into clozapine, which, in addition to activating DREADDs, acts as an antagonist at various neurotransmitter receptors, suggesting potential off-target effects of CNO on animal physiology and behaviors. Recently, second-generation DREADD agonists compound 21 (C21) and JHU37160 (J60) have been developed, but their off-target effects are not fully understood. OBJECTIVES The present studies assessed the effect of novel DREADD ligands on reward-seeking behavior. METHODS We first tested the possible effect of acute i.p. injection of low-to-moderate (0.1, 0.3, 1, 3 mg/kg) of CNO, C21, and J60 on motivated reward-seeking behavior in wild-type mice. We then examined whether a high dose (10 mg/kg) of these drugs might be able to alter responding. RESULTS Low-to-moderate doses of all drugs and a high dose of CNO or C21 did not alter operant lick responding for a reward under a progressive ratio schedule of reinforcement, in which the number of operant lick responses to obtain a reward increases after each reward collection. However, high-dose J60 resulted in a total lack of responding that was later observed in an open field arena to be due to a sedative effect. CONCLUSIONS This study provides definitive evidence that commonly used doses of CNO, C21, and J60 have negligible off-target effects on motivated reward-seeking but urges caution when using high doses of J60 due to sedative effects.
Collapse
Affiliation(s)
- Yoshiatsu Aomine
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan
| | - Yoshinobu Oyama
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan
| | - Koki Sakurai
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan
- Laboratory of Protein Profiling and Functional Proteomics, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Tom Macpherson
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan
| | - Takaaki Ozawa
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita, Osaka, Japan.
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan.
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Suita, Osaka, Japan.
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan.
| |
Collapse
|
9
|
Shimizu M, Yoshimura M, Baba K, Ikeda N, Nonaka Y, Maruyama T, Onaka T, Ueta Y. Deschloroclozapine exhibits an exquisite agonistic effect at lower concentration compared to clozapine-N-oxide in hM3Dq expressing chemogenetically modified rats. Front Neurosci 2023; 17:1301515. [PMID: 38099201 PMCID: PMC10720889 DOI: 10.3389/fnins.2023.1301515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/03/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Within the realm of chemogenetics, a particular form of agonists targeting designer receptors exclusively activated by designer drugs (DREADDs) has emerged. Deschloroclozapine (DCZ), a recently introduced DREADDs agonist, demonstrates remarkable potency in activating targeted neurons at a lower dosage compared to clozapine-N-oxide (CNO). Methods We conducted a comparative analysis of the effects of subcutaneously administered CNO (1 mg/kg) and DCZ (0.1 mg/kg) in our transgenic rats expressing hM3Dq and mCherry exclusively in oxytocin (OXT) neurons. Results and Discussion Notably, DCZ exhibited a swift and robust elevation of serum OXT, surpassing the effects of CNO, with a significant increase in the area under the curve (AUC) up to 3 hours post-administration. Comprehensive assessment of brain neuronal activity, using Fos as an indicator, revealed comparable effects between CNO and DCZ. Additionally, in a neuropathic pain model, both CNO and DCZ increased the mechanical nociceptive and thermal thresholds; however, the DCZ-treated group exhibited a significantly accelerated onset of the effects, aligning harmoniously with the observed alterations in serum OXT concentration following DCZ administration. These findings emphasize the remarkable efficacy of DCZ in rats, suggesting its equivalent or potentially superior performance to CNO at considerably lower dosages, thus positioning it as a promising contender among DREADDs agonists.
Collapse
Affiliation(s)
- Makiko Shimizu
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Mitsuhiro Yoshimura
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuhiko Baba
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Naofumi Ikeda
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuki Nonaka
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
10
|
Jo A, Deniz S, Cherian S, Xu J, Futagi D, DeVries SH, Zhu Y. Modular interneuron circuits control motion sensitivity in the mouse retina. Nat Commun 2023; 14:7746. [PMID: 38008788 PMCID: PMC10679153 DOI: 10.1038/s41467-023-43382-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/08/2023] [Indexed: 11/28/2023] Open
Abstract
Neural computations arise from highly precise connections between specific types of neurons. Retinal ganglion cells (RGCs) with similar stratification patterns are positioned to receive similar inputs but often display different response properties. In this study, we used intersectional mouse genetics to achieve single-cell type labeling and identified an object motion sensitive (OMS) AC type, COMS-AC(counter-OMS AC). Optogenetic stimulation revealed that COMS-AC makes glycinergic synapses with the OMS-insensitive HD2p-RGC, while chemogenetic inactivation showed that COMS-AC provides inhibitory control to HD2p-RGC during local motion. This local inhibition, combined with the inhibitory drive from TH2-AC during global motion, explains the OMS-insensitive feature of HD2p-RGC. In contrast, COMS-AC fails to make synapses with W3(UHD)-RGC, allowing it to exhibit OMS under the control of VGlut3-AC and TH2-AC. These findings reveal modular interneuron circuits that endow structurally similar RGC types with different responses and present a mechanism for redundancy-reduction in the retina to expand coding capacity.
Collapse
Affiliation(s)
- Andrew Jo
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sercan Deniz
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Suraj Cherian
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jian Xu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Daiki Futagi
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Steven H DeVries
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Yongling Zhu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
11
|
Lawson KA, Ruiz CM, Mahler SV. A head-to-head comparison of two DREADD agonists for suppressing operant behavior in rats via VTA dopamine neuron inhibition. Psychopharmacology (Berl) 2023; 240:2101-2110. [PMID: 37530882 PMCID: PMC10794001 DOI: 10.1007/s00213-023-06429-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/18/2023] [Indexed: 08/03/2023]
Abstract
RATIONALE Designer receptors exclusively activated by designer drugs (DREADDs) are a tool for "remote control" of defined neuronal populations during behavior. These receptors are inert unless bound by an experimenter-administered designer drug, commonly clozapine-n-oxide (CNO). However, questions have emerged about the suitability of CNO as a systemically administered DREADD agonist. OBJECTIVES Second-generation agonists such as JHU37160 (J60) have been developed, which may have more favorable properties than CNO. Here we sought to directly compare effects of CNO (0, 1, 5, & 10 mg/kg, i.p.) and J60 (0, 0.03, 0.3, & 3 mg/kg, i.p.) on operant food pursuit. METHODS Male and female TH:Cre + rats and their wildtype (WT) littermates received cre-dependent hM4Di-mCherry vector injections into ventral tegmental area (VTA), causing inhibitory DREADD expression in VTA dopamine neurons of TH:Cre + rats. All rats were trained to stably lever press for palatable food on a fixed ratio 10 schedule, and doses of both agonists were tested on separate days in counterbalanced order. RESULTS All three CNO doses reduced operant rewards earned in rats with DREADDs, and no CNO dose had behavioral effects in WT controls. The highest J60 dose tested significantly reduced responding in DREADD rats, but this dose also increased responding in WTs, indicating non-specific effects. The magnitude of CNO and J60 effects in TH:Cre + rats were correlated and were present in both sexes. CONCLUSIONS Findings demonstrate the usefulness of directly comparing DREADD agonists when optimizing behavioral chemogenetics, and highlight the importance of proper controls, regardless of the DREADD agonist employed.
Collapse
Affiliation(s)
- Kate A Lawson
- Department of Neurobiology and Behavior, University of California Irvine, 1132 McGaugh Hall, Irvine, CA, 92697, USA.
| | - Christina M Ruiz
- Department of Neurobiology and Behavior, University of California Irvine, 1132 McGaugh Hall, Irvine, CA, 92697, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California Irvine, 1132 McGaugh Hall, Irvine, CA, 92697, USA
| |
Collapse
|
12
|
Pereira MJ, Ayana R, Holt MG, Arckens L. Chemogenetic manipulation of astrocyte activity at the synapse- a gateway to manage brain disease. Front Cell Dev Biol 2023; 11:1193130. [PMID: 37534103 PMCID: PMC10393042 DOI: 10.3389/fcell.2023.1193130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Astrocytes are the major glial cell type in the central nervous system (CNS). Initially regarded as supportive cells, it is now recognized that this highly heterogeneous cell population is an indispensable modulator of brain development and function. Astrocytes secrete neuroactive molecules that regulate synapse formation and maturation. They also express hundreds of G protein-coupled receptors (GPCRs) that, once activated by neurotransmitters, trigger intracellular signalling pathways that can trigger the release of gliotransmitters which, in turn, modulate synaptic transmission and neuroplasticity. Considering this, it is not surprising that astrocytic dysfunction, leading to synaptic impairment, is consistently described as a factor in brain diseases, whether they emerge early or late in life due to genetic or environmental factors. Here, we provide an overview of the literature showing that activation of genetically engineered GPCRs, known as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), to specifically modulate astrocyte activity partially mimics endogenous signalling pathways in astrocytes and improves neuronal function and behavior in normal animals and disease models. Therefore, we propose that expressing these genetically engineered GPCRs in astrocytes could be a promising strategy to explore (new) signalling pathways which can be used to manage brain disorders. The precise molecular, functional and behavioral effects of this type of manipulation, however, differ depending on the DREADD receptor used, targeted brain region and timing of the intervention, between healthy and disease conditions. This is likely a reflection of regional and disease/disease progression-associated astrocyte heterogeneity. Therefore, a thorough investigation of the effects of such astrocyte manipulation(s) must be conducted considering the specific cellular and molecular environment characteristic of each disease and disease stage before this has therapeutic applicability.
