301
|
Abstract
Pharmacology, the chemical control of physiology, emerged as an offshoot of physiology when the physiologists using chemicals to probe physiological systems became more interested in the probes than the systems. Pharmacologists were always, and in many ways still are, bound to study drugs in systems they do not fully understand. Under these circumstances, null methods were the main ways in which conclusions about biologically active molecules were made. However, as understanding of the basic mechanisms of cellular function and biochemical systems were elucidated, so too did the understanding of how drugs affected these systems. Over the past 20 years, new ideas have emerged in the field that have completely changed and revitalized it; these are described herein. It will be seen how null methods in isolated tissues gave way to, first biochemical radioligand binding studies, and then to a wide array of functional assay technologies that can measure the effects of molecules on drug targets. In addition, the introduction of molecular dynamics, the appreciation of the allosteric nature of receptors, protein X-ray crystal structures, genetic manipulations in the form of knock-out and knock-in systems and Designer Receptors Exclusively Activated by Designer Drugs have revolutionized pharmacology.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
302
|
Hua R, Wang X, Chen X, Wang X, Huang P, Li P, Mei W, Li H. Calretinin Neurons in the Midline Thalamus Modulate Starvation-Induced Arousal. Curr Biol 2018; 28:3948-3959.e4. [PMID: 30528578 DOI: 10.1016/j.cub.2018.11.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/29/2018] [Accepted: 11/06/2018] [Indexed: 11/17/2022]
Abstract
Orchestration of sleep and feeding behavior is essential for organismal health and survival. Although sleep deprivation promotes feeding and starvation suppresses sleep, the underlying neural mechanisms remain largely unknown. Here, we showed that starvation in mice potently promoted arousal and activated calretinin neurons (CR+) in the paraventricular thalamus (PVT). Direct activation of PVTCR+ neurons promoted arousal, and their activity was necessary for starvation-induced sleep suppression. Specifically, the PVTCR+-bed nucleus of the stria terminalis (BNST) circuit rapidly initiated arousal. Selective inhibition of BNST-projecting PVT neurons opposed arousal during starvation. Taken together, our results define a cell-type-specific neural circuitry modulating starvation-induced arousal and coordinating the conflict between sleeping and feeding.
Collapse
Affiliation(s)
- Ruifang Hua
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; Henan Key Laboratory of Immunology and Targeted Therapy, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Xu Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xinfeng Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xinxin Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Pengcheng Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Pengcheng Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wei Mei
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Haohong Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.
| |
Collapse
|
303
|
Ketamine and selective activation of parvalbumin interneurons inhibit stress-induced dendritic spine elimination. Transl Psychiatry 2018; 8:272. [PMID: 30531859 PMCID: PMC6288154 DOI: 10.1038/s41398-018-0321-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022] Open
Abstract
Stress is a major risk factor for the onset of many psychiatric diseases. In rodent models, chronic stress induces depression and impairs excitatory neurotransmission. However, little is known about the effect of stress on synaptic circuitry during the development of behavioral symptoms. Using two-photon transcranial imaging, we studied the effect of repeated restraint stress on dendritic spine plasticity in the frontal cortex in vivo. We found that restraint stress induced dendritic spine loss by decreasing the rate of spine formation and increasing the rate of spine elimination. The N-methyl-D-aspartate receptor antagonist ketamine inhibited stress-induced spine loss mainly by protecting mushroom spines from elimination. Ketamine also induced re-formation of spines in close proximity to previously stress-eliminated spines. Electrophysiological and in vivo imaging experiments showed that ketamine enhanced activity of parvalbumin (PV) interneurons under stress and counterbalanced the stress-induced net loss of PV axonal boutons. In addition, selective chemogenetic excitation of PV interneurons mimicked the protective effects of ketamine on dendritic spines against stress. Collectively, our data provide new insights on the effects of ketamine on synaptic circuitry under stress and a possible mechanism to counteract stress-induced synaptic impairments through PV interneuron activation.
Collapse
|
304
|
Assaf F, Schiller Y. A chemogenetic approach for treating experimental Parkinson's disease. Mov Disord 2018; 34:469-479. [DOI: 10.1002/mds.27554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 09/28/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Fadi Assaf
- The Rappaport Faculty of MedicineTechnion–Israel Institute of Technology Haifa Israel
| | - Yitzhak Schiller
- The Rappaport Faculty of MedicineTechnion–Israel Institute of Technology Haifa Israel
- Department of NeurologyRambam Medical Center Haifa Israel
| |
Collapse
|
305
|
Input-Specific Synaptic Location and Function of the α5 GABA A Receptor Subunit in the Mouse CA1 Hippocampal Neurons. J Neurosci 2018; 39:788-801. [PMID: 30523065 DOI: 10.1523/jneurosci.0567-18.2018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 10/01/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022] Open
Abstract
Hippocampus-dependent learning processes are coordinated via a large diversity of GABAergic inhibitory mechanisms. The α5 subunit-containing GABAA receptor (α5-GABAAR) is abundantly expressed in the hippocampus populating primarily the extrasynaptic domain of CA1 pyramidal cells, where it mediates tonic inhibitory conductance and may cause functional deficits in synaptic plasticity and hippocampus-dependent memory. However, little is known about synaptic expression of the α5-GABAAR and, accordingly, its location site-specific function. We examined the cell- and synapse-specific distribution of the α5-GABAAR in the CA1 stratum oriens/alveus (O/A) using a combination of immunohistochemistry, whole-cell patch-clamp recordings and optogenetic stimulation in hippocampal slices obtained from mice of either sex. In addition, the input-specific role of the α5-GABAAR in spatial learning and anxiety-related behavior was studied using behavioral testing and chemogenetic manipulations. We demonstrate that α5-GABAAR is preferentially targeted to the inhibitory synapses made by the vasoactive intestinal peptide (VIP)- and calretinin-positive terminals onto dendrites of somatostatin-expressing interneurons. In contrast, synapses made by the parvalbumin-positive inhibitory inputs to O/A interneurons showed no or little α5-GABAAR. Inhibiting the α5-GABAAR in control mice in vivo improved spatial learning but also induced anxiety-like behavior. Inhibiting the α5-GABAAR in mice with inactivated CA1 VIP input could still improve spatial learning and was not associated with anxiety. Together, these data indicate that the α5-GABAAR-mediated phasic inhibition via VIP input to interneurons plays a predominant role in the regulation of anxiety while the α5-GABAAR tonic inhibition via this subunit may control spatial learning.SIGNIFICANCE STATEMENT The α5-GABAAR subunit exhibits high expression in the hippocampus, and regulates the induction of synaptic plasticity and the hippocampus-dependent mnemonic processes. In CA1 principal cells, this subunit occupies mostly extrasynaptic sites and mediates tonic inhibition. Here, we provide evidence that, in CA1 somatostatin-expressing interneurons, the α5-GABAAR subunit is targeted to synapses formed by the VIP- and calretinin-expressing inputs, and plays a specific role in the regulation of anxiety-like behavior.
Collapse
|
306
|
A circuit from hippocampal CA2 to lateral septum disinhibits social aggression. Nature 2018; 564:213-218. [PMID: 30518859 PMCID: PMC6364572 DOI: 10.1038/s41586-018-0772-0] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/12/2018] [Indexed: 01/06/2023]
Abstract
Although the hippocampus is known to be important for declarative memory, how hippocampal output regulates motivated behaviors, such as social aggression, is less well understood. Here we report that hippocampal CA2 pyramidal neurons, which are important for social memory, promote social aggression. This action depends on CA2 output to the lateral septum that is selectively enhanced immediately prior to attack. Activation of lateral septum by CA2 recruits a circuit that disinhibits a subnucleus of the ventro-medial hypothalamus known to trigger attack. The social hormone arginine-vasopressin enhances social aggression by acting on arginine-vasopressin 1b receptors on CA2 presynaptic terminals in lateral septum to facilitate excitatory synaptic transmission. In this manner, release of vasopressin in lateral septum, driven by an animal’s internal state, may serve as a modulatory control that determines whether CA2 activity leads to declarative memory of a social encounter or proceeds to promote motivated social aggression.
Collapse
|
307
|
Robinson E. Psychopharmacology: From serendipitous discoveries to rationale design, but what next? Brain Neurosci Adv 2018; 2:2398212818812629. [PMID: 32166162 PMCID: PMC7058199 DOI: 10.1177/2398212818812629] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Indexed: 12/15/2022] Open
Abstract
Psychopharmacology really developed as a discipline from the mid-20th century with the discovery of a number of new classes of psychoactive drugs which could modify behaviour. These drugs were discovered as a consequence of clinical observations of patients, often being treated for other conditions. These serendipitous discoveries were the start of an era of drug development which has led to the antidepressants, antipsychotics, anxiolytics and mood stabilisers used today. Subsequent research focused on understanding why these drugs were effective, and used this information to develop a second generation of drugs that were more selective for their therapeutic targets, and therefore had reduced side effects and improved safety and tolerability. After a period of decline in new discoveries and withdrawal of the majority of the major pharmaceutical companies from active development programmes in psychiatry, new avenues are emerging fuelling renewed interest in this area.
