1
|
Iguchi Y, Benton R, Kobayashi K. A chemogenetic technology using insect Ionotropic Receptors to stimulate target cell populations in the mammalian brain. Neurosci Res 2025; 214:56-61. [PMID: 39532176 DOI: 10.1016/j.neures.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Chemogenetics uses artificially-engineered proteins to modify the activity of cells, notably neurons, in response to small molecules. Although a common set of chemogenetic tools are the G protein-coupled receptor-based DREADDs, there has been great hope for ligand-gated, ion channel-type chemogenetic tools that directly impact neuronal excitability. We have devised such a technology by exploiting insect Ionotropic Receptors (IRs), a highly divergent subfamily of ionotropic glutamate receptors that evolved to detect diverse environmental chemicals. Here, we review a series of studies developing and applying this "IR-mediated neuronal activation" (IRNA) technology with the Drosophila melanogaster IR84a/IR8a complex, which detects phenyl-containing ligands. We also discuss how variants of IRNA could be produced by modifying the composition of the IR complex, using natural or engineered subunits, which would enable artificial activation of different cell populations in the brain in response to distinct chemicals.
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
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne CH-1015, Switzerland
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan.
| |
Collapse
|
2
|
McLaurin KA, Illenberger JM, Li H, Booze RM, Mactutus CF. SEX-DEPENDENT MODULATION OF BEHAVIORAL ALLOCATION VIA VENTRAL TEGMENTAL AREA-NUCLEUS ACCUMBENS SHELL CIRCUITRY. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637318. [PMID: 39990454 PMCID: PMC11844387 DOI: 10.1101/2025.02.09.637318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Diagnostic criteria for substance use disorder, cocaine type (i.e., cocaine use disorder), outlined in the 5 th edition of the Diagnostic and Statistical Manual, imply that the disorder arises, at least in part, from the maladaptive allocation of behavior to drug use. To date, however, the neural circuits involved in the allocation of behavior have not been systematically evaluated. Herein, a chemogenetics approach (i.e., designer receptors exclusively activated by designer drugs (DREADDs)) was utilized in combination with a concurrent choice self-administration experimental paradigm to evaluate the role of the mesolimbic neurocircuit in the allocation of behavior. Pharmacological activation of hM3D(G q ) DREADDs in neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens (AcbSh) induced a sex-dependent shift in the allocation of behavior in rodents transduced with DREADDs. Specifically, male DREADDs animals exhibited a robust increase in responding for a natural (i.e., sucrose) reward following pharmacological activation of the VTA-AcbSh circuit; female DREADDs rodents, in sharp contrast, displayed a prominent decrease in drug-reinforced (i.e., cocaine) responding. The sequential activation of hM3D(G q ) and KORD DREADDs within the same neuronal population validated the role of the VTA-AcbSh circuit in reinforced responding for concurrently available natural and drug rewards. Collectively, the VTA-AcbSh circuit is fundamentally involved in behavioral allocation affording a key target for the development of novel pharmacotherapies.
Collapse
|
3
|
Zhang H, Zheng Z, Chen X, Xu L, Guo C, Wang J, Cui Y, Yang F. RADICAL: a rationally designed ion channel activated by ligand for chemogenetics. Protein Cell 2025; 16:136-142. [PMID: 39225378 PMCID: PMC11786721 DOI: 10.1093/procel/pwae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Heng Zhang
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Kidney Disease Center of the First Affiliated Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Zhiwei Zheng
- Department of Neurobiology, Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain–Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiaoying Chen
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Kidney Disease Center of the First Affiliated Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Lizhen Xu
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Kidney Disease Center of the First Affiliated Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Chen Guo
- Department of Neurobiology, Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain–Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiawei Wang
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Kidney Disease Center of the First Affiliated Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Yihui Cui
- Department of Neurobiology, Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain–Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fan Yang
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Kidney Disease Center of the First Affiliated Hospital and Department of Biophysics, Zhejiang University School of Medicine, Hangzhou 311121, China
| |
Collapse
|
4
|
Pan Y, Pan C, Mao L, Yu P. Neuromodulation with chemicals: Opportunities and challenges. FUNDAMENTAL RESEARCH 2025; 5:55-62. [PMID: 40166084 PMCID: PMC11955035 DOI: 10.1016/j.fmre.2024.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/02/2025] Open
Abstract
Chemicals play a crucial role in neurophysiological and neuropathological processes. By regulating the concentration of specific chemicals, receptors on the neuron cell membrane can be modulated to activate or inhibit, thereby influencing specific ion channels and facilitating neuromodulation. This review introduces several chemical modulation techniques, such as microinjection, electrode/nanoparticle-based chemical delivery methods, in situ electrochemical synthesis and chemogenetics. While these techniques show promise in expanding the application of chemical neuromodulation, they currently exhibit different degrees of shortcomings and room for improvement. This review summarizes the opportunities and challenges for chemical neuromodulation methods and provide an outlook for their prospects in the future.
Collapse
Affiliation(s)
- Yifei Pan
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Cong Pan
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ping Yu
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
5
|
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
|
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
|
Gao H, Wang J, Zhang R, Luo T. Recent advances in neural mechanism of general anesthesia induced unconsciousness: insights from optogenetics and chemogenetics. Front Pharmacol 2024; 15:1360864. [PMID: 38655183 PMCID: PMC11035785 DOI: 10.3389/fphar.2024.1360864] [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: 12/24/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
For over 170 years, general anesthesia has played a crucial role in clinical practice, yet a comprehensive understanding of the neural mechanisms underlying the induction of unconsciousness by general anesthetics remains elusive. Ongoing research into these mechanisms primarily centers around the brain nuclei and neural circuits associated with sleep-wake. In this context, two sophisticated methodologies, optogenetics and chemogenetics, have emerged as vital tools for recording and modulating the activity of specific neuronal populations or circuits within distinct brain regions. Recent advancements have successfully employed these techniques to investigate the impact of general anesthesia on various brain nuclei and neural pathways. This paper provides an in-depth examination of the use of optogenetic and chemogenetic methodologies in studying the effects of general anesthesia on specific brain nuclei and pathways. Additionally, it discusses in depth the advantages and limitations of these two methodologies, as well as the issues that must be considered for scientific research applications. By shedding light on these facets, this paper serves as a valuable reference for furthering the accurate exploration of the neural mechanisms underlying general anesthesia. It aids researchers and clinicians in effectively evaluating the applicability of these techniques in advancing scientific research and clinical practice.
Collapse
Affiliation(s)
- Hui Gao
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jingyi Wang
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rui Zhang
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Tao Luo
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
8
|
Xiao W, Li P, Kong F, Kong J, Pan A, Long L, Yan X, Xiao B, Gong J, Wan L. Unraveling the Neural Circuits: Techniques, Opportunities and Challenges in Epilepsy Research. Cell Mol Neurobiol 2024; 44:27. [PMID: 38443733 PMCID: PMC10914928 DOI: 10.1007/s10571-024-01458-5] [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: 12/25/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024]
Abstract
Epilepsy, a prevalent neurological disorder characterized by high morbidity, frequent recurrence, and potential drug resistance, profoundly affects millions of people globally. Understanding the microscopic mechanisms underlying seizures is crucial for effective epilepsy treatment, and a thorough understanding of the intricate neural circuits underlying epilepsy is vital for the development of targeted therapies and the enhancement of clinical outcomes. This review begins with an exploration of the historical evolution of techniques used in studying neural circuits related to epilepsy. It then provides an extensive overview of diverse techniques employed in this domain, discussing their fundamental principles, strengths, limitations, as well as their application. Additionally, the synthesis of multiple techniques to unveil the complexity of neural circuits is summarized. Finally, this review also presents targeted drug therapies associated with epileptic neural circuits. By providing a critical assessment of methodologies used in the study of epileptic neural circuits, this review seeks to enhance the understanding of these techniques, stimulate innovative approaches for unraveling epilepsy's complexities, and ultimately facilitate improved treatment and clinical translation for epilepsy.
Collapse
Affiliation(s)
- Wenjie Xiao
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Peile Li
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Fujiao Kong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jingyi Kong
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaoe Gong
- Department of Neurology, Hunan Children's Hospital, Changsha, Hunan Province, China.