Collapse
Affiliation(s)
- Maria João Pereira
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Rajagopal Ayana
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Instituto de Investigação e Inovação em Saúde (i3S), Laboratory of Synapse Biology, Universidade do Porto, Porto, Portugal
| | - Lutgarde Arckens
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
13
|
Lee DG, McLachlan CA, Nogueira R, Kwon O, Carey AE, House G, Lagani GD, LaMay D, Fusi S, Chen JL. PERIRHINAL CORTEX LEARNS A PREDICTIVE MAP (INTERNAL MODEL) OF THE TASK ENVIRONMENT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.532214. [PMID: 36993645 PMCID: PMC10055158 DOI: 10.1101/2023.03.17.532214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Goal-directed tasks involve acquiring an internal model, known as a predictive map, of relevant stimuli and associated outcomes to guide behavior. Here, we identified neural signatures of a predictive map of task behavior in perirhinal cortex (Prh). Mice learned to perform a tactile working memory task by classifying sequential whisker stimuli over multiple training stages. Chemogenetic inactivation demonstrated that Prh is involved in task learning. Chronic two-photon calcium imaging, population analysis, and computational modeling revealed that Prh encodes stimulus features as sensory prediction errors. Prh forms stable stimulus-outcome associations that expand in a retrospective manner and generalize as animals learn new contingencies. Stimulus-outcome associations are linked to prospective network activity encoding possible expected outcomes. This link is mediated by cholinergic signaling to guide task performance, demonstrated by acetylcholine imaging and perturbation. We propose that Prh combines error-driven and map-like properties to acquire a predictive map of learned task behavior.
Collapse
Affiliation(s)
- David G Lee
- Department of Biomedical Engineering, Boston University, Boston MA 02215, USA
- Center for Neurophotonics, Boston University, Boston MA 02215, USA
| | - Caroline A McLachlan
- Center for Neurophotonics, Boston University, Boston MA 02215, USA
- Department of Biology, Boston University, Boston MA 02215, USA
| | - Ramon Nogueira
- Center for Theoretical Neuroscience, Columbia University, New York, NY 10027, USA
- Department of Neuroscience, Columbia University, New York NY 10027, USA
| | - Osung Kwon
- Center for Neurophotonics, Boston University, Boston MA 02215, USA
- Department of Biology, Boston University, Boston MA 02215, USA
| | - Alanna E Carey
- Center for Neurophotonics, Boston University, Boston MA 02215, USA
- Department of Biology, Boston University, Boston MA 02215, USA
| | - Garrett House
- Department of Biology, Boston University, Boston MA 02215, USA
| | - Gavin D Lagani
- Department of Biology, Boston University, Boston MA 02215, USA
| | - Danielle LaMay
- Department of Biology, Boston University, Boston MA 02215, USA
| | - Stefano Fusi
- Center for Theoretical Neuroscience, Columbia University, New York, NY 10027, USA
- Department of Neuroscience, Columbia University, New York NY 10027, USA
| | - Jerry L Chen
- Department of Biomedical Engineering, Boston University, Boston MA 02215, USA
- Center for Neurophotonics, Boston University, Boston MA 02215, USA
- Department of Biology, Boston University, Boston MA 02215, USA
- Center for Systems Neuroscience, Boston University, Boston MA 02215, USA
| |
Collapse
|
14
|
Lawson KA, Ruiz CM, Mahler SV. A head-to-head comparison of two DREADD agonists for suppressing operant behavior in rats via VTA dopamine neuron inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.27.534429. [PMID: 37034819 PMCID: PMC10081263 DOI: 10.1101/2023.03.27.534429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Rationale Designer receptors exclusively activated by designer drugs (DREADDs) are a tool for "remote control" of defined neuronal populations during behavior. These receptors are inert unless bound by an experimenter-administered designer drug, most commonly clozapine-n-oxide (CNO). However, questions have emerged about the suitability of CNO as a systemically administered DREADD agonist. Objectives Second-generation agonists such as JHU37160 (J60) have been developed, which may have more favorable properties than CNO. Here we sought to directly compare effects of CNO (0, 1, 5, & 10 mg/kg, i.p.) and J60 (0, 0.03, 0.3, & 3 mg/kg, i.p.) on operant food pursuit. Methods Male and female TH:Cre+ rats and their wildtype (WT) littermates received cre-dependent hM4Di-mCherry vector injections into ventral tegmental area (VTA), causing inhibitory DREADD expression in VTA dopamine neurons in TH:Cre+ rats. Rats were trained to stably lever press for palatable food on a fixed ratio 10 schedule, and doses of both agonists were tested on separate days in a counterbalanced order. Results All three CNO doses reduced operant food seeking in rats with DREADDs, and no CNO dose had behavioral effects in WT controls. The highest tested J60 dose significantly reduced responding in DREADD rats, but this dose also increased responding in WTs, indicating non-specific effects. The magnitude of CNO and J60 effects in TH:Cre+ rats were correlated and were present in both sexes. Conclusions Findings demonstrate the usefulness of directly comparing DREADD agonists when optimizing behavioral chemogenetics, and highlight the importance of proper controls, regardless of the DREADD agonist employed.
Collapse
Affiliation(s)
- Kate A Lawson
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA USA
| | - Christina M Ruiz
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA USA
| |
Collapse
|
15
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine regulates epithelial fluid secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533208. [PMID: 36993541 PMCID: PMC10055254 DOI: 10.1101/2023.03.17.533208] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Tuft cells are solitary chemosensory epithelial cells that can sense lumenal stimuli at mucosal barriers and secrete effector molecules to regulate the physiology and immune state of their surrounding tissue. In the small intestine, tuft cells detect parasitic worms (helminths) and microbe-derived succinate, and signal to immune cells to trigger a Type 2 immune response that leads to extensive epithelial remodeling spanning several days. Acetylcholine (ACh) from airway tuft cells has been shown to stimulate acute changes in breathing and mucocilliary clearance, but its function in the intestine is unknown. Here we show that tuft cell chemosensing in the intestine leads to release of ACh, but that this does not contribute to immune cell activation or associated tissue remodeling. Instead, tuft cell-derived ACh triggers immediate fluid secretion from neighboring epithelial cells into the intestinal lumen. This tuft cell-regulated fluid secretion is amplified during Type 2 inflammation, and helminth clearance is delayed in mice lacking tuft cell ACh. The coupling of the chemosensory function of tuft cells with fluid secretion creates an epithelium-intrinsic response unit that effects a physiological change within seconds of activation. This response mechanism is shared by tuft cells across tissues, and serves to regulate the epithelial secretion that is both a hallmark of Type 2 immunity and an essential component of homeostatic maintenance at mucosal barriers.
Collapse
Affiliation(s)
- Tyler E. Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M. Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Derek B. Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew B. Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret M. McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Darshan N. Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - John W. McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kaitlyn A. Barrow
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Lucille M. Rich
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, California, USA
| | - Jason S. Debley
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | | | - Michael R. Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
16
|
Traut J, Mengual JP, Meijer EJ, McKillop LE, Alfonsa H, Hoerder-Suabedissen A, Song SH, Fehér KD, Riemann D, Molnar Z, Akerman CJ, Vyazovskiy VV, Krone LB. Effects of clozapine-N-oxide and compound 21 on sleep in laboratory mice. eLife 2023; 12:e84740. [PMID: 36892930 PMCID: PMC9998087 DOI: 10.7554/elife.84740] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 01/03/2023] [Indexed: 03/10/2023] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are chemogenetic tools for remote control of targeted cell populations using chemical actuators that bind to modified receptors. Despite the popularity of DREADDs in neuroscience and sleep research, potential effects of the DREADD actuator clozapine-N-oxide (CNO) on sleep have never been systematically tested. Here, we show that intraperitoneal injections of commonly used CNO doses (1, 5, and 10 mg/kg) alter sleep in wild-type male laboratory mice. Using electroencephalography (EEG) and electromyography (EMG) to analyse sleep, we found a dose-dependent suppression of rapid eye movement (REM) sleep, changes in EEG spectral power during non-REM (NREM) sleep, and altered sleep architecture in a pattern previously reported for clozapine. Effects of CNO on sleep could arise from back-metabolism to clozapine or binding to endogenous neurotransmitter receptors. Interestingly, we found that the novel DREADD actuator, compound 21 (C21, 3 mg/kg), similarly modulates sleep despite a lack of back-metabolism to clozapine. Our results demonstrate that both CNO and C21 can modulate sleep of mice not expressing DREADD receptors. This implies that back-metabolism to clozapine is not the sole mechanism underlying side effects of chemogenetic actuators. Therefore, any chemogenetic experiment should include a DREADD-free control group injected with the same CNO, C21, or newly developed actuator. We suggest that electrophysiological sleep assessment could serve as a sensitive tool to test the biological inertness of novel chemogenetic actuators.
Collapse
Affiliation(s)
- Janine Traut
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
| | - Jose Prius Mengual
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
- The Kavli Institute for Nanoscience DiscoveryOxfordUnited Kingdom
| | - Elise J Meijer
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
- The Kavli Institute for Nanoscience DiscoveryOxfordUnited Kingdom
| | - Laura E McKillop
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
| | - Hannah Alfonsa
- Department of Pharmacology, University of OxfordOxfordUnited Kingdom
| | | | - Seo Ho Song
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
| | - Kristoffer D Fehér
- Geneva University Hospitals (HUG), Division of Psychiatric SpecialtiesGenevaSwitzerland
- University Hospital of Psychiatry and Psychotherapy, University of BernBernSwitzerland
| | - Dieter Riemann
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
| | - Zoltan Molnar
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Colin J Akerman
- Department of Pharmacology, University of OxfordOxfordUnited Kingdom
| | - Vladyslav V Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
- The Kavli Institute for Nanoscience DiscoveryOxfordUnited Kingdom
| | - Lukas B Krone
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of OxfordOxfordUnited Kingdom
- The Kavli Institute for Nanoscience DiscoveryOxfordUnited Kingdom
- University Hospital of Psychiatry and Psychotherapy, University of BernBernSwitzerland
- Centre for Experimental Neurology, University of BernBernSwitzerland
| |
Collapse
|
17
|
Goutaudier R, Joly F, Mallet D, Bartolomucci M, Guicherd D, Carcenac C, Vossier F, Dufourd T, Boulet S, Deransart C, Chovelon B, Carnicella S. Hypodopaminergic state of the nigrostriatal pathway drives compulsive alcohol use. Mol Psychiatry 2023; 28:463-474. [PMID: 36376463 PMCID: PMC9812783 DOI: 10.1038/s41380-022-01848-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022]
Abstract
The neurobiological mechanisms underlying compulsive alcohol use, a cardinal feature of alcohol use disorder, remain elusive. The key modulator of motivational processes, dopamine (DA), is suspected to play an important role in this pathology, but its exact role remains to be determined. Here, we found that rats expressing compulsive-like alcohol use, operationalized as punishment-resistant self-administration, showed a decrease in DA levels restricted to the dorsolateral territories of the striatum, the main output structure of the nigrostriatal DA pathway. We then causally demonstrated that chemogenetic-induced selective hypodopaminergia of this pathway resulted in compulsive-like alcohol self-administration in otherwise resilient rats, accompanied by the emergence of alcohol withdrawal-like motivational impairments (i.e., impaired motivation for a natural reinforcer). Finally, the use of the monoamine stabilizer OSU6162, previously reported to correct hypodopaminergic states, transiently decreased compulsive-like alcohol self-administration in vulnerable rats. These results suggest a potential critical role of tonic nigrostriatal hypodopaminergic states in alcohol addiction and provide new insights into our understanding of the neurobiological mechanisms underlying compulsive alcohol use.