Collapse
Affiliation(s)
- Emma Robinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
308
|
Chemogenetic inactivation of the dorsal hippocampus and medial prefrontal cortex, individually and concurrently, impairs object recognition and spatial memory consolidation in female mice. Neurobiol Learn Mem 2018; 156:103-116. [PMID: 30408525 PMCID: PMC7310386 DOI: 10.1016/j.nlm.2018.11.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/25/2018] [Accepted: 11/03/2018] [Indexed: 01/23/2023]
Abstract
The dorsal hippocampus (DH) and medial prefrontal cortex (mPFC) are brain regions essential for processing and storing episodic memory. In rodents, the DH has a well-established role in supporting the consolidation of episodic-like memory in tasks such as object recognition and object placement. However, the role of the mPFC in the consolidation of episodic-like memory tasks remains controversial. Therefore, the present study examined involvement of the DH and mPFC, alone and in combination, in object and spatial recognition memory consolidation in ovariectomized female mice. To this end, we utilized two types of inhibitory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to inactivate the DH alone, the mPFC alone, or both brain regions concurrently immediately after object training to assess the role of each region in the consolidation of object recognition and spatial memories. Our results using single and multiplexed DREADDS suggest that excitatory activity in the DH and mPFC, alone or in combination, is required for the successful consolidation of object recognition and spatial memories. Together, these studies provide critical insight into how the DH and mPFC work in concert to facilitate memory consolidation in female mice.
Collapse
|
309
|
Maharjan DM, Dai YY, Glantz EH, Jadhav SP. Disruption of dorsal hippocampal - prefrontal interactions using chemogenetic inactivation impairs spatial learning. Neurobiol Learn Mem 2018; 155:351-360. [PMID: 30179661 PMCID: PMC6365178 DOI: 10.1016/j.nlm.2018.08.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 01/29/2023]
Abstract
The hippocampus (HPC) and prefrontal cortex (PFC) are both necessary for learning and memory-guided behavior. Multiple direct and indirect anatomical projections connect the two regions, and HPC - PFC functional interactions are mediated by diverse physiological network patterns, thought to sub serve various memory processes. Disconnection experiments using contralateral inactivation approaches have established the role of direct, ipsilateral projections from ventral and intermediate HPC (vHPC and iHPC) to PFC in spatial memory. However, numerous studies have also prominently implicated physiological interactions between dorsal HPC (dHPC) and PFC regions in spatial memory tasks, and recent reports have identified direct dHPC - PFC connections. Whether dHPC - PFC interactions are necessary for spatial learning and memory has yet to be tested. Here, we used a chemogenetic inactivation approach using virally-expressed DREADDs (designer receptors exclusively activated by designer drugs) in rats to investigate the role of dHPC - PFC interactions in learning a hippocampal - dependent spatial alternation task. We implemented a rapid learning paradigm for a continuous W-track spatial alternation task comprising two components: an outbound, working memory component, and an inbound, spatial reference memory component. We investigated the effect of contralateral inactivation of dHPC and PFC on learning this task as compared with naïve and vehicle injection controls, as well as ipsilateral inactivation of the same regions. Contralateral dHPC - PFC inactivation selectively led to a significant impairment in learning the spatial working memory task compared to control groups, but did not impair learning of the spatial reference memory task. Ipsilateral inactivation animals showed similar learning rates as animals in the control groups. In a separate experiment, we confirmed that bilateral inactivation of PFC also leads to an impairment in learning the spatial working memory task. Our results thus demonstrate that dHPC - PFC interactions are necessary for spatial alternation learning in novel tasks. In addition, they provide crucial evidence to support the view that physiological interactions between dHPC and PFC play a key role in spatial learning and memory.
Collapse
Affiliation(s)
- Dennis M Maharjan
- Department of Psychology, Neuroscience Program, and Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Yu Y Dai
- Undergraduate Program in Neuroscience, Brandeis University, Waltham, MA 02453, USA
| | - Ethan H Glantz
- Undergraduate Program in Neuroscience, Brandeis University, Waltham, MA 02453, USA
| | - Shantanu P Jadhav
- Department of Psychology, Neuroscience Program, and Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA.
| |
Collapse
|
310
|
Dystonia: Are animal models relevant in therapeutics? Rev Neurol (Paris) 2018; 174:608-614. [DOI: 10.1016/j.neurol.2018.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023]
|
311
|
Wang L, Chen SR, Ma H, Chen H, Hittelman WN, Pan HL. Regulating nociceptive transmission by VGluT2-expressing spinal dorsal horn neurons. J Neurochem 2018; 147:526-540. [PMID: 30203849 DOI: 10.1111/jnc.14588] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 11/28/2022]
Abstract
Vesicular glutamate transporter-2 (VGluT2) mediates the uptake of glutamate into synaptic vesicles in neurons. Spinal cord dorsal horn interneurons are highly heterogeneous and molecularly diverse. The functional significance of VGluT2-expressing dorsal horn neurons in physiological and pathological pain conditions has not been explicitly demonstrated. Designer receptors exclusively activated by designer drugs (DREADDs) are a powerful chemogenetic tool to reversibly control neuronal excitability and behavior. Here, we used transgenic mice with Cre recombinase expression driven by the VGluT2 promoter, combined with the chemogenetic approach, to determine the contribution of VGluT2-expressing dorsal horn neurons to nociceptive regulation. Adeno-associated viral vectors expressing double-floxed Cre-dependent Gαq-coupled human M3 muscarinic receptor DREADD (hM3D)-mCherry or Gαi-coupled κ-opioid receptor DREADD (KORD)-IRES-mCitrine were microinjected into the superficial spinal dorsal horn of VGluT2-Cre mice. Immunofluorescence labeling showed that VGluT2 was predominantly expressed in lamina II excitatory interneurons. Activation of excitatory hM3D in VGluT2-expressing neurons with clozapine N-oxide caused a profound increase in neuronal firing and synaptic glutamate release. Conversely, activation of inhibitory KORD in VGluT2-expressing neurons with salvinorin B markedly inhibited neuronal activity and synaptic glutamate release. In addition, chemogenetic stimulation of VGluT2-expressing neurons increased mechanical and thermal sensitivities in naive mice, whereas chemogenetic silencing of VGluT2-expressing neurons reversed pain hypersensitivity induced by tissue inflammation and peripheral nerve injury. These findings indicate that VGluT2-expressing excitatory neurons play a crucial role in mediating nociceptive transmission in the spinal dorsal horn. Targeting glutamatergic dorsal horn neurons with inhibitory DREADDs may be a new strategy for treating inflammatory pain and neuropathic pain.
Collapse
Affiliation(s)
- Li Wang
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huijie Ma
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Walter N Hittelman
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
312
|
Sleep and Wakefulness Are Controlled by Ventral Medial Midbrain/Pons GABAergic Neurons in Mice. J Neurosci 2018; 38:10080-10092. [PMID: 30282729 DOI: 10.1523/jneurosci.0598-18.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 01/10/2023] Open
Abstract
Sleep-wake behavior is controlled by a wide range of neuronal populations in the mammalian brain. Although the ventral midbrain/pons (VMP) area is suggested to participate in sleep-wake regulation, the neuronal mechanisms have remained unclear. Here, we found that nonspecific cell ablation or selective ablation of GABAergic neurons by expressing diphtheria toxin fragment A in the VMP in male mice induced a large increase in wakefulness that lasted at least 4 weeks. In contrast, selective ablation of dopaminergic neurons in the VMP had little effect on wakefulness. Chemogenetic inhibition of VMP GABAergic neurons also markedly increased wakefulness. The wake-promoting effect of the VMP GABAergic neuron ablation or inhibition was attenuated to varying degrees by the administration of dopamine D1 or D2/3 receptor antagonists and abolished by the administration of both antagonists together. In contrast, chemogenetic activation of VMP GABAergic neurons very strongly increased slow-wave sleep and reduced wakefulness. These findings suggest that VMP GABAergic neurons regulate dopaminergic actions in the sleep-wake behavior of mice.SIGNIFICANCE STATEMENT Current understanding of the neuronal mechanisms and populations that regulate sleep-wake behavior is incomplete. Here, we identified a GABAergic ventral midbrain/pons area that is necessary for controlling the daily amount of sleep and wakefulness in mice. We also found that these inhibitory neurons control wakefulness by suppressing dopaminergic systems. Surprisingly, activation of these neurons strongly induced slow-wave sleep while suppressing wakefulness. Our study reveals a new brain mechanism critical for sleep-wake regulation.
Collapse
|
313
|
Restoring wild-type-like CA1 network dynamics and behavior during adulthood in a mouse model of schizophrenia. Nat Neurosci 2018; 21:1412-1420. [PMID: 30224804 DOI: 10.1038/s41593-018-0225-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/12/2018] [Indexed: 01/16/2023]
Abstract
Schizophrenia is a severely debilitating neurodevelopmental disorder. Establishing a causal link between circuit dysfunction and particular behavioral traits that are relevant to schizophrenia is crucial to shed new light on the mechanisms underlying the pathology. We studied an animal model of the human 22q11 deletion syndrome, the mutation that represents the highest genetic risk of developing schizophrenia. We observed a desynchronization of hippocampal neuronal assemblies that resulted from parvalbumin interneuron hypoexcitability. Rescuing parvalbumin interneuron excitability with pharmacological or chemogenetic approaches was sufficient to restore wild-type-like CA1 network dynamics and hippocampal-dependent behavior during adulthood. In conclusion, our data provide insights into the network dysfunction underlying schizophrenia and highlight the use of reverse engineering to restore physiological and behavioral phenotypes in an animal model of neurodevelopmental disorder.
