401
|
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
|
402
|
Role of Recombinant DNA Technology to Improve Life. Int J Genomics 2016; 2016:2405954. [PMID: 28053975 PMCID: PMC5178364 DOI: 10.1155/2016/2405954] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/21/2016] [Accepted: 11/06/2016] [Indexed: 12/26/2022] Open
Abstract
In the past century, the recombinant DNA technology was just an imagination that desirable characteristics can be improved in the living bodies by controlling the expressions of target genes. However, in recent era, this field has demonstrated unique impacts in bringing advancement in human life. By virtue of this technology, crucial proteins required for health problems and dietary purposes can be produced safely, affordably, and sufficiently. This technology has multidisciplinary applications and potential to deal with important aspects of life, for instance, improving health, enhancing food resources, and resistance to divergent adverse environmental effects. Particularly in agriculture, the genetically modified plants have augmented resistance to harmful agents, enhanced product yield, and shown increased adaptability for better survival. Moreover, recombinant pharmaceuticals are now being used confidently and rapidly attaining commercial approvals. Techniques of recombinant DNA technology, gene therapy, and genetic modifications are also widely used for the purpose of bioremediation and treating serious diseases. Due to tremendous advancement and broad range of application in the field of recombinant DNA technology, this review article mainly focuses on its importance and the possible applications in daily life.
Collapse
|
403
|
Developmentally defined forebrain circuits regulate appetitive and aversive olfactory learning. Nat Neurosci 2016; 20:20-23. [PMID: 27918532 PMCID: PMC5191939 DOI: 10.1038/nn.4452] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022]
Abstract
Postnatal and adult neurogenesis are region- and modality-specific, but the significance of developmentally distinct neuronal populations remains unclear. We demonstrate that chemogenetic inactivation of a subset of forebrain and olfactory neurons generated at birth disrupts responses to an aversive odor. In contrast, novel appetitive odor learning is sensitive to inactivation of adult born neurons, unveiling that developmentally defined sets of neurons may differentially participate in hedonic aspects of sensory learning.
Collapse
|
404
|
Baslow MH, Cain CK, Sears R, Wilson DA, Bachman A, Gerum S, Guilfoyle DN. Stimulation-induced transient changes in neuronal activity, blood flow and N-acetylaspartate content in rat prefrontal cortex: a chemogenetic fMRS-BOLD study. NMR IN BIOMEDICINE 2016; 29:1678-1687. [PMID: 27696530 PMCID: PMC5123928 DOI: 10.1002/nbm.3629] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/11/2016] [Accepted: 08/29/2016] [Indexed: 06/01/2023]
Abstract
Brain activation studies in humans have shown the dynamic nature of neuronal N-acetylaspartate (NAA) and N-acetylaspartylglutamate (NAAG) based on changes in their MRS signals in response to stimulation. These studies demonstrated that upon visual stimulation there was a focal increase in cerebral blood flow (CBF) and a decrease in NAA or in the total of NAA and NAAG signals in the visual cortex, and that these changes were reversed upon cessation of stimulation. In the present study we have developed an animal model in order to explore the relationships between brain stimulation, neuronal activity, CBF and NAA. We use "designer receptor exclusively activated by designer drugs" (DREADDs) technology for site-specific neural activation, a local field potential electrophysiological method for measurement of changes in the rate of neuronal activity, functional MRS for measurement of changes in NAA and a blood oxygenation level-dependent (BOLD) MR technique for evaluating changes in CBF. We show that stimulation of the rat prefrontal cortex using DREADDs results in the following: (i) an increase in level of neuronal activity; (ii) an increase in BOLD and (iii) a decrease in the NAA signal. These findings show for the first time the tightly coupled relationships between stimulation, neuron activity, CBF and NAA dynamics in brain, and also provide the first demonstration of the novel inverse stimulation-NAA phenomenon in an animal model.
Collapse
Affiliation(s)
- Morris H. Baslow
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Christopher K. Cain
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, New York University Langone School of Medicine, 560 1 Avenue, New York, NY, 10016, USA
| | - Robert Sears
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, New York University Langone School of Medicine, 560 1 Avenue, New York, NY, 10016, USA
- Department of Neuroscience & Physiology, New York University Langone School of Medicine, 560 1 Avenue, New York, NY, 10016, USA
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
- Department of Child & Adolescent Psychiatry, New York University Langone School of Medicine, 560 1 Avenue, New York, NY, 10016, USA
- Department of Neuroscience & Physiology, New York University Langone School of Medicine, 560 1 Avenue, New York, NY, 10016, USA
| | - Alvin Bachman
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Scott Gerum
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - David N. Guilfoyle
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| |
Collapse
|
405
|
Dimidschstein J, Chen Q, Tremblay R, Rogers SL, Saldi GA, Guo L, Xu C, Liu R, Lu C, Chu J, Avery MC, Rashid SM, Baek M, Jacob AL, Smith GB, Wilson DE, Kosche G, Kruglikov I, Rusielewicz T, Kotak VC, Mowery TM, Anderson SA, Callaway EM, Dasen JS, Fitzpatrick D, Fossati V, Long MA, Noggle S, Reynolds JH, Sanes DH, Rudy B, Feng G, Fishell G. A viral strategy for targeting and manipulating interneurons across vertebrate species. Nat Neurosci 2016; 19:1743-1749. [PMID: 27798629 PMCID: PMC5348112 DOI: 10.1038/nn.4430] [Citation(s) in RCA: 359] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022]
Abstract
A fundamental impediment to understanding the brain is the availability of inexpensive and robust methods for targeting and manipulating specific neuronal populations. The need to overcome this barrier is pressing because there are considerable anatomical, physiological, cognitive and behavioral differences between mice and higher mammalian species in which it is difficult to specifically target and manipulate genetically defined functional cell types. In particular, it is unclear the degree to which insights from mouse models can shed light on the neural mechanisms that mediate cognitive functions in higher species, including humans. Here we describe a novel recombinant adeno-associated virus that restricts gene expression to GABAergic interneurons within the telencephalon. We demonstrate that the viral expression is specific and robust, allowing for morphological visualization, activity monitoring and functional manipulation of interneurons in both mice and non-genetically tractable species, thus opening the possibility to study GABAergic function in virtually any vertebrate species.
Collapse
Affiliation(s)
- J Dimidschstein
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
- Center for Genomics & Systems Biology, New York University, Abu Dhabi, UAE
| | - Q Chen
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - R Tremblay
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - SL Rogers
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - GA Saldi
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - L Guo
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
- Center for Genomics & Systems Biology, New York University, Abu Dhabi, UAE
| | - C Xu
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
- Center for Genomics & Systems Biology, New York University, Abu Dhabi, UAE
| | - R Liu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - C Lu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - J Chu
- Department of Psychiatry, Children’s Hospital of Philadelphia and UPenn School of Medicine, Philadelphia, PA 19104, USA
| | - MC Avery
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - SM Rashid
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - M Baek
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - AL Jacob
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - GB Smith
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - DE Wilson
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - G Kosche
- NYU Neuroscience Institute and the Department of Otolaryngology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - I Kruglikov
- New York Stem Cell Foundation, New York, NY10023, USA
| | - T Rusielewicz
- New York Stem Cell Foundation, New York, NY10023, USA
| | - VC Kotak
- NYU Center for Neural Science, New York University, New York, NY 10003, USA
| | - TM Mowery
- NYU Center for Neural Science, New York University, New York, NY 10003, USA
| | - SA Anderson
- Department of Psychiatry, Children’s Hospital of Philadelphia and UPenn School of Medicine, Philadelphia, PA 19104, USA
| | - EM Callaway
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - JS Dasen
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - D Fitzpatrick
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - V Fossati
- New York Stem Cell Foundation, New York, NY10023, USA
| | - MA Long
- NYU Neuroscience Institute and the Department of Otolaryngology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - S Noggle
- New York Stem Cell Foundation, New York, NY10023, USA
| | - JH Reynolds
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - DH Sanes
- NYU Center for Neural Science, New York University, New York, NY 10003, USA
| | - B Rudy
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - G Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - G Fishell
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University Langone Medical Center, New York, NY 10016, USA
- Center for Genomics & Systems Biology, New York University, Abu Dhabi, UAE
| |
Collapse
|
406
|
Direct dorsal hippocampal-prelimbic cortex connections strengthen fear memories. Nat Neurosci 2016; 20:52-61. [PMID: 27869801 PMCID: PMC5191950 DOI: 10.1038/nn.4443] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/26/2016] [Indexed: 01/26/2023]
Abstract
The ability to regulate the consolidation and strengthening of memories for threatening experiences is critical for mental health, and its dysregulation may lead to psychopathologies. Re-exposure to the context in which the threat was experienced can either increase or decrease fear response through distinct processes known, respectively, as reconsolidation or extinction. Using a context retrieval-dependent memory enhancement paradigm in rats, we report that memory strengthens through the activation of direct projections from the dorsal hippocampus (dHC) to the prelimbic (PL) cortex and of critical PL molecular mechanisms, which are not required for extinction. Furthermore, while a sustained PL BDNF expression is required for memory consolidation, retrieval engages PL BDNF to regulate the excitatory and inhibitory synaptic proteins neuroligin 1 and neuroligin 2, which promote memory strengthening while inhibiting extinction. Thus, context retrieval-mediated fear memory enhancement results from a concerted action of mechanisms that strengthen memory through reconsolidation while suppressing extinction.