Collapse
Affiliation(s)
- Raphaël Goutaudier
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Fanny Joly
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - David Mallet
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Magali Bartolomucci
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Denis Guicherd
- grid.410529.b0000 0001 0792 4829Service de Biochimie, Biologie Moléculaire, Toxicologie Environnementale, CHU de Grenoble-Alpes Site Nord − Institut de Biologie et de Pathologie, F-38041 Grenoble, France
| | - Carole Carcenac
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédérique Vossier
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Thibault Dufourd
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Sabrina Boulet
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Colin Deransart
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Benoit Chovelon
- grid.410529.b0000 0001 0792 4829Service de Biochimie, Biologie Moléculaire, Toxicologie Environnementale, CHU de Grenoble-Alpes Site Nord − Institut de Biologie et de Pathologie, F-38041 Grenoble, France ,grid.4444.00000 0001 2112 9282Univ. Grenoble Alpes, CNRS, DPM, 38000 Grenoble, France
| | - Sebastien Carnicella
- Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000, Grenoble, France.
| |
Collapse
|
18
|
Cushnie AK, Bullock DN, Manea AM, Tang W, Zimmermann J, Heilbronner SR. The use of chemogenetic actuator ligands in nonhuman primate DREADDs-fMRI. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 4:100072. [PMID: 36691404 PMCID: PMC9860110 DOI: 10.1016/j.crneur.2022.100072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/01/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are engineered receptors that allow for genetically targeted, reversible manipulation of cellular activity via systemic drug administration. DREADD induced manipulations are initiated via the binding of an actuator ligand. Therefore, the use of DREADDs is contingent on the availability of actuator ligands. Actuator ligands low-dose clozapine (CLZ) and deschloroclozapine (DCZ) are highly selective for DREADDs, and, upon binding, induce physiological and behavioral changes in rodents and nonhuman primates (NHPs). Despite this reported specificity, both CLZ and DCZ have partial affinity for a variety of endogenous receptors and can induce dose-specific changes even in naïve animals. As such, this study aimed to examine the effects of CLZ and DCZ on resting-state functional connectivity (rs-FC) and intrinsic neural timescales (INTs) in naïve NHPs. In doing so, we evaluated whether CLZ and DCZ - in the absence of DREADDs - are inert by examining these ligands' effects on the intrinsic functional properties of the brain. Low-dose DCZ did not induce consistent changes in rs-FC or INTs prior to the expression of DREADDs; however, a high dose resulted in subject-specific changes in rs-FC and INTs. In contrast, CLZ administration induced consistent changes in rs-FC and INTs prior to DREADD expression in our subjects. Our results caution against the use of CLZ by explicitly demonstrating the impact of off-target effects that can confound experimental results. Altogether, these data endorse the use of low dose DCZ for future DREADD-based experiments.
Collapse
Affiliation(s)
- Adriana K. Cushnie
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Daniel N. Bullock
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ana M.G. Manea
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Tang
- Department of Computer Science, Program in Neuroscience, Indiana University Bloomington, Bloomington, IN, 47408, USA
| | - Jan Zimmermann
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sarah R. Heilbronner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
19
|
Zhang S, Gumpper RH, Huang XP, Liu Y, Krumm BE, Cao C, Fay JF, Roth BL. Molecular basis for selective activation of DREADD-based chemogenetics. Nature 2022; 612:354-362. [PMID: 36450989 DOI: 10.1038/s41586-022-05489-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/27/2022] [Indexed: 12/02/2022]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) represent a powerful chemogenetic technology for the remote control of neuronal activity and cellular signalling1-4. The muscarinic receptor-based DREADDs are the most widely used chemogenetic tools in neuroscience research. The Gq-coupled DREADD (hM3Dq) is used to enhance neuronal activity, whereas the Gi/o-coupled DREADD (hM4Di) is utilized to inhibit neuronal activity5. Here we report four DREADD-related cryogenic electron microscopy high-resolution structures: a hM3Dq-miniGq complex and a hM4Di-miniGo complex bound to deschloroclozapine; a hM3Dq-miniGq complex bound to clozapine-N-oxide; and a hM3R-miniGq complex bound to iperoxo. Complemented with mutagenesis, functional and computational simulation data, our structures reveal key details of the recognition of DREADD chemogenetic actuators and the molecular basis for activation. These findings should accelerate the structure-guided discovery of next-generation chemogenetic tools.
Collapse
Affiliation(s)
- Shicheng Zhang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan H Gumpper
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xi-Ping Huang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yongfeng Liu
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brian E Krumm
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Can Cao
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonathan F Fay
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
20
|
Chemogenetic activation of VGLUT3-expressing neurons decreases movement. Eur J Pharmacol 2022; 935:175298. [PMID: 36198338 DOI: 10.1016/j.ejphar.2022.175298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) are responsible for the storage of glutamate into secretory vesicles. The VGLUT3 isoform is mainly expressed in neurons that secrete other classical neurotransmitters, including the cholinergic interneurons in the striatum, and VGLUT3-expressing neurons often secrete two distinct neurotransmitters. VGLUT3 is discretely distributed throughout the brain and is found in subpopulations of spinal cord interneurons, in subset of neurons in the dorsal root ganglion, and in Merkel cells. Mice with a global loss of VGLUT3 are hyperactive and the modulation of specific VGLUT3-expressing circuits can lead to changes in movement. In this study, we tested the hypothesis that increased activity of VGLUT3-expressing neurons is associated with decreased movement. Using a mouse line expressing excitatory designer receptor exclusively activated by designer drugs (hM3Dq-DREADD) on VGLUT3-expressing neurons, we showed that activation of hM3Dq signalling acutely decreased locomotor activity. This decreased locomotion was likely not due to circuit changes mediated by glutamate nor acetylcholine released from VGLUT3-expressing neurons, as activation of hM3Dq signalling in mice that do not release glutamate or acetylcholine from VGLUT3-expressing neurons also decreased locomotor activity. This suggests that other neurotransmitters are likely driving this hypoactive phenotype. We used these mouse lines to compare the effects of DREADD agonists in vivo. We observed that clozapine-N-oxide (CNO), clozapine, compound 21 and perlapine show small differences in the speed at which they prompt behavioural responses but the four of them are selective DREADD ligands.
Collapse
|
21
|
Abstract
The fundamental commonality across pharmacotherapies for the epilepsies
is the modulation of neuronal excitability. This poses a clear
challenge—patterned neuronal excitation is essential to normal
function, thus disrupting this activity leads to side effects.
Moreover, the efficacy of current pharmacotherapy remains incomplete
despite decades of drug development. Approaches that allow for the
selective targeting of critical populations of cells and particular
pathways in the brain have the potential to both avoid side effects
and improve efficacy. Chemogenetic methods, which combine the
selective expression of designer receptors with designer drugs, have
rapidly grown in use in the neurosciences, including in epilepsy. This
review will briefly highlight the history of chemogenetics, their
applications to date in epilepsy, and the potential (and potential
hurdles to overcome) for future translation.
Collapse
Affiliation(s)
- Patrick A. Forcelli
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
- Department of Neuroscience, Georgetown University, Washington, DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| |
Collapse
|
22
|
Dean JG, Fields CW, Brito MA, Silverstein BH, Rybicki-Kler C, Fryzel AM, Groenhout T, Liu T, Mashour GA, Pal D. Inactivation of Prefrontal Cortex Attenuates Behavioral Arousal Induced by Stimulation of Basal Forebrain During Sevoflurane Anesthesia. Anesth Analg 2022; 134:1140-1152. [PMID: 35436248 PMCID: PMC9093733 DOI: 10.1213/ane.0000000000006011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cholinergic stimulation of prefrontal cortex (PFC) can reverse anesthesia. Conversely, inactivation of PFC can delay emergence from anesthesia. PFC receives cholinergic projections from basal forebrain, which contains wake-promoting neurons. However, the role of basal forebrain cholinergic neurons in arousal from the anesthetized state requires refinement, and it is currently unknown whether the arousal-promoting effect of basal forebrain is mediated through PFC. To address these gaps in knowledge, we implemented a novel approach to the use of chemogenetic stimulation and tested the role of basal forebrain cholinergic neurons in behavioral arousal during sevoflurane anesthesia. Next, we investigated the effect of tetrodotoxin-mediated inactivation of PFC on behavioral arousal produced by electrical stimulation of basal forebrain during sevoflurane anesthesia.