Collapse
|
314
|
Alvarsson A, Stanley SA. Remote control of glucose-sensing neurons to analyze glucose metabolism. Am J Physiol Endocrinol Metab 2018; 315:E327-E339. [PMID: 29812985 PMCID: PMC6171010 DOI: 10.1152/ajpendo.00469.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/25/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
The central nervous system relies on a continual supply of glucose, and must be able to detect glucose levels and regulate peripheral organ functions to ensure that its energy requirements are met. Specialized glucose-sensing neurons, first described half a century ago, use glucose as a signal and modulate their firing rates as glucose levels change. Glucose-excited neurons are activated by increasing glucose concentrations, while glucose-inhibited neurons increase their firing rate as glucose concentrations fall and decrease their firing rate as glucose concentrations rise. Glucose-sensing neurons are present in multiple brain regions and are highly expressed in hypothalamic regions, where they are involved in functions related to glucose homeostasis. However, the roles of glucose-sensing neurons in healthy and disease states remain poorly understood. Technologies that can rapidly and reversibly activate or inhibit defined neural populations provide invaluable tools to investigate how specific neural populations regulate metabolism and other physiological roles. Optogenetics has high temporal and spatial resolutions, requires implants for neural stimulation, and is suitable for modulating local neural populations. Chemogenetics, which requires injection of a synthetic ligand, can target both local and widespread populations. Radio- and magnetogenetics offer rapid neural activation in localized or widespread neural populations without the need for implants or injections. These tools will allow us to better understand glucose-sensing neurons and their metabolism-regulating circuits.
Collapse
Affiliation(s)
- Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sarah A Stanley
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| |
Collapse
|
315
|
Thompson KJ, Khajehali E, Bradley SJ, Navarrete JS, Huang XP, Slocum S, Jin J, Liu J, Xiong Y, Olsen RHJ, Diberto JF, Boyt KM, Pina MM, Pati D, Molloy C, Bundgaard C, Sexton PM, Kash TL, Krashes MJ, Christopoulos A, Roth BL, Tobin AB. DREADD Agonist 21 Is an Effective Agonist for Muscarinic-Based DREADDs in Vitro and in Vivo. ACS Pharmacol Transl Sci 2018; 1:61-72. [PMID: 30868140 PMCID: PMC6407913 DOI: 10.1021/acsptsci.8b00012] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Indexed: 02/07/2023]
Abstract
![]()
Chemogenetic tools such as designer
receptors exclusively activated
by designer drugs (DREADDs) are routinely used to modulate neuronal
and non-neuronal signaling and activity in a relatively noninvasive
manner. The first generation of DREADDs were templated from the human
muscarinic acetylcholine receptor family and are relatively insensitive
to the endogenous agonist acetylcholine but instead are activated
by clozapine-N-oxide (CNO). Despite the undisputed
success of CNO as an activator of muscarinic DREADDs, it has been
known for some time that CNO is subject to a low rate of metabolic
conversion to clozapine, raising the need for alternative chemical
actuators of muscarinic-based DREADDs. Here we show that DREADD agonist 21 (C21) (11-(1-piperazinyl)-5H-dibenzo[b,e][1,4]diazepine)
is a potent and selective agonist at both excitatory (hM3Dq) and inhibitory
(hM4Di) DREADDs and has excellent bioavailability, pharmacokinetic
properties, and brain penetrability. We also show that C21-induced
activation of hM3Dq and hM4Di in vivo can modulate
bidirectional feeding in defined circuits in mice. These results indicate
that C21 represents an alternative to CNO for in vivo studies where metabolic conversion of CNO to clozapine is a concern.
Collapse
Affiliation(s)
- Karen J Thompson
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| | - Elham Khajehali
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sophie J Bradley
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| | - Jovana S Navarrete
- Diabetes, Endocrinology, and Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States.,National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Xi Ping Huang
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Samuel Slocum
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Jian Jin
- Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029, United States
| | - Jing Liu
- Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029, United States
| | - Yan Xiong
- Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029, United States
| | - Reid H J Olsen
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Jeffrey F Diberto
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Kristen M Boyt
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Melanie M Pina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Dipanwita Pati
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Colin Molloy
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| | - Christoffer Bundgaard
- Neuroscience, Eli Lilly & Co., Erl Wood Manor, Windlesham, Surrey GU20 6PH, United Kingdom
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas L Kash
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States.,National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC2751, United States
| | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, United Kingdom
| |
Collapse
|
316
|
van der Peet PL, Gunawan C, Abdul-Ridha A, Ma S, Scott DJ, Gundlach AL, Bathgate RAD, White JM, Williams SJ. Gram scale preparation of clozapine N-oxide (CNO), a synthetic small molecule actuator for muscarinic acetylcholine DREADDs. MethodsX 2018; 5:257-267. [PMID: 30038895 PMCID: PMC6053635 DOI: 10.1016/j.mex.2018.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/14/2018] [Indexed: 11/30/2022] Open
Abstract
Chemogenetics uses engineered proteins that are controlled by small molecule actuators, allowing in vivo functional studies of proteins with temporal and dose control, and include Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). One major class of DREADDs are mutated muscarinic receptors that are unresponsive to acetylcholine, and are activated by administration of clozapine N-oxide (CNO). However, CNO is available in only small amounts and large scale studies involving animals and multiple cohorts are prohibitively expensive for many investigators. The precursor, clozapine, is also expensive when purchased from specialist suppliers. Here we report: A simple extraction method of clozapine from commercial tablets; A simple preparation of CNO from clozapine, and for the first time its single-crystal X-ray structure; and That the CNO prepared by this method specifically activates the DREADD receptor hM3Dq in vivo.
This method provides large quantities of CNO suitable for large-scale DREADD applications that is identical to commercial material.
Collapse
Affiliation(s)
- Phillip L van der Peet
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Christian Gunawan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Alaa Abdul-Ridha
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
| | - Sherie Ma
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Victoria 3010 Australia
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Ross A D Bathgate
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Victoria 3010 Australia
| | - Jonathan M White
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Spencer J Williams
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
317
|
Nuno-Perez A, Tchenio A, Mameli M, Lecca S. Lateral Habenula Gone Awry in Depression: Bridging Cellular Adaptations With Therapeutics. Front Neurosci 2018; 12:485. [PMID: 30083090 PMCID: PMC6064733 DOI: 10.3389/fnins.2018.00485] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022] Open
Abstract
Depression is a highly heterogeneous disease characterized by symptoms spanning from anhedonia and behavioral despair to social withdrawal and learning deficit. Such diversity of behavioral phenotypes suggests that discrete neural circuits may underlie precise aspects of the disease, rendering its treatment an unmet challenge for modern neuroscience. Evidence from humans and animal models indicate that the lateral habenula (LHb), an epithalamic center devoted to processing aversive stimuli, is aberrantly affected during depression. This raises the hypothesis that rescuing maladaptations within this nucleus may be a potential way to, at least partially, treat aspects of mood disorders. In this review article, we will discuss pre-clinical and clinical evidence highlighting the role of LHb and its cellular adaptations in depression. We will then describe interventional approaches aiming to rescue LHb dysfunction and ultimately ameliorate depressive symptoms. Altogether, we aim to merge the mechanistic-, circuit-, and behavioral-level knowledge obtained about LHb maladaptations in depression to build a general framework that might prove valuable for potential therapeutic interventions.
Collapse
Affiliation(s)
- Alvaro Nuno-Perez
- Department of Fundamental Neuroscience, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Anna Tchenio
- Department of Fundamental Neuroscience, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Manuel Mameli
- Department of Fundamental Neuroscience, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,INSERM, UMR-S 839, Paris, France
| | - Salvatore Lecca
- Department of Fundamental Neuroscience, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
318
|
Considerations for the use of virally delivered genetic tools for in-vivo circuit analysis and behavior in mutant mice: a practical guide to optogenetics. Behav Pharmacol 2018; 28:598-609. [PMID: 29099403 DOI: 10.1097/fbp.0000000000000361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Optogenetics was the method of the year in 2010 according to Nature Neuroscience. Since then, this method has become widespread, the use of virally delivered genetic tools has extended to other fields such as pharmacogenetics, and optogenetic techniques have become frequently applied in genetically manipulated animals for in-vivo circuit analysis and behavioral studies. However, several issues should be taken into consideration when planning such experiments. We aimed to summarize the critical points concerning optogenetic manipulation of a specific brain area in mutant mice. First, the appropriate vector should be chosen to allow optimal optogenetic manipulation. Adeno-associated viral vectors are the most common carriers with different available serotypes. Light-sensitive channels are available in many forms, and the expression of the delivered genetic material can be influenced in many ways. Second, selecting the adequate stimulation protocol is also essential. The pattern, intensity, and timing could be determinative parameters. Third, the mutant strain might have a phenotype that influences the observed behavior. In conclusion, detailed preliminary experiments and numerous control groups are required to choose the best vector and stimulation protocol and to ensure that the mutant animals do not have a specific phenotype that can influence the examined behavior.