Collapse
|
407
|
Prasad AA, McNally GP. Ventral Pallidum Output Pathways in Context-Induced Reinstatement of Alcohol Seeking. J Neurosci 2016; 36:11716-11726. [PMID: 27852779 PMCID: PMC6705636 DOI: 10.1523/jneurosci.2580-16.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/24/2016] [Accepted: 09/26/2016] [Indexed: 11/21/2022] Open
Abstract
Ventral pallidum (VP) is a well-established locus for the reinforcing effects of drugs of abuse and reinstatement of drug seeking. However, VP neurons are at the origin of multiple output pathways, with strong projections to ventral tegmental area (VTA), subthalamic nucleus (STN), lateral hypothalamus, among others, and the roles of these VP output pathways in reinstatement of drug seeking remain poorly understood. Here we addressed these issues using a combination of neuroanatomical tracing and chemogenetic approaches. First, using dual-retrograde tracing, we show that VP neurons projecting to either VTA or STN are recruited during context-induced reinstatement of extinguished alcohol seeking in rats. Then, using chemogenetics, we show modulation of context-induced reinstatement and reacquisition of alcohol seeking via designer receptors exclusively activated by designer drugs excitation or inhibition of the VP. To determine the causal roles of VP → VTA and VP → STN pathways in context-induced reinstatement and reacquisition we used a chemogenetic disconnection approach and show that silencing either the VP → VTA or VP → STN pathways is sufficient to reduce both reinstatement and reacquisition of alcohol seeking. Moreover, these disconnections also each reduced responding and motivation during a progressive ratio test but had no effect on locomotor activity. Together, these results show that multiple ventral pallidal output pathways contribute to relapse to alcohol seeking. SIGNIFICANCE STATEMENT Ventral pallidum (VP) serves important roles in reward and motivation and is a critical node in the neural circuitry for reinstatement of drug seeking. Despite being a common locus for different forms of reinstatement, fundamental aspects of neural circuitry for these VP contributions to reinstatement of drug seeking remain unknown. Here we used a combination of neuroanatomical tracing and chemogenetic approaches to map the VP output pathways for context-induced reinstatement and reacquisition of alcohol seeking. We show that VP output pathways to the subthalamic nucleus and also to the ventral tegmental area are necessary for these forms of reinstatement.
Collapse
Affiliation(s)
- Asheeta A Prasad
- School of Psychology, University of New South Wales, Sydney, 2052 NSW, Australia
| | - Gavan P McNally
- School of Psychology, University of New South Wales, Sydney, 2052 NSW, Australia
| |
Collapse
|
408
|
Fliervoet LAL, Mastrobattista E. Drug delivery with living cells. Adv Drug Deliv Rev 2016; 106:63-72. [PMID: 27129442 DOI: 10.1016/j.addr.2016.04.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/25/2022]
Abstract
The field of drug delivery has grown tremendously in the past few decades by developing a wide range of advanced drug delivery systems. An interesting category is cell-based drug delivery, which includes encapsulation of drugs inside cells or attached to the surface and subsequent transportation through the body. Another approach involves genetic engineering of cells to secrete therapeutic molecules in a controlled way. The next-generation systems integrate expertise from synthetic biology to generate therapeutic gene networks for highly advanced sensory and output devices. These developments are very exciting for the drug delivery field and could radically change the way we administer biological medicines to chronically ill patients. This review is covering the use of living cells, either as transport system or production-unit, to deliver therapeutic molecules and bioactive proteins inside the body. It describes a wide range of approaches in cell-based drug delivery and highlights exceptional examples.
Collapse
Affiliation(s)
- Lies A L Fliervoet
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands.
| |
Collapse
|
409
|
Zou D, Chen L, Deng D, Jiang D, Dong F, McSweeney C, Zhou Y, Liu L, Chen G, Wu Y, Mao Y. DREADD in parvalbumin interneurons of the dentate gyrus modulates anxiety, social interaction and memory extinction. Curr Mol Med 2016; 16:91-102. [PMID: 26733123 PMCID: PMC4997952 DOI: 10.2174/1566524016666151222150024] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/03/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022]
Abstract
Parvalbumin (PV)-positive interneurons in the hippocampus play a critical role in animal memory, such as spatial working memory. However, how PV-positive interneurons in the subregions of the hippocampus affect animal behaviors remains poorly defined. Here, we achieved specific and reversible activation of PV-positive interneurons using designer receptors exclusively activated by designer drugs (DREADD) technology. Inducible DREADD expression was demonstrated in vitro in cultured neurons, in which co-transfection of the hM3D-Gq-mCherry vector with a Cre plasmid resulted in a cellular response to hM3Dq ligand clozapine-N-oxide (CNO) stimulation. In addition, the dentate gyrus (DG) of PV-Cre mice received bilateral injection of control lentivirus or lentivirus expressing double floxed hM3D-Gq-mCherry. Selective activation of PV-positive interneurons in the DG did not affect locomotor activity or depression-related behavior in mice. Interestingly, stimulation of PV-positive interneurons induced an anxiolytic effect. Activation of PVpositive interneurons appears to impair social interaction to novelty, but has no effect on social motivation. However, this defect is likely due to the anxiolytic effect as the exploratory behavior of mice expressing hM3DGq is significantly increased. Mice expressing hM3D-Gq did not affect novel object recognition. Activation of PV-positive interneurons in the DG maintains intact cued and contextual fear memory but facilitates fear extinction. Collectively, our results demonstrated that proper control of PV interneurons activity in the DG is critical for regulation of the anxiety, social interaction and fear extinction. These results improve our fundamental understanding of the physiological role of PV-positive interneurons in the hippocampus.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Y Mao
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
410
|
Too bored to stay awake. Nat Neurosci 2016; 19:1274-6. [DOI: 10.1038/nn.4383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
411
|
Bradley SJ, Tobin AB. Design of Next-Generation G Protein-Coupled Receptor Drugs: Linking Novel Pharmacology and In Vivo Animal Models. Annu Rev Pharmacol Toxicol 2016; 56:535-59. [PMID: 26738479 DOI: 10.1146/annurev-pharmtox-011613-140012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the fact that G protein-coupled receptors (GPCRs) are the most successful drug targets in history, this supergene family of cell surface receptors has yet to be fully exploited as targets in the treatment of human disease. Here, we present optimism that this may change in the future by reviewing the substantial progress made in the understanding of GPCR molecular pharmacology that has generated an extensive toolbox of ligand types that include orthosteric, allosteric, and bitopic ligands, many of which show signaling bias. We discuss how combining these advances with recently described transgenic, chemical genetic, and optogenetic animal models will provide the framework to allow for the rational design of next-generation GPCR drugs that possess increased therapeutic efficacy and decreased adverse/toxic responses.
Collapse
Affiliation(s)
- Sophie J Bradley
- MRC Toxicology Unit, University of Leicester, Leicester LE1 9HN United Kingdom; ,
| | - Andrew B Tobin
- MRC Toxicology Unit, University of Leicester, Leicester LE1 9HN United Kingdom; ,
| |
Collapse
|
412
|
Ross B, Mehta S, Zhang J. Molecular tools for acute spatiotemporal manipulation of signal transduction. Curr Opin Chem Biol 2016; 34:135-142. [PMID: 27639090 DOI: 10.1016/j.cbpa.2016.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023]
Abstract
The biochemical activities involved in signal transduction in cells are under tight spatiotemporal regulation. To study the effects of the spatial patterning and temporal dynamics of biochemical activities on downstream signaling, researchers require methods to manipulate signaling pathways acutely and rapidly. In this review, we summarize recent developments in the design of three broad classes of molecular tools for perturbing signal transduction, classified by their type of input signal: chemically induced, optically induced, and magnetically induced.