Collapse
Affiliation(s)
- Jon G Dean
- From the Departments of Anesthesiology.,Molecular and Integrative Physiology.,Center for Consciousness Science
| | | | - Michael A Brito
- From the Departments of Anesthesiology.,Center for Consciousness Science.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | | - George A Mashour
- From the Departments of Anesthesiology.,Center for Consciousness Science.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| | - Dinesh Pal
- From the Departments of Anesthesiology.,Molecular and Integrative Physiology.,Center for Consciousness Science.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
23
|
Du M, Santiago A, Akiz C, Aoki C. GABAergic interneurons' feedback inhibition of dorsal raphe-projecting pyramidal neurons of the medial prefrontal cortex suppresses feeding of adolescent female mice undergoing activity-based anorexia. Brain Struct Funct 2022; 227:2127-2151. [PMID: 35635653 DOI: 10.1007/s00429-022-02507-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/30/2022] [Indexed: 12/19/2022]
Abstract
Anorexia Nervosa (AN) is characterized by voluntary food restriction, excessive exercise and extreme body weight loss. AN is particularly prevalent among adolescent females experiencing stress-induced anxiety. We used the animal model, activity-based anorexia (ABA), which captures these characteristics of AN, to reveal the neurobiology underlying individual differences in AN vulnerability. Dorsal raphe (DR) regulates feeding and is recruited when coping inescapable stress. Through chemogenetic activation, we investigated the role of mPFC pyramidal neurons projecting to DR (mPFC→DR) in adolescent female mice's decision to eat or exercise following ABA induction. Although the DREADD ligand C21 could activate 44% of the mPFC→DR neurons, this did not generate significant group mean difference in the amount of food intake, compared to control ABA mice without chemogenetic activation. However, analysis of individuals' responses to C21 revealed a significant, positive correlation between food intake and mPFC→DR neurons that co-express cFos, a marker for neuronal activity. cFos expression by GABAergic interneurons (GABA-IN) in mPFC was significantly greater than that for the control ABA mice, indicating recruitment of GABA-IN by mPFC→DR neurons. Electron microscopic immunohistochemistry revealed that GABAergic innervation is 60% greater for the PFC→DR neurons than adjacent Layer 5 pyramidal neurons without projections to DR. Moreover, individual differences in this innervation correlated negatively with food intake specifically on the day of C21 administration. We propose that C21 activates two antagonistic pathways: (1) PFC→DR pyramidal neurons that promote food intake; and (2) GABA-IN in the mPFC that dampen food intake through feedback inhibition of mPFC→DR neurons.
Collapse
Affiliation(s)
- Muzi Du
- Center for Neural Science, New York University, New York, NY, 10003, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Adrienne Santiago
- Center for Neural Science, New York University, New York, NY, 10003, USA.,New York State Psychiatric Institute, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Cenk Akiz
- Center for Neural Science, New York University, New York, NY, 10003, USA
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, NY, 10003, USA. .,Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
24
|
Claes M, De Groef L, Moons L. The DREADDful Hurdles and Opportunities of the Chronic Chemogenetic Toolbox. Cells 2022; 11:1110. [PMID: 35406674 PMCID: PMC8998042 DOI: 10.3390/cells11071110] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/10/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022] Open
Abstract
The chronic character of chemogenetics has been put forward as one of the assets of the technique, particularly in comparison to optogenetics. Yet, the vast majority of chemogenetic studies have focused on acute applications, while repeated, long-term neuromodulation has only been booming in the past few years. Unfortunately, together with the rising number of studies, various hurdles have also been uncovered, especially in relation to its chronic application. It becomes increasingly clear that chronic neuromodulation warrants caution and that the effects of acute neuromodulation cannot be extrapolated towards chronic experiments. Deciphering the underlying cellular and molecular causes of these discrepancies could truly unlock the chronic chemogenetic toolbox and possibly even pave the way for chemogenetics towards clinical application. Indeed, we are only scratching the surface of what is possible with chemogenetic research. For example, most investigations are concentrated on behavioral read-outs, whereas dissecting the underlying molecular signature after (chronic) neuromodulation could reveal novel insights in terms of basic neuroscience and deregulated neural circuits. In this review, we highlight the hurdles associated with the use of chemogenetic experiments, as well as the unexplored research questions for which chemogenetics offers the ideal research platform, with a particular focus on its long-term application.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
| | - Lies De Groef
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
- Laboratory of Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
25
|
Miura Y, Senoo A, Doura T, Kiyonaka S. Chemogenetics of cell surface receptors: beyond genetic and pharmacological approaches. RSC Chem Biol 2022; 3:269-287. [PMID: 35359495 PMCID: PMC8905536 DOI: 10.1039/d1cb00195g] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/20/2022] [Indexed: 11/29/2022] Open
Abstract
Cell surface receptors transmit extracellular information into cells. Spatiotemporal regulation of receptor signaling is crucial for cellular functions, and dysregulation of signaling causes various diseases. Thus, it is highly desired to control receptor functions with high spatial and/or temporal resolution. Conventionally, genetic engineering or chemical ligands have been used to control receptor functions in cells. As the alternative, chemogenetics has been proposed, in which target proteins are genetically engineered to interact with a designed chemical partner with high selectivity. The engineered receptor dissects the function of one receptor member among a highly homologous receptor family in a cell-specific manner. Notably, some chemogenetic strategies have been used to reveal the receptor signaling of target cells in living animals. In this review, we summarize the developing chemogenetic methods of transmembrane receptors for cell-specific regulation of receptor signaling. We also discuss the prospects of chemogenetics for clinical applications.
Collapse
Affiliation(s)
- Yuta Miura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Akinobu Senoo
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Shigeki Kiyonaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| |
Collapse
|
26
|
Characterization of DREADD receptor expression and function in rhesus macaques trained to discriminate ethanol. Neuropsychopharmacology 2022; 47:857-865. [PMID: 34654906 PMCID: PMC8882175 DOI: 10.1038/s41386-021-01181-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/10/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022]
Abstract
Circuit manipulation has been a staple technique in neuroscience to identify how the brain functions to control complex behaviors. Chemogenetics, including designer receptors exclusively activated by designer drugs (DREADDs), have proven to be a powerful tool for the reversible modulation of discrete brain circuitry without the need for implantable devices, thereby making them especially useful in awake and unrestrained animals. This study used a DREADD approach to query the role of the nucleus accumbens (NAc) in mediating the interoceptive effects of 1.0 g/kg ethanol (i.g.) in rhesus monkeys (n = 7) using a drug discrimination procedure. After training, stereotaxic surgery was performed to introduce an AAV carrying the human muscarinic 4 receptor DREADD (hM4Di) bilaterally into the NAc. The hypothesis was that decreasing the output of the NAc by activation of hM4Di with the DREADD actuator, clozapine-n-oxide (CNO), would potentiate the discriminative stimulus effect of ethanol (i.e., a leftward shift the ethanol dose discrimination curve). The results showed individual variability shifts of the ethanol dose-response determination under DREADD activation. Characterization of the expression and function of hM4Di with MRI, immunohistochemical, and electrophysiological techniques found the selectivity of NAc transduction was proportional to behavioral effect. Specifically, the proportion of hM4Di expression restricted to the NAc was associated with the potency of the discriminative stimulus effects of ethanol. Together, these experiments highlight the NAc in mediating the interoceptive effects of ethanol, provide a framework for validation of chemogenetic tools in primates, and underscore the importance of robust within-subjects examination of DREADD expression for interpretation of behavioral findings.
Collapse
|
27
|
Applications of chemogenetics in non-human primates. Curr Opin Pharmacol 2022; 64:102204. [DOI: 10.1016/j.coph.2022.102204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/10/2022] [Accepted: 02/11/2022] [Indexed: 11/23/2022]
|
28
|
Rodd ZA, Engleman EA, Truitt WA, Burke AR, Molosh AI, Bell RL, Hauser SR. CNO administration increases dopamine and glutamate in the medial prefrontal cortex of wistar rats: Further concerns for the validity of the CNO-activated DREADD procedure. Neuroscience 2022; 491:176-184. [DOI: 10.1016/j.neuroscience.2022.03.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/03/2022] [Accepted: 03/23/2022] [Indexed: 12/14/2022]
|
29
|
Cox SS, Kearns AM, Woods SK, Brown BJ, Brown SJ, Reichel CM. The role of the anterior insular during targeted helping behavior in male rats. Sci Rep 2022; 12:3315. [PMID: 35228625 PMCID: PMC8885669 DOI: 10.1038/s41598-022-07365-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/17/2022] [Indexed: 12/30/2022] Open
Abstract
Empathy, the understanding of the emotional state of others, can be examined across species using the Perception Action Model, where shared affect promotes an action by "Observers" to aid a distressed "Target". The anterior insula (AI) has garnered interest in empathic behavior due to its role integrating sensory and emotional information of self and other. In the following studies, the AI was inhibited pharmacologically and chemogenetically during targeted helping. We demonstrate the insula is active during, and is necessary for the maintenance of, targeted helping. Analysis of ultrasonic vocalizations revealed distress calls from Targets increased when Observers' helping was attenuated due to insula inhibition. Targets' elevated distress was directly correlated to Observers' diminished helping behavior, suggesting emotional transfer between Observer and Target is blunted following Observer AI inhibition. Finally, the AI may selectively blunt targeted helping, as social exploration did not change in a social reward place conditioning task. These studies help further establish the anterior insula as a critical node in the empathic brain during targeted helping, even in the absence of direct social contact.