Collapse
|
319
|
Cell reprogramming approaches in gene- and cell-based therapies for Parkinson's disease. J Control Release 2018; 286:114-124. [PMID: 30026082 DOI: 10.1016/j.jconrel.2018.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
Degeneration of dopamine (DA) neurons in the substantia nigra pars compacta is the pathological hallmark of Parkinson's disease (PD). In PD multiple pathogenic mechanisms initiate and drive this neurodegenerative process, making the development of effective treatments challenging. To date, PD patients are primarily treated with dopaminergic drugs able to temporarily enhance DA levels, therefore relieving motor symptoms. However, the drawbacks of these therapies including the inability to alter disease progression are constantly supporting the search for alternative treatment approaches. Over the past years efforts have been put into the development of new therapeutic strategies based on the delivery of therapeutic genes using viral vectors or transplantation of DA neurons for cell-based DA replacement. Here, past achievements and recent advances in gene- and cell-based therapies for PD are outlined. We discuss how current gene and cell therapy strategies hold great promise for the treatment of PD and how the use of stem cells and recent developments in cellular reprogramming could contribute to open a new avenue in PD therapy.
Collapse
|
320
|
Denaxa M, Neves G, Burrone J, Pachnis V. Homeostatic Regulation of Interneuron Apoptosis During Cortical Development. J Exp Neurosci 2018; 12:1179069518784277. [PMID: 30013387 PMCID: PMC6043931 DOI: 10.1177/1179069518784277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/29/2018] [Indexed: 12/31/2022] Open
Abstract
The mammalian cortex consists of two main neuronal types: the principal excitatory pyramidal neurons (PNs) and the inhibitory interneurons (INs). The interplay between these two neuronal populations – which drive excitation and inhibition (E/I balance), respectively – is crucial for controlling the overall activity in the brain. A number of neurological and psychiatric disorders have been associated with changes in E/I balance. It is not surprising, therefore, that neural networks employ several different mechanisms to maintain their firing rates at a stable level, collectively referred as homeostatic forms of plasticity. Here, we share our views on how the size of IN populations may provide an early homeostatic checkpoint for controlling brain activity. In a recent paper published in Cell Reports, we demonstrate that the extent of IN apoptosis during a critical early postnatal period is plastic, cell type specific, and can be reduced in a cell-autonomous manner by acute increases in neuronal activity. We propose that a critical interplay between the physiological state of the network and its cellular units fine-tunes the size of IN populations with the aim of stabilizing network activity.
Collapse
Affiliation(s)
- Myrto Denaxa
- Development and Function of Cortical Interneurons Lab, BSRC Al. Fleming, Athens, Greece
| | - Guilherme Neves
- Centre for Developmental Neurobiology, King's College London, London, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology, King's College London, London, UK
| | - Vassilis Pachnis
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
321
|
Bell AH, Bultitude JH. Methods matter: A primer on permanent and reversible interference techniques in animals for investigators of human neuropsychology. Neuropsychologia 2018; 115:211-219. [PMID: 28943365 PMCID: PMC6018620 DOI: 10.1016/j.neuropsychologia.2017.09.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/07/2017] [Accepted: 09/19/2017] [Indexed: 12/05/2022]
Abstract
The study of patients with brain lesions has contributed greatly to our understanding of the biological bases of human cognition, but this approach also has several unavoidable limitations. Research that uses animal models complements and extends human neuropsychology by addressing many of these limitations. In this review, we provide an overview of permanent and reversible animal lesion techniques for researchers of human neuropsychology, with the aim of highlighting how these methods provide a valuable adjunct to behavioural, neuroimaging, physiological, and clinical investigations in humans. Research in animals has provided important lessons about how the limitations of one or more techniques, or differences in their mechanism of action, has impacted upon the understanding of brain organisation and function. These cautionary tales highlight the importance of striving for a thorough understanding of how any intereference technique works (whether in animal or human), and for how to best use animal research to clarify the precise mechanisms underlying temporary lesion methods in humans.
Collapse
Affiliation(s)
- Andrew H Bell
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK; Department of Experimental Psychology, University of Oxford, Oxford, UK.
| | - Janet H Bultitude
- Department of Psychology, University of Bath, Bath, UK; Centre for Pain Research, University of Bath, Bath, UK; The Centre for Functional Magnetic Resonance Imaging of the Brain, University of Oxford, Oxford, UK
| |
Collapse
|
322
|
Luo J, Qian A, Oetjen LK, Yu W, Yang P, Feng J, Xie Z, Liu S, Yin S, Dryn D, Cheng J, Riehl TE, Zholos AV, Stenson WF, Kim BS, Hu H. TRPV4 Channel Signaling in Macrophages Promotes Gastrointestinal Motility via Direct Effects on Smooth Muscle Cells. Immunity 2018; 49:107-119.e4. [PMID: 29958798 DOI: 10.1016/j.immuni.2018.04.021] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 03/06/2018] [Accepted: 04/18/2018] [Indexed: 01/09/2023]
Abstract
Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.
Collapse
Affiliation(s)
- Jialie Luo
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aihua Qian
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai 200025, China
| | - Landon K Oetjen
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Weihua Yu
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - Pu Yang
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jing Feng
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zili Xie
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shenbin Liu
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shijin Yin
- College of Pharmacy, South-Central University for Nationalities, Wuhan, Hubei 430073, China
| | - Dari Dryn
- Department of Biophysics, Institute of Biology, Taras Shevchenko National University of Kyiv, Kyiv 03022, Ukraine
| | - Jizhong Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Terrence E Riehl
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander V Zholos
- Department of Biophysics, Institute of Biology, Taras Shevchenko National University of Kyiv, Kyiv 03022, Ukraine
| | - William F Stenson
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian S Kim
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Hongzhen Hu
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
323
|
Barnett SC, Perry BAL, Dalrymple-Alford JC, Parr-Brownlie LC. Optogenetic stimulation: Understanding memory and treating deficits. Hippocampus 2018; 28:457-470. [DOI: 10.1002/hipo.22960] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/24/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023]
Affiliation(s)
- S. C. Barnett
- Department of Psychology; University of Canterbury; Christchurch 8041 New Zealand
- Brain Research New Zealand; New Zealand
| | - B. A. L. Perry
- Department of Psychology; University of Canterbury; Christchurch 8041 New Zealand
- Brain Research New Zealand; New Zealand
| | - J. C. Dalrymple-Alford
- Department of Psychology; University of Canterbury; Christchurch 8041 New Zealand
- Brain Research New Zealand; New Zealand
- New Zealand Brain Research Institute; Christchurch New Zealand
| | - L. C. Parr-Brownlie
- Brain Research New Zealand; New Zealand
- Department of Anatomy, School of Biomedical Science; Brain Health Research Centre, University of Otago; Dunedin New Zealand
| |
Collapse
|
324
|
Priyadarshini M, Kotlo KU, Dudeja PK, Layden BT. Role of Short Chain Fatty Acid Receptors in Intestinal Physiology and Pathophysiology. Compr Physiol 2018; 8:1091-1115. [PMID: 29978895 DOI: 10.1002/cphy.c170050] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nutrient sensing is a mechanism for organisms to sense their environment. In larger animals, including humans, the intestinal tract is a major site of nutrient sensing for the body, not surprisingly, as this is the central location where nutrients are absorbed. In the gut, bacterial fermentation results in generation of short chain fatty acids (SCFAs), a class of nutrients, which are sensed by specific membrane bound receptors, FFA2, FFA3, GPR109a, and Olfr78. These receptors are expressed uniquely throughout the gut and signal through distinct mechanisms. To date, the emerging data suggests a role of these receptors in normal and pathological conditions. The overall function of these receptors is to regulate aspects of intestinal motility, hormone secretion, maintenance of the epithelial barrier, and immune cell function. Besides in intestinal health, a prominent role of these receptors has emerged in modulation of inflammatory and immune responses during pathological conditions. Moreover, these receptors are being revealed to interact with the gut microbiota. This review article updates the current body of knowledge on SCFA sensing receptors in the gut and their roles in intestinal health and disease as well as in whole body energy homeostasis. © 2017 American Physiological Society. Compr Physiol 8:1091-1115, 2018.
Collapse
Affiliation(s)
- Medha Priyadarshini
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Illinois, USA
| | - Kumar U Kotlo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Illinois, USA
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Illinois, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
325
|
Wei J, Zhong P, Qin L, Tan T, Yan Z. Chemicogenetic Restoration of the Prefrontal Cortex to Amygdala Pathway Ameliorates Stress-Induced Deficits. Cereb Cortex 2018; 28:1980-1990. [PMID: 28498919 PMCID: PMC6018994 DOI: 10.1093/cercor/bhx104] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/27/2017] [Indexed: 01/11/2023] Open
Abstract
Corticosteroid stress hormones exert a profound impact on cognitive and emotional processes. Understanding the neuronal circuits that are altered by chronic stress is important for counteracting the detrimental effects of stress in a brain region- and cell type-specific manner. Using the chemogenetic tool, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), which enables the remote, noninvasive and long-lasting modulation of cellular activity and signal transduction in discrete neuronal populations in vivo, we sought to identify the specific pathways that play an essential role in stress responses. We found that prolonged severe stress induced the diminished glutamatergic projection from pyramidal neurons in prefrontal cortex (PFC) to GABAergic interneurons in basolateral amygdala (BLA), leading to the loss of feedforward inhibition and ensuing hyperexcitability of BLA principal neurons, which caused a variety of behavioral abnormalities. Activating PFC pyramidal neurons with hM3D(Gq) DREADD restored the functional connection between PFC and BLA in stressed animals, resulting in the rescue of recognition memory, normalization of locomotor activity and reduction of aggressive behaviors. Inhibiting BLA principal neurons directly with hM4D(Gi) DREADD also blocked BLA hyperactivity and aggressive behaviors in stressed animals. These results have offered an effective avenue to counteract the stress-induced disruption of circuitry homeostasis.