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China.
| |
Collapse
|
9
|
Middleton SJ, Hu H, Perez-Sanchez J, Zuberi S, McGrath Williams J, Weir GA, Bennett DL. GluCl.Cre ON enables selective inhibition of molecularly defined pain circuits. Pain 2023; 164:2780-2791. [PMID: 37366588 PMCID: PMC10652717 DOI: 10.1097/j.pain.0000000000002976] [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: 10/03/2022] [Revised: 02/17/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023]
Abstract
ABSTRACT Insight into nociceptive circuits will ultimately build our understanding of pain processing and aid the development of analgesic strategies. Neural circuit analysis has been advanced greatly by the development of optogenetic and chemogenetic tools, which have allowed function to be ascribed to discrete neuronal populations. Neurons of the dorsal root ganglion, which include nociceptors, have proved challenging targets for chemogenetic manipulation given specific confounds with commonly used DREADD technology. We have developed a cre/lox dependant version of the engineered glutamate-gated chloride channel (GluCl) to restrict and direct its expression to molecularly defined neuronal populations. We have generated GluCl.Cre ON that selectively renders neurons expressing cre-recombinase susceptible to agonist-induced silencing. We have functionally validated our tool in multiple systems in vitro, and subsequently generated viral vectors and tested its applicability in vivo. Using Nav1.8 Cre mice to restrict AAV-GluCl.Cre ON to nociceptors, we demonstrate effective silencing of electrical activity in vivo and concomitant hyposensitivity to noxious thermal and noxious mechanical pain, whereas light touch and motor function remained intact. We also demonstrated that our strategy can effectively silence inflammatory-like pain in a chemical pain model. Collectively, we have generated a novel tool that can be used to selectively silence defined neuronal circuits in vitro and in vivo. We believe that this addition to the chemogenetic tool box will facilitate further understanding of pain circuits and guide future therapeutic development.
Collapse
Affiliation(s)
- Steven J. Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Huimin Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Jimena Perez-Sanchez
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Sana Zuberi
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Greg A. Weir
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Grzelka K, Wilhelms H, Dodt S, Dreisow ML, Madara JC, Walker SJ, Wu C, Wang D, Lowell BB, Fenselau H. A synaptic amplifier of hunger for regaining body weight in the hypothalamus. Cell Metab 2023; 35:770-785.e5. [PMID: 36965483 PMCID: PMC10160008 DOI: 10.1016/j.cmet.2023.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 12/15/2022] [Accepted: 03/01/2023] [Indexed: 03/27/2023]
Abstract
Restricting caloric intake effectively reduces body weight, but most dieters fail long-term adherence to caloric deficit and eventually regain lost weight. Hypothalamic circuits that control hunger drive critically determine body weight; yet, how weight loss sculpts these circuits to motivate food consumption until lost weight is regained remains unclear. Here, we probe the contribution of synaptic plasticity in discrete excitatory afferents on hunger-promoting AgRP neurons. We reveal a crucial role for activity-dependent, remarkably long-lasting amplification of synaptic activity originating from paraventricular hypothalamus thyrotropin-releasing (PVHTRH) neurons in long-term body weight control. Silencing PVHTRH neurons inhibits the potentiation of excitatory input to AgRP neurons and diminishes concomitant regain of lost weight. Brief stimulation of the pathway is sufficient to enduringly potentiate this glutamatergic hunger synapse and triggers an NMDAR-dependent gaining of body weight that enduringly persists. Identification of this activity-dependent synaptic amplifier provides a previously unrecognized target to combat regain of lost weight.
Collapse
Affiliation(s)
- Katarzyna Grzelka
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Hannah Wilhelms
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Stephan Dodt
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Marie-Luise Dreisow
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Samuel J Walker
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chen Wu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daqing Wang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA.
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
11
|
Zhao R, Grunke SD, Wood CA, Perez GA, Comstock M, Li MH, Singh AK, Park KW, Jankowsky JL. Activity disruption causes degeneration of entorhinal neurons in a mouse model of Alzheimer's circuit dysfunction. eLife 2022; 11:e83813. [PMID: 36468693 PMCID: PMC9873254 DOI: 10.7554/elife.83813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are characterized by selective vulnerability of distinct cell populations; however, the cause for this specificity remains elusive. Here, we show that entorhinal cortex layer 2 (EC2) neurons are unusually vulnerable to prolonged neuronal inactivity compared with neighboring regions of the temporal lobe, and that reelin + stellate cells connecting EC with the hippocampus are preferentially susceptible within the EC2 population. We demonstrate that neuronal death after silencing can be elicited through multiple independent means of activity inhibition, and that preventing synaptic release, either alone or in combination with electrical shunting, is sufficient to elicit silencing-induced degeneration. Finally, we discovered that degeneration following synaptic silencing is governed by competition between active and inactive cells, which is a circuit refinement process traditionally thought to end early in postnatal life. Our data suggests that the developmental window for wholesale circuit plasticity may extend into adulthood for specific brain regions. We speculate that this sustained potential for remodeling by entorhinal neurons may support lifelong memory but renders them vulnerable to prolonged activity changes in disease.
Collapse
Affiliation(s)
- Rong Zhao
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Stacy D Grunke
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Caleb A Wood
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Gabriella A Perez
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Melissa Comstock
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Ming-Hua Li
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Anand K Singh
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Kyung-Won Park
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| | - Joanna L Jankowsky
- Department of Neuroscience, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
- Departments of Neurology, Neurosurgery, and Molecular and Cellular Biology, Huffington Center on Aging, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
12
|
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
|
13
|
Fifel K, El Farissi A, Cherasse Y, Yanagisawa M. Motivational and Valence-Related Modulation of Sleep/Wake Behavior are Mediated by Midbrain Dopamine and Uncoupled from the Homeostatic and Circadian Processes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200640. [PMID: 35794435 PMCID: PMC9403635 DOI: 10.1002/advs.202200640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Motivation and its hedonic valence are powerful modulators of sleep/wake behavior, yet its underlying mechanism is still poorly understood. Given the well-established role of midbrain dopamine (mDA) neurons in encoding motivation and emotional valence, here, neuronal mechanisms mediating sleep/wake regulation are systematically investigated by DA neurotransmission. It is discovered that mDA mediates the strong modulation of sleep/wake states by motivational valence. Surprisingly, this modulation can be uncoupled from the classically employed measures of circadian and homeostatic processes of sleep regulation. These results establish the experimental foundation for an additional new factor of sleep regulation. Furthermore, an electroencephalographic marker during wakefulness at the theta range is identified that can be used to reliably track valence-related modulation of sleep. Taken together, this study identifies mDA signaling as an important neural substrate mediating sleep modulation by motivational valence.
Collapse
Affiliation(s)
- Karim Fifel
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| | - Amina El Farissi
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8577Japan
| |
Collapse
|
14
|
Mueller JS, Tescarollo FC, Sun H. DREADDs in Epilepsy Research: Network-Based Review. Front Mol Neurosci 2022; 15:863003. [PMID: 35465094 PMCID: PMC9021489 DOI: 10.3389/fnmol.2022.863003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Epilepsy can be interpreted as altered brain rhythms from overexcitation or insufficient inhibition. Chemogenetic tools have revolutionized neuroscience research because they allow "on demand" excitation or inhibition of neurons with high cellular specificity. Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are the most frequently used chemogenetic techniques in epilepsy research. These engineered muscarinic receptors allow researchers to excite or inhibit targeted neurons with exogenous ligands. As a result, DREADDs have been applied to investigate the underlying cellular and network mechanisms of epilepsy. Here, we review the existing literature that has applied DREADDs to understand the pathophysiology of epilepsy. The aim of this review is to provide a general introduction to DREADDs with a focus on summarizing the current main findings in experimental epilepsy research using these techniques. Furthermore, we explore how DREADDs may be applied therapeutically as highly innovative treatments for epilepsy.
Collapse
Affiliation(s)
| | | | - Hai Sun
- Department of Neurosurgery, Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
15
|
Kolesov DV, Sokolinskaya EL, Lukyanov KA, Bogdanov AM. Molecular Tools for Targeted Control of Nerve Cell Electrical Activity. Part II. Acta Naturae 2021; 13:17-32. [PMID: 35127143 PMCID: PMC8807539 DOI: 10.32607/actanaturae.11415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 05/14/2021] [Indexed: 01/01/2023] Open
Abstract
In modern life sciences, the issue of a specific, exogenously directed manipulation of a cell's biochemistry is a highly topical one. In the case of electrically excitable cells, the aim of the manipulation is to control the cells' electrical activity, with the result being either excitation with subsequent generation of an action potential or inhibition and suppression of the excitatory currents. The techniques of electrical activity stimulation are of particular significance in tackling the most challenging basic problem: figuring out how the nervous system of higher multicellular organisms functions. At this juncture, when neuroscience is gradually abandoning the reductionist approach in favor of the direct investigation of complex neuronal systems, minimally invasive methods for brain tissue stimulation are becoming the basic element in the toolbox of those involved in the field. In this review, we describe three approaches that are based on the delivery of exogenous, genetically encoded molecules sensitive to external stimuli into the nervous tissue. These approaches include optogenetics (overviewed in Part I), as well as chemogenetics and thermogenetics (described here, in Part II), which is significantly different not only in the nature of the stimuli and structure of the appropriate effector proteins, but also in the details of experimental applications. The latter circumstance is an indication that these are rather complementary than competing techniques.