Collapse
Affiliation(s)
- Stewart S Cox
- Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building Suite 416a, Charleston, SC, 29425, USA.
| | - Angela M Kearns
- Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building Suite 416a, Charleston, SC, 29425, USA
| | - Samuel K Woods
- Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building Suite 416a, Charleston, SC, 29425, USA
| | - Brogan J Brown
- Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building Suite 416a, Charleston, SC, 29425, USA
| | - Samantha J Brown
- Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building Suite 416a, Charleston, SC, 29425, USA
| | - Carmela M Reichel
- Medical University of South Carolina, 173 Ashley Avenue, Basic Science Building Suite 416a, Charleston, SC, 29425, USA.
| |
Collapse
|
30
|
Godoy LD, Prizon T, Rossignoli MT, Leite JP, Liberato JL. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Front Integr Neurosci 2022; 16:765324. [PMID: 35250498 PMCID: PMC8891758 DOI: 10.3389/fnint.2022.765324] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Parvalbumin is a calcium-binding protein present in inhibitory interneurons that play an essential role in regulating many physiological processes, such as intracellular signaling and synaptic transmission. Changes in parvalbumin expression are deeply related to epilepsy, which is considered one of the most disabling neuropathologies. Epilepsy is a complex multi-factor group of disorders characterized by periods of hypersynchronous activity and hyperexcitability within brain networks. In this scenario, inhibitory neurotransmission dysfunction in modulating excitatory transmission related to the loss of subsets of parvalbumin-expressing inhibitory interneuron may have a prominent role in disrupted excitability. Some studies also reported that parvalbumin-positive interneurons altered function might contribute to psychiatric comorbidities associated with epilepsy, such as depression, anxiety, and psychosis. Understanding the epileptogenic process and comorbidities associated with epilepsy have significantly advanced through preclinical and clinical investigation. In this review, evidence from parvalbumin altered function in epilepsy and associated psychiatric comorbidities were explored with a translational perspective. Some advances in potential therapeutic interventions are highlighted, from current antiepileptic and neuroprotective drugs to cutting edge modulation of parvalbumin subpopulations using optogenetics, designer receptors exclusively activated by designer drugs (DREADD) techniques, transcranial magnetic stimulation, genome engineering, and cell grafting. Creating new perspectives on mechanisms and therapeutic strategies is valuable for understanding the pathophysiology of epilepsy and its psychiatric comorbidities and improving efficiency in clinical intervention.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- João Pereira Leite,
| | - José Luiz Liberato
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: José Luiz Liberato,
| |
Collapse
|
31
|
Grossman CD, Bari BA, Cohen JY. Serotonin neurons modulate learning rate through uncertainty. Curr Biol 2022; 32:586-599.e7. [PMID: 34936883 PMCID: PMC8825708 DOI: 10.1016/j.cub.2021.12.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 10/11/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
Abstract
Regulating how fast to learn is critical for flexible behavior. Learning about the consequences of actions should be slow in stable environments, but accelerate when that environment changes. Recognizing stability and detecting change are difficult in environments with noisy relationships between actions and outcomes. Under these conditions, theories propose that uncertainty can be used to modulate learning rates ("meta-learning"). We show that mice behaving in a dynamic foraging task exhibit choice behavior that varied as a function of two forms of uncertainty estimated from a meta-learning model. The activity of dorsal raphe serotonin neurons tracked both types of uncertainty in the foraging task as well as in a dynamic Pavlovian task. Reversible inhibition of serotonin neurons in the foraging task reproduced changes in learning predicted by a simulated lesion of meta-learning in the model. We thus provide a quantitative link between serotonin neuron activity, learning, and decision making.
Collapse
Affiliation(s)
- Cooper D Grossman
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Bilal A Bari
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Jeremiah Y Cohen
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
32
|
Desloovere J, Boon P, Larsen LE, Goossens MG, Delbeke J, Carrette E, Wadman W, Vonck K, Raedt R. Chemogenetic Seizure Control with Clozapine and the Novel Ligand JHU37160 Outperforms the Effects of Levetiracetam in the Intrahippocampal Kainic Acid Mouse Model. Neurotherapeutics 2022; 19:342-351. [PMID: 34862591 PMCID: PMC9130374 DOI: 10.1007/s13311-021-01160-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 01/03/2023] Open
Abstract
Expression of inhibitory designer receptors exclusively activated by designer drugs (DREADDs) on excitatory hippocampal neurons in the hippocampus represents a potential new therapeutic strategy for drug-resistant epilepsy. To overcome the limitations of the commonly used DREADD agonist clozapine, we investigated the efficacy of the novel DREADD ligand JHU37160 in chemogenetic seizure suppression in the intrahippocampal kainic acid (IHKA) mouse model for temporal lobe epilepsy (TLE). In addition, seizure-suppressing effects of chemogenetics were compared to the commonly used anti-epileptic drug (AED), levetiracetam (LEV). Therefore, an adeno-associated viral vector was injected in the sclerotic hippocampus of IHKA mice to induce expression of a tagged inhibitory DREADD hM4Di or only a tag (control) specifically in excitatory neurons using the CamKIIα promoter. Subsequently, animals were treated with LEV (800 mg/kg), clozapine (0.1 mg/kg), and DREADD ligand JHU37160 (0.1 mg/kg) and the effect on spontaneous seizures was investigated. Clozapine and JHU37160-mediated chemogenetic treatment both suppressed seizures in DREADD-expressing IHKA mice. Clozapine treatment suppressed seizures up to 34 h after treatment, and JHU37160 effects lasted for 26 h after injection. Moreover, both compounds reduced the length of seizures that did occur after treatment up to 28 h and 18 h after clozapine and JHU37160, respectively. No seizure-suppressing effects were found in control animals using these ligands. Chemogenetic seizure treatment suppressed seizures during the first 30 min after injection, and seizures remained suppressed during 8 h following treatment. Chemogenetics thus outperformed effects of levetiracetam (p < 0.001), which suppressed seizure frequency with a maximum of 55 ± 9% for up to 1.5 h (p < 0.05). Only chemogenetic and not levetiracetam treatment affected the length of seizures after treatment (p < 0.001). These results show that the chemogenetic therapeutic strategy with either clozapine or JHU37160 effectively suppresses spontaneous seizures in the IHKA mouse model, confirming JHU37160 as an effective DREADD ligand. Moreover, chemogenetic therapy outperforms the effects of levetiracetam, indicating its potential to suppress drug-resistant seizures.
Collapse
Affiliation(s)
- Jana Desloovere
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Paul Boon
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Lars Emil Larsen
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Medical Image and Signal Processing, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | | | - Jean Delbeke
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Evelien Carrette
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Wytse Wadman
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Kristl Vonck
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Robrecht Raedt
- 4BRAIN, Department of Neurology, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
33
|
Fazekas CL, Bellardie M, Török B, Sipos E, Tóth B, Baranyi M, Sperlágh B, Dobos-Kovács M, Chaillou E, Zelena D. Pharmacogenetic excitation of the median raphe region affects social and depressive-like behavior and core body temperature in male mice. Life Sci 2021; 286:120037. [PMID: 34637795 DOI: 10.1016/j.lfs.2021.120037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/27/2021] [Accepted: 10/05/2021] [Indexed: 11/29/2022]
Abstract
AIMS Median raphe region (MRR) is an important bottom-up regulatory center for various behaviors as well as vegetative functions, but detailed descriptions and links between the two are still largely unexplored. METHODS Pharmacogenetics was used to study the role of MRR in social (sociability, social interaction, resident intruder test) and emotional behavior (forced swim test) parallel with some vegetative changes (biotelemetry: core body temperature). Additionally, to validate pharmacogenetics, the effect of clozapine-N-oxide (CNO), the ligand of the artificial receptor, was studied by measuring (i) serum and brainstem concentrations of CNO and clozapine; (ii) MRR stimulation induced neurotransmitter release in hippocampus; (iii) CNO induced changes in body temperature and locomotor activity. KEY FINDINGS MRR stimulation decreased locomotion, increased friendly social behavior in the resident intruder test and enhanced depressive-like behavior. The latter was accompanied by diminished decrease in core body temperature. Thirty minutes after CNO injection clozapine was predominant in the brainstem. Nonetheless, peripheral CNO injection was able to induce glutamate release in the hippocampus. CNO had no immediate (<30 min) or chronic (repeated injections) effect on the body temperature or locomotion. SIGNIFICANCE We confirmed the role of MRR in locomotion, social and depressive-like behavior. Most interestingly, only depressive-like behavior was accompanied by changed body temperature regulation, which was also observed in human depressive disorders previously. This indicates clinical relevance of our findings. Despite low penetration, CNO acts centrally, but does not influence the examined basic parameters, being suitable for repeated behavioral testing.
Collapse
Affiliation(s)
- Csilla Lea Fazekas
- Institute of Experimental Medicine, Budapest, Hungary; János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary.
| | - Manon Bellardie
- Institute of Experimental Medicine, Budapest, Hungary; INRAE Centre Val de Loire, CNRS, IFCE, Université de Tours, UMR 85 Physiologie de la Reproduction et des Comportements, France
| | - Bibiána Török
- Institute of Experimental Medicine, Budapest, Hungary; János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Eszter Sipos
- Institute of Experimental Medicine, Budapest, Hungary
| | - Blanka Tóth
- Budapest University of Technology and Economics, Faculty of Chemical Technology and Biotechnology, Department of Inorganic and Analytical Chemistry, Budapest, Hungary
| | - Mária Baranyi
- Institute of Experimental Medicine, Budapest, Hungary
| | | | | | - Elodie Chaillou
- INRAE Centre Val de Loire, CNRS, IFCE, Université de Tours, UMR 85 Physiologie de la Reproduction et des Comportements, France
| | - Dóra Zelena
- Institute of Experimental Medicine, Budapest, Hungary; Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
34
|
Sternson SM, Bleakman D. Chemogenetics: drug-controlled gene therapies for neural circuit disorders. ACTA ACUST UNITED AC 2021; 6:1079-1094. [PMID: 34422319 PMCID: PMC8376173 DOI: 10.18609/cgti.2020.112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Many patients with nervous system disorders have considerable unmet clinical needs or suffer debilitating drug side effects. A major limitation of exiting treatment approaches is that traditional small molecule pharmacotherapy lacks sufficient specificity to effectively treat many neurological diseases. Chemogenetics is a new gene therapy technology that targets an engineered receptor to cell types involved in nervous system dysfunction, enabling highly selective drug-controlled neuromodulation. Here, we discuss chemogenetic platforms and considerations for their potential application as human nervous system therapies.
Collapse
Affiliation(s)
- Scott M Sternson
- Janelia Research Campus, HHMI, 19700 Helix Dr. Ashburn, VA 20147, USA
| | - David Bleakman
- Redpin Therapeutics, 1329, Madison Avenue, Suite 125, New York, NY 10029, USA
| |
Collapse
|
35
|
Van Steenbergen V, Bareyre FM. Chemogenetic approaches to unravel circuit wiring and related behavior after spinal cord injury. Exp Neurol 2021; 345:113839. [PMID: 34389362 DOI: 10.1016/j.expneurol.2021.113839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/30/2021] [Accepted: 08/08/2021] [Indexed: 01/21/2023]
Abstract
A critical shortcoming of the central nervous system is its limited ability to repair injured nerve connections. Trying to overcome this limitation is not only relevant to understand basic neurobiological principles but also holds great promise to advance therapeutic strategies related, in particular, to spinal cord injury (SCI). With barely any SCI patients re-gaining complete neurological function, there is a high need to understand how we could target and improve spinal plasticity to re-establish neuronal connections into a functional network. The development of chemogenetic tools has proven to be of great value to understand functional circuit wiring before and after injury and to correlate novel circuit formation with behavioral outcomes. This review covers commonly used chemogenetic approaches based on metabotropic receptors and their use to improve our understanding of circuit wiring following spinal cord injury.