Collapse
Affiliation(s)
- Jing Wei
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Luye Qin
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Tao Tan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, USA
- Medical Research, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| |
Collapse
|
326
|
Salay LD, Ishiko N, Huberman AD. A midline thalamic circuit determines reactions to visual threat. Nature 2018; 557:183-189. [DOI: 10.1038/s41586-018-0078-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/23/2018] [Indexed: 01/27/2023]
|
327
|
Booth MJ, Restrepo Schild V, Downs FG, Bayley H. Functional aqueous droplet networks. MOLECULAR BIOSYSTEMS 2018; 13:1658-1691. [PMID: 28766622 DOI: 10.1039/c7mb00192d] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Droplet interface bilayers (DIBs), comprising individual lipid bilayers between pairs of aqueous droplets in an oil, are proving to be a useful tool for studying membrane proteins. Recently, attention has turned to the elaboration of networks of aqueous droplets, connected through functionalized interface bilayers, with collective properties unachievable in droplet pairs. Small 2D collections of droplets have been formed into soft biodevices, which can act as electronic components, light-sensors and batteries. A substantial breakthrough has been the development of a droplet printer, which can create patterned 3D droplet networks of hundreds to thousands of connected droplets. The 3D networks can change shape, or carry electrical signals through defined pathways, or express proteins in response to patterned illumination. We envisage using functional 3D droplet networks as autonomous synthetic tissues or coupling them with cells to repair or enhance the properties of living tissues.
Collapse
Affiliation(s)
- Michael J Booth
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK.
| | | | | | | |
Collapse
|
328
|
Jayaraj ND, Bhattacharyya BJ, Belmadani AA, Ren D, Rathwell CA, Hackelberg S, Hopkins BE, Gupta HR, Miller RJ, Menichella DM. Reducing CXCR4-mediated nociceptor hyperexcitability reverses painful diabetic neuropathy. J Clin Invest 2018. [PMID: 29533926 DOI: 10.1172/jci92117] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Painful diabetic neuropathy (PDN) is an intractable complication of diabetes that affects 25% of patients. PDN is characterized by neuropathic pain and small-fiber degeneration, accompanied by dorsal root ganglion (DRG) nociceptor hyperexcitability and loss of their axons within the skin. The molecular mechanisms underlying DRG nociceptor hyperexcitability and small-fiber degeneration in PDN are unknown. We hypothesize that chemokine CXCL12/CXCR4 signaling is central to this mechanism, as we have shown that CXCL12/CXCR4 signaling is necessary for the development of mechanical allodynia, a pain hypersensitivity behavior common in PDN. Focusing on DRG neurons expressing the sodium channel Nav1.8, we applied transgenic, electrophysiological, imaging, and chemogenetic techniques to test this hypothesis. In the high-fat diet mouse model of PDN, we were able to prevent and reverse mechanical allodynia and small-fiber degeneration by limiting CXCR4 signaling or neuronal excitability. This study reveals that excitatory CXCR4/CXCL12 signaling in Nav1.8-positive DRG neurons plays a critical role in the pathogenesis of mechanical allodynia and small-fiber degeneration in a mouse model of PDN. Hence, we propose that targeting CXCR4-mediated DRG nociceptor hyperexcitability is a promising therapeutic approach for disease-modifying treatments for this currently intractable and widespread affliction.
Collapse
Affiliation(s)
| | | | - Abdelhak A Belmadani
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dongjun Ren
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Craig A Rathwell
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Brittany E Hopkins
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Herschel R Gupta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Richard J Miller
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniela M Menichella
- Department of Neurology and.,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
329
|
Yun S, Reynolds RP, Petrof I, White A, Rivera PD, Segev A, Gibson AD, Suarez M, DeSalle MJ, Ito N, Mukherjee S, Richardson DR, Kang CE, Ahrens-Nicklas RC, Soler I, Chetkovich DM, Kourrich S, Coulter DA, Eisch AJ. Stimulation of entorhinal cortex-dentate gyrus circuitry is antidepressive. Nat Med 2018; 24:658-666. [PMID: 29662202 PMCID: PMC5948139 DOI: 10.1038/s41591-018-0002-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 01/26/2018] [Indexed: 12/23/2022]
Abstract
Major Depressive Disorder (MDD) is considered a “circuitopathy”, and brain stimulation therapies hold promise for ameliorating MDD symptoms, including hippocampal dysfunction. It is unknown if stimulation of upstream hippocampal circuitry, such as the entorhinal cortex (Ent), is antidepressive, although Ent stimulation improves learning and memory in lab animals and humans. Here we show molecular targeting (Ent-specific knockdown of a psychosocial stress-induced protein) and chemogenetic stimulation of Ent neurons induce antidepressive-like effects in mice. Mechanistically, we show that Ent stimulation-induced antidepressive-like behavior relies on the generation of new hippocampal neurons. Thus, controlled stimulation of Ent hippocampal afferents is antidepressive via increased hippocampal neurogenesis. These findings emphasize the power and potential of Ent glutamatergic afferent stimulation - previously well known for the ability to influence learning and memory - for MDD treatment.
Collapse
Affiliation(s)
- Sanghee Yun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Ryan P Reynolds
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Iraklis Petrof
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Alicia White
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Phillip D Rivera
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, Massachusetts General Hospital for Children, Charlestown, MA, USA
| | - Amir Segev
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adam D Gibson
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Maiko Suarez
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Matthew J DeSalle
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Naoki Ito
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Oriental Medicine Research Center, Kitasato University, Tokyo, Japan
| | - Shibani Mukherjee
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Devon R Richardson
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Catherine E Kang
- Department of Neurology and Clinical Neurological Sciences, Northwestern University, Chicago, IL, USA
| | | | - Ivan Soler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Dane M Chetkovich
- Department of Neurology and Clinical Neurological Sciences, Northwestern University, Chicago, IL, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Douglas A Coulter
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Amelia J Eisch
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA. .,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
330
|
Berglind F, Andersson M, Kokaia M. Dynamic interaction of local and transhemispheric networks is necessary for progressive intensification of hippocampal seizures. Sci Rep 2018; 8:5669. [PMID: 29618778 PMCID: PMC5884800 DOI: 10.1038/s41598-018-23659-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/02/2018] [Indexed: 12/02/2022] Open
Abstract
The detailed mechanisms of progressive intensification of seizures often occurring in epilepsy are not well understood. Animal models of kindling, with progressive intensification of stimulation-induced seizures, have been previously used to investigate alterations in neuronal networks, but has been obscured by limited recording capabilities during electrical stimulations. Remote networks in kindling have been studied by physical deletions of the connected structures or pathways, inevitably leading to structural reorganisations and related adverse effects. We used optogenetics to circumvent the above-mentioned problems inherent to electrical kindling, and chemogenetics to temporarily inhibit rather than ablate the remote interconnected networks. Progressively intensifying afterdischarges (ADs) were induced by repetitive photoactivation of principal neurons in the hippocampus of anaesthetized transgenic mice expressing ChR2. This allowed, during the stimulation, to reveal dynamic increases in local field potentials (LFPs), which coincided with the start of AD intensification. Furthermore, chemogenetic functional inhibition of contralateral hippocampal neurons via hM4D(Gi) receptors abrogated AD progression. These findings demonstrate that, during repeated activation, local circuits undergo acute plastic changes with appearance of additional network discharges (LFPs), leading to transhemispheric recruitment of contralateral dentate gyrus, which seems to be necessary for progressive intensification of ADs.
Collapse
Affiliation(s)
- Fredrik Berglind
- Epilepsy Centre, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - My Andersson
- Epilepsy Centre, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Merab Kokaia
- Epilepsy Centre, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden.
| |
Collapse
|
331
|
Abstract
Optogenetics and chemogenetics provide the ability to modulate neurons in a type- and region-specific manner. These powerful techniques are useful to test hypotheses regarding the neural circuit mechanisms of general anesthetic end points such as hypnosis and analgesia. With both techniques, a genetic strategy is used to target expression of light-sensitive ion channels (opsins) or designer receptors exclusively activated by designer drugs in specific neurons. Optogenetics provides precise temporal control of neuronal firing with light pulses, whereas chemogenetics provides the ability to modulate neuronal firing for several hours with the single administration of a designer drug. This chapter provides an overview of neuronal targeting and experimental strategies and highlights the important advantages and disadvantages of each technique.
Collapse
Affiliation(s)
- Ksenia Vlasov
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Christa J Van Dort
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts Institute of Technology, Cambridge, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ken Solt
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts Institute of Technology, Cambridge, MA, United States; Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
332
|
Tian B, Xu S, Rogers JA, Cestellos-Blanco S, Yang P, Carvalho-de-Souza JL, Bezanilla F, Liu J, Bao Z, Hjort M, Cao Y, Melosh N, Lanzani G, Benfenati F, Galli G, Gygi F, Kautz R, Gorodetsky AA, Kim SS, Lu TK, Anikeeva P, Cifra M, Krivosudský O, Havelka D, Jiang Y. Roadmap on semiconductor-cell biointerfaces. Phys Biol 2018; 15:031002. [PMID: 29205173 PMCID: PMC6599646 DOI: 10.1088/1478-3975/aa9f34] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This roadmap outlines the role semiconductor-based materials play in understanding the complex biophysical dynamics at multiple length scales, as well as the design and implementation of next-generation electronic, optoelectronic, and mechanical devices for biointerfaces. The roadmap emphasizes the advantages of semiconductor building blocks in interfacing, monitoring, and manipulating the activity of biological components, and discusses the possibility of using active semiconductor-cell interfaces for discovering new signaling processes in the biological world.
Collapse
Affiliation(s)
- Bozhi Tian
- Department of Chemistry, University of Chicago, Chicago, IL 60637, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
333
|
The DREADD agonist clozapine N-oxide (CNO) is reverse-metabolized to clozapine and produces clozapine-like interoceptive stimulus effects in rats and mice. Sci Rep 2018; 8:3840. [PMID: 29497149 PMCID: PMC5832819 DOI: 10.1038/s41598-018-22116-z] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/15/2018] [Indexed: 12/01/2022] Open
Abstract
Clozapine-N-oxide (CNO) has long been the ligand of choice for selectively activating Designer Receptors Exclusively Activated by Designer Drugs (DREADDs). However, recent studies have challenged the long-held assertion that CNO is otherwise pharmacologically inert. The present study aimed to 1) determine whether CNO is reverse-metabolized to its parent compound clozapine in mice (as has recently been reported in rats), and 2) determine whether CNO exerts clozapine-like interoceptive stimulus effects in rats and/or mice. Following administration of 10.0 mg/kg CNO, pharmacokinetic analyses replicated recent reports of back-conversion to clozapine in rats and revealed that this phenomenon also occurs in mice. In rats and mice trained to discriminate 1.25 mg/kg clozapine from vehicle, CNO (1.0–20.0 mg/kg) produced partial substitution for the clozapine stimulus on average, with full substitution being detected in some individual animals of both species at doses frequently used to activate DREADDs. The present demonstration that CNO is converted to clozapine and exerts clozapine-like behavioral effects in both mice and rats further emphasizes the need for appropriate control groups in studies employing DREADDs, and highlights the utility of the drug discrimination procedure as a tool with which to screen the off-target effects of novel DREADD agonists.
Collapse
|
334
|
|
335
|
Adekunbi DA, Li XF, Lass G, Shetty K, Adegoke OA, Yeo SH, Colledge WH, Lightman SL, O'Byrne KT. Kisspeptin neurones in the posterodorsal medial amygdala modulate sexual partner preference and anxiety in male mice. J Neuroendocrinol 2018; 30:e12572. [PMID: 29356147 PMCID: PMC5873280 DOI: 10.1111/jne.12572] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 01/05/2018] [Accepted: 01/16/2018] [Indexed: 01/27/2023]
Abstract
The posterodorsal medial amygdala (MePD) is a neural site in the limbic brain involved in regulating emotional and sexual behaviours. There is, however, limited information available on the specific neuronal cell type in the MePD functionally mediating these behaviours in rodents. The recent discovery of a significant kisspeptin neurone population in the MePD has raised interest in the possible role of kisspeptin and its cognate receptor in sexual behaviour. The present study therefore tested the hypothesis that the MePD kisspeptin neurone population is involved in regulating attraction towards opposite sex conspecifics, sexual behaviour, social interaction and the anxiety response by selectively stimulating these neurones using the novel pharmacosynthetic DREADDs (designer receptors exclusively activated by designer drugs) technique. Adult male Kiss-Cre mice received bilateral stereotaxic injections of a stimulatory DREADD viral construct (AAV-hSyn-DIO-hM3 D(Gq)-mCherry) targeted to the MePD, with subsequent activation by i.p. injection of clozapine-N-oxide (CNO). Socio-sexual behaviours were assessed in a counter-balanced fashion after i.p. injection of either saline or CNO (5 mg kg-1 ). Selective activation of MePD kisspeptin neurones by CNO significantly increased the time spent by male mice in investigating an oestrous female, as well as the duration of social interaction. Additionally, after CNO injection, the mice appeared less anxious, as indicated by a longer exploratory time in the open arms of the elevated plus maze. However, levels of copulatory behaviour were comparable between CNO and saline-treated controls. These data indicate that DREADD-induced activation of MePD kisspeptin neurones enhances both sexual partner preference in males and social interaction and also decreases anxiety, suggesting a key role played by MePD kisspeptin in sexual motivation and social behaviour.
Collapse
Affiliation(s)
- D. A. Adekunbi
- Division of Women's HealthFaculty of Life Sciences and MedicineKing's College LondonLondonUK
- Department of PhysiologyCollege of MedicineUniversity of LagosLagosNigeria
| | - X. F. Li
- Division of Women's HealthFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - G. Lass
- Division of Women's HealthFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - K. Shetty
- Division of Women's HealthFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - O. A. Adegoke
- Department of PhysiologyCollege of MedicineUniversity of LagosLagosNigeria
| | - S. H. Yeo
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - W. H. Colledge
- Reproductive Physiology GroupDepartment of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - S. L. Lightman
- Henry Wellcome Laboratory for Integrative Neuroscience and EndocrinologyUniversity of BristolBristolUK
| | - K. T. O'Byrne
- Division of Women's HealthFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| |
Collapse
|
336
|
Hurni N, Kolodziejczak M, Tomasello U, Badia J, Jacobshagen M, Prados J, Dayer A. Transient Cell-intrinsic Activity Regulates the Migration and Laminar Positioning of Cortical Projection Neurons. Cereb Cortex 2018; 27:3052-3063. [PMID: 28334356 DOI: 10.1093/cercor/bhx059] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 02/23/2017] [Indexed: 12/21/2022] Open
Abstract
Neocortical microcircuits are built during development and require the coordinated assembly of excitatory glutamatergic projection neurons (PNs) into functional networks. Neuronal migration is an essential step in this process. In addition to cell-intrinsic mechanisms, external cues including neurotransmitters regulate cortical neuron migration, suggesting that early activity could influence this process. Here, we aimed to investigate the role of cell-intrinsic activity in migrating PNs in vivo using a designer receptor exclusively activated by a designer drug (DREADD) chemogenetic approach. In utero electroporation was used to specifically express the human M3 muscarinic cholinergic Gq-coupled receptor (hM3Dq) in PNs and calcium activity, migratory dynamics, gene expression, and laminar positioning of PNs were assessed following embryonic DREADD activation. We found that transient embryonic DREADD activation induced premature branching and transcriptional changes in migrating PNs leading to a persistent laminar mispositioning of superficial layer PNs into deep cortical layers without affecting expression of layer-specific molecular identity markers. In addition, live imaging approaches indicated that embryonic DREADD activation increased calcium transients in migrating PNs and altered their migratory dynamics by increasing their pausing time. Taken together, these results support the idea that increased cell-intrinsic activity during migration acts as a stop signal for migrating cortical PNs.
Collapse
Affiliation(s)
- Nicolas Hurni
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Marta Kolodziejczak
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Ugo Tomasello
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Joan Badia
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Moritz Jacobshagen
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Julien Prados
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Alexandre Dayer
- Department of Psychiatry, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
337
|
Campbell EJ, Marchant NJ. The use of chemogenetics in behavioural neuroscience: receptor variants, targeting approaches and caveats. Br J Pharmacol 2018; 175:994-1003. [PMID: 29338070 DOI: 10.1111/bph.14146] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/13/2017] [Accepted: 12/27/2017] [Indexed: 12/18/2022] Open
Abstract
The last decade has seen major advances in neuroscience tools allowing us to selectively modulate cellular pathways in freely moving animals. Chemogenetic approaches such as designer receptors exclusively activated by designer drugs (DREADDs) permit the remote control of neuronal function by systemic drug administration. These approaches have dramatically advanced our understanding of the neural control of behaviour. Here, we review the different techniques and genetic approaches available for the restriction of chemogenetic receptors to defined neuronal populations. We highlight the use of a dual virus approach to target specific circuitries and the effectiveness of different routes of administration of designer drugs. Finally, we discuss the potential caveats associated with DREADDs including off-target effects of designer drugs, the effects of chronic chemogenetic receptor activation and the issue of collateral projections associated with DREADD activation and inhibition.
Collapse
Affiliation(s)
- Erin J Campbell
- The Florey Institute of Neuroscience and Mental Health, Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Nathan J Marchant
- The Florey Institute of Neuroscience and Mental Health, Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy & Neurosciences, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
338
|
Wolff SB, Ölveczky BP. The promise and perils of causal circuit manipulations. Curr Opin Neurobiol 2018; 49:84-94. [PMID: 29414070 DOI: 10.1016/j.conb.2018.01.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/27/2017] [Accepted: 01/15/2018] [Indexed: 02/07/2023]
Abstract
The development of increasingly sophisticated methods for recording and manipulating neural activity is revolutionizing neuroscience. By probing how activity patterns in different types of neurons and circuits contribute to behavior, these tools can help inform mechanistic models of brain function and explain the roles of distinct circuit elements. However, in systems where functions are distributed over large networks, interpreting causality experiments can be challenging. Here we review common assumptions underlying circuit manipulations in behaving animals and discuss the strengths and limitations of different approaches.
Collapse
Affiliation(s)
- Steffen Be Wolff
- Department of Organismic and Evolutionary Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Bence P Ölveczky
- Department of Organismic and Evolutionary Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
339
|
Kesby JP, Eyles DW, McGrath JJ, Scott JG. Dopamine, psychosis and schizophrenia: the widening gap between basic and clinical neuroscience. Transl Psychiatry 2018; 8:30. [PMID: 29382821 PMCID: PMC5802623 DOI: 10.1038/s41398-017-0071-9] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/10/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022] Open
Abstract
The stagnation in drug development for schizophrenia highlights the need for better translation between basic and clinical research. Understanding the neurobiology of schizophrenia presents substantial challenges but a key feature continues to be the involvement of subcortical dopaminergic dysfunction in those with psychotic symptoms. Our contemporary knowledge regarding dopamine dysfunction has clarified where and when dopaminergic alterations may present in schizophrenia. For example, clinical studies have shown patients with schizophrenia show increased presynaptic dopamine function in the associative striatum, rather than the limbic striatum as previously presumed. Furthermore, subjects deemed at high risk of developing schizophrenia show similar presynaptic dopamine abnormalities in the associative striatum. Thus, our view of subcortical dopamine function in schizophrenia continues to evolve as we accommodate this newly acquired information. However, basic research in animal models has been slow to incorporate these clinical findings. For example, psychostimulant-induced locomotion, the commonly utilised phenotype for positive symptoms in rodents, is heavily associated with dopaminergic activation in the limbic striatum. This anatomical misalignment has brought into question how we assess positive symptoms in animal models and represents an opportunity for improved translation between basic and clinical research. The current review focuses on the role of subcortical dopamine dysfunction in psychosis and schizophrenia. We present and discuss alternative phenotypes that may provide a more translational approach to assess the neurobiology of positive symptoms in schizophrenia. Incorporation of recent clinical findings is essential if we are to develop meaningful translational animal models.
Collapse
Affiliation(s)
- JP Kesby
- 0000 0000 9320 7537grid.1003.2Queensland Brain Institute, The University of Queensland, St. Lucia, QLD Australia ,0000 0000 9320 7537grid.1003.2Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD Australia
| | - DW Eyles
- 0000 0000 9320 7537grid.1003.2Queensland Brain Institute, The University of Queensland, St. Lucia, QLD Australia ,0000 0004 0606 3563grid.417162.7Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD Australia
| | - JJ McGrath
- 0000 0000 9320 7537grid.1003.2Queensland Brain Institute, The University of Queensland, St. Lucia, QLD Australia ,0000 0004 0606 3563grid.417162.7Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD Australia ,0000 0001 1956 2722grid.7048.bNational Centre for Register-based Research, Aarhus University, Aarhus C, Denmark
| | - JG Scott
- 0000 0000 9320 7537grid.1003.2Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD Australia ,0000 0004 0606 3563grid.417162.7Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD Australia ,0000 0001 0688 4634grid.416100.2Metro North Mental Health, Royal Brisbane and Women’s Hospital, Herston, QLD Australia
| |
Collapse
|
340
|
Abstract
The programming of new functions into mammalian cells has tremendous application in research and medicine. Continued improvements in the capacity to sequence and synthesize DNA have rapidly increased our understanding of mechanisms of gene function and regulation on a genome-wide scale and have expanded the set of genetic components available for programming cell biology. The invention of new research tools, including targetable DNA-binding systems such as CRISPR/Cas9 and sensor-actuator devices that can recognize and respond to diverse chemical, mechanical, and optical inputs, has enabled precise control of complex cellular behaviors at unprecedented spatial and temporal resolution. These tools have been critical for the expansion of synthetic biology techniques from prokaryotic and lower eukaryotic hosts to mammalian systems. Recent progress in the development of genome and epigenome editing tools and in the engineering of designer cells with programmable genetic circuits is expanding approaches to prevent, diagnose, and treat disease and to establish personalized theranostic strategies for next-generation medicines. This review summarizes the development of these enabling technologies and their application to transforming mammalian synthetic biology into a distinct field in research and medicine.
Collapse
Affiliation(s)
- Joshua B Black
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708; , .,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708
| | - Pablo Perez-Pinera
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; .,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708; , .,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina 27708.,Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
341
|
Pati S, Sood A, Mukhopadhyay S, Vaidya VA. Acute pharmacogenetic activation of medial prefrontal cortex excitatory neurons regulates anxiety-like behaviour. J Biosci 2018. [DOI: 10.1007/s12038-018-9732-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
342
|
Gomez JL, Bonaventura J, Lesniak W, Mathews WB, Sysa-Shah P, Rodriguez LA, Ellis RJ, Richie CT, Harvey BK, Dannals RF, Pomper MG, Bonci A, Michaelides M. Chemogenetics revealed: DREADD occupancy and activation via converted clozapine. Science 2018; 357:503-507. [PMID: 28774929 DOI: 10.1126/science.aan2475] [Citation(s) in RCA: 710] [Impact Index Per Article: 101.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/03/2017] [Indexed: 01/01/2023]
Abstract
The chemogenetic technology DREADD (designer receptors exclusively activated by designer drugs) is widely used for remote manipulation of neuronal activity in freely moving animals. DREADD technology posits the use of "designer receptors," which are exclusively activated by the "designer drug" clozapine N-oxide (CNO). Nevertheless, the in vivo mechanism of action of CNO at DREADDs has never been confirmed. CNO does not enter the brain after systemic drug injections and shows low affinity for DREADDs. Clozapine, to which CNO rapidly converts in vivo, shows high DREADD affinity and potency. Upon systemic CNO injections, converted clozapine readily enters the brain and occupies central nervous system-expressed DREADDs, whereas systemic subthreshold clozapine injections induce preferential DREADD-mediated behaviors.
Collapse
Affiliation(s)
- Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD 21224, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD 21224, USA
| | - Wojciech Lesniak
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - William B Mathews
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Polina Sysa-Shah
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Lionel A Rodriguez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD 21224, USA
| | - Randall J Ellis
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD 21224, USA
| | - Christopher T Richie
- Optogenetics and Transgenic Technology Core, NIDA Intramural Research Program, Baltimore, MD 21224, USA
| | - Brandon K Harvey
- Optogenetics and Transgenic Technology Core, NIDA Intramural Research Program, Baltimore, MD 21224, USA
| | - Robert F Dannals
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Martin G Pomper
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Antonello Bonci
- Synaptic Plasticity Section, NIDA Intramural Research Program, Baltimore, MD 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse (NIDA) Intramural Research Program, Baltimore, MD 21224, USA. .,Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
343
|
Wu Q, Han Y, Tong Q. Current Genetic Techniques in Neural Circuit Control of Feeding and Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:211-233. [PMID: 30390293 DOI: 10.1007/978-981-13-1286-1_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The current epidemic of obesity and its associated metabolic syndromes imposes unprecedented challenges to our society. Despite intensive research focus on obesity pathogenesis, an effective therapeutic strategy to treat and cure obesity is still lacking. The obesity development is due to a disturbed homeostatic control of feeding and energy expenditure, both of which are controlled by an intricate neural network in the brain. Given the inherent complexity of brain networks in controlling feeding and energy expenditure, the understanding of brain-based pathophysiology for obesity development is limited. One key limiting factor in dissecting neural pathways for feeding and energy expenditure is unavailability of techniques that can be used to effectively reduce the complexity of the brain network to a tractable paradigm, based on which a strong hypothesis can be tested. Excitingly, emerging techniques have been involved to be able to link specific groups of neurons and neural pathways to behaviors (i.e., feeding and energy expenditure). In this chapter, novel techniques especially those based on animal models and viral vector approaches will be discussed. We hope that this chapter will provide readers with a basis that can help to understand the literatures using these techniques and with a guide to apply these exciting techniques to investigate brain mechanisms underlying feeding and energy expenditure.
Collapse
Affiliation(s)
- Qi Wu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA. .,Children's Nutrition Research Center, Research Service of Department of Agriculture of USA, Houston, TX, USA.
| | - Yong Han
- Department of Pediatrics, Baylor College of Medicine, USDA-ARS, Houston, TX, USA
| | - Qingchun Tong
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
344
|
Abstract
For many years, the complexity and multifactorial nature of brain-immune interactions limited our ability to dissect their underlying mechanisms. An especially challenging question was how the brain controls immunity, since the repertoire of techniques to control the brain's activity was extremely limited. New tools, such as optogenetics and chemogenetics (e.g., DREADDs), developed over the last decade, opened new frontiers in neuroscience with major implications for neuroimmunology. These tools enable mapping the causal effects of activating/attenuating defined neurons in the brain, on the immune system. Here, we present a detailed experimental protocol for the analysis of brain-immune interactions, based on chemogenetic or optogenetic manipulation of defined neuronal populations in the brain, and the subsequent analysis of immune cells. Such detailed and systematic dissection of brain-immune interactions has the potential to revolutionize our understanding of how mental and neurological states affect health and disease.