Collapse
Affiliation(s)
- Brian Ross
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
413
|
The Medial Orbitofrontal Cortex Regulates Sensitivity to Outcome Value. J Neurosci 2016; 36:4600-13. [PMID: 27098701 DOI: 10.1523/jneurosci.4253-15.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/08/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED An essential component of goal-directed decision-making is the ability to maintain flexible responding based on the value of a given reward, or "reinforcer." The medial orbitofrontal cortex (mOFC), a subregion of the ventromedial prefrontal cortex, is uniquely positioned to regulate this process. We trained mice to nose poke for food reinforcers and then stimulated this region using CaMKII-driven Gs-coupled designer receptors exclusively activated by designer drugs (DREADDs). In other mice, we silenced the neuroplasticity-associated neurotrophin brain-derived neurotrophic factor (BDNF). Activation of Gs-DREADDs increased behavioral sensitivity to reinforcer devaluation, whereas Bdnf knockdown blocked sensitivity. These changes were accompanied by modifications in breakpoint ratios in a progressive ratio task, and they were recapitulated in Bdnf(+/-)mice. Replacement of BDNF selectively in the mOFC in Bdnf(+/-)mice rescued behavioral deficiencies, as well as phosphorylation of extracellular-signal regulated kinase 1/2 (ERK1/2). Thus, BDNF expression in the mOFC is both necessary and sufficient for the expression of typical effort allocation relative to an anticipated reinforcer. Additional experiments indicated that expression of the immediate-early gene c-fos was aberrantly elevated in the Bdnf(+/-)dorsal striatum, and BDNF replacement in the mOFC normalized expression. Also, systemic administration of an MAP kinase kinase inhibitor increased breakpoint ratios, whereas the addition of discrete cues bridging the response-outcome contingency rescued breakpoints in Bdnf(+/-)mice. We argue that BDNF-ERK1/2 in the mOFC is a key regulator of "online" goal-directed action selection. SIGNIFICANCE STATEMENT Goal-directed response selection often involves predicting the consequences of one's actions and the value of potential payoffs. Lesions or chemogenetic inactivation of the medial orbitofrontal cortex (mOFC) in rats induces failures in retrieving outcome identity memories (Bradfield et al., 2015), suggesting that the healthy mOFC serves to access outcome value information when it is not immediately observable and thereby guide goal-directed decision-making. Our findings suggest that the mOFC also bidirectionally regulates effort allocation for a given reward and that expression of the neurotrophin BDNF in the mOFC is both necessary and sufficient for mice to sustain stable representations of reinforcer value.
Collapse
|
414
|
Wess J. Use of Designer G Protein-Coupled Receptors to Dissect Metabolic Pathways. Trends Endocrinol Metab 2016; 27:600-603. [PMID: 27381463 PMCID: PMC4992635 DOI: 10.1016/j.tem.2016.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/03/2016] [Accepted: 04/04/2016] [Indexed: 10/21/2022]
Abstract
G protein-coupled receptors (GPCRs) regulate virtually all metabolic processes, including glucose and energy homeostasis. Recently, the use of designer GPCRs referred to as designer receptors exclusively activated by designer drug (DREADDs) has made it possible to dissect metabolically relevant GPCR signaling pathways in a temporally and spatially controlled fashion in vivo.
Collapse
Affiliation(s)
- Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg 8A, 8 Center Drive MSC 0810, Bethesda, MD 20892, USA.
| |
Collapse
|
415
|
Iyer SM, Vesuna S, Ramakrishnan C, Huynh K, Young S, Berndt A, Lee SY, Gorini CJ, Deisseroth K, Delp SL. Optogenetic and chemogenetic strategies for sustained inhibition of pain. Sci Rep 2016; 6:30570. [PMID: 27484850 PMCID: PMC4971509 DOI: 10.1038/srep30570] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/04/2016] [Indexed: 01/02/2023] Open
Abstract
Spatially targeted, genetically-specific strategies for sustained inhibition of nociceptors may help transform pain science and clinical management. Previous optogenetic strategies to inhibit pain have required constant illumination, and chemogenetic approaches in the periphery have not been shown to inhibit pain. Here, we show that the step-function inhibitory channelrhodopsin, SwiChR, can be used to persistently inhibit pain for long periods of time through infrequent transdermally delivered light pulses, reducing required light exposure by >98% and resolving a long-standing limitation in optogenetic inhibition. We demonstrate that the viral expression of the hM4D receptor in small-diameter primary afferent nociceptor enables chemogenetic inhibition of mechanical and thermal nociception thresholds. Finally, we develop optoPAIN, an optogenetic platform to non-invasively assess changes in pain sensitivity, and use this technique to examine pharmacological and chemogenetic inhibition of pain.
Collapse
Affiliation(s)
| | - Sam Vesuna
- Bioengineering, Stanford University, Stanford, CA 94305, USA
| | | | - Karen Huynh
- Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Stephanie Young
- Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Andre Berndt
- Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Soo Yeun Lee
- Bioengineering, Stanford University, Stanford, CA 94305, USA
| | | | - Karl Deisseroth
- Bioengineering, Stanford University, Stanford, CA 94305, USA
- Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Scott L. Delp
- Bioengineering, Stanford University, Stanford, CA 94305, USA
- Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
416
|
Adenosine A2A receptors in the olfactory bulb suppress rapid eye movement sleep in rodents. Brain Struct Funct 2016; 222:1351-1366. [DOI: 10.1007/s00429-016-1281-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/26/2016] [Indexed: 12/25/2022]
|
417
|
Romer SH, Seedle K, Turner SM, Li J, Baccei ML, Crone SA. Accessory respiratory muscles enhance ventilation in ALS model mice and are activated by excitatory V2a neurons. Exp Neurol 2016; 287:192-204. [PMID: 27456268 DOI: 10.1016/j.expneurol.2016.05.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/09/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023]
Abstract
Inspiratory accessory respiratory muscles (ARMs) enhance ventilation when demands are high, such as during exercise and/or pathological conditions. Despite progressive degeneration of phrenic motor neurons innervating the diaphragm, amyotrophic lateral sclerosis (ALS) patients and rodent models are able to maintain ventilation at early stages of disease. In order to assess the contribution of ARMs to respiratory compensation in ALS, we examined the activity of ARMs and ventilation throughout disease progression in SOD1G93A ALS model mice at rest using a combination of electromyography and unrestrained whole body plethysmography. Increased ARM activity, accompanied by increased ventilation, is observed beginning at the onset of symptoms. However, ARM recruitment fails to occur at rest at late stages of disease, even though the same ARMs are used for other behaviors. Using a chemogenetic approach, we demonstrate that a glutamatergic class of neurons in the brainstem and spinal cord, the V2a class, is sufficient to drive increased ARM activity at rest in healthy mice. Additionally, we reveal pathology in the medial reticular formation of the brainstem of SOD1G93A mice using immunohistochemistry and confocal imaging. Both spinal and brainstem V2a neurons degenerate in ALS model mice, accompanied by regional activation of astrocytes and microglia. These results establish inspiratory ARM recruitment as one of the compensatory mechanisms that maintains breathing at early stages of disease and indicate that V2a neuron degeneration may contribute to ARM failure at late stages of disease.
Collapse
Affiliation(s)
- Shannon H Romer
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Kari Seedle
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Sarah M Turner
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Jie Li
- Pain Research Center, Dept. of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267
| | - Mark L Baccei
- Pain Research Center, Dept. of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267
| | - Steven A Crone
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229.
| |
Collapse
|
418
|
Shemesh Y, Forkosh O, Mahn M, Anpilov S, Sztainberg Y, Manashirov S, Shlapobersky T, Elliott E, Tabouy L, Ezra G, Adler ES, Ben-Efraim YJ, Gil S, Kuperman Y, Haramati S, Dine J, Eder M, Deussing JM, Schneidman E, Yizhar O, Chen A. Ucn3 and CRF-R2 in the medial amygdala regulate complex social dynamics. Nat Neurosci 2016; 19:1489-1496. [DOI: 10.1038/nn.4346] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022]
|
419
|
Avaliani N, Andersson M, Runegaard AH, Woldbye D, Kokaia M. DREADDs suppress seizure-like activity in a mouse model of pharmacoresistant epileptic brain tissue. Gene Ther 2016; 23:760-766. [PMID: 27416078 DOI: 10.1038/gt.2016.56] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Epilepsy is a neurological disorder with a prevalence of ≈1% of general population. Available antiepileptic drugs (AEDs) have multiple side effects and are ineffective in 30% of patients. Therefore, development of effective treatment strategies is highly needed, requiring drug-screening models that are relevant and reliable. We investigated novel chemogenetic approach, using DREADDs (designer receptors exclusively activated by designer drugs) as possible inhibitor of epileptiform activity in organotypic hippocampal slice cultures (OHSCs). The OHSCs are characterized by increased overall excitability and closely resemble features of human epileptic tissue. Studies suggest that chemically induced epileptiform activity in rat OHSCs is pharmacoresistant to most of AEDs. However, high-frequency electric stimulus train-induced bursting (STIB) in OHSCs is responsive to carbamazepine and phenytoin. We investigated whether inhibitory DREADD, hM4Di, would be effective in suppressing STIB in OHSC. hM4Di is a mutated muscarinic receptor selectively activated by otherwise inert clozapine-N-oxide, which leads to hyperpolarization in neurons. We demonstrated that this hyperpolarization effectively suppresses STIB in mouse OHSCs. As we also found that STIB in mouse OHSCs is resistant to common AED, valproic acid, collectively our findings suggest that DREADD-based strategy may be effective in suppressing epileptiform activity in a pharamcoresitant epileptic brain tissue.
Collapse
Affiliation(s)
- N Avaliani
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - M Andersson
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - A H Runegaard
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - D Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - M Kokaia
- Epilepsy Centre, Experimental Epilepsy Group, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| |
Collapse
|
420
|
Lim JHA, Stafford BK, Nguyen PL, Lien BV, Wang C, Zukor K, He Z, Huberman AD. Neural activity promotes long-distance, target-specific regeneration of adult retinal axons. Nat Neurosci 2016; 19:1073-84. [PMID: 27399843 PMCID: PMC5708130 DOI: 10.1038/nn.4340] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/01/2016] [Indexed: 11/23/2022]
Abstract
Axons in the mammalian central nervous system (CNS) fail to regenerate after injury. Here we show that if retinal ganglion cell (RGC) activity is increased by visual stimulation or using chemogenetics, their axons regenerate. We also show that if enhancement of neural activity is combined with elevation of the cell growth-promoting pathway involving mammalian target of rapamycin (mTOR), RGC axons regenerate the long distances necessary to re-innervate the brain. Analysis of genetically-labeled RGCs revealed this regrowth can be target specific: RGC axons navigated back to their correct visual targets and avoided targets incorrect for their function. Moreover, these regenerated connections were successful in partially rescuing a subset of visual behaviors. Our findings indicate that combining neural activity with activation of mTOR can serve as powerful tool for enhancing axon regeneration and they highlight the remarkable capacity of CNS neurons to re-establish accurate circuit connections in the adult brain.
Collapse
Affiliation(s)
- Jung-Hwan A Lim
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Benjamin K Stafford
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Phong L Nguyen
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Brian V Lien
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, USA
| | - Chen Wang
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine Zukor
- Animal, Dairy and Veterinary Sciences Department, Utah State University, Logan, Utah, USA
| | - Zhigang He
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, USA.,Department of Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
421
|
Chemogenetic approach to model hypofrontality. Med Hypotheses 2016; 93:113-6. [PMID: 27372868 DOI: 10.1016/j.mehy.2016.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/06/2016] [Accepted: 05/25/2016] [Indexed: 11/21/2022]
Abstract
Clinical evidence suggests that the prefrontal cortex (PFC) is hypofunctional in disorders including schizophrenia, drug addiction, and attention-deficit/hyperactivity disorder (ADHD). In schizophrenia, hypofrontality has been further suggested to cause both the negative and cognitive symptoms, and overactivity of dopamine neurons that project to subcortical areas. The latter may contribute to the development of positive symptoms of the disorder. Nevertheless, what causes hypofrontality and how it alters dopamine transmission in subcortical structures remain unclear due, in part, to the difficulty in modeling hypofrontality using previous techniques (e.g. PFC lesioning, focal cooling, repeated treatment with psychotomimetic drugs). We propose that the use of designer receptors exclusively activated by designer drugs (DREADDs) chemogenetic technique will allow precise interrogations of PFC functions. Combined with electrophysiological recordings, we can investigate the effects of PFC hypofunction on activity of dopamine neurons. Importantly, from a drug target discovery perspective, the use of DREADDs will enable us to examine whether chemogenetically enhancing PFC activity will reverse the behavioral abnormalities associated with PFC hypofunction and dopamine neuron overactivity, and also explore druggable targets for the treatment of schizophrenia and other disorders associated with abnormalities via modulation of the G-protein coupled receptor signaling pathway. In conclusion, the use of the DREADDs technique has several advantages over other previously employed strategies to simulate PFC hypofunction not only in terms of disease modeling but also from the viewpoint of drug target discovery.
Collapse
|
422
|
Ausländer S, Fussenegger M. Engineering Gene Circuits for Mammalian Cell-Based Applications. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a023895. [PMID: 27194045 DOI: 10.1101/cshperspect.a023895] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Synthetic gene switches are basic building blocks for the construction of complex gene circuits that transform mammalian cells into useful cell-based machines for next-generation biotechnological and biomedical applications. Ligand-responsive gene switches are cellular sensors that are able to process specific signals to generate gene product responses. Their involvement in complex gene circuits results in sophisticated circuit topologies that are reminiscent of electronics and that are capable of providing engineered cells with the ability to memorize events, oscillate protein production, and perform complex information-processing tasks. Microencapsulated mammalian cells that are engineered with closed-loop gene networks can be implanted into mice to sense disease-related input signals and to process this information to produce a custom, fine-tuned therapeutic response that rebalances animal metabolism. Progress in gene circuit design, in combination with recent breakthroughs in genome engineering, may result in tailored engineered mammalian cells with great potential for future cell-based therapies.
Collapse
Affiliation(s)
- Simon Ausländer
- Department of Biosystems Science and Engineering, ETH Zurich, CH-4058 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, CH-4058 Basel, Switzerland Faculty of Science, University of Basel, CH-4058 Basel, Switzerland
| |
Collapse
|
423
|
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
|
424
|
Schukur L, Fussenegger M. Engineering of synthetic gene circuits for (re-)balancing physiological processes in chronic diseases. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:402-22. [DOI: 10.1002/wsbm.1345] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/12/2016] [Accepted: 04/26/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Lina Schukur
- Department of Biosystems Science and Engineering; ETH Zurich; Basel Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering; ETH Zurich; Basel Switzerland
- Faculty of Science; University of Basel; Basel Switzerland
| |
Collapse
|
425
|
Rössler OG, Thiel G. Specificity of Stress-Responsive Transcription Factors Nrf2, ATF4, and AP-1. J Cell Biochem 2016; 118:127-140. [PMID: 27278863 DOI: 10.1002/jcb.25619] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/07/2016] [Indexed: 01/15/2023]
Abstract
Cellular stress leads to an upregulation of gene transcription. We asked if there is a specificity in the activation of the stress-responsive transcription factors Nrf2, ATF4, and AP-1/c-Jun, or if activation of these proteins is a redundant cellular answer toward extracellular stressors. Here, we show that oxidative stress, induced by stimulation of the cells with the oxidant arsenite, strongly activated gene transcription via the stress-responsive element (StRE), while phorbol ester or tunicamycin, activators of AP-1/c-Jun or ATF4, respectively, activated AP-1 or nutrient-sensing response element-mediated transcription. Preincubation of the cells with N-acetyl-cysteine or overexpression of thioredoxin selectively attenuated arsenite-induced upregulation of StRE-regulated transcription. Expression of either dominant-negative or constitutively active mutants of Nrf2, ATF4, or c-Jun confirmed that distinct transcription units are regulated by these transcription factors. Physiological stimuli involving the activation of either Gαq-coupled designer receptors or the protein kinases c-Jun N-terminal protein kinase or p38 strongly stimulated transcription via AP-1/c-Jun, with minimal effects on Nrf2 or ATF4-responsive promoters. Thus, activation of transcription by extracellular signaling molecules shows specificity at the level of the chemical nature of the signaling molecule, at the level of the intracellular transduction process, and at the level of signal-responsive transcription factors. J. Cell. Biochem. 118: 127-140, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Oliver G Rössler
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, Building 44, D-66421, Homburg, Germany
| | - Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, Building 44, D-66421, Homburg, Germany
| |
Collapse
|
426
|
Roman CW, Derkach VA, Palmiter RD. Genetically and functionally defined NTS to PBN brain circuits mediating anorexia. Nat Commun 2016; 7:11905. [PMID: 27301688 PMCID: PMC4912612 DOI: 10.1038/ncomms11905] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 05/11/2016] [Indexed: 11/10/2022] Open
Abstract
The central nervous system controls food consumption to maintain metabolic homoeostasis. In response to a meal, visceral signals from the gut activate neurons in the nucleus of the solitary tract (NTS) via the vagus nerve. These NTS neurons then excite brain regions known to mediate feeding behaviour, such as the lateral parabrachial nucleus (PBN). We previously described a neural circuit for appetite suppression involving calcitonin gene-related protein (CGRP)-expressing PBN (CGRPPBN) neurons; however, the molecular identity of the inputs to these neurons was not established. Here we identify cholecystokinin (CCK) and noradrenergic, dopamine β-hydroxylase (DBH)-expressing NTS neurons as two separate populations that directly excite CGRPPBN neurons. When these NTS neurons are activated using optogenetic or chemogenetic methods, food intake decreases and with chronic stimulation mice lose body weight. Our optogenetic results reveal that CCK and DBH neurons in the NTS directly engage CGRPPBN neurons to promote anorexia. Neurons in the nucleus of the solitary tract (NTS) are known to receive visceral signals from the gut during feeding. Here, the authors identify two populations of CCK- and DBH-expressing NTS neurons that work to suppress food intake when activated via opto- or chemogenetic stimulation.
Collapse
Affiliation(s)
- Carolyn W Roman
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
| | - Victor A Derkach
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, USA
| | - Richard D Palmiter
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
427
|
Milligan G, Shimpukade B, Ulven T, Hudson BD. Complex Pharmacology of Free Fatty Acid Receptors. Chem Rev 2016; 117:67-110. [PMID: 27299848 DOI: 10.1021/acs.chemrev.6b00056] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are historically the most successful family of drug targets. In recent times it has become clear that the pharmacology of these receptors is far more complex than previously imagined. Understanding of the pharmacological regulation of GPCRs now extends beyond simple competitive agonism or antagonism by ligands interacting with the orthosteric binding site of the receptor to incorporate concepts of allosteric agonism, allosteric modulation, signaling bias, constitutive activity, and inverse agonism. Herein, we consider how evolving concepts of GPCR pharmacology have shaped understanding of the complex pharmacology of receptors that recognize and are activated by nonesterified or "free" fatty acids (FFAs). The FFA family of receptors is a recently deorphanized set of GPCRs, the members of which are now receiving substantial interest as novel targets for the treatment of metabolic and inflammatory diseases. Further understanding of the complex pharmacology of these receptors will be critical to unlocking their ultimate therapeutic potential.
Collapse
Affiliation(s)
- Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow G12 8QQ, Scotland, United Kingdom
| | - Bharat Shimpukade
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark , Campusvej 55, DK-5230 Odense M, Denmark
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark , Campusvej 55, DK-5230 Odense M, Denmark
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow G12 8QQ, Scotland, United Kingdom
| |
Collapse
|
428
|
Lindsley CW, Emmitte KA, Hopkins CR, Bridges TM, Gregory KJ, Niswender CM, Conn PJ. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem Rev 2016; 116:6707-41. [PMID: 26882314 PMCID: PMC4988345 DOI: 10.1021/acs.chemrev.5b00656] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allosteric modulation of GPCRs has initiated a new era of basic and translational discovery, filled with therapeutic promise yet fraught with caveats. Allosteric ligands stabilize unique conformations of the GPCR that afford fundamentally new receptors, capable of novel pharmacology, unprecedented subtype selectivity, and unique signal bias. This review provides a comprehensive overview of the basics of GPCR allosteric pharmacology, medicinal chemistry, drug metabolism, and validated approaches to address each of the major challenges and caveats. Then, the review narrows focus to highlight recent advances in the discovery of allosteric ligands for metabotropic glutamate receptor subtypes 1-5 and 7 (mGlu1-5,7) highlighting key concepts ("molecular switches", signal bias, heterodimers) and practical solutions to enable the development of tool compounds and clinical candidates. The review closes with a section on late-breaking new advances with allosteric ligands for other GPCRs and emerging data for endogenous allosteric modulators.
Collapse
Affiliation(s)
- Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Corey R. Hopkins
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville VIC 3052, Australia
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
429
|
Jiang X, Lachance M, Rossignol E. Involvement of cortical fast-spiking parvalbumin-positive basket cells in epilepsy. PROGRESS IN BRAIN RESEARCH 2016; 226:81-126. [PMID: 27323940 DOI: 10.1016/bs.pbr.2016.04.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GABAergic interneurons of the parvalbumin-positive fast-spiking basket cells subtype (PV INs) are important regulators of cortical network excitability and of gamma oscillations, involved in signal processing and cognition. Impaired development or function of PV INs has been associated with epilepsy in various animal models of epilepsy, as well as in some genetic forms of epilepsy in humans. In this review, we provide an overview of some of the experimental data linking PV INs dysfunction with epilepsy, focusing on disorders of the specification, migration, maturation, synaptic function, or connectivity of PV INs. Furthermore, we reflect on the potential therapeutic use of cell-type specific stimulation of PV INs within active networks and on the transplantation of PV INs precursors in the treatment of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- X Jiang
- Université de Montréal, Montréal, QC, Canada; CHU Ste-Justine Research Center, Montréal, QC, Canada
| | - M Lachance
- CHU Ste-Justine Research Center, Montréal, QC, Canada
| | - E Rossignol
- Université de Montréal, Montréal, QC, Canada; CHU Ste-Justine Research Center, Montréal, QC, Canada.
| |
Collapse
|
430
|
Zhu H, Aryal DK, Olsen RHJ, Urban DJ, Swearingen A, Forbes S, Roth BL, Hochgeschwender U. Cre-dependent DREADD (Designer Receptors Exclusively Activated by Designer Drugs) mice. Genesis 2016; 54:439-46. [PMID: 27194399 DOI: 10.1002/dvg.22949] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 11/09/2022]
Abstract
DREADDs, designer receptors exclusively activated by designer drugs, are engineered G protein-coupled receptors (GPCR) which can precisely control GPCR signaling pathways (for example, Gq, Gs, and Gi). This chemogenetic technology for control of GPCR signaling has been successfully applied in a variety of in vivo studies, including in mice, to remotely control GPCR signaling, for example, in neurons, glia cells, pancreatic β-cells, or cancer cells. In order to fully explore the in vivo applications of the DREADD technology, we generated hM3Dq and hM4Di strains of mice which allow for Cre recombinase-mediated restricted expression of these pathway-selective DREADDs. With the many Cre driver lines now available, these DREADD lines will be applicable to studying a wide array of research and preclinical questions. genesis 54:439-446, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hu Zhu
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina
| | - Dipendra K Aryal
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina
| | - Reid H J Olsen
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina
| | - Daniel J Urban
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina
| | | | - Stacy Forbes
- Neurotransgenic Laboratory, Duke University, Durham, North Carolina
| | - Bryan L Roth
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina
| | | |
Collapse
|
431
|
Sciolino NR, Plummer NW, Chen YW, Alexander GM, Robertson SD, Dudek SM, McElligott ZA, Jensen P. Recombinase-Dependent Mouse Lines for Chemogenetic Activation of Genetically Defined Cell Types. Cell Rep 2016; 15:2563-73. [PMID: 27264177 DOI: 10.1016/j.celrep.2016.05.034] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/05/2016] [Accepted: 05/06/2016] [Indexed: 10/21/2022] Open
Abstract
Chemogenetic technologies, including the mutated human Gq-coupled M3 muscarinic receptor (hM3Dq), have greatly facilitated our ability to directly link changes in cellular activity to altered physiology and behavior. Here, we extend the hM3Dq toolkit with recombinase-responsive mouse lines that permit hM3Dq expression in virtually any cell type. These alleles encode a fusion protein designed to increase effective expression levels by concentrating hM3Dq to the cell body and dendrites. To illustrate their broad utility, we targeted three different genetically defined cell populations: noradrenergic neurons of the compact, bilateral locus coeruleus and two dispersed populations, Camk2a+ neurons and GFAP+ glia. In all three populations, we observed reproducible expression and confirmed that activation of hM3Dq is sufficient to dose-dependently evoke phenotypic changes, without extreme phenotypes associated with hM3Dq overexpression. These alleles offer the ability to non-invasively control activity of diverse cell types to uncover their function and dysfunction at any developmental stage.
Collapse
Affiliation(s)
- Natale R Sciolino
- Neurobiology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC 27709, USA
| | - Nicholas W Plummer
- Neurobiology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC 27709, USA
| | - Yu-Wei Chen
- Neurobiology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC 27709, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC 27709, USA
| | - Sabrina D Robertson
- Biotechnology Program, Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Serena M Dudek
- Neurobiology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC 27709, USA
| | - Zoe A McElligott
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA; Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Patricia Jensen
- Neurobiology Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
432
|
Ma S, Allocca G, Ong-Pålsson EKE, Singleton CE, Hawkes D, McDougall SJ, Williams SJ, Bathgate RAD, Gundlach AL. Nucleus incertus promotes cortical desynchronization and behavioral arousal. Brain Struct Funct 2016; 222:515-537. [PMID: 27206427 DOI: 10.1007/s00429-016-1230-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 04/26/2016] [Indexed: 01/09/2023]
Abstract
Arousal and vigilance are essential for survival and relevant regulatory neural circuits lie within the brainstem, hypothalamus and forebrain. The nucleus incertus (NI) is a distinct site within the pontine periventricular gray, containing a substantial population of GABAergic neurons with long-range, ascending projections. Existing neuroanatomical data and functional studies in anesthetized rats, suggest the NI is a central component of a midline behavioral control network well positioned to modulate arousal, vigilance and exploratory navigation, yet none of these roles have been established experimentally. We used a chemogenetic approach-clozapine-N-oxide (CNO) activation of virally delivered excitatory hM3Dq-DREADDs-to activate the NI in rats and examined the behavioral and physiological effects, relative to effects in naïve rats and appropriate viral-treated controls. hM3Dq activation by CNO resulted in long-lasting depolarization of NI neurons with action potentials, in vitro. Peripheral injection of CNO significantly increased c-Fos immunoreactivity in the NI and promoted cortical electroencephalograph (EEG) desynchronization. These brain changes were associated with heightened arousal, and increased locomotor activity in the homecage and in a novel environment. Furthermore, NI activation altered responses in a fear conditioning paradigm, reflected by increased head-scanning, vigilant behaviors during conditioned fear recall. These findings provide direct evidence that the NI promotes general arousal via a broad behavioral activation circuit and support early hypotheses, based on its connectivity, that the NI is a modulator of cognition and attention, and emotional and motivated behaviors.