Collapse
Affiliation(s)
- Valérie Van Steenbergen
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany; Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg-Martinsried, Germany.
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, 81377 Munich, Germany; Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, 82152 Planegg-Martinsried, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
36
|
Klink PC, Aubry JF, Ferrera VP, Fox AS, Froudist-Walsh S, Jarraya B, Konofagou EE, Krauzlis RJ, Messinger A, Mitchell AS, Ortiz-Rios M, Oya H, Roberts AC, Roe AW, Rushworth MFS, Sallet J, Schmid MC, Schroeder CE, Tasserie J, Tsao DY, Uhrig L, Vanduffel W, Wilke M, Kagan I, Petkov CI. Combining brain perturbation and neuroimaging in non-human primates. Neuroimage 2021; 235:118017. [PMID: 33794355 PMCID: PMC11178240 DOI: 10.1016/j.neuroimage.2021.118017] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Brain perturbation studies allow detailed causal inferences of behavioral and neural processes. Because the combination of brain perturbation methods and neural measurement techniques is inherently challenging, research in humans has predominantly focused on non-invasive, indirect brain perturbations, or neurological lesion studies. Non-human primates have been indispensable as a neurobiological system that is highly similar to humans while simultaneously being more experimentally tractable, allowing visualization of the functional and structural impact of systematic brain perturbation. This review considers the state of the art in non-human primate brain perturbation with a focus on approaches that can be combined with neuroimaging. We consider both non-reversible (lesions) and reversible or temporary perturbations such as electrical, pharmacological, optical, optogenetic, chemogenetic, pathway-selective, and ultrasound based interference methods. Method-specific considerations from the research and development community are offered to facilitate research in this field and support further innovations. We conclude by identifying novel avenues for further research and innovation and by highlighting the clinical translational potential of the methods.
Collapse
Affiliation(s)
- P Christiaan Klink
- Department of Vision & Cognition, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands.
| | - Jean-François Aubry
- Physics for Medicine Paris, Inserm U1273, CNRS UMR 8063, ESPCI Paris, PSL University, Paris, France
| | - Vincent P Ferrera
- Department of Neuroscience & Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Andrew S Fox
- Department of Psychology & California National Primate Research Center, University of California, Davis, CA, USA
| | | | - Béchir Jarraya
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France; Foch Hospital, UVSQ, Suresnes, France
| | - Elisa E Konofagou
- Ultrasound and Elasticity Imaging Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Richard J Krauzlis
- Laboratory of Sensorimotor Research, National Eye Institute, Bethesda, MD, USA
| | - Adam Messinger
- Laboratory of Brain and Cognition, National Institute of Mental Health, Bethesda, MD, USA
| | - Anna S Mitchell
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom
| | - Michael Ortiz-Rios
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Hiroyuki Oya
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neurosurgery, University of Iowa, Iowa city, IA, USA
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, United Kingdom
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | | | - Jérôme Sallet
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom; Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France; Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Michael Christoph Schmid
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, CH-1700 Fribourg, Switzerland
| | - Charles E Schroeder
- Nathan Kline Institute, Orangeburg, NY, USA; Columbia University, New York, NY, USA
| | - Jordy Tasserie
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Doris Y Tsao
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience; Howard Hughes Medical Institute; Computation and Neural Systems, Caltech, Pasadena, CA, USA
| | - Lynn Uhrig
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, Neurosciences Department, KU Leuven Medical School, Leuven, Belgium; Leuven Brain Institute, KU Leuven, Leuven Belgium; Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Melanie Wilke
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Department of Cognitive Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Igor Kagan
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.
| | - Christopher I Petkov
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom.
| |
Collapse
|
37
|
Morelli C, Castaldi L, Brown SJ, Streich LL, Websdale A, Taberner FJ, Cerreti B, Barenghi A, Blum KM, Sawitzke J, Frank T, Steffens LK, Doleschall B, Serrao J, Ferrarini D, Lechner SG, Prevedel R, Heppenstall PA. Identification of a population of peripheral sensory neurons that regulates blood pressure. Cell Rep 2021; 35:109191. [PMID: 34077727 PMCID: PMC8187988 DOI: 10.1016/j.celrep.2021.109191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/14/2020] [Accepted: 05/09/2021] [Indexed: 02/08/2023] Open
Abstract
The vasculature is innervated by a network of peripheral afferents that sense and regulate blood flow. Here, we describe a system of non-peptidergic sensory neurons with cell bodies in the spinal ganglia that regulate vascular tone in the distal arteries. We identify a population of mechanosensitive neurons, marked by tropomyosin receptor kinase C (TrkC) and tyrosine hydroxylase in the dorsal root ganglia, which projects to blood vessels. Local stimulation of TrkC neurons decreases vessel diameter and blood flow, whereas systemic activation increases systolic blood pressure and heart rate variability via the sympathetic nervous system. Ablation of the neurons provokes variability in local blood flow, leading to a reduction in systolic blood pressure, increased heart rate variability, and ultimately lethality within 48 h. Thus, a population of TrkC+ sensory neurons forms part of a sensory-feedback mechanism that maintains cardiovascular homeostasis through the autonomic nervous system. TrkC+/Th+ DRG neurons project to blood vessels Local stimulation of TrkC+ DRG neurons decreases vessel diameter and blood flow Systemic activation of TrkC+ DRG neurons increases blood pressure and heart rate Ablation of TrkC+ neurons dysregulates cardiovascular homeostasis and is lethal
Collapse
Affiliation(s)
- Chiara Morelli
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany; Collaboration for joint PhD degree between EMBL Heidelberg, Heidelberg, Germany, and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Laura Castaldi
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Sam J Brown
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy
| | - Lina L Streich
- Collaboration for joint PhD degree between EMBL Heidelberg, Heidelberg, Germany, and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Francisco J Taberner
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | | | | | - Kevin M Blum
- Center for Regenerative Medicine, the Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Biomedical Engineering, the Ohio State University, Columbus, OH, USA
| | | | - Tessa Frank
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy
| | - Laura K Steffens
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Joana Serrao
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy
| | | | - Stefan G Lechner
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Robert Prevedel
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany; Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Paul A Heppenstall
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany.
| |
Collapse
|
38
|
Ozawa A, Arakawa H. Chemogenetics drives paradigm change in the investigation of behavioral circuits and neural mechanisms underlying drug action. Behav Brain Res 2021; 406:113234. [PMID: 33741409 PMCID: PMC8110310 DOI: 10.1016/j.bbr.2021.113234] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Recent developments in chemogenetic approaches to the investigation of brain function have ushered in a paradigm change in the strategy for drug and behavior research and clinical drug-based medications. As the nature of the drug action is based on humoral regulation, it is a challenge to identify the neuronal mechanisms responsible for the expression of certain targeted behavior induced by drug application. The development of chemogenetic approaches has allowed researchers to control neural activities in targeted neurons through a toolbox, including engineered G protein-coupled receptors or ligand-gated ion channels together with exogenously inert synthetic ligands. This review provides a brief overview of the chemogenetics toolbox with an emphasis on the DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) technique used in rodent models, which is applicable to the investigation of how specific neural circuits regulate behavioral processes. The use of chemogenetics has had a significant impact on basic neuroscience for a better understanding of the relationships between brain activity and the expression of behaviors with cell- and circuit-specific orders. Furthermore, chemogenetics is potentially a useful tool to deconstruct the neuropathological mechanisms of mental diseases and its regulation by drug, and provide us with transformative therapeutics with medication. We also review recent findings in the use of chemogenetic techniques to uncover functional circuit connections of serotonergic neurons in rodent models.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, USA
| | - Hiroyuki Arakawa
- Department of Psychology, Tokiwa University, Mito, Ibaraki, Japan; Department of Systems Physiology, University of Ryukyus, Faculty of Medicine, Nakagami District, Okinawa, Japan.
| |
Collapse
|
39
|
Guo W, Wan X, Ma L, Zhang J, Hashimoto K. Abnormalities in the composition of the gut microbiota in mice after repeated administration of DREADD ligands. Brain Res Bull 2021; 173:66-73. [PMID: 34004259 DOI: 10.1016/j.brainresbull.2021.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/06/2021] [Accepted: 05/13/2021] [Indexed: 02/08/2023]
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are known as genetically modified G-protein-coupled receptors (GPCRs), which can be activated by synthetic ligands such as clozapine N-oxide (CNO) and DREADD agonist 21 (compound 21: C21). The brain-gut-microbiota axis has a crucial role in bidirectional interactions between the brain and the gastrointestinal microbiota. In this study, we investigated whether repeated administration of CNO or C21 could influence the gut microbiota and short-chain fatty acids (SCFAs) in feces of adult mice. Repeated administration of CNO or C21 as drinking water did not alter the α- and β-diversity of gut microbiota in mice compared with control mice. However, we found significant changes in relative abundance for several bacteria in the CNO (or C21) group at the taxonomic level compared to the control group. The linear discriminant analysis effect size (LEfSe) algorithm distinguished the family Prevotellaceae, the genus Anaerocolumna, the genus Prevotella, and the genus Frisingicoccus, these four specific microbial markers for the CNO group relative to the control group. In addition, the LEfSe algorithm identified the family Clostridiaceae, the genus Faecalicatena and the genus Marinisporobacter, these three bacteria of different taxonomic as potential microbial markers for the C21 group relative to the control group. In contrast, repeated administration of CNO (or C21) did not alter SCFAs in feces samples of adult mice. The data suggest that repeated administration of CNO or C21 contributes to an unusual organization of the gut microbiota in adult mice. Therefore, abnormalities in the composition of gut microbiota by repeated dosing of DREADD ligands should be taken into consideration for behavioral and biological functions in rodents treated with DREADD ligands.