Collapse
Affiliation(s)
- Ben Korin
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
345
|
Donthamsetti PC, Winter N, Schönberger M, Levitz J, Stanley C, Javitch JA, Isacoff EY, Trauner D. Optical Control of Dopamine Receptors Using a Photoswitchable Tethered Inverse Agonist. J Am Chem Soc 2017; 139:18522-18535. [PMID: 29166564 PMCID: PMC5942546 DOI: 10.1021/jacs.7b07659] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Family A G protein-coupled receptors (GPCRs) control diverse biological processes and are of great clinical relevance. Their archetype rhodopsin becomes naturally light sensitive by binding covalently to the photoswitchable tethered ligand (PTL) retinal. Other GPCRs, however, neither bind covalently to ligands nor are light sensitive. We sought to impart the logic of rhodopsin to light-insensitive Family A GPCRs in order to enable their remote control in a receptor-specific, cell-type-specific, and spatiotemporally precise manner. Dopamine receptors (DARs) are of particular interest for their roles in motor coordination, appetitive, and aversive behavior, as well as neuropsychiatric disorders such as Parkinson's disease, schizophrenia, mood disorders, and addiction. Using an azobenzene derivative of the well-known DAR ligand 2-(N-phenethyl-N-propyl)amino-5-hydroxytetralin (PPHT), we were able to rapidly, reversibly, and selectively block dopamine D1 and D2 receptors (D1R and D2R) when the PTL was conjugated to an engineered cysteine near the dopamine binding site. Depending on the site of tethering, the ligand behaved as either a photoswitchable tethered neutral antagonist or inverse agonist. Our results indicate that DARs can be chemically engineered for selective remote control by light and provide a template for precision control of Family A GPCRs.
Collapse
Affiliation(s)
- Prashant C. Donthamsetti
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Nils Winter
- Department of Chemistry and Center for Integrated Protein Science, Ludwig-Maximilians-Universität, Butenandtstraβe 5-13, Munich 81377, Germany
| | - Matthias Schönberger
- Department of Chemistry and Center for Integrated Protein Science, Ludwig-Maximilians-Universität, Butenandtstraβe 5-13, Munich 81377, Germany
| | - Joshua Levitz
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Cherise Stanley
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Jonathan A. Javitch
- Departments of Psychiatry and Pharmacology, Columbia University, New York, New York 10027, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Ehud Y. Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720, United States
- Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Dirk Trauner
- Department of Chemistry and Center for Integrated Protein Science, Ludwig-Maximilians-Universität, Butenandtstraβe 5-13, Munich 81377, Germany
- Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
346
|
Gutruf P, Rogers JA. Implantable, wireless device platforms for neuroscience research. Curr Opin Neurobiol 2017; 50:42-49. [PMID: 29289027 DOI: 10.1016/j.conb.2017.12.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/13/2017] [Accepted: 12/12/2017] [Indexed: 01/19/2023]
Abstract
Recently developed classes of ultraminiaturized wireless devices provide powerful capabilities in neuroscience research, as implantable light sources for simulation/inhibition via optogenetics, as integrated microfluidic systems for programmed pharmacological delivery and as multimodal sensors for physiological measurements. These platforms leverage basic advances in biocompatible materials, semiconductor device designs and systems engineering concepts to afford modes of operation that are qualitatively distinct from those of conventional approaches that tether animals to external hardware by means of optical fibers, electrical cables and/or fluidic tubing. Neuroscience studies that exploit the unique features of these technologies enable insights into neural function through targeted stimulation, inhibition and recording, with spatially and genetically precise manipulation of neural circuit activity. Experimental possibilities include studies in naturalistic, three dimensional environments, investigations of pair-wise or group related social interactions and many other scenarios of interest that cannot be addressed using traditional hardware.
Collapse
Affiliation(s)
- Philipp Gutruf
- Departments of Materials Science and Engineering, Biomedical Engineering, Chemistry, Neurological Surgery, Mechanical Engineering, Electrical Engineering and Computer Science, Simpson Querrey Institute & Feinberg Medical School, 2145 Sheridan Road, Evanston, IL 60208, United States
| | - John A Rogers
- Departments of Materials Science and Engineering, Biomedical Engineering, Chemistry, Neurological Surgery, Mechanical Engineering, Electrical Engineering and Computer Science, Simpson Querrey Institute & Feinberg Medical School, 2145 Sheridan Road, Evanston, IL 60208, United States.
| |
Collapse
|
347
|
Seabrook TA, Dhande OS, Ishiko N, Wooley VP, Nguyen PL, Huberman AD. Strict Independence of Parallel and Poly-synaptic Axon-Target Matching during Visual Reflex Circuit Assembly. Cell Rep 2017; 21:3049-3064. [PMID: 29241535 PMCID: PMC6333306 DOI: 10.1016/j.celrep.2017.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/04/2017] [Accepted: 11/10/2017] [Indexed: 12/19/2022] Open
Abstract
The use of sensory information to drive specific behaviors relies on circuits spanning long distances that wire up through a range of axon-target recognition events. Mechanisms assembling poly-synaptic circuits and the extent to which parallel pathways can "cross-wire" to compensate for loss of one another remain unclear and are crucial to our understanding of brain development and models of regeneration. In the visual system, specific retinal ganglion cells (RGCs) project to designated midbrain targets connected to downstream circuits driving visuomotor reflexes. Here, we deleted RGCs connecting to pupillary light reflex (PLR) midbrain targets and discovered that axon-target matching is tightly regulated. RGC axons of the eye-reflex pathway avoided vacated PLR targets. Moreover, downstream PLR circuitry is maintained; hindbrain and peripheral components retained their proper connectivity and function. These findings point to a model in which poly-synaptic circuit development reflects independent, highly stringent wiring of each parallel pathway and downstream station.
Collapse
Affiliation(s)
- Tania A Seabrook
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Onkar S Dhande
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Nao Ishiko
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Victoria P Wooley
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Phong L Nguyen
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94304, USA; Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94303, USA; Bio-X, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
348
|
Functional networks and network perturbations in rodents. Neuroimage 2017; 163:419-436. [DOI: 10.1016/j.neuroimage.2017.09.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 11/16/2022] Open
|
349
|
Swanson AM, DePoy LM, Gourley SL. Inhibiting Rho kinase promotes goal-directed decision making and blocks habitual responding for cocaine. Nat Commun 2017; 8:1861. [PMID: 29187752 PMCID: PMC5707361 DOI: 10.1038/s41467-017-01915-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 10/25/2017] [Indexed: 01/04/2023] Open
Abstract
The prelimbic prefrontal cortex is necessary for associating actions with their consequences, enabling goal-directed decision making. We find that the strength of action–outcome conditioning correlates with dendritic spine density in prelimbic cortex, suggesting that new action–outcome learning involves dendritic spine plasticity. To test this, we inhibited the cytoskeletal regulatory factor Rho kinase. We find that the inhibitor fasudil enhances action–outcome memory, resulting in goal-directed behavior in mice that would otherwise express stimulus-response habits. Fasudil transiently reduces prelimbic cortical dendritic spine densities during a period of presumed memory consolidation, but only when paired with new learning. Fasudil also blocks habitual responding for cocaine, an effect that persists over time, across multiple contexts, and depends on actin polymerization. We suggest that Rho kinase inhibition promotes goal-oriented action selection by augmenting the plasticity of prelimbic cortical dendritic spines during the formation of new action–outcome memories. Action-outcome learning requires the prelimbic prefrontal cortex. Here the authors report that fasudil, a Rho kinase inhibitor, reduces dendritic spine densities on prelimbic neurons in an activity-dependent manner, stimulating goal-directed actions, and reducing habitual responding for cocaine.
Collapse
Affiliation(s)
- Andrew M Swanson
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA
| | - Lauren M DePoy
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA. .,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
350
|
Mitra A, Raichle ME. How networks communicate: propagation patterns in spontaneous brain activity. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0546. [PMID: 27574315 DOI: 10.1098/rstb.2015.0546] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2016] [Indexed: 12/18/2022] Open
Abstract
Initially regarded as 'noise', spontaneous (intrinsic) activity accounts for a large portion of the brain's metabolic cost. Moreover, it is now widely known that infra-slow (less than 0.1 Hz) spontaneous activity, measured using resting state functional magnetic resonance imaging of the blood oxygen level-dependent (BOLD) signal, is correlated within functionally defined resting state networks (RSNs). However, despite these advances, the temporal organization of spontaneous BOLD fluctuations has remained elusive. By studying temporal lags in the resting state BOLD signal, we have recently shown that spontaneous BOLD fluctuations consist of remarkably reproducible patterns of whole brain propagation. Embedded in these propagation patterns are unidirectional 'motifs' which, in turn, give rise to RSNs. Additionally, propagation patterns are markedly altered as a function of state, whether physiological or pathological. Understanding such propagation patterns will likely yield deeper insights into the role of spontaneous activity in brain function in health and disease.This article is part of the themed issue 'Interpreting blood oxygen level-dependent: a dialogue between cognitive and cellular neuroscience'.
Collapse
Affiliation(s)
- Anish Mitra
- Department of Radiology, Washington University, St Louis, MO 63110, USA
| | - Marcus E Raichle
- Department of Radiology, Washington University, St Louis, MO 63110, USA Department of Neurology, Washington University, St Louis, MO 63110, USA
| |
Collapse
|