Collapse
Affiliation(s)
- D. V. Kolesov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - E. L. Sokolinskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - K. A. Lukyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - A. M. Bogdanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| |
Collapse
|
16
|
Simpson S, Chen Y, Wellmeyer E, Smith LC, Aragon Montes B, George O, Kimbrough A. The Hidden Brain: Uncovering Previously Overlooked Brain Regions by Employing Novel Preclinical Unbiased Network Approaches. Front Syst Neurosci 2021; 15:595507. [PMID: 33967705 PMCID: PMC8097000 DOI: 10.3389/fnsys.2021.595507] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
A large focus of modern neuroscience has revolved around preselected brain regions of interest based on prior studies. While there are reasons to focus on brain regions implicated in prior work, the result has been a biased assessment of brain function. Thus, many brain regions that may prove crucial in a wide range of neurobiological problems, including neurodegenerative diseases and neuropsychiatric disorders, have been neglected. Advances in neuroimaging and computational neuroscience have made it possible to make unbiased assessments of whole-brain function and identify previously overlooked regions of the brain. This review will discuss the tools that have been developed to advance neuroscience and network-based computational approaches used to further analyze the interconnectivity of the brain. Furthermore, it will survey examples of neural network approaches that assess connectivity in clinical (i.e., human) and preclinical (i.e., animal model) studies and discuss how preclinical studies of neurodegenerative diseases and neuropsychiatric disorders can greatly benefit from the unbiased nature of whole-brain imaging and network neuroscience.
Collapse
Affiliation(s)
- Sierra Simpson
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Yueyi Chen
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States.,Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Emma Wellmeyer
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Lauren C Smith
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Brianna Aragon Montes
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Olivier George
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Adam Kimbrough
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Inflammation, Immunology, and Infectious Disease, West Lafayette, IN, United States
| |
Collapse
|
17
|
Buckingham SD, Mann HJ, Hearnden OK, Sattelle DB. Turning a Drug Target into a Drug Candidate: A New Paradigm for Neurological Drug Discovery? Bioessays 2020; 42:e2000011. [PMID: 32776366 DOI: 10.1002/bies.202000011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/26/2020] [Indexed: 11/11/2022]
Abstract
The conventional paradigm for developing new treatments for disease mainly involves either the discovery of new drug targets, or finding new, improved drugs for old targets. However, an ion channel found only in invertebrates offers the potential of a completely new paradigm in which an established drug target can be re-engineered to serve as a new candidate therapeutic agent. The L-glutamate-gated chloride channels (GluCls) of invertebrates are absent from vertebrate genomes, offering the opportunity to introduce this exogenous, inhibitory, L-glutamate receptor into vertebrate neuronal circuits either as a tool with which to study neural networks, or a candidate therapy. Epileptic seizures can involve L-glutamate-induced hyper-excitation and toxicity. Variant GluCls, with their inhibitory responses to L-glutamate, when engineered into human neurons, might counter the excitotoxic effects of excess L-glutamate. In reviewing recent studies on model organisms, it appears that this approach might offer a new paradigm for the development of candidate therapeutics for epilepsy.
Collapse
Affiliation(s)
- Steven D Buckingham
- School of Biological and Chemical Sciences, Queen Mary University London, Mile End Road, London, E1 4NS, UK.,UCL Respiratory, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Harry-Jack Mann
- UCL Respiratory, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Olivia K Hearnden
- UCL Respiratory, University College London, 5 University Street, London, WC1E 6JF, UK
| | - David B Sattelle
- UCL Respiratory, University College London, 5 University Street, London, WC1E 6JF, UK
| |
Collapse
|
18
|
Melonakos ED, Moody OA, Nikolaeva K, Kato R, Nehs CJ, Solt K. Manipulating Neural Circuits in Anesthesia Research. Anesthesiology 2020; 133:19-30. [PMID: 32349073 PMCID: PMC8351362 DOI: 10.1097/aln.0000000000003279] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The neural circuits underlying the distinct endpoints that define general anesthesia remain incompletely understood. It is becoming increasingly evident, however, that distinct pathways in the brain that mediate arousal and pain are involved in various endpoints of general anesthesia. To critically evaluate this growing body of literature, familiarity with modern tools and techniques used to study neural circuits is essential. This Readers' Toolbox article describes four such techniques: (1) electrical stimulation, (2) local pharmacology, (3) optogenetics, and (4) chemogenetics. Each technique is explained, including the advantages, disadvantages, and other issues that must be considered when interpreting experimental results. Examples are provided of studies that probe mechanisms of anesthesia using each technique. This information will aid researchers and clinicians alike in interpreting the literature and in evaluating the utility of these techniques in their own research programs.
Collapse
Affiliation(s)
- Eric D. Melonakos
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Olivia A. Moody
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ksenia Nikolaeva
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Risako Kato
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Christa J. Nehs
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
19
|
Chemogenetics a robust approach to pharmacology and gene therapy. Biochem Pharmacol 2020; 175:113889. [DOI: 10.1016/j.bcp.2020.113889] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
|
20
|
Luo L, Callaway EM, Svoboda K. Genetic Dissection of Neural Circuits: A Decade of Progress. Neuron 2019; 98:256-281. [PMID: 29673479 DOI: 10.1016/j.neuron.2018.03.040] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 01/24/2023]
Abstract
Tremendous progress has been made since Neuron published our Primer on genetic dissection of neural circuits 10 years ago. Since then, cell-type-specific anatomical, neurophysiological, and perturbation studies have been carried out in a multitude of invertebrate and vertebrate organisms, linking neurons and circuits to behavioral functions. New methods allow systematic classification of cell types and provide genetic access to diverse neuronal types for studies of connectivity and neural coding during behavior. Here we evaluate key advances over the past decade and discuss future directions.
Collapse
Affiliation(s)
- Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Karel Svoboda
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| |
Collapse
|
21
|
Basbaum A. Chemogenetic management of neuropathic pain. Brain 2019; 140:2522-2525. [PMID: 28969393 DOI: 10.1093/brain/awx227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Allan Basbaum
- Department of Anatomy, University California San Francisco, San Francisco, CA 94901, USA
| |
Collapse
|
22
|
Stanley SA, Friedman JM. Electromagnetic Regulation of Cell Activity. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034322. [PMID: 30249601 DOI: 10.1101/cshperspect.a034322] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability to observe the effects of rapidly and reversibly regulating cell activity in targeted cell populations has provided numerous physiologic insights. Over the last decade, a wide range of technologies have emerged for regulating cellular activity using optical, chemical, and, more recently, electromagnetic modalities. Electromagnetic fields can freely penetrate cells and tissue and their energy can be absorbed by metal particles. When released, the absorbed energy can in turn gate endogenous or engineered receptors and ion channels to regulate cell activity. In this manner, electromagnetic fields acting on external nanoparticles have been used to exert mechanical forces on cell membranes and organelles to generate heat and interact with thermally activated proteins or to induce receptor aggregation and intracellular signaling. More recently, technologies using genetically encoded nanoparticles composed of the iron storage protein, ferritin, have been used for targeted, temporal control of cell activity in vitro and in vivo. These tools provide a means for noninvasively modulating gene expression, intracellular organelles, such as endosomes, and whole-cell activity both in vitro and in freely moving animals. The use of magnetic fields interacting with external or genetically encoded nanoparticles thus provides a rapid noninvasive means for regulating cell activity.
Collapse
Affiliation(s)
- Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Rockefeller University, New York, New York 10065.,Howard Hughes Medical Institute, New York, New York 10065
| |
Collapse
|
23
|
A Photoactivatable Botulinum Neurotoxin for Inducible Control of Neurotransmission. Neuron 2019; 101:863-875.e6. [PMID: 30704911 DOI: 10.1016/j.neuron.2019.01.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/14/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
Regulated secretion is critical for diverse biological processes ranging from immune and endocrine signaling to synaptic transmission. Botulinum and tetanus neurotoxins, which specifically proteolyze vesicle fusion proteins involved in regulated secretion, have been widely used as experimental tools to block these processes. Genetic expression of these toxins in the nervous system has been a powerful approach for disrupting neurotransmitter release within defined circuitry, but their current utility in the brain and elsewhere remains limited by lack of spatial and temporal control. Here we engineered botulinum neurotoxin B so that it can be activated with blue light. We demonstrate the utility of this approach for inducibly disrupting excitatory neurotransmission, providing a first-in-class optogenetic tool for persistent, light-triggered synaptic inhibition. In addition to blocking neurotransmitter release, this approach will have broad utility for conditionally disrupting regulated secretion of diverse bioactive molecules, including neuropeptides, neuromodulators, hormones, and immune molecules. VIDEO ABSTRACT.
Collapse
|
24
|
Aldrin-Kirk P, Björklund T. Practical Considerations for the Use of DREADD and Other Chemogenetic Receptors to Regulate Neuronal Activity in the Mammalian Brain. Methods Mol Biol 2019; 1937:59-87. [PMID: 30706390 DOI: 10.1007/978-1-4939-9065-8_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chemogenetics is the process of genetically expressing a macromolecule receptor capable of modulating the activity of the cell in response to selective chemical ligand. This chapter will cover the chemogenetic technologies that are available to date, focusing on the commonly available engineered or otherwise modified ligand-gated ion channels and G-protein-coupled receptors in the context of neuromodulation. First, we will give a brief overview of each chemogenetic approach as well as in vitro/in vivo applications, then we will list their strengths and weaknesses. Finally, we will provide tips for ligand application in each case.Each technology has specific limitations that make them more or less suitable for different applications in neuroscience although we will focus mainly on the most commonly used and versatile family named designer receptors exclusively activated by designer drugs or DREADDs. We here describe the most common cases where these can be implemented and provide tips on how and where these technologies can be applied in the field of neuroscience.
Collapse
Affiliation(s)
- Patrick Aldrin-Kirk
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| |
Collapse
|
25
|
Lv ZM, Zhao RJ, Zhi XS, Huang Y, Chen JY, Song NN, Su CJ, Ding YQ. Expression of DCX and Transcription Factor Profiling in Photothrombosis-Induced Focal Ischemia in Mice. Front Cell Neurosci 2018; 12:455. [PMID: 30524246 PMCID: PMC6262056 DOI: 10.3389/fncel.2018.00455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 11/09/2018] [Indexed: 11/26/2022] Open
Abstract
Adult neurogenesis is present in the dentate gyrus and the subventricular zone in mammalian brain under physiological conditions. Recently, adult neurogenesis has also been reported in other brain regions after brain injury. In this study, we established a focal striatal ischemic model in adult mice via photothrombosis (PT) and investigated how focal ischemia elicits neurogenesis in the striatum. We found that astrocytes and microglia increased in early post-ischemic stage, followed by a 1-week late-onset of doublecortin (DCX) expression in the striatum. The number of DCX-positive neurons reached the peak level at day 7, but they were still observed at day 28 post-ischemia. Moreover, Rbp-J (a key effector of Notch signaling) deletion in astrocytes has been reported to promote the neuron regeneration after brain ischemia, and we provided the change of gene expression profile in the striatum of astrocyte-specific Rbp-J knockout (KO) mice glial fibrillary acidic protein (GFAP-CreER:Rbp-Jfl/fl), which may help to clarify detailed potential mechanisms for the post-ischemic neurogenesis in the striatum.
Collapse
Affiliation(s)
- Zhu-Man Lv
- Department of Basic Medicine, Institute of Neurosciences, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Rong-Jian Zhao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Xiao-Song Zhi
- Center for Stem Cells and Medicine, Department of Cell Biology, Second Military Medical University, Shanghai, China
| | - Ying Huang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Jia-Yin Chen
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Ning-Ning Song
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Chang-Jun Su
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Yu-Qiang Ding
- Department of Basic Medicine, Institute of Neurosciences, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Schoderboeck L, Wicky HE, Abraham WC, Hughes SM. Genetic Targeting and Chemogenetic Inhibition of Newborn Neurons. Hum Gene Ther Methods 2018; 29:259-268. [PMID: 30526082 DOI: 10.1089/hgtb.2018.182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of this study was to develop a method to silence a very specific set of cells in a spatially and temporally refined manner. Here, an approach is presented that combines the use of a transgenic mouse line, expressing cre recombinase under a nestin promoter, with lentiviral delivery of a floxed, ivermectin (IVM)-gated chloride channel construct to the dentate gyrus. This approach was used to express an IVM-sensitive chloride channel in newly born granule cells in adult mouse brains, and its ability to silence neuronal activity was tested by analyzing the effect on immediate early gene expression in vitro in cre-transgenic primary neuronal cultures. IVM treatment of cells expressing the chloride channel prevented gabazine-induced expression of the immediate early gene product EGR1, while cells expressing a control inactive channel or no channel retained their EGR1 response. Thus, a genetic strategy is presented for targeting a specific neurogenic niche for transgene expression in the adult mouse brain, and proof of principle is shown that it can be used in vitro as a method for silencing neuronal activity.
Collapse
Affiliation(s)
- Lucia Schoderboeck
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Department of Psychology, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre and Brain Research New Zealand, and University of Otago, Dunedin, New Zealand.,Genetics Otago, University of Otago, Dunedin, New Zealand
| | - Hollie E Wicky
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre and Brain Research New Zealand, and University of Otago, Dunedin, New Zealand.,Genetics Otago, University of Otago, Dunedin, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre and Brain Research New Zealand, and University of Otago, Dunedin, New Zealand
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre and Brain Research New Zealand, and University of Otago, Dunedin, New Zealand.,Genetics Otago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
27
|
Abstract
Chemogenetic technologies enable selective pharmacological control of specific cell populations. An increasing number of approaches have been developed that modulate different signaling pathways. Selective pharmacological control over G protein-coupled receptor signaling, ion channel conductances, protein association, protein stability, and small molecule targeting allows modulation of cellular processes in distinct cell types. Here, we review these chemogenetic technologies and instances of their applications in complex tissues in vivo and ex vivo.
Collapse
Affiliation(s)
- Deniz Atasoy
- Department of Physiology, School of Medicine and Regenerative-Restorative Medicine Research Center (REMER), Istanbul Medipol University , Istanbul , Turkey ; and Janelia Research Campus, Howard Hughes Medical Institute , Ashburn, Virginia
| | - Scott M Sternson
- Department of Physiology, School of Medicine and Regenerative-Restorative Medicine Research Center (REMER), Istanbul Medipol University , Istanbul , Turkey ; and Janelia Research Campus, Howard Hughes Medical Institute , Ashburn, Virginia
| |
Collapse
|
28
|
Alvarsson A, Stanley SA. Remote control of glucose-sensing neurons to analyze glucose metabolism. Am J Physiol Endocrinol Metab 2018; 315:E327-E339. [PMID: 29812985 PMCID: PMC6171010 DOI: 10.1152/ajpendo.00469.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/25/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
The central nervous system relies on a continual supply of glucose, and must be able to detect glucose levels and regulate peripheral organ functions to ensure that its energy requirements are met. Specialized glucose-sensing neurons, first described half a century ago, use glucose as a signal and modulate their firing rates as glucose levels change. Glucose-excited neurons are activated by increasing glucose concentrations, while glucose-inhibited neurons increase their firing rate as glucose concentrations fall and decrease their firing rate as glucose concentrations rise. Glucose-sensing neurons are present in multiple brain regions and are highly expressed in hypothalamic regions, where they are involved in functions related to glucose homeostasis. However, the roles of glucose-sensing neurons in healthy and disease states remain poorly understood. Technologies that can rapidly and reversibly activate or inhibit defined neural populations provide invaluable tools to investigate how specific neural populations regulate metabolism and other physiological roles. Optogenetics has high temporal and spatial resolutions, requires implants for neural stimulation, and is suitable for modulating local neural populations. Chemogenetics, which requires injection of a synthetic ligand, can target both local and widespread populations. Radio- and magnetogenetics offer rapid neural activation in localized or widespread neural populations without the need for implants or injections. These tools will allow us to better understand glucose-sensing neurons and their metabolism-regulating circuits.
Collapse
Affiliation(s)
- Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sarah A Stanley
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| |
Collapse
|
29
|
Munshi R, Qadri SM, Pralle A. Transient Magnetothermal Neuronal Silencing Using the Chloride Channel Anoctamin 1 (TMEM16A). Front Neurosci 2018; 12:560. [PMID: 30154692 PMCID: PMC6103273 DOI: 10.3389/fnins.2018.00560] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/25/2018] [Indexed: 12/15/2022] Open
Abstract
Determining the role and necessity of specific neurons in a network calls for precisely timed, reversible removal of these neurons from the circuit via remotely triggered transient silencing. Previously, we have shown that alternating magnetic field mediated heating of magnetic nanoparticles, bound to neurons, expressing temperature-sensitive cation channels TRPV1 remotely activates these neurons, evoking behavioral responses in mice. Here, we demonstrate how to apply magnetic nanoparticle heating to silence target neurons. Rat hippocampal neuronal cultures were transfected to express the temperature gated chloride channel, anoctamin 1 (TMEM16A). Spontaneous firing was suppressed within seconds of alternating magnetic field application to anoctamin 1 (TMEM16A) channel expressing, magnetic nanoparticle decorated neurons. Five seconds of magnetic field application leads to 12 s of silencing, with a latency of 2 s and an average suppression ratio of more than 80%. Immediately following the silencing period spontaneous activity resumed. The method provides a promising avenue for tether free, remote, transient neuronal silencing in vivo for both scientific and therapeutic applications.
Collapse
Affiliation(s)
| | | | - Arnd Pralle
- Department of Physics, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
30
|
Thompson KJ, Khajehali E, Bradley SJ, Navarrete JS, Huang XP, Slocum S, Jin J, Liu J, Xiong Y, Olsen RHJ, Diberto JF, Boyt KM, Pina MM, Pati D, Molloy C, Bundgaard C, Sexton PM, Kash TL, Krashes MJ, Christopoulos A, Roth BL, Tobin AB. DREADD Agonist 21 Is an Effective Agonist for Muscarinic-Based DREADDs in Vitro and in Vivo. ACS Pharmacol Transl Sci 2018; 1:61-72. [PMID: 30868140 PMCID: PMC6407913 DOI: 10.1021/acsptsci.8b00012] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Indexed: 02/07/2023]
Abstract
![]()
Chemogenetic tools such as designer
receptors exclusively activated
by designer drugs (DREADDs) are routinely used to modulate neuronal
and non-neuronal signaling and activity in a relatively noninvasive
manner. The first generation of DREADDs were templated from the human
muscarinic acetylcholine receptor family and are relatively insensitive
to the endogenous agonist acetylcholine but instead are activated
by clozapine-N-oxide (CNO). Despite the undisputed
success of CNO as an activator of muscarinic DREADDs, it has been
known for some time that CNO is subject to a low rate of metabolic
conversion to clozapine, raising the need for alternative chemical
actuators of muscarinic-based DREADDs. Here we show that DREADD agonist 21 (C21) (11-(1-piperazinyl)-5H-dibenzo[b,e][1,4]diazepine)
is a potent and selective agonist at both excitatory (hM3Dq) and inhibitory
(hM4Di) DREADDs and has excellent bioavailability, pharmacokinetic
properties, and brain penetrability. We also show that C21-induced
activation of hM3Dq and hM4Di in vivo can modulate
bidirectional feeding in defined circuits in mice. These results indicate
that C21 represents an alternative to CNO for in vivo studies where metabolic conversion of CNO to clozapine is a concern.
Collapse
Affiliation(s)
- Karen J Thompson
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| | - Elham Khajehali
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sophie J Bradley
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| | - Jovana S Navarrete
- Diabetes, Endocrinology, and Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States.,National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Xi Ping Huang
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Samuel Slocum
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Jian Jin
- Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029, United States
| | - Jing Liu
- Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029, United States
| | - Yan Xiong
- Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029, United States
| | - Reid H J Olsen
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Jeffrey F Diberto
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Kristen M Boyt
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Melanie M Pina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Dipanwita Pati
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Colin Molloy
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| | - Christoffer Bundgaard
- Neuroscience, Eli Lilly & Co., Erl Wood Manor, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas L Kash
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States.,National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| |
Collapse
|
31
|
Machado NLS, Abbott SBG, Resch JM, Zhu L, Arrigoni E, Lowell BB, Fuller PM, Fontes MAP, Saper CB. A Glutamatergic Hypothalamomedullary Circuit Mediates Thermogenesis, but Not Heat Conservation, during Stress-Induced Hyperthermia. Curr Biol 2018; 28:2291-2301.e5. [PMID: 30017482 DOI: 10.1016/j.cub.2018.05.064] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/11/2018] [Accepted: 05/22/2018] [Indexed: 11/30/2022]
Abstract
Stress elicits a variety of autonomic responses, including hyperthermia (stress fever) in humans and animals. In this present study, we investigated the circuit basis for thermogenesis and heat conservation during this response. We first demonstrated the glutamatergic identity of the dorsal hypothalamic area (DHAVglut2) neurons that innervate the raphe pallidus nucleus (RPa) to regulate core temperature (Tc) and mediate stress-induced hyperthermia. Then, using chemogenetic and optogenetic methods to manipulate this hypothalamomedullary circuit, we found that activation of DHAVglut2 neurons potently drove an increase in Tc, but surprisingly, stress-induced hyperthermia was only reduced by about one-third when they were inhibited. Further investigation showed that DHAVglut2 neurons activate brown adipose tissue (BAT) but do not cause vasoconstriction, instead allowing reflex tail artery vasodilation as a response to BAT-induced hyperthermia. Retrograde rabies virus tracing revealed projections from DHAVglut2 neurons to RPaVglut3, but not to RPaGABA neurons, and identified a set of inputs to DHAVglut2 → RPa neurons that are likely to mediate BAT activation. The dissociation of the DHAVglut2 thermogenic pathway from the thermoregulatory vasoconstriction (heat-conserving) pathway may explain stress flushing (skin vasodilation but a feeling of being too hot) during stressful times.
Collapse
Affiliation(s)
- Natalia L S Machado
- Department of Neurology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA; Department of Physiology and Biophysics, Federal University of Minas Gerais, Antonio Carlos Avenue, Belo Horizonte 31270-901, Brazil
| | - Stephen B G Abbott
- Department of Neurology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA; Department of Pharmacology, University of Virginia, Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Jon M Resch
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA
| | - Lin Zhu
- Department of Neurology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA
| | - Elda Arrigoni
- Department of Neurology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA
| | - Patrick M Fuller
- Department of Neurology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA
| | - Marco A P Fontes
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Antonio Carlos Avenue, Belo Horizonte 31270-901, Brazil
| | - Clifford B Saper
- Department of Neurology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Blackfan Circle, Boston, MA 02215, USA.
| |
Collapse
|
32
|
Campbell EJ, Marchant NJ. The use of chemogenetics in behavioural neuroscience: receptor variants, targeting approaches and caveats. Br J Pharmacol 2018; 175:994-1003. [PMID: 29338070 DOI: 10.1111/bph.14146] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/13/2017] [Accepted: 12/27/2017] [Indexed: 12/18/2022] Open
Abstract
The last decade has seen major advances in neuroscience tools allowing us to selectively modulate cellular pathways in freely moving animals. Chemogenetic approaches such as designer receptors exclusively activated by designer drugs (DREADDs) permit the remote control of neuronal function by systemic drug administration. These approaches have dramatically advanced our understanding of the neural control of behaviour. Here, we review the different techniques and genetic approaches available for the restriction of chemogenetic receptors to defined neuronal populations. We highlight the use of a dual virus approach to target specific circuitries and the effectiveness of different routes of administration of designer drugs. Finally, we discuss the potential caveats associated with DREADDs including off-target effects of designer drugs, the effects of chronic chemogenetic receptor activation and the issue of collateral projections associated with DREADD activation and inhibition.
Collapse
Affiliation(s)
- Erin J Campbell
- The Florey Institute of Neuroscience and Mental Health, Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Nathan J Marchant
- The Florey Institute of Neuroscience and Mental Health, Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Hippocampus-driven feed-forward inhibition of the prefrontal cortex mediates relapse of extinguished fear. Nat Neurosci 2018; 21:384-392. [PMID: 29403033 DOI: 10.1038/s41593-018-0073-9] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/19/2017] [Indexed: 12/18/2022]
Abstract
The medial prefrontal cortex (mPFC) has been implicated in the extinction of emotional memories, including conditioned fear. We found that ventral hippocampal (vHPC) projections to the infralimbic (IL) cortex recruited parvalbumin-expressing interneurons to counter the expression of extinguished fear and promote fear relapse. Whole-cell recordings ex vivo revealed that optogenetic activation of vHPC input to amygdala-projecting pyramidal neurons in the IL was dominated by feed-forward inhibition. Selectively silencing parvalbumin-expressing, but not somatostatin-expressing, interneurons in the IL eliminated vHPC-mediated inhibition. In behaving rats, pharmacogenetic activation of vHPC→IL projections impaired extinction recall, whereas silencing IL projectors diminished fear renewal. Intra-IL infusion of GABA receptor agonists or antagonists, respectively, reproduced these effects. Together, our findings describe a previously unknown circuit mechanism for the contextual control of fear, and indicate that vHPC-mediated inhibition of IL is an essential neural substrate for fear relapse.