Collapse
Affiliation(s)
- Wei Guo
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Xiayun Wan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Li Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Jiancheng Zhang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
40
|
Santiago AN, Makowicz EA, Du M, Aoki C. Food Restriction Engages Prefrontal Corticostriatal Cells and Local Microcircuitry to Drive the Decision to Run versus Conserve Energy. Cereb Cortex 2021; 31:2868-2885. [PMID: 33497440 DOI: 10.1093/cercor/bhaa394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/31/2022] Open
Abstract
Food restriction (FR) evokes running, which may promote adaptive foraging in times of food scarcity, but can become lethal if energy expenditure exceeds caloric availability. Here, we demonstrate that chemogenetic activation of either the general medial prefrontal cortex (mPFC) pyramidal cell population, or the subpopulation projecting to dorsal striatum (DS) drives running specifically during hours preceding limited food availability, and not during ad libitum food availability. Conversely, suppression of mPFC pyramidal cells generally, or targeting mPFC-to-DS cells, reduced wheel running specifically during FR and not during ad libitum food access. Post mortem c-Fos analysis and electron microscopy of mPFC layer 5 revealed distinguishing characteristics of mPFC-to-DS cells, when compared to neighboring non-DS-projecting pyramidal cells: 1) greater recruitment of GABAergic activity and 2) less axo-somatic GABAergic innervation. Together, these attributes position the mPFC-to-DS subset of pyramidal cells to dominate mPFC excitatory outflow, particularly during FR, revealing a specific and causal role for mPFC-to-DS control of the decision to run during food scarcity. Individual differences in GABAergic activity correlate with running response to further support this interpretation. FR enhancement of PFC-to-DS activity may influence neural circuits both in studies using FR to motivate animal behavior and in human conditions hallmarked by FR.
Collapse
Affiliation(s)
- Adrienne N Santiago
- Center for Neural Science, New York University, 4 Washington place, New York, NY 10003, USA
| | - Emily A Makowicz
- Center for Neural Science, New York University, 4 Washington place, New York, NY 10003, USA.,Hunter College, City University of New York, 695 Park Ave, New York, NY, 10065, USA
| | - Muzi Du
- Center for Neural Science, New York University, 4 Washington place, New York, NY 10003, USA.,Langone Neuroscience Institute, New York University, 435 East 30th St, New York, NY 10016, USA
| | - Chiye Aoki
- Center for Neural Science, New York University, 4 Washington place, New York, NY 10003, USA.,New York University Shanghai, 1555 Century Ave, Pudong, Shanghai 200122, China
| |
Collapse
|
41
|
Boehm MA, Bonaventura J, Gomez JL, Solís O, Stein EA, Bradberry CW, Michaelides M. Translational PET applications for brain circuit mapping with transgenic neuromodulation tools. Pharmacol Biochem Behav 2021; 204:173147. [PMID: 33549570 PMCID: PMC8297666 DOI: 10.1016/j.pbb.2021.173147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023]
Abstract
Transgenic neuromodulation tools have transformed the field of neuroscience over the past two decades by enabling targeted manipulation of neuronal populations and circuits with unprecedented specificity. Chemogenetic and optogenetic neuromodulation systems are among the most widely used and allow targeted control of neuronal activity through the administration of a selective compound or light, respectively. Innovative genetic targeting strategies are utilized to transduce specific cells to express transgenic receptors and opsins capable of manipulating neuronal activity. These allow mapping of neuroanatomical projection sites and link cellular manipulations with brain circuit functions and behavior. As these tools continue to expand knowledge of the nervous system in preclinical models, developing translational applications for human therapies is becoming increasingly possible. However, new strategies for implementing and monitoring transgenic tools are needed for safe and effective use in translational research and potential clinical applications. A major challenge for such applications is the need to track the location and function of chemogenetic receptors and opsins in vivo, and new developments in positron emission tomography (PET) imaging techniques offer promising solutions. The goal of this review is to summarize current research combining transgenic tools with PET for in vivo mapping and manipulation of brain circuits and to propose future directions for translational applications.
Collapse
Affiliation(s)
- Matthew A Boehm
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States; Department of Neuroscience, Brown University, Providence, RI 02906, United States.
| | - Jordi Bonaventura
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Juan L Gomez
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Oscar Solís
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Elliot A Stein
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Charles W Bradberry
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Michael Michaelides
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States; Department of Psychiatry & Behavioral Sciences, Johns Hopkins Medicine, Baltimore, MD, 21205, United States.
| |
Collapse
|
42
|
Kumar V. Designed Synthesis of Diversely Substituted Hydantoins and Hydantoin-Based Hybrid Molecules: A Personal Account. Synlett 2021. [DOI: 10.1055/a-1480-6474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractHydantoin and its analogues such as thiohydantoin and iminohydantoin have received substantial attention from both a chemical and a biological point of view. Several compounds of this class have shown useful pharmacological activities such as anticonvulsant, antitumor, antiarrhythmic, and herbicidal properties that have led, in some cases, to clinical applications. Because of these broad-spectrum activities, intensive research efforts have been dedicated in industry and academia to the synthesis and structural modifications of hydantoin and its derivatives. Realizing the importance of hydantoin in organic and medicinal chemistry, we also initiated a research program that successfully designed and developed new routes and methods for the formation of hydantoin, thiohydantoin, and iminohydantoin substituted at various positions, particularly at the N-1 position without following a protection–deprotection strategy. Because combinations of two or more pharmacophoric groups can lead to hybrid molecules that display a mixed mechanism of action on biological targets, we extended our developed strategy to the syntheses of new types of hydantoin-based hybrid molecules by combining hydantoin with a triazole, isoxazoline, or phosphate scaffold as a second pharmacophore to exploit their diverse biological functions.1 Introduction2 Chemistry and Properties2.1 Physical Properties2.2 Chemical Properties2.3 Biological Properties3 General Synthetic Methods4 Synthesis of Diversely Substituted Hydantoins5 Synthesis of Diversely Substituted Thiohydantoins6 Synthesis of Diversely Substituted Iminohydantoins7 Fused or Bicyclic (Thio)hydantoins8 Di- or Multivalent (Thio)hydantoins9 Hydantoin-Based Hybrid Molecules9.1 Hydantoin–Isooxazoline Hybrids9.2 Hydantoin–Triazole Hybrids9.3 Hydantoin–Phosphate Hybrids: Phosphorylated Hydantoins10 Summary and Outlook
Collapse
|
43
|
Meister J, Wang L, Pydi SP, Wess J. Chemogenetic approaches to identify metabolically important GPCR signaling pathways: Therapeutic implications. J Neurochem 2021; 158:603-620. [PMID: 33540469 DOI: 10.1111/jnc.15314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 12/16/2022]
Abstract
DREADDs (Designer Receptors Exclusively Activated by a Designer Drug) are designer G protein-coupled receptors (GPCRs) that are widely used in the neuroscience field to modulate neuronal activity. In this review, we will focus on DREADD studies carried out with genetically engineered mice aimed at elucidating signaling pathways important for maintaining proper glucose and energy homeostasis. The availability of muscarinic receptor-based DREADDs endowed with selectivity for one of the four major classes of heterotrimeric G proteins (Gs , Gi , Gq , and G12 ) has been instrumental in dissecting the physiological and pathophysiological roles of distinct G protein signaling pathways in metabolically important cell types. The novel insights gained from this work should inform the development of novel classes of drugs useful for the treatment of several metabolic disorders including type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| |
Collapse
|
44
|
Goossens MG, Boon P, Wadman W, Van den Haute C, Baekelandt V, Verstraete AG, Vonck K, Larsen LE, Sprengers M, Carrette E, Desloovere J, Meurs A, Delbeke J, Vanhove C, Raedt R. Long-term chemogenetic suppression of seizures in a multifocal rat model of temporal lobe epilepsy. Epilepsia 2021; 62:659-670. [PMID: 33570167 DOI: 10.1111/epi.16840] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 01/14/2023]
Abstract
OBJECTIVE One third of epilepsy patients do not become seizure-free using conventional medication. Therefore, there is a need for alternative treatments. Preclinical research using designer receptors exclusively activated by designer drugs (DREADDs) has demonstrated initial success in suppressing epileptic activity. Here, we evaluated whether long-term chemogenetic seizure suppression could be obtained in the intraperitoneal kainic acid rat model of temporal lobe epilepsy, when DREADDs were selectively expressed in excitatory hippocampal neurons. METHODS Epileptic male Sprague Dawley rats received unilateral hippocampal injections of adeno-associated viral vector encoding the inhibitory DREADD hM4D(Gi), preceded by a cell-specific promotor targeting excitatory neurons. The effect of clozapine-mediated DREADD activation on dentate gyrus evoked potentials and spontaneous electrographic seizures was evaluated. Animals were systemically treated with single (.1 mg/kg/24 h) or repeated (.1 mg/kg/6 h) injections of clozapine. In addition, long-term continuous release of clozapine and olanzapine (2.8 mg/kg/7 days) using implantable minipumps was evaluated. All treatments were administered during the chronic epileptic phase and between 1.5 and 13.5 months after viral transduction. RESULTS In the DREADD group, dentate gyrus evoked potentials were inhibited after clozapine treatment. Only in DREADD-expressing animals, clozapine reduced seizure frequency during the first 6 h postinjection. When administered repeatedly, seizures were suppressed during the entire day. Long-term treatment with clozapine and olanzapine both resulted in significant seizure-suppressing effects for multiple days. Histological analysis revealed DREADD expression in both hippocampi and some cortical regions. However, lesions were also detected at the site of vector injection. SIGNIFICANCE This study shows that inhibition of the hippocampus using chemogenetics results in potent seizure-suppressing effects in the intraperitoneal kainic acid rat model, even 1 year after viral transduction. Despite a need for further optimization, chemogenetic neuromodulation represents a promising treatment prospect for temporal lobe epilepsy.
Collapse
Affiliation(s)
| | - Paul Boon
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Wytse Wadman
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Center for Molecular Medicine and Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Leuven Viral Vector Core, Center for Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Center for Molecular Medicine and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Alain G Verstraete
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kristl Vonck
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Lars E Larsen
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Mathieu Sprengers
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Evelien Carrette
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Jana Desloovere
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Alfred Meurs
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Jean Delbeke
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- IBiTech, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Robrecht Raedt
- 4BRAIN, Department of Head and Skin, Ghent University, Ghent, Belgium
| |
Collapse
|
45
|
Chemogenetic manipulation of astrocytic activity: Is it possible to reveal the roles of astrocytes? Biochem Pharmacol 2021; 186:114457. [PMID: 33556341 DOI: 10.1016/j.bcp.2021.114457] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 01/08/2023]
Abstract
Astrocytes are the major glial cells in the central nervous system, but unlike neurons, they do not produce action potentials. For many years, astrocytes were considered supporting cells in the central nervous system (CNS). Technological advances over the last two decades are changing the face of glial research. Accumulating data from recent investigations show that astrocytes display transient calcium spikes and regulate synaptic transmission by releasing transmitters called gliotransmitters. Many new powerful technologies are used to interfere with astrocytic activity, in order to obtain a better understanding of the roles of astrocytes in the brain. Among these technologies, chemogenetics has recently been used frequently. In this review, we will summarize new functions of astrocytes in the brain that have been revealed using this cutting-edge technique. Moreover, we will discuss the possibilities and challenges of manipulating astrocytic activity using this technology.
Collapse
|
46
|
Maldonado R, Calvé P, García-Blanco A, Domingo-Rodriguez L, Senabre E, Martín-García E. Vulnerability to addiction. Neuropharmacology 2021; 186:108466. [PMID: 33482225 DOI: 10.1016/j.neuropharm.2021.108466] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
Addiction is a chronic brain disease that has dramatic health and socioeconomic consequences worldwide. Multiple approaches have been used for decades to clarify the neurobiological basis of this disease and to identify novel potential treatments. This review summarizes the main brain networks involved in the vulnerability to addiction and specific innovative technological approaches to investigate these neural circuits. First, the evolution of the definition of addiction across the Diagnostic and Statistical Manual of Mental Disorders (DSM) is revised. We next discuss several innovative experimental techniques that, combined with behavioral approaches, have allowed recent critical advances in understanding the neural circuits involved in addiction, including DREADDs, calcium imaging, and electrophysiology. All these techniques have been used to investigate specific neural circuits involved in vulnerability to addiction and have been extremely useful to clarify the neurobiological basis of each specific component of the addictive process. These novel tools targeting specific brain regions are of great interest to further understand the different aspects of this complex disease. This article is part of the special issue on 'Vulnerabilities to Substance Abuse.'.
Collapse
Affiliation(s)
- R Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain; Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| | - P Calvé
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - A García-Blanco
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - L Domingo-Rodriguez
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - E Senabre
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - E Martín-García
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
47
|
Hu F, Morris PJ, Bonaventura J, Fan H, Mathews WB, Holt DP, Lam S, Boehm M, Dannals RF, Pomper MG, Michaelides M, Horti AG. 18F-labeled radiotracers for in vivo imaging of DREADD with positron emission tomography. Eur J Med Chem 2020; 213:113047. [PMID: 33280897 DOI: 10.1016/j.ejmech.2020.113047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/08/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022]
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADD) are a preclinical chemogenetic approach with clinical potential for various disorders. In vivo visualization of DREADDs has been achieved with positron emission tomography (PET) using 11C radiotracers. The objective of this study was to develop DREADD radiotracers labeled with 18F for a longer isotope half-life. A series of non-radioactive fluorinated analogs of clozapine with a wide range of in vitro binding affinities for the hM3Dq and hM4Di DREADD receptors has been synthesized for PET. Compound [18F]7b was radiolabeled via a modified 18F-deoxyfluorination protocol with a commercial ruthenium reagent. [18F]7b demonstrated encouraging PET imaging properties in a DREADD hM3Dq transgenic mouse model, whereas the radiotracer uptake in the wild type mouse brain was low. [18F]7b is a promising long-lived alternative to the DREADD radiotracers [11C]clozapine ([11C]CLZ) and [11C]deschloroclozapine ([11C]DCZ).
Collapse
Affiliation(s)
- Feng Hu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Patrick J Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Hong Fan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - William B Mathews
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Daniel P Holt
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Sherry Lam
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Matthew Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Robert F Dannals
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Martin G Pomper
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA; Department of Psychiatry and behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 221205, USA
| | - Andrew G Horti
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
48
|
Kahn JB, Port RG, Anderson SA, Coulter DA. Modular, Circuit-Based Interventions Rescue Hippocampal-Dependent Social and Spatial Memory in a 22q11.2 Deletion Syndrome Mouse Model. Biol Psychiatry 2020; 88:710-718. [PMID: 32682567 PMCID: PMC7554065 DOI: 10.1016/j.biopsych.2020.04.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/09/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND 22q11.2 deletion syndrome (22qDS) manifests with myriad symptoms, including multiple neuropsychiatric disorders. Complications associated with the polygenic haploinsufficiency make 22qDS symptoms particularly difficult to manage with traditional therapeutic approaches. However, the varying mechanistic consequences often culminate to generate inappropriate regulation of neuronal circuit activity. We explored whether managing this aberrant activity in adults could be a therapeutically beneficial strategy. METHODS To assess and dissect hippocampal circuit function, we performed functional imaging in acute slices and targeted eloquent circuits (specific subcircuits tied to specific behavioral tasks) to provide relevant behavioral outputs. For example, the ventral and dorsal CA1 regions critically support social and spatial discrimination, respectively. We focally introduced chemogenetic constructs in 34 control and 24 22qDS model mice via adeno-associated viral vectors, driven by excitatory neuron-specific promoter elements, to manipulate circuit recruitment in an on-demand fashion. RESULTS 22qDS model mice exhibited CA1 excitatory ensemble hyperexcitability and concomitant behavioral deficits in both social and spatial memory. Remarkably, acute chemogenetic inhibition of pyramidal cells successfully corrected memory deficits and did so in a regionally specific manner: ventrally targeted constructs rescued only social behavior, while those expressed dorsally selectively affected spatial memory. Additionally, manipulating activity in control mice could recapitulate the memory deficits in a regionally specific manner. CONCLUSIONS These data suggest that retuning activity dysregulation can rescue function in disease-altered circuits, even in the face of a polygenetic haploinsufficiency with a strong developmental component. Targeting circuit excitability in a focal, modular manner may prove to be an effective therapeutic for treatment-resistant symptoms of mental illness.
Collapse
Affiliation(s)
- Julia B. Kahn
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell G. Port
- Departments of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Stewart A. Anderson
- Departments of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Douglas A. Coulter
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Departments of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| |
Collapse
|
49
|
Upright NA, Baxter MG. Effect of chemogenetic actuator drugs on prefrontal cortex-dependent working memory in nonhuman primates. Neuropsychopharmacology 2020; 45:1793-1798. [PMID: 32193513 PMCID: PMC7608232 DOI: 10.1038/s41386-020-0660-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The most common chemogenetic neuromodulatory system, designer receptors exclusively activated by designer drugs (DREADDs), uses a non-endogenous actuator ligand to activate a modified muscarinic acetylcholine receptor that is insensitive to acetylcholine. It is crucial in studies using these systems to test the potential effects of DREADD actuators prior to any DREADD transduction, so that effects of DREADDs can be attributed to the chemogenetic system rather than the actuator drug, particularly in experiments using nonhuman primates. We investigated working memory performance after injections of three DREADD actuators, clozapine, olanzapine, and deschloroclozapine, in four male rhesus monkeys tested in a spatial delayed response task before any DREADD transduction took place. Performance at 0.1 mg/kg clozapine and 0.1 mg/kg deschloroclozapine did not differ from vehicle in any of the four subjects. 0.2 mg/kg clozapine impaired working memory function in three of the four monkeys. Two monkeys were impaired after 0.1 mg/kg olanzapine and two were impaired after 0.3 mg/kg deschloroclozapine. We speculate that the unique neuropharmacology of prefrontal cortex function makes the primate prefrontal cortex especially vulnerable to off-target effects of DREADD actuator drugs with affinity for endogenous monoaminergic receptor systems. These findings underscore the importance of within-subject controls for DREADD actuator drugs in the specific tasks under study to confirm that effects following DREADD receptor transduction are not owing to the actuator drug itself. They also suggest that off-target effects of DREADD actuators may limit translational applications of chemogenetic neuromodulation.
Collapse
Affiliation(s)
- Nicholas A Upright
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mark G Baxter
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
50
|
Goutaudier R, Coizet V, Carcenac C, Carnicella S. Compound 21, a two-edged sword with both DREADD-selective and off-target outcomes in rats. PLoS One 2020; 15:e0238156. [PMID: 32946510 PMCID: PMC7500623 DOI: 10.1371/journal.pone.0238156] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) represent a technical revolution in integrative neuroscience. However, the first used ligands exhibited dose-dependent selectivity for their molecular target, leading to potential unspecific effects. Compound 21 (C21) was recently proposed as an alternative, but in vivo characterization of its properties is not sufficient yet. Here, we evaluated its potency to selectively modulate the activity of nigral dopaminergic (DA) neurons through the canonical DREADD receptor hM4Di using TH-Cre rats. In males, 1 mg.kg-1 of C21 strongly increased nigral neurons activity in control animals, indicative of a significant off-target effect. Reducing the dose to 0.5 mg.kg-1 circumvented this unspecific effect, while activated the inhibitory DREADDs and selectively reduced nigral neurons firing. In females, 0.5 mg.kg-1 of C21 induced a transient and residual off-target effect that may mitigated the inhibitory DREADDs-mediated effect. This study raises up the necessity to test selectivity and efficacy of chosen ligands for each new experimental condition.
Collapse
Affiliation(s)
- Raphaël Goutaudier
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| | - Véronique Coizet
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| | - Carole Carcenac
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| | - Sebastien Carnicella
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| |
Collapse
|