Collapse
Affiliation(s)
- Sherie Ma
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia. .,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| | - Giancarlo Allocca
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Emma K E Ong-Pålsson
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Caitlin E Singleton
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - David Hawkes
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, Australia
| | - Stuart J McDougall
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Spencer J Williams
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Ross A D Bathgate
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia. .,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia. .,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
433
|
Todd TP, Mehlman ML, Keene CS, DeAngeli NE, Bucci DJ. Retrosplenial cortex is required for the retrieval of remote memory for auditory cues. ACTA ACUST UNITED AC 2016; 23:278-88. [PMID: 27194795 PMCID: PMC4880149 DOI: 10.1101/lm.041822.116] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/07/2016] [Indexed: 11/25/2022]
Abstract
The restrosplenial cortex (RSC) has a well-established role in contextual and spatial learning and memory, consistent with its known connectivity with visuo-spatial association areas. In contrast, RSC appears to have little involvement with delay fear conditioning to an auditory cue. However, all previous studies have examined the contribution of the RSC to recently acquired auditory fear memories. Since neocortical regions have been implicated in the permanent storage of remote memories, we examined the contribution of the RSC to remotely acquired auditory fear memories. In Experiment 1, retrieval of a remotely acquired auditory fear memory was impaired when permanent lesions (either electrolytic or neurotoxic) were made several weeks after initial conditioning. In Experiment 2, using a chemogenetic approach, we observed impairments in the retrieval of remote memory for an auditory cue when the RSC was temporarily inactivated during testing. In Experiment 3, after injection of a retrograde tracer into the RSC, we observed labeled cells in primary and secondary auditory cortices, as well as the claustrum, indicating that the RSC receives direct projections from auditory regions. Overall our results indicate the RSC has a critical role in the retrieval of remotely acquired auditory fear memories, and we suggest this is related to the quality of the memory, with less precise memories being RSC dependent.
Collapse
Affiliation(s)
- Travis P Todd
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Max L Mehlman
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Christopher S Keene
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Nicole E DeAngeli
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - David J Bucci
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| |
Collapse
|
434
|
Whissell PD, Tohyama S, Martin LJ. The Use of DREADDs to Deconstruct Behavior. Front Genet 2016; 7:70. [PMID: 27242888 PMCID: PMC4868840 DOI: 10.3389/fgene.2016.00070] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/12/2016] [Indexed: 02/03/2023] Open
Abstract
A central goal in understanding brain function is to link specific cell populations to behavioral outputs. In recent years, the selective targeting of specific neural circuits has been made possible with the development of new experimental approaches, including chemogenetics. This technique allows for the control of molecularly defined subsets of cells through engineered G protein-coupled receptors (GPCRs), which have the ability to activate or silence neuronal firing. Through chemogenetics, neural circuits are being linked to behavioral outputs at an unprecedented rate. Further, the coupling of chemogenetics with imaging techniques to monitor neural activity in freely moving animals now makes it possible to deconstruct the complex whole-brain networks that are fundamental to behavioral states. In this review, we highlight a specific chemogenetic application known as DREADDs (designer receptors exclusively activated by designer drugs). DREADDs are used ubiquitously to modulate GPCR activity in vivo and have been widely applied in the basic sciences, particularly in the field of behavioral neuroscience. Here, we focus on the impact and utility of DREADD technology in dissecting the neural circuitry of various behaviors including memory, cognition, reward, feeding, anxiety and pain. By using DREADDs to monitor the electrophysiological, biochemical, and behavioral outputs of specific neuronal types, researchers can better understand the links between brain activity and behavior. Additionally, DREADDs are useful in studying the pathogenesis of disease and may ultimately have therapeutic potential.
Collapse
Affiliation(s)
- Paul D Whissell
- Department of Psychology, University of Toronto Toronto, ON, Canada
| | - Sarasa Tohyama
- Department of Psychology, University of Toronto Mississauga Mississauga, ON, Canada
| | - Loren J Martin
- Department of Psychology, University of Toronto Mississauga Mississauga, ON, Canada
| |
Collapse
|
435
|
Montgomery KL, Iyer SM, Christensen AJ, Deisseroth K, Delp SL. Beyond the brain: Optogenetic control in the spinal cord and peripheral nervous system. Sci Transl Med 2016; 8:337rv5. [DOI: 10.1126/scitranslmed.aad7577] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/18/2016] [Indexed: 12/12/2022]
|
436
|
Breese GR, Knapp DJ. Persistent adaptation by chronic alcohol is facilitated by neuroimmune activation linked to stress and CRF. Alcohol 2016; 52:9-23. [PMID: 27139233 PMCID: PMC4855305 DOI: 10.1016/j.alcohol.2016.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/10/2015] [Accepted: 01/24/2016] [Indexed: 01/11/2023]
Abstract
This review updates the conceptual basis for the association of alcohol abuse with an insidious adaptation that facilitates negative affect during withdrawal from chronic intermittent alcohol (CIA) exposure - a change that later supports sensitization of stress-induced anxiety following alcohol abstinence. The finding that a CRF1-receptor antagonist (CRF1RA) minimized CIA withdrawal-induced negative affect supported an association of alcohol withdrawal with a stress mechanism. The finding that repeated stresses or multiple CRF injections into selected brain sites prior to a single 5-day chronic alcohol (CA) exposure induced anxiety during withdrawal provided critical support for a linkage of CIA withdrawal with stress. The determination that CRF1RA injection into positive CRF-sensitive brain sites prevented CIA withdrawal-induced anxiety provided support that neural path integration maintains the persistent CIA adaptation. Based upon reports that stress increases neuroimmune function, an effort was undertaken to test whether cytokines would support the adaptation induced by stress/CA exposure. Twenty-four hours after withdrawal from CIA, cytokine mRNAs were found to be increased in cortex as well as other sites in brain. Further, repeated cytokine injections into previously identified brain sites substituted for stress and CRF induction of anxiety during CA withdrawal. Discovery that a CRF1RA prevented the brain cytokine mRNA increase induced by CA withdrawal provided critical evidence for CRF involvement in this neuroimmune induction after CA withdrawal. However, the CRF1RA did not block the stress increase in cytokine mRNA increases in controls. The latter data supported the hypothesis that distinct mechanisms linked to stress and CA withdrawal can support common neuroimmune functions within a brain site. As evidence evolves concerning neural involvement in brain neuroimmune function, a better understanding of the progressive adaptation associated with CIA exposure will advance new knowledge that could possibly lead to strategies to combat alcohol abuse.
Collapse
Affiliation(s)
- George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; The UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA.
| | - Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA
| |
Collapse
|
437
|
Lateral Hypothalamus GABAergic Neurons Modulate Consummatory Behaviors Regardless of the Caloric Content or Biological Relevance of the Consumed Stimuli. Neuropsychopharmacology 2016; 41:1505-12. [PMID: 26442599 PMCID: PMC4832010 DOI: 10.1038/npp.2015.304] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/03/2015] [Accepted: 10/01/2015] [Indexed: 02/05/2023]
Abstract
It was recently reported that activation of a subset of lateral hypothalamus (LH) GABAergic neurons induced both appetitive (food-seeking) and consummatory (eating) behaviors in vGat-ires-cre mice, while inhibition or deletion of GABAergic neurons blunted these behaviors. As food and caloric-dense liquid solutions were used, the data reported suggest that these LH GABAergic neurons may modulate behaviors that function to maintain homeostatic caloric balance. Here we report that chemogenetic activation of this GABAergic population in vGat-ires-cre mice increased consummatory behavior directed at any available stimulus, including those entailing calories (food, sucrose, and ethanol), those that do not (saccharin and water), and those lacking biological relevance (wood). Chemogenetic inhibition of these neurons attenuated consummatory behaviors. These data indicate that LH GABAergic neurons modulate consummatory behaviors regardless of the caloric content or biological relevance of the consumed stimuli.
Collapse
|
438
|
Abstract
Optogenetic methodology enables direct targeting of specific neural circuit elements for inhibition or excitation while spanning timescales from the acute (milliseconds) to the chronic (many days or more). Although the impact of this temporal versatility and cellular specificity has been greater for basic science than clinical research, it is natural to ask whether the dynamic patterns of neural circuit activity discovered to be causal in adaptive or maladaptive behaviors could become targets for treatment of neuropsychiatric diseases. Here, we consider the landscape of ideas related to therapeutic targeting of circuit dynamics. Specifically, we highlight optical, ultrasonic, and magnetic concepts for the targeted control of neural activity, preclinical/clinical discovery opportunities, and recently reported optogenetically guided clinical outcomes.
Collapse
Affiliation(s)
| | - Emily Ferenczi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
439
|
Hu J, Stern M, Gimenez LE, Wanka L, Zhu L, Rossi M, Meister J, Inoue A, Beck-Sickinger AG, Gurevich VV, Wess J. A G Protein-biased Designer G Protein-coupled Receptor Useful for Studying the Physiological Relevance of Gq/11-dependent Signaling Pathways. J Biol Chem 2016; 291:7809-7820. [PMID: 26851281 PMCID: PMC4824988 DOI: 10.1074/jbc.m115.702282] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/26/2016] [Indexed: 01/14/2023] Open
Abstract
Designerreceptorsexclusivelyactivated by adesignerdrug (DREADDs) are clozapine-N-oxide-sensitive designer G protein-coupled receptors (GPCRs) that have emerged as powerful novel chemogenetic tools to study the physiological relevance of GPCR signaling pathways in specific cell types or tissues. Like endogenous GPCRs, clozapine-N-oxide-activated DREADDs do not only activate heterotrimeric G proteins but can also trigger β-arrestin-dependent (G protein-independent) signaling. To dissect the relative physiological relevance of G protein-mediatedversusβ-arrestin-mediated signaling in different cell types or physiological processes, the availability of G protein- and β-arrestin-biased DREADDs would be highly desirable. In this study, we report the development of a mutationally modified version of a non-biased DREADD derived from the M3muscarinic receptor that can activate Gq/11with high efficacy but lacks the ability to interact with β-arrestins. We also demonstrate that this novel DREADD is activein vivoand that cell type-selective expression of this new designer receptor can provide novel insights into the physiological roles of G protein (Gq/11)-dependentversusβ-arrestin-dependent signaling in hepatocytes. Thus, this novel Gq/11-biased DREADD represents a powerful new tool to study the physiological relevance of Gq/11-dependent signaling in distinct tissues and cell types, in the absence of β-arrestin-mediated cellular effects. Such studies should guide the development of novel classes of functionally biased ligands that show high efficacy in various pathophysiological conditions but display a reduced incidence of side effects.
Collapse
Affiliation(s)
- Jianxin Hu
- From the Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew Stern
- From the Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Luis E Gimenez
- the Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Lizzy Wanka
- the Institute of Biochemistry, University of Leipzig, Leipzig 04103, Germany
| | - Lu Zhu
- From the Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Mario Rossi
- From the Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Jaroslawna Meister
- From the Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Asuka Inoue
- the Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Miyagi 980-8578, Japan, and the Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology (PRESTO), Kawaguchi, Saitama 332-0012, Japan
| | | | | | - Jürgen Wess
- From the Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892,
| |
Collapse
|
440
|
Abstract
Unraveling the complex network of neural circuits that form the nervous system demands tools that can manipulate specific circuits. The recent evolution of genetic tools to target neural circuits allows an unprecedented precision in elucidating their function. Here we describe two general approaches for achieving circuit specificity. The first uses the genetic identity of a cell, such as a transcription factor unique to a circuit, to drive expression of a molecule that can manipulate cell function. The second uses the spatial connectivity of a circuit to achieve specificity: one genetic element is introduced at the origin of a circuit and the other at its termination. When the two genetic elements combine within a neuron, they can alter its function. These two general approaches can be combined to allow manipulation of neurons with a specific genetic identity by introducing a regulatory gene into the origin or termination of the circuit. We consider the advantages and disadvantages of both these general approaches with regard to specificity and efficacy of the manipulations. We also review the genetic techniques that allow gain- and loss-of-function within specific neural circuits. These approaches introduce light-sensitive channels (optogenetic) or drug sensitive channels (chemogenetic) into neurons that form specific circuits. We compare these tools with others developed for circuit-specific manipulation and describe the advantages of each. Finally, we discuss how these tools might be applied for identification of the neural circuits that mediate behavior and for repair of neural connections.
Collapse
Affiliation(s)
- Hong Geun Park
- Burke Medical Research Institute, White Plains, NY, USA.
| | - Jason B Carmel
- Burke Medical Research Institute, White Plains, NY, USA
- Brain and Mind Research Institute and Departments of Neurology and Pediatrics, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
441
|
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
|
442
|
Nation HL, Nicoleau M, Kinsman BJ, Browning KN, Stocker SD. DREADD-induced activation of subfornical organ neurons stimulates thirst and salt appetite. J Neurophysiol 2016; 115:3123-9. [PMID: 27030736 DOI: 10.1152/jn.00149.2016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/25/2016] [Indexed: 01/19/2023] Open
Abstract
The subfornical organ (SFO) plays a pivotal role in body fluid homeostasis through its ability to integrate neurohumoral signals and subsequently alter behavior, neuroendocrine function, and autonomic outflow. The purpose of the present study was to evaluate whether selective activation of SFO neurons using virally mediated expression of Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) stimulated thirst and salt appetite. Male C57BL/6 mice (12-15 wk) received an injection of rAAV2-CaMKII-HA-hM3D(Gq)-IRES-mCitrine targeted at the SFO. Two weeks later, acute injection of clozapine N-oxide (CNO) produced dose-dependent increases in water intake of mice with DREADD expression in the SFO. CNO also stimulated the ingestion of 0.3 M NaCl. Acute injection of CNO significantly increased the number of Fos-positive nuclei in the SFO of mice with robust DREADD expression. Furthermore, in vivo single-unit recordings demonstrate that CNO significantly increases the discharge frequency of both ANG II- and NaCl-responsive neurons. In vitro current-clamp recordings confirm that bath application of CNO produces a significant membrane depolarization and increase in action potential frequency. In a final set of experiments, chronic administration of CNO approximately doubled 24-h water intake without an effect on salt appetite. These findings demonstrate that DREADD-induced activation of SFO neurons stimulates thirst and that DREADDs are a useful tool to acutely or chronically manipulate neuronal circuits influencing body fluid homeostasis.
Collapse
Affiliation(s)
- Haley L Nation
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania; and
| | - Marvin Nicoleau
- Department of Biochemistry and Molecular Biology, Franklin & Marshall College, Lancaster, Pennsylvania
| | - Brian J Kinsman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania; and
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania; and
| | - Sean D Stocker
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania; and
| |
Collapse
|
443
|
Bolognini D, Tobin AB, Milligan G, Hodge D. The Pharmacology and Function of Receptors for Short-Chain Fatty Acids. Mol Pharmacol 2016; 89:388-98. [PMID: 26719580 DOI: 10.1124/mol.115.102301] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/29/2015] [Indexed: 02/14/2025] Open
Abstract
Despite some blockbuster G protein-coupled receptor (GPCR) drugs, only a small fraction (∼ 15%) of the more than 390 nonodorant GPCRs have been successfully targeted by the pharmaceutical industry. One way that this issue might be addressed is via translation of recent deorphanization programs that have opened the prospect of extending the reach of new medicine design to novel receptor types with potential therapeutic value. Prominent among these receptors are those that respond to short-chain free fatty acids of carbon chain length 2-6. These receptors, FFA2 (GPR43) and FFA3 (GPR41), are each predominantly activated by the short-chain fatty acids acetate, propionate, and butyrate, ligands that originate largely as fermentation by-products of anaerobic bacteria in the gut. However, the presence of FFA2 and FFA3 on pancreatic β-cells, FFA3 on neurons, and FFA2 on leukocytes and adipocytes means that the biologic role of these receptors likely extends beyond the widely accepted role of regulating peptide hormone release from enteroendocrine cells in the gut. Here, we review the physiologic roles of FFA2 and FFA3, the recent development and use of receptor-selective pharmacological tool compounds and genetic models available to study these receptors, and present evidence of the potential therapeutic value of targeting this emerging receptor pair.
Collapse
Affiliation(s)
- Daniele Bolognini
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, Scotland, United Kingdom (D.B., G.M.); and Medical Research Council Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.B.T., C.E.M.)
| | - Andrew B Tobin
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, Scotland, United Kingdom (D.B., G.M.); and Medical Research Council Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.B.T., C.E.M.)
| | - Graeme Milligan
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, Scotland, United Kingdom (D.B., G.M.); and Medical Research Council Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.B.T., C.E.M.)
| | - Daryl Hodge
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, Scotland, United Kingdom (D.B., G.M.); and Medical Research Council Toxicology Unit, University of Leicester, Leicester, United Kingdom (A.B.T., C.E.M.)
| |
Collapse
|
444
|
Chemogenetic Inactivation of Dorsal Anterior Cingulate Cortex Neurons Disrupts Attentional Behavior in Mouse. Neuropsychopharmacology 2016; 41. [PMID: 26224620 PMCID: PMC4748426 DOI: 10.1038/npp.2015.229] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Attention is disrupted commonly in psychiatric disorders, yet mechanistic insight remains limited. Deficits in this function are associated with dorsal anterior cingulate cortex (dACC) excitotoxic lesions and pharmacological disinhibition; however, a causal relationship has not been established at the cellular level. Moreover, this association has not yet been examined in a genetically tractable species such as mice. Here, we reveal that dACC neurons causally contribute to attention processing by combining a chemogenetic approach that reversibly suppresses neural activity with a translational, touchscreen-based attention task in mice. We virally expressed inhibitory hM4Di DREADD (designer receptor exclusively activated by a designer drug) in dACC neurons, and examined the effects of this inhibitory action with the attention-based five-choice serial reaction time task. DREADD inactivation of the dACC neurons during the task significantly increased omission and correct response latencies, indicating that the neuronal activities of dACC contribute to attention and processing speed. Selective inactivation of excitatory neurons in the dACC not only increased omission, but also decreased accuracy. The effect of inactivating dACC neurons was selective to attention as response control, motivation, and locomotion remain normal. This finding suggests that dACC excitatory neurons play a principal role in modulating attention to task-relevant stimuli. This study establishes a foundation to chemogenetically dissect specific cell-type and circuit mechanisms underlying attentional behaviors in a genetically tractable species.
Collapse
|
445
|
Zhao W, Ho L, Wang J, Bi W, Yemul S, Ward L, Freire D, Mazzola P, Brathwaite J, Mezei M, Sanchez R, Elder GA, Pasinetti GM. In Silico Modeling of Novel Drug Ligands for Treatment of Concussion Associated Tauopathy. J Cell Biochem 2016; 117:2241-8. [PMID: 26910498 DOI: 10.1002/jcb.25521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/19/2016] [Indexed: 11/07/2022]
Abstract
The objective of this study was to develop an in silico screening model for characterization of potential novel ligands from commercial drug libraries able to functionally activate certain olfactory receptors (ORs), which are members of the class A rhodopsin-like family of G protein couple receptors (GPCRs), in the brain of murine models of concussion. We previously found that concussions may significantly influence expression of certain ORs, for example, OR4M1 in subjects with a history of concussion/traumatic brain injury (TBI). In this study, we built a 3-D OR4M1 model and used it in in silico screening of potential novel ligands from commercial drug libraries. We report that in vitro activation of OR4M1 with the commercially available ZINC library compound 10915775 led to a significant attenuation of abnormal tau phosphorylation in embryonic cortico-hippocampal neuronal cultures derived from NSE-OR4M1 transgenic mice, possibly through modulation of the JNK signaling pathway. The attenuation of abnormal tau phosphorylation was rather selective since ZINC10915775 significantly decreased tau phosphorylation on tau Ser202/T205 (AT8 epitope) and tau Thr212/Ser214 (AT100 epitope), but not on tau Ser396/404 (PHF-1 epitope). Moreover, no response of ZINC10915775 was found in control hippocampal neuronal cultures derived from wild type littermates. Our in silico model provides novel means to pharmacologically modulate select ubiquitously expressed ORs in the brain through high affinity ligand activation to prevent and eventually to treat concussion induced down regulation of ORs and subsequent cascade of tau pathology. J. Cell. Biochem. 117: 2241-2248, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York.,Geriatric Research Education Clinical Center at James J. Peters VA Medical Center, Bronx, New York
| | - Lap Ho
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York
| | - Jun Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York.,Geriatric Research Education Clinical Center at James J. Peters VA Medical Center, Bronx, New York
| | - Weina Bi
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York
| | - Shrishailam Yemul
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York
| | - Libby Ward
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York
| | - Daniel Freire
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York
| | - Paolo Mazzola
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York.,School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Justin Brathwaite
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York
| | - Mihaly Mezei
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York.,Experimental Therapeutics Institute, Icahn School of Medicine at Mount Sinai, New York
| | - Roberto Sanchez
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York.,Experimental Therapeutics Institute, Icahn School of Medicine at Mount Sinai, New York
| | - Gregory A Elder
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York.,Geriatric Research Education Clinical Center at James J. Peters VA Medical Center, Bronx, New York.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
446
|
In vivo imaging identifies temporal signature of D1 and D2 medium spiny neurons in cocaine reward. Proc Natl Acad Sci U S A 2016; 113:2726-31. [PMID: 26831103 DOI: 10.1073/pnas.1521238113] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The reinforcing and rewarding properties of cocaine are attributed to its ability to increase dopaminergic transmission in nucleus accumbens (NAc). This action reinforces drug taking and seeking and leads to potent and long-lasting associations between the rewarding effects of the drug and the cues associated with its availability. The inability to extinguish these associations is a key factor contributing to relapse. Dopamine produces these effects by controlling the activity of two subpopulations of NAc medium spiny neurons (MSNs) that are defined by their predominant expression of either dopamine D1 or D2 receptors. Previous work has demonstrated that optogenetically stimulating D1 MSNs promotes reward, whereas stimulating D2 MSNs produces aversion. However, we still lack a clear understanding of how the endogenous activity of these cell types is affected by cocaine and encodes information that drives drug-associated behaviors. Using fiber photometry calcium imaging we define D1 MSNs as the specific population of cells in NAc that encodes information about drug associations and elucidate the temporal profile with which D1 activity is increased to drive drug seeking in response to contextual cues. Chronic cocaine exposure dysregulates these D1 signals to both prevent extinction and facilitate reinstatement of drug seeking to drive relapse. Directly manipulating these D1 signals using designer receptors exclusively activated by designer drugs prevents contextual associations. Together, these data elucidate the responses of D1- and D2-type MSNs in NAc to acute cocaine and during the formation of context-reward associations and define how prior cocaine exposure selectively dysregulates D1 signaling to drive relapse.
Collapse
|
447
|
New Technologies for Elucidating Opioid Receptor Function. Trends Pharmacol Sci 2016; 37:279-289. [PMID: 26833118 DOI: 10.1016/j.tips.2016.01.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/22/2015] [Accepted: 01/04/2016] [Indexed: 01/25/2023]
Abstract
Recent advances in technology, including high resolution crystal structures of opioid receptors, novel chemical tools, and new genetic approaches have provided an unparalleled palette of tools for deconstructing opioid receptor actions in vitro and in vivo. Here we provide a brief description of our understanding of opioid receptor function from both molecular and atomic perspectives, as well as their role in neural circuits in vivo. We then show how insights into the molecular details of opioid actions can facilitate the creation of functionally selective (biased) and photoswitchable opioid ligands. Finally, we describe how newly engineered opioid receptor-based chemogenetic and optogenetic tools, and new mouse lines, are expanding and transforming our understanding of opioid function and, perhaps, paving the way for new therapeutics.
Collapse
|
448
|
Butcher AJ, Bradley SJ, Prihandoko R, Brooke SM, Mogg A, Bourgognon JM, Macedo-Hatch T, Edwards JM, Bottrill AR, Challiss RAJ, Broad LM, Felder CC, Tobin AB. An Antibody Biosensor Establishes the Activation of the M1 Muscarinic Acetylcholine Receptor during Learning and Memory. J Biol Chem 2016; 291:8862-75. [PMID: 26826123 PMCID: PMC4861454 DOI: 10.1074/jbc.m115.681726] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Indexed: 11/15/2022] Open
Abstract
Establishing the in vivo activation status of G protein-coupled receptors would not only indicate physiological roles of G protein-coupled receptors but would also aid drug discovery by establishing drug/receptor engagement. Here, we develop a phospho-specific antibody-based biosensor to detect activation of the M1 muscarinic acetylcholine receptor (M1 mAChR) in vitro and in vivo. Mass spectrometry phosphoproteomics identified 14 sites of phosphorylation on the M1 mAChR. Phospho-specific antibodies to four of these sites established that serine at position 228 (Ser228) on the M1 mAChR showed extremely low levels of basal phosphorylation that were significantly up-regulated by orthosteric agonist stimulation. In addition, the M1 mAChR-positive allosteric modulator, 1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid, enhanced acetylcholine-mediated phosphorylation at Ser228. These data supported the hypothesis that phosphorylation at Ser228 was an indicator of M1 mAChR activation. This was further supported in vivo by the identification of phosphorylated Ser228 on the M1 mAChR in the hippocampus of mice following administration of the muscarinic ligands xanomeline and 1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid. Finally, Ser228 phosphorylation was seen to increase in the CA1 region of the hippocampus following memory acquisition, a response that correlated closely with up-regulation of CA1 neuronal activity. Thus, determining the phosphorylation status of the M1 mAChR at Ser228 not only provides a means of establishing receptor activation following drug treatment both in vitro and in vivo but also allows for the mapping of the activation status of the M1 mAChR in the hippocampus following memory acquisition thereby establishing a link between M1 mAChR activation and hippocampus-based memory and learning.
Collapse
Affiliation(s)
| | | | | | | | - Adrian Mogg
- Eli Lilly and Co. Neuroscience, Erl Wood Manor, Windlesham, Surrey GU20 6PH, United Kingdom
| | | | | | | | - Andrew R Bottrill
- Protein and Nucleic Acid Chemistry Laboratory, University of Leicester, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - R A John Challiss
- the Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Lisa M Broad
- Eli Lilly and Co. Neuroscience, Erl Wood Manor, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Christian C Felder
- Eli Lilly and Co. Neuroscience, Lilly Corporate Center, Indianapolis, Indiana 46285, and
| | | |
Collapse
|
449
|
Mejias-Aponte CA. Specificity and impact of adrenergic projections to the midbrain dopamine system. Brain Res 2016; 1641:258-73. [PMID: 26820641 DOI: 10.1016/j.brainres.2016.01.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 01/11/2016] [Accepted: 01/20/2016] [Indexed: 12/18/2022]
Abstract
Dopamine (DA) is a neuromodulator that regulates different brain circuits involved in cognitive functions, motor coordination, and emotions. Dysregulation of DA is associated with many neurological and psychiatric disorders such as Parkinson's disease and substance abuse. Several lines of research have shown that the midbrain DA system is regulated by the central adrenergic system. This review focuses on adrenergic interactions with midbrain DA neurons. It discusses the current neuroanatomy including source of adrenergic innervation, type of synapses, and adrenoceptors expression. It also discusses adrenergic regulation of DA cell activity and neurotransmitter release. Finally, it reviews several neurological and psychiatric disorders where changes in adrenergic system are associated with dysregulation of the midbrain DA system. This article is part of a Special Issue entitled SI: Noradrenergic System.
Collapse
Affiliation(s)
- Carlos A Mejias-Aponte
- National Institute on Drug Abuse Histology Core, Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Biomedical Research Center, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA.
| |
Collapse
|
450
|
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
|