Collapse
|
34
|
Spampanato J, Gibson A, Dudek FE. The antihelminthic moxidectin enhances tonic GABA currents in rodent hippocampal pyramidal neurons. J Neurophysiol 2018; 119:1693-1698. [PMID: 29364072 DOI: 10.1152/jn.00587.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Macrocyclic lactones (MLs) are commonly used treatments for parasitic worm and insect infections in humans, livestock, and companion animals. MLs target the invertebrate glutamate-activated chloride channel that is not present in vertebrates. MLs are not entirely inert in vertebrates, though; they have been reported to have activity in heterologous expression systems consisting of ligand-gated ion channels that are present in the mammalian central nervous system (CNS). However, these compounds are typically not able to reach significant concentrations in the CNS because of the activity of the blood-brain barrier P-glycoprotein extrusion system. Despite this, these compounds are able to reach low levels in the CNS that may be useful in the design of novel "designer" ligand-receptor systems that can be used to directly investigate neuronal control of behavior in mammals and have potential for use in treating human neurological diseases. To determine whether MLs might affect neurons in intact brains, we investigated the activity of the ML moxidectin (MOX) at native GABA receptors. Specifically, we recorded tonic and phasic miniature inhibitory postsynaptic currents (mIPSCs) in ex vivo brain slices. Our data show that MOX potentiated tonic GABA currents in a dose-dependent manner but had no concomitant effects on phasic GABA currents (i.e., MOX had no effect on the amplitude, frequency, or decay kinetics of mIPSCs). These studies indicate that behavioral experiments that implement a ML-based novel ligand-receptor system should take care to control for potential effects of the ML on native tonic GABA receptors. NEW & NOTEWORTHY We have identified a novel mechanism of action in the mammalian central nervous system for the antihelminthic moxidectin, commonly prescribed to animals worldwide and currently being evaluated for use in humans. Specifically, moxidectin applied to rodent brain slices selectively enhanced the tonic GABA conductance of hippocampal pyramidal neurons.
Collapse
Affiliation(s)
- Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine , Salt Lake City, Utah
| | - Anne Gibson
- Department of Neurosurgery, University of Utah School of Medicine , Salt Lake City, Utah
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine , Salt Lake City, Utah
| |
Collapse
|
35
|
Neuromodulation Using Optogenetics and Related Technologies. Neuromodulation 2018. [DOI: 10.1016/b978-0-12-805353-9.00035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Devarakonda K, Stanley S. Investigating metabolic regulation using targeted neuromodulation. Ann N Y Acad Sci 2017; 1411:83-95. [PMID: 29106710 DOI: 10.1111/nyas.13468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/11/2017] [Accepted: 08/18/2017] [Indexed: 01/13/2023]
Abstract
The central nervous system (CNS) plays a vital role in regulating energy balance and metabolism. Over the last 50 years, studies in animal models have allowed us to identify critical CNS regions involved in these processes and even crucial cell populations. Now, techniques for genetically and anatomically targeted manipulation of specific neural populations using light (optogenetic), ligands (chemogenetic), or magnetic fields (radiogenetic/magnetogenetic) allow detailed investigation of circuits involved in metabolic regulation. In this review, we provide a brief overview of recent studies using light- and magnetic field-regulated neural activity to investigate the neural circuits contributing to metabolic control.
Collapse
Affiliation(s)
- Kavya Devarakonda
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah Stanley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
37
|
Weir GA, Middleton SJ, Clark AJ, Daniel T, Khovanov N, McMahon SB, Bennett DL. Using an engineered glutamate-gated chloride channel to silence sensory neurons and treat neuropathic pain at the source. Brain 2017; 140:2570-2585. [PMID: 28969375 PMCID: PMC5841150 DOI: 10.1093/brain/awx201] [Citation(s) in RCA: 40] [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: 03/15/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022] Open
Abstract
See Basbaum (doi:10.1093/brain/awx227) for a scientific commentary on this article. Peripheral neuropathic pain arises as a consequence of injury to sensory neurons; the development of ectopic activity in these neurons is thought to be critical for the induction and maintenance of such pain. Local anaesthetics and anti-epileptic drugs can suppress hyperexcitability; however, these drugs are complicated by unwanted effects on motor, central nervous system and cardiac function, and alternative more selective treatments to suppress hyperexcitability are therefore required. Here we show that a glutamate-gated chloride channel modified to be activated by low doses of ivermectin (but not glutamate) is highly effective in silencing sensory neurons and reversing neuropathic pain-related hypersensitivity. Activation of the glutamate-gated chloride channel expressed in either rodent or human induced pluripotent stem cell-derived sensory neurons in vitro potently inhibited their response to both electrical and algogenic stimuli. We have shown that silencing is achieved both at nerve terminals and the soma and is independent of membrane hyperpolarization and instead likely mediated by lowering of the membrane resistance. Using intrathecal adeno-associated virus serotype 9-based delivery, the glutamate-gated chloride channel was successfully targeted to mouse sensory neurons in vivo, resulting in high level and long-lasting expression of the channel selectively in sensory neurons. This enabled reproducible and reversible modulation of thermal and mechanical pain thresholds in vivo; analgesia was observed for 3 days after a single systemic dose of ivermectin. We did not observe any motor or proprioceptive deficits and noted no reduction in cutaneous afferent innervation or upregulation of the injury marker ATF3 following prolonged glutamate-gated chloride channel expression. Established mechanical and cold pain-related hypersensitivity generated by the spared nerve injury model of neuropathic pain was reversed by ivermectin treatment. The efficacy of ivermectin in ameliorating behavioural hypersensitivity was mirrored at the cellular level by a cessation of ectopic activity in sensory neurons. These findings demonstrate the importance of aberrant afferent input in the maintenance of neuropathic pain and the potential for targeted chemogenetic silencing as a new treatment modality in neuropathic pain.
Collapse
Affiliation(s)
- Greg A Weir
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alex J Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Tarun Daniel
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | | | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Wiegert JS, Mahn M, Prigge M, Printz Y, Yizhar O. Silencing Neurons: Tools, Applications, and Experimental Constraints. Neuron 2017; 95:504-529. [PMID: 28772120 DOI: 10.1016/j.neuron.2017.06.050] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022]
Abstract
Reversible silencing of neuronal activity is a powerful approach for isolating the roles of specific neuronal populations in circuit dynamics and behavior. In contrast with neuronal excitation, for which the majority of studies have used a limited number of optogenetic and chemogenetic tools, the number of genetically encoded tools used for inhibition of neuronal activity has vastly expanded. Silencing strategies vary widely in their mechanism of action and in their spatial and temporal scales. Although such manipulations are commonly applied, the design and interpretation of neuronal silencing experiments present unique challenges, both technically and conceptually. Here, we review the most commonly used tools for silencing neuronal activity and provide an in-depth analysis of their mechanism of action and utility for particular experimental applications. We further discuss the considerations that need to be given to experimental design, analysis, and interpretation of collected data. Finally, we discuss future directions for the development of new silencing approaches in neuroscience.
Collapse
Affiliation(s)
- J Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, Falkenried 94, 20251 Hamburg, Germany
| | - Mathias Mahn
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Matthias Prigge
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoav Printz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ofer Yizhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
39
|
Resolving Behavioral Output via Chemogenetic Designer Receptors Exclusively Activated by Designer Drugs. J Neurosci 2017; 36:9268-82. [PMID: 27605603 DOI: 10.1523/jneurosci.1333-16.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) have proven to be highly effective neuromodulatory tools for the investigation of neural circuits underlying behavioral outputs. They exhibit a number of advantages: they rely on cell-specific manipulations through canonical intracellular signaling pathways, they are easy and cost-effective to implement in a laboratory setting, and they are easily scalable for single-region or full-brain manipulations. On the other hand, DREADDs rely on ligand-G-protein-coupled receptor interactions, leading to coarse temporal dynamics. In this review we will provide a brief overview of DREADDs, their implementation, and the advantages and disadvantages of their use in animal systems. We also will provide numerous examples of their use across a broad variety of biomedical research fields.
Collapse
|
40
|
Forcelli PA. Applications of optogenetic and chemogenetic methods to seizure circuits: Where to go next? J Neurosci Res 2017; 95:2345-2356. [PMID: 28791729 DOI: 10.1002/jnr.24135] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/05/2017] [Accepted: 07/14/2017] [Indexed: 01/02/2023]
Abstract
Epilepsy is the quintessential circuit disorder, with seizure activity propagating through anatomically constrained pathways. These pathways, necessary for normal sensory, motor, and cognitive function, are hijacked during seizures. Understanding the network architecture at the level of both local microcircuits and distributed macrocircuits may provide new therapeutic avenues for the treatment of epilepsy. Over the past decade, optogenetic and chemogenetic tools have enabled previously impossible levels of functional circuit mapping in neuroscience. In this review, examples of the application of optogenetics and chemogenetics to epilepsy are raised, the comparative strengths and weaknesses of these approaches are discussed for both preclinical and translational applications, and recent applications of these approaches in other areas of neuroscience are highlighted. These points are raised in an effort to highlight the potential of these methods to address additional unanswered questions in epilepsy.
Collapse
Affiliation(s)
- Patrick A Forcelli
- Department of Pharmacology & Physiology, Department of Neuroscience, and Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine, Washington, DC
| |
Collapse
|
41
|
Islam R, Zhang Y, Xu L, Sah P, Lynch JW. A Chemogenetic Receptor That Enhances the Magnitude and Frequency of Glycinergic Inhibitory Postsynaptic Currents without Inducing a Tonic Chloride Flux. ACS Chem Neurosci 2017; 8:460-467. [PMID: 27958714 DOI: 10.1021/acschemneuro.6b00382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The gene transfer-mediated expression of inhibitory ion channels in nociceptive neurons holds promise for treating intractable pain. Chemogenetics, which involves expressing constructs activated by biologically inert molecules, is of particular interest as it permits tunable neuromodulation. However, current chloride-permeable chemogenetic constructs are problematic as they mediate a tonic chloride influx which over time would deplete the chloride electrochemical gradient and reduce inhibitory efficacy. Inflammatory pain sensitization can be caused by prostaglandin E2-mediated inhibition of glycinergic inhibitory postsynaptic currents in spinal nociceptive neurons. We developed a highly conducting (100 pS) inhibitory chemogenetic construct based on a human glycine receptor (α1Y279F,A288G) with high ivermectin sensitivity. When virally infected into spinal neurons, 10 nM ivermectin increased the magnitude and frequency of glycinergic postsynaptic currents without activating a tonic chloride flux. The construct should thus produce analgesia. Its human origin and the well-established biocompatibility of its ligand suggest it may be suited to human use.
Collapse
Affiliation(s)
- Robiul Islam
- Queensland Brain Institute and ‡School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yan Zhang
- Queensland Brain Institute and ‡School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Li Xu
- Queensland Brain Institute and ‡School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Pankaj Sah
- Queensland Brain Institute and ‡School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joseph W. Lynch
- Queensland Brain Institute and ‡School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
42
|
Islam R, Keramidas A, Xu L, Durisic N, Sah P, Lynch JW. Ivermectin-Activated, Cation-Permeable Glycine Receptors for the Chemogenetic Control of Neuronal Excitation. ACS Chem Neurosci 2016; 7:1647-1657. [PMID: 27611437 DOI: 10.1021/acschemneuro.6b00168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The ability to control neuronal activation is rapidly advancing our understanding of brain function and is widely viewed as having eventual therapeutic application. Although several highly effective optogenetic, optochemical genetic, and chemogenetic techniques have been developed for this purpose, new approaches may provide better solutions for addressing particular questions and would increase the number of neuronal populations that can be controlled independently. An early chemogenetic neuronal silencing method employed a glutamate receptor Cl- channel engineered for activation by 1-3 nM ivermectin. This construct has been validated in vivo. Here, we sought to develop cation-permeable ivermectin-gated receptors that were either maximally Ca2+-permeable so as to induce neuro-excitotoxic cell death or minimally Ca2+-permeable so as to depolarize neurons with minimal excitotoxic risk. Our constructs were based on the human α1 glycine receptor Cl- channel due to its high conductance, human origin, high ivermectin sensitivity (once mutated), and because pore mutations that render it permeable to Na+ alone or Na+ plus Ca2+ are well characterized. We developed a Ca2+-impermeable excitatory receptor by introducing the F207A/P-2'Δ/A-1'E/T13'V/A288G mutations and a Ca2+-permeable excitatory receptor by introducing the F207A/A-1'E/A288G mutations. The latter receptor efficiently induces cell death and strongly depolarizes neurons at nanomolar ivermectin concentrations.
Collapse
Affiliation(s)
- Robiul Islam
- Queensland
Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Angelo Keramidas
- Queensland
Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Li Xu
- Queensland
Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nela Durisic
- Queensland
Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Pankaj Sah
- Queensland
Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joseph W. Lynch
- Queensland
Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
- School
of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
43
|
McEown K, Takata Y, Cherasse Y, Nagata N, Aritake K, Lazarus M. Chemogenetic inhibition of the medial prefrontal cortex reverses the effects of REM sleep loss on sucrose consumption. eLife 2016; 5. [PMID: 27919319 PMCID: PMC5140266 DOI: 10.7554/elife.20269] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/14/2016] [Indexed: 01/29/2023] Open
Abstract
Rapid eye movement (REM) sleep loss is associated with increased consumption of weight-promoting foods. The prefrontal cortex (PFC) is thought to mediate reward anticipation. However, the precise role of the PFC in mediating reward responses to highly palatable foods (HPF) after REM sleep deprivation is unclear. We selectively reduced REM sleep in mice over a 25–48 hr period and chemogenetically inhibited the medial PFC (mPFC) by using an altered glutamate-gated and ivermectin-gated chloride channel that facilitated neuronal inhibition through hyperpolarizing infected neurons. HPF consumption was measured while the mPFC was inactivated and REM sleep loss was induced. We found that REM sleep loss increased HPF consumption compared to control animals. However, mPFC inactivation reversed the effect of REM sleep loss on sucrose consumption without affecting fat consumption. Our findings provide, for the first time, a causal link between REM sleep, mPFC function and HPF consumption. DOI:http://dx.doi.org/10.7554/eLife.20269.001
Collapse
Affiliation(s)
- Kristopher McEown
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yohko Takata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Nanae Nagata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Kosuke Aritake
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
44
|
Obenhaus HA, Rozov A, Bertocchi I, Tang W, Kirsch J, Betz H, Sprengel R. Causal Interrogation of Neuronal Networks and Behavior through Virally Transduced Ivermectin Receptors. Front Mol Neurosci 2016; 9:75. [PMID: 27625595 PMCID: PMC5004486 DOI: 10.3389/fnmol.2016.00075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/10/2016] [Indexed: 01/14/2023] Open
Abstract
The causal interrogation of neuronal networks involved in specific behaviors requires the spatially and temporally controlled modulation of neuronal activity. For long-term manipulation of neuronal activity, chemogenetic tools provide a reasonable alternative to short-term optogenetic approaches. Here we show that virus mediated gene transfer of the ivermectin (IVM) activated glycine receptor mutant GlyRα1AG can be used for the selective and reversible silencing of specific neuronal networks in mice. In the striatum, dorsal hippocampus, and olfactory bulb, GlyRα1AG promoted IVM dependent effects in representative behavioral assays. Moreover, GlyRα1AG mediated silencing had a strong and reversible impact on neuronal ensemble activity and c-Fos activation in the olfactory bulb. Together our results demonstrate that long-term, reversible and re-inducible neuronal silencing via GlyRα1AG is a promising tool for the interrogation of network mechanisms underlying the control of behavior and memory formation.
Collapse
Affiliation(s)
- Horst A Obenhaus
- Sprengel Research Group, Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany
| | - Andrei Rozov
- OpenLab of Neurobiology, Kazan Federal UniversityKazan, Russia; Division of Neuro- and Sensory Physiology, Institute of Physiology and Pathophysiology, Heidelberg UniversityHeidelberg, Germany
| | - Ilaria Bertocchi
- Sprengel Research Group, Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany
| | - Wannan Tang
- Sprengel Research Group, Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany
| | - Joachim Kirsch
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University Heidelberg, Germany
| | - Heinrich Betz
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany
| | - Rolf Sprengel
- Sprengel Research Group, Department of Molecular Neurobiology, Max Planck Institute for Medical ResearchHeidelberg, Germany; Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg UniversityHeidelberg, Germany
| |
Collapse
|
45
|
Zhao R, Grunke SD, Keralapurath MM, Yetman MJ, Lam A, Lee TC, Sousounis K, Jiang Y, Swing DA, Tessarollo L, Ji D, Jankowsky JL. Impaired Recall of Positional Memory following Chemogenetic Disruption of Place Field Stability. Cell Rep 2016; 16:793-804. [PMID: 27373150 PMCID: PMC4956499 DOI: 10.1016/j.celrep.2016.06.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/03/2016] [Accepted: 06/03/2016] [Indexed: 11/06/2022] Open
Abstract
The neural network of the temporal lobe is thought to provide a cognitive map of our surroundings. Functional analysis of this network has been hampered by coarse tools that often result in collateral damage to other circuits. We developed a chemogenetic system to temporally control electrical input into the hippocampus. When entorhinal input to the perforant path was acutely silenced, hippocampal firing patterns became destabilized and underwent extensive remapping. We also found that spatial memory acquired prior to neural silencing was impaired by loss of input through the perforant path. Together, our experiments show that manipulation of entorhinal activity destabilizes spatial coding and disrupts spatial memory. Moreover, we introduce a chemogenetic model for non-invasive neuronal silencing that offers multiple advantages over existing strategies in this setting.
Collapse
Affiliation(s)
- Rong Zhao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stacy D Grunke
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Michael J Yetman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander Lam
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Cognitive Science, Rice University, Houston, TX 77251, USA
| | - Tang-Cheng Lee
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Yongying Jiang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Deborah A Swing
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Daoyun Ji
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Joanna L Jankowsky
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Abstract
To understand brain function, it is essential that we discover how cellular signaling specifies normal and pathological brain function. In this regard, chemogenetic technologies represent valuable platforms for manipulating neuronal and non-neuronal signal transduction in a cell-type-specific fashion in freely moving animals. Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-based chemogenetic tools are now commonly used by neuroscientists to identify the circuitry and cellular signals that specify behavior, perceptions, emotions, innate drives, and motor functions in species ranging from flies to nonhuman primates. Here I provide a primer on DREADDs highlighting key technical and conceptual considerations and identify challenges for chemogenetics going forward.
Collapse
|
47
|
Heusser SA, Yoluk Ö, Klement G, Riederer EA, Lindahl E, Howard RJ. Functional characterization of neurotransmitter activation and modulation in a nematode model ligand-gated ion channel. J Neurochem 2016; 138:243-53. [PMID: 27102368 DOI: 10.1111/jnc.13644] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/21/2016] [Accepted: 04/07/2016] [Indexed: 11/29/2022]
Abstract
The superfamily of pentameric ligand-gated ion channels includes neurotransmitter receptors that mediate fast synaptic transmission in vertebrates, and are targets for drugs including alcohols, anesthetics, benzodiazepines, and anticonvulsants. However, the mechanisms of ion channel opening, gating, and modulation in these receptors leave many open questions, despite their pharmacological importance. Subtle conformational changes in both the extracellular and transmembrane domains are likely to influence channel opening, but have been difficult to characterize given the limited structural data available for human membrane proteins. Recent crystal structures of a modified Caenorhabditis elegans glutamate-gated chloride channel (GluCl) in multiple states offer an appealing model system for structure-function studies. However, the pharmacology of the crystallographic GluCl construct is not well established. To establish the functional relevance of this system, we used two-electrode voltage-clamp electrophysiology in Xenopus oocytes to characterize activation of crystallographic and native-like GluCl constructs by L-glutamate and ivermectin. We also tested modulation by ethanol and other anesthetic agents, and used site-directed mutagenesis to explore the role of a region of Loop F which was implicated in ligand gating by molecular dynamics simulations. Our findings indicate that the crystallographic construct functionally models concentration-dependent agonism and allosteric modulation of pharmacologically relevant receptors. Specific substitutions at residue Leu174 in loop F altered direct L-glutamate activation, consistent with computational evidence for this region's role in ligand binding. These insights demonstrate conservation of activation and modulation properties in this receptor family, and establish a framework for GluCl as a model system, including new possibilities for drug discovery. In this study, we elucidate the validity of a modified glutamate-gated chloride channel (GluClcryst ) as a structurally accessible model for GABAA receptors. In contrast to native-like controls, GluClcryst exhibits classical activation by its neurotransmitter ligand L-glutamate. The modified channel is also sensitive to allosteric modulators associated with human GABAA receptors, and to site-directed mutations predicted to alter channel opening.
Collapse
Affiliation(s)
- Stephanie A Heusser
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Özge Yoluk
- Swedish e-Science Research Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Göran Klement
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Erika A Riederer
- Department of Chemistry, Skidmore College, Saratoga Springs, NY, USA
| | - Erik Lindahl
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden.,Swedish e-Science Research Center, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rebecca J Howard
- Department of Chemistry, Skidmore College, Saratoga Springs, NY, USA
| |
Collapse
|
48
|
Sizemore RJ, Seeger-Armbruster S, Hughes SM, Parr-Brownlie LC. Viral vector-based tools advance knowledge of basal ganglia anatomy and physiology. J Neurophysiol 2016; 115:2124-46. [PMID: 26888111 PMCID: PMC4869490 DOI: 10.1152/jn.01131.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/16/2016] [Indexed: 01/07/2023] Open
Abstract
Viral vectors were originally developed to deliver genes into host cells for therapeutic potential. However, viral vector use in neuroscience research has increased because they enhance interpretation of the anatomy and physiology of brain circuits compared with conventional tract tracing or electrical stimulation techniques. Viral vectors enable neuronal or glial subpopulations to be labeled or stimulated, which can be spatially restricted to a single target nucleus or pathway. Here we review the use of viral vectors to examine the structure and function of motor and limbic basal ganglia (BG) networks in normal and pathological states. We outline the use of viral vectors, particularly lentivirus and adeno-associated virus, in circuit tracing, optogenetic stimulation, and designer drug stimulation experiments. Key studies that have used viral vectors to trace and image pathways and connectivity at gross or ultrastructural levels are reviewed. We explain how optogenetic stimulation and designer drugs used to modulate a distinct pathway and neuronal subpopulation have enhanced our mechanistic understanding of BG function in health and pathophysiology in disease. Finally, we outline how viral vector technology may be applied to neurological and psychiatric conditions to offer new treatments with enhanced outcomes for patients.
Collapse
Affiliation(s)
- Rachel J Sizemore
- Department of Anatomy, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Sonja Seeger-Armbruster
- Department of Physiology, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand; and
| | - Stephanie M Hughes
- Department of Biochemistry, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Louise C Parr-Brownlie
- Department of Anatomy, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand;
| |
Collapse
|
49
|
Abstract
Advances in neuro-technology for mapping, manipulating, and monitoring molecularly defined cell types are rapidly advancing insight into neural circuits that regulate appetite. Here, we review these important tools and their applications in circuits that control food seeking and consumption. Technical capabilities provided by these tools establish a rigorous experimental framework for research into the neurobiology of hunger.
Collapse
Affiliation(s)
- Scott M Sternson
- Janelia Research Campus, HHMI, 19700 Helix Drive, Ashburn, VA 20147, USA.
| | - Deniz Atasoy
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810 Istanbul, Turkey
| | - J Nicholas Betley
- Janelia Research Campus, HHMI, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Fredrick E Henry
- Janelia Research Campus, HHMI, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Shengjin Xu
- Janelia Research Campus, HHMI, 19700 Helix Drive, Ashburn, VA 20147, USA
| |
Collapse
|
50
|
Auffenberg E, Jurik A, Mattusch C, Stoffel R, Genewsky A, Namendorf C, Schmid RM, Rammes G, Biel M, Uhr M, Moosmang S, Michalakis S, Wotjak CT, Thoeringer CK. Remote and reversible inhibition of neurons and circuits by small molecule induced potassium channel stabilization. Sci Rep 2016; 6:19293. [PMID: 26757616 PMCID: PMC4725838 DOI: 10.1038/srep19293] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 12/09/2015] [Indexed: 01/12/2023] Open
Abstract
Manipulating the function of neurons and circuits that translate electrical and chemical signals into behavior represents a major challenges in neuroscience. In addition to optogenetic methods using light-activatable channels, pharmacogenetic methods with ligand induced modulation of cell signaling and excitability have been developed. However, they are largely based on ectopic expression of exogenous or chimera proteins. Now, we describe the remote and reversible expression of a Kir2.1 type potassium channel using the chemogenetic technique of small molecule induced protein stabilization. Based on shield1-mediated shedding of a destabilizing domain fused to a protein of interest and inhibition of protein degradation, this principle has been adopted for biomedicine, but not in neuroscience so far. Here, we apply this chemogenetic approach in brain research for the first time in order to control a potassium channel in a remote and reversible manner. We could show that shield1-mediated ectopic Kir2.1 stabilization induces neuronal silencing in vitro and in vivo in the mouse brain. We also validated this novel pharmacogenetic method in different neurobehavioral paradigms.The DD-Kir2.1 may complement the existing portfolio of pharmaco- and optogenetic techniques for specific neuron manipulation, but it may also provide an example for future applications of this principle in neuroscience research.
Collapse
Affiliation(s)
- Eva Auffenberg
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Angela Jurik
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Corinna Mattusch
- Institute of Anesthesiology, Technical University of Munich, Germany
| | - Rainer Stoffel
- Max Planck Institute of Psychiatry, Department of Stress Physiology and Neurogenetics, Munich, Germany
| | - Andreas Genewsky
- Max Planck Institute of Psychiatry, Department of Stress Physiology and Neurogenetics, Munich, Germany
| | - Christian Namendorf
- Max Planck Institute of Psychiatry, Department of Stress Physiology and Neurogenetics, Munich, Germany
| | - Roland M Schmid
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Gerhard Rammes
- Institute of Anesthesiology, Technical University of Munich, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich (CIPSM) and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University of Munich, Germany
| | - Manfred Uhr
- Max Planck Institute of Psychiatry, Department of Stress Physiology and Neurogenetics, Munich, Germany
| | - Sven Moosmang
- Institute of Pharmacology, Technical University of Munich, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich (CIPSM) and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University of Munich, Germany
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Department of Stress Physiology and Neurogenetics, Munich, Germany
| | - Christoph K Thoeringer
- Department of Internal Medicine II, Klinikum rechts der Isar, Technical University of Munich, Germany
| |
Collapse
|