251
|
Liu K, Wang L. Optogenetics: Therapeutic spark in neuropathic pain. Bosn J Basic Med Sci 2019; 19:321-327. [PMID: 30995901 DOI: 10.17305/bjbms.2019.4114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/11/2019] [Indexed: 01/14/2023] Open
Abstract
Optogenetics is an emerging field, which uses light and molecular genetics to manipulate the activity of live cells by expressing light-sensitive proteins. With the discovery of bacteriorhodopsin, a light-sensitive bacterial protein, in 1971 Oesterhelt and Stoeckenius laid the pavement of optogenetics. However, the cross-integration of different disciplines is a little more than a decade old. The toolbox contains fluorescent sensors and optogenetic actuators that enable visualization of signaling events and manipulation of cellular activities, respectively. Neuropathic pain is pain caused either by damage or disease that affects the somatosensory system. The exact mechanism for neuropathic pain is not known, however proposed mechanisms include immune reactions, ion channel expressions, and inflammation. Current regimen for the disease provides about 50% relief for only 40-60% of patients. Recent in vivo and in vitro studies demonstrate the potential therapeutic applications of optogenetics by manipulating the activity of neurons. This review summarizes the basic concept, therapeutic applications for neuropathy, and potential of optogenetics to reach from bench to bedside in the near future.
Collapse
Affiliation(s)
- Kang Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| | | |
Collapse
|
252
|
Pedro MP, Salinas Parra N, Gutkind JS, Iglesias-Bartolome R. Activation of G-Protein Coupled Receptor-Gαi Signaling Increases Keratinocyte Proliferation and Reduces Differentiation, Leading to Epidermal Hyperplasia. J Invest Dermatol 2019; 140:1195-1203.e3. [PMID: 31707029 DOI: 10.1016/j.jid.2019.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/07/2019] [Accepted: 10/19/2019] [Indexed: 12/28/2022]
Abstract
G-protein coupled receptors (GPCRs) and their associated heterotrimeric G proteins impinge on pathways that control epithelial cell self-renewal and differentiation. Although it is known that Gαs protein signaling regulates skin homeostasis in vivo, the role of GPCR-coupled Gαi proteins in the skin is unclear. Here, by using a chemogenetic approach, we demonstrate that GPCR-Gαi activation can regulate keratinocyte proliferation and differentiation and that overactivation of Gαi-signaling in the basal compartment of the mouse skin can lead to epidermal hyperplasia. Our results expand our understanding of the role of GPCR-cAMP signaling in skin homeostasis and reveal overlapping and divergent roles of the cAMP-regulating heterotrimeric Gαs and Gαi proteins in keratinocytes.
Collapse
Affiliation(s)
- M Pilar Pedro
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Natalia Salinas Parra
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
253
|
Torre-Muruzabal T, Devoght J, Van den Haute C, Brône B, Van der Perren A, Baekelandt V. Chronic nigral neuromodulation aggravates behavioral deficits and synaptic changes in an α-synuclein based rat model for Parkinson's disease. Acta Neuropathol Commun 2019; 7:160. [PMID: 31640762 PMCID: PMC6805517 DOI: 10.1186/s40478-019-0814-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/22/2019] [Indexed: 01/26/2023] Open
Abstract
Aggregation of alpha-synuclein (α-SYN) is the pathological hallmark of several diseases named synucleinopathies, including Parkinson's disease (PD), which is the most common neurodegenerative motor disorder. Alpha-SYN has been linked to synaptic function both in physiological and pathological conditions. However, the exact link between neuronal activity, α-SYN toxicity and disease progression in PD is not clear. In this study, we aimed to investigate the effect of chronic neuromodulation in an α-SYN-based rat model for PD using chemogenetics. To do this, we expressed excitatory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) combined with mutant A53T α-SYN, using two different recombinant adeno-associated viral (rAAV) vectors (serotypes 2/7 and 2/8) in rat substantia nigra (SN) and investigated the effect on motor behavior, synapses and neuropathology. We found that chronic neuromodulation aggravates motor deficits induced by α-SYN, without altering dopaminergic neurodegeneration. In addition, neuronal activation led to changes in post-translational modification and subcellular localization of α-SYN, linking neuronal activity to the pathophysiological role of α-SYN in PD.
Collapse
Affiliation(s)
- Teresa Torre-Muruzabal
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | | | - Chris Van den Haute
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
- KU Leuven, Leuven Viral Vector Core, Leuven, Belgium
| | | | - Anke Van der Perren
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Veerle Baekelandt
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
254
|
Abstract
While significant advances have been achieved with non-living synthetic cells built from the bottom-up, less progress has been made with the fabrication of synthetic tissues built from such cells. Synthetic tissues comprise patterned three-dimensional (3D) collections of communicating compartments. They can include both biological and synthetic parts and may incorporate features that do more than merely mimic nature. 3D-printed materials based on droplet-interface bilayers are the basis of the most advanced synthetic tissues and are being developed for several applications, including the controlled release of therapeutic agents and the repair of damaged organs. Current goals include the ability to manipulate synthetic tissues by remote signaling and the formation of hybrid structures with fabricated or natural living tissues.
Collapse
|
255
|
Raper J, Murphy L, Richardson R, Romm Z, Kovacs-Balint Z, Payne C, Galvan A. Chemogenetic Inhibition of the Amygdala Modulates Emotional Behavior Expression in Infant Rhesus Monkeys. eNeuro 2019; 6:ENEURO.0360-19.2019. [PMID: 31541000 PMCID: PMC6791827 DOI: 10.1523/eneuro.0360-19.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/13/2019] [Indexed: 01/12/2023] Open
Abstract
Manipulation of neuronal activity during the early postnatal period in monkeys has been largely limited to permanent lesion studies, which can be impacted by developmental plasticity leading to reorganization and compensation from other brain structures that can interfere with the interpretations of results. Chemogenetic tools, such as DREADDs (designer receptors exclusively activated by designer drugs), can transiently and reversibly activate or inactivate brain structures, avoiding the pitfalls of permanent lesions to better address important developmental neuroscience questions. We demonstrate that inhibitory DREADDs in the amygdala can be used to manipulate socioemotional behavior in infant monkeys. Two infant rhesus monkeys (1 male, 1 female) received AAV5-hSyn-HA-hM4Di-IRES-mCitrine injections bilaterally in the amygdala at 9 months of age. DREADD activation after systemic administration of either clozapine-N-oxide or low-dose clozapine resulted in decreased freezing and anxiety on the human intruder paradigm and changed the looking patterns on a socioemotional attention eye-tracking task, compared with vehicle administration. The DREADD-induced behaviors were reminiscent of, but not identical to, those seen after permanent amygdala lesions in infant monkeys, such that neonatal lesions produce a more extensive array of behavioral changes in response to the human intruder task that were not seen with DREADD-evoked inhibition of this region. Our results may help support the notion that the more extensive behavior changes seen after early lesions are manifested from brain reorganization that occur after permanent damage. The current study provides a proof of principle that DREADDs can be used in young infant monkeys to transiently and reversibly manipulate behavior.
Collapse
Affiliation(s)
- Jessica Raper
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia 30329
| | - Lauren Murphy
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Department of Psychology, Emory University, Atlanta, Georgia 30329
| | - Rebecca Richardson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | - Zoe Romm
- Drexel University, Philadelphia, Pennsylvania 19104
| | - Zsofia Kovacs-Balint
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
| | | | - Adriana Galvan
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329
- Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia 30329
| |
Collapse
|
256
|
Arimura D, Shinohara K, Takahashi Y, Sugimura YK, Sugimoto M, Tsurugizawa T, Marumo K, Kato F. Primary Role of the Amygdala in Spontaneous Inflammatory Pain- Associated Activation of Pain Networks - A Chemogenetic Manganese-Enhanced MRI Approach. Front Neural Circuits 2019; 13:58. [PMID: 31632244 PMCID: PMC6779784 DOI: 10.3389/fncir.2019.00058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic pain is a major health problem, affecting 10–30% of the population in developed countries. While chronic pain is defined as “a persistent complaint of pain lasting for more than the usual period for recovery,” recently accumulated lines of evidence based on human brain imaging have revealed that chronic pain is not simply a sustained state of nociception, but rather an allostatic state established through gradually progressing plastic changes in the central nervous system. To visualize the brain activity associated with spontaneously occurring pain during the shift from acute to chronic pain under anesthetic-free conditions, we used manganese-enhanced magnetic resonance imaging (MEMRI) with a 9.4-T scanner to visualize neural activity-dependent accumulation of manganese in the brains of mice with hind paw inflammation. Time-differential analysis between 2- and 6-h after formalin injection to the left hind paw revealed a significantly increased MEMRI signal in various brain areas, including the right insular cortex, right nucleus accumbens, right globus pallidus, bilateral caudate putamen, right primary/secondary somatosensory cortex, bilateral thalamus, right amygdala, bilateral substantial nigra, and left ventral tegmental area. To analyze the role of the right amygdala in these post-formalin MEMRI signals, we repeatedly inhibited right amygdala neurons during this 2–6-h period using the “designer receptors exclusively activated by designer drugs” (DREADD) technique. Pharmacological activation of inhibitory DREADDs expressed in the right amygdala significantly attenuated MEMRI signals in the bilateral infralimbic cortex, bilateral nucleus accumbens, bilateral caudate putamen, right globus pallidus, bilateral ventral tegmental area, and bilateral substantia nigra, suggesting that the inflammatory pain-associated activation of these structures depends on the activity of the right amygdala and DREADD-expressing adjacent structures. In summary, the combined use of DREADD and MEMRI is a promising approach for revealing regions associated with spontaneous pain-associated brain activities and their causal relationships.
Collapse
Affiliation(s)
- Daigo Arimura
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Department of Orthopaedics, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Kei Shinohara
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Department of Orthopaedics, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Yukari Takahashi
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Yae K Sugimura
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Mariko Sugimoto
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Tomokazu Tsurugizawa
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan.,NeuroSpin, CEA-Saclay, Gif-sur-Yvette, France
| | - Keishi Marumo
- Department of Orthopaedics, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Fusao Kato
- Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
257
|
Rao S, Chen R, LaRocca AA, Christiansen MG, Senko AW, Shi CH, Chiang PH, Varnavides G, Xue J, Zhou Y, Park S, Ding R, Moon J, Feng G, Anikeeva P. Remotely controlled chemomagnetic modulation of targeted neural circuits. NATURE NANOTECHNOLOGY 2019; 14:967-973. [PMID: 31427746 PMCID: PMC6778020 DOI: 10.1038/s41565-019-0521-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 07/03/2019] [Indexed: 05/20/2023]
Abstract
Connecting neural circuit output to behaviour can be facilitated by the precise chemical manipulation of specific cell populations1,2. Engineered receptors exclusively activated by designer small molecules enable manipulation of specific neural pathways3,4. However, their application to studies of behaviour has thus far been hampered by a trade-off between the low temporal resolution of systemic injection versus the invasiveness of implanted cannulae or infusion pumps2. Here, we developed a remotely controlled chemomagnetic modulation-a nanomaterials-based technique that permits the pharmacological interrogation of targeted neural populations in freely moving subjects. The heat dissipated by magnetic nanoparticles (MNPs) in the presence of alternating magnetic fields (AMFs) triggers small-molecule release from thermally sensitive lipid vesicles with a 20 s latency. Coupled with the chemogenetic activation of engineered receptors, this technique permits the control of specific neurons with temporal and spatial precision. The delivery of chemomagnetic particles to the ventral tegmental area (VTA) allows the remote modulation of motivated behaviour in mice. Furthermore, this chemomagnetic approach activates endogenous circuits by enabling the regulated release of receptor ligands. Applied to an endogenous dopamine receptor D1 (DRD1) agonist in the nucleus accumbens (NAc), a brain area involved in mediating social interactions, chemomagnetic modulation increases sociability in mice. By offering a temporally precise control of specified ligand-receptor interactions in neurons, this approach may facilitate molecular neuroscience studies in behaving organisms.
Collapse
Affiliation(s)
- Siyuan Rao
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ritchie Chen
- Simons Center for Social Brain, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Ava A LaRocca
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael G Christiansen
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Health Sciences and Technology at the Swiss Federal Institute of Technology in Zürich (ETHZ), Zürich, Switzerland
| | - Alexander W Senko
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cindy H Shi
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Po-Han Chiang
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Georgios Varnavides
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jian Xue
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Zhou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Seongjun Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ruihua Ding
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Junsang Moon
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
258
|
Gomez-Ramirez M, More AI, Friedman NG, Hochgeschwender U, Moore CI. The BioLuminescent-OptoGenetic in vivo response to coelenterazine is proportional, sensitive, and specific in neocortex. J Neurosci Res 2019; 98:471-480. [PMID: 31544973 DOI: 10.1002/jnr.24498] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/11/2019] [Accepted: 07/03/2019] [Indexed: 12/20/2022]
Abstract
BioLuminescent (BL) light production can modulate neural activity and behavior through co-expressed OptoGenetic (OG) elements, an approach termed "BL-OG." Yet, the relationship between BL-OG effects and bioluminescent photon emission has not been characterized in vivo. Further, the degree to which BL-OG effects strictly depend on optogenetic mechanisms driven by bioluminescent photons is unknown. Crucial to every neuromodulation method is whether the activator shows a dynamic concentration range driving robust, selective, and nontoxic effects. We systematically tested the effects of four key components of the BL-OG mechanism (luciferin, oxidized luciferin, luciferin vehicle, and bioluminescence), and compared these against effects induced by the Luminopsin-3 (LMO3) BL-OG molecule, a fusion of slow burn Gaussia luciferase (sbGLuc) and Volvox ChannelRhodopsin-1 (VChR1). We performed combined bioluminescence imaging and electrophysiological recordings while injecting specific doses of Coelenterazine (substrate for sbGluc), Coelenteramide (CTM, the oxidized product of CTZ), or CTZ vehicle. CTZ robustly drove activity in mice expressing LMO3, with photon production proportional to firing rate. In contrast, low and moderate doses of CTZ, CTM, or vehicle did not modulate activity in mice that did not express LMO3. We also failed to find bioluminescence effects on neural activity in mice expressing an optogenetically nonsensitive LMO3 variant. We observed weak responses to the highest dose of CTZ in control mice, but these effects were significantly smaller than those observed in the LMO3 group. These results show that in neocortex in vivo, there is a large CTZ range wherein BL-OG effects are specific to its active chemogenetic mechanism.
Collapse
Affiliation(s)
| | - Alexander I More
- Department of Neuroscience, Brown University, Providence, Rhode Island
| | - Nina G Friedman
- Department of Neuroscience, Brown University, Providence, Rhode Island
| | - Ute Hochgeschwender
- College of Medicine and Neuroscience Program, Central Michigan University, Mount Pleasant, Michigan
| | | |
Collapse
|
259
|
Hinton EA, Li DC, Allen AG, Gourley SL. Social Isolation in Adolescence Disrupts Cortical Development and Goal-Dependent Decision-Making in Adulthood, Despite Social Reintegration. eNeuro 2019; 6:ENEURO.0318-19.2019. [PMID: 31527057 PMCID: PMC6757188 DOI: 10.1523/eneuro.0318-19.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 12/18/2022] Open
Abstract
The social environment influences neurodevelopment. Investigations using rodents to study this phenomenon commonly isolate subjects, then assess neurobehavioral consequences while animals are still isolated. This approach precludes one from dissociating the effects of on-going versus prior isolation, hindering our complete understanding of the consequences of social experience during particular developmental periods. Here, we socially isolated adolescent mice from postnatal day (P)31 to P60, then re-housed them into social groups. We tested their ability to select actions based on expected outcomes using multiple reinforcer devaluation and instrumental contingency degradation techniques. Social isolation in adolescence (but not adulthood) weakened instrumental response updating, causing mice to defer to habit-like behaviors. Habit biases were associated with glucocorticoid insufficiency in adolescence, oligodendrocyte marker loss throughout cortico-striatal regions, and dendritic spine and synaptic marker excess in the adult orbitofrontal cortex (OFC). Artificial, chemogenetic stimulation of the ventrolateral OFC in typical, healthy mice recapitulated response biases following isolation, causing habit-like behaviors. Meanwhile, correcting dendritic architecture by inhibiting the cytoskeletal regulatory protein ROCK remedied instrumental response updating defects in socially isolated mice. Our findings suggest that adolescence is a critical period during which social experience optimizes one's ability to seek and attain goals later in life. Age-typical dendritic spine elimination appears to be an essential factor, and in its absence, organisms may defer to habit-based behaviors.
Collapse
Affiliation(s)
- Elizabeth A Hinton
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, 30329
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
- Department of Psychiatry, Emory University, Atlanta, GA, 30329
| | - Dan C Li
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, 30329
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
- Department of Psychiatry, Emory University, Atlanta, GA, 30329
| | - Aylet G Allen
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
| | - Shannon L Gourley
- Graduate Program in Neuroscience, Emory University, Atlanta, GA, 30329
- Center for Translational and Social Neuroscience, Emory University, Atlanta, GA, 30329
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
- Department of Pediatrics, Emory University, Atlanta, GA, 30329
- Department of Psychiatry, Emory University, Atlanta, GA, 30329
| |
Collapse
|
260
|
Abstract
Over the past decade, basic sleep research investigating the circuitry controlling sleep and wakefulness has been boosted by pharmacosynthetic approaches, including chemogenetic techniques using designed receptors exclusively activated by designer drugs (DREADD). DREADD offers a series of tools that selectively control neuronal activity as a way to probe causal relationship between neuronal sub-populations and the regulation of the sleep-wake cycle. Following the path opened by optogenetics, DREADD tools applied to discrete neuronal sub-populations in numerous brain areas quickly made their contribution to the discovery and the expansion of our understanding of critical brain structures involved in a wide variety of behaviors and in the control of vigilance state architecture.
Collapse
|
261
|
Steiner PJ, Bedewitz MA, Medina‐Cucurella AV, Cutler SR, Whitehead TA. A yeast surface display platform for plant hormone receptors: Toward directed evolution of new biosensors. AIChE J 2019. [DOI: 10.1002/aic.16767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Paul J. Steiner
- Department of Chemical and Biological Engineering University of Colorado Boulder Colorado
| | - Matthew A. Bedewitz
- Department of Chemical and Biological Engineering University of Colorado Boulder Colorado
| | | | - Sean R. Cutler
- Department of Botany and Plant Sciences University of California Riverside California
| | - Timothy A. Whitehead
- Department of Chemical and Biological Engineering University of Colorado Boulder Colorado
- Department of Chemical Engineering and Materials Science Michigan State University East Lansing Michigan
| |
Collapse
|
262
|
Üner AG, Keçik O, Quaresma PGF, De Araujo TM, Lee H, Li W, Kim HJ, Chung M, Bjørbæk C, Kim YB. Role of POMC and AgRP neuronal activities on glycaemia in mice. Sci Rep 2019; 9:13068. [PMID: 31506541 PMCID: PMC6736943 DOI: 10.1038/s41598-019-49295-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/20/2019] [Indexed: 11/23/2022] Open
Abstract
Leptin regulates both feeding and glycaemia primarily through its receptors expressed on agouti-related peptide (AgRP) and pro-opiomelanocortin-expressing (POMC) neurons; however, it is unknown whether activity of these neuronal populations mediates the regulation of these processes. To determine this, we injected Cre-dependent designer receptors exclusively activated by designer drugs (DREADD) viruses into the hypothalamus of normoglycaemic and diabetic AgRP-ires-cre and POMC-cre mice to chemogenetically activate or inhibit these neuronal populations. Despite robust changes in food intake, activation or inhibition of AgRP neurons did not affect glycaemia, while activation caused significant (P = 0.014) impairment in insulin sensitivity. Stimulation of AgRP neurons in diabetic mice reversed leptin’s ability to inhibit feeding but did not counter leptin’s ability to lower blood glucose levels. Notably, the inhibition of POMC neurons stimulated feeding while decreasing glucose levels in normoglycaemic mice. The findings suggest that leptin’s effects on feeding by AgRP neurons are mediated by changes in neuronal firing, while the control of glucose balance by these cells is independent of chemogenetic activation or inhibition. The firing-dependent glucose lowering mechanism within POMC neurons is a potential target for the development of novel anti-diabetic medicines.
Collapse
Affiliation(s)
- Aykut Göktürk Üner
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA.,Department of Physiology, Faculty of Veterinary Medicine, Adnan Menderes University, Efeler, Aydin, 09010, Turkey
| | - Onur Keçik
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Paula G F Quaresma
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Thiago M De Araujo
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Hyon Lee
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Wenjing Li
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Hyun Jeong Kim
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Michelle Chung
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Christian Bjørbæk
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA
| | - Young-Bum Kim
- Department of Endocrinology, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
263
|
Wang L, Pydi SP, Cui Y, Zhu L, Meister J, Gavrilova O, Berdeaux R, Fortin JP, Bence KK, Vernochet C, Wess J. Selective activation of G s signaling in adipocytes causes striking metabolic improvements in mice. Mol Metab 2019; 27:83-91. [PMID: 31272886 PMCID: PMC6717953 DOI: 10.1016/j.molmet.2019.06.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/30/2019] [Accepted: 06/16/2019] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Given the worldwide epidemics of obesity and type 2 diabetes, novel antidiabetic and appetite-suppressing drugs are urgently needed. Adipocytes play a central role in the regulation of whole-body glucose and energy homeostasis. The goal of this study was to examine the metabolic effects of acute and chronic activation of Gs signaling selectively in adipocytes (activated Gs stimulates cAMP production), both in lean and obese mice. METHODS To address this question, we generated a novel mutant mouse strain (adipo-GsD mice) that expressed a Gs-coupled designer G protein-coupled receptor (Gs DREADD or short GsD) selectively in adipocytes. Importantly, the GsD receptor can only be activated by administration of an exogenous agent (CNO) that is otherwise pharmacologically inert. The adipo-GsD mice were maintained on either regular chow or a high-fat diet and then subjected to a comprehensive series of metabolic tests. RESULTS Pharmacological (CNO) activation of the GsD receptor in adipocytes of adipo-GsD mice caused profound improvements in glucose homeostasis and protected mice against the metabolic deficits associated with the consumption of a calorie-rich diet. Moreover, chronic activation of Gs signaling in adipocytes led to a striking increase in energy expenditure and reduced food intake, resulting in a decrease in body weight and fat mass when mice consumed a calorie-rich diet. CONCLUSION Systematic studies with a newly developed mouse model enabled us to assess the metabolic consequences caused by acute or chronic activation of Gs signaling selectively in adipocytes. Most strikingly, chronic activation of this pathway led to reduced body fat mass and restored normal glucose homeostasis in obese mice. These findings are of considerable relevance for the development of novel antidiabetic and anti-obesity drugs.
Collapse
Affiliation(s)
- Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology and Center for Metabolic and Degenerative Diseases at the Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth) and Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Jean-Philippe Fortin
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, MA, 02139, USA
| | - Kendra K Bence
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, MA, 02139, USA
| | - Cecile Vernochet
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, MA, 02139, USA.
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA.
| |
Collapse
|
264
|
Kahn JB, Port RG, Yue C, Takano H, Coulter DA. Circuit-based interventions in the dentate gyrus rescue epilepsy-associated cognitive dysfunction. Brain 2019; 142:2705-2721. [PMID: 31363737 PMCID: PMC6736326 DOI: 10.1093/brain/awz209] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022] Open
Abstract
Temporal lobe epilepsy is associated with significant structural pathology in the hippocampus. In the dentate gyrus, the summative effect of these pathologies is massive hyperexcitability in the granule cells, generating both increased seizure susceptibility and cognitive deficits. To date, therapeutic approaches have failed to improve the cognitive symptoms in fully developed, chronic epilepsy. As the dentate's principal signalling population, the granule cells' aggregate excitability has the potential to provide a mechanistically-independent downstream target. We examined whether normalizing epilepsy-associated granule cell hyperexcitability-without correcting the underlying structural circuit disruptions-would constitute an effective therapeutic approach for cognitive dysfunction. In the systemic pilocarpine mouse model of temporal lobe epilepsy, the epileptic dentate gyrus excessively recruits granule cells in behavioural contexts, not just during seizure events, and these mice fail to perform on a dentate-mediated spatial discrimination task. Acutely reducing dorsal granule cell hyperactivity in chronically epileptic mice via either of two distinct inhibitory chemogenetic receptors rescued behavioural performance such that they responded comparably to wild type mice. Furthermore, recreating granule cell hyperexcitability in control mice via excitatory chemogenetic receptors, without altering normal circuit anatomy, recapitulated spatial memory deficits observed in epileptic mice. However, making the granule cells overly quiescent in both epileptic and control mice again disrupted behavioural performance. These bidirectional manipulations reveal that there is a permissive excitability window for granule cells that is necessary to support successful behavioural performance. Chemogenetic effects were specific to the targeted dorsal hippocampus, as hippocampal-independent and ventral hippocampal-dependent behaviours remained unaffected. Fos expression demonstrated that chemogenetics can modulate granule cell recruitment via behaviourally relevant inputs. Rather than driving cell activity deterministically or spontaneously, chemogenetic intervention merely modulates the behaviourally permissive activity window in which the circuit operates. We conclude that restoring appropriate principal cell tuning via circuit-based therapies, irrespective of the mechanisms generating the disease-related hyperactivity, is a promising translational approach.
Collapse
Affiliation(s)
- Julia B Kahn
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell G Port
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Cuiyong Yue
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hajime Takano
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas A Coulter
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Research Institute of the Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
265
|
Zhan J, Komal R, Keenan WT, Hattar S, Fernandez DC. Non-invasive Strategies for Chronic Manipulation of DREADD-controlled Neuronal Activity. J Vis Exp 2019. [PMID: 31498301 DOI: 10.3791/59439] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Chemogenetic strategies have emerged as reliable tools for remote control of neuronal activity. Among these, designer receptors exclusively activated by designer drugs (DREADDs) have become the most popular chemogenetic approach used in modern neuroscience. Most studies deliver the ligand clozapine-N-oxide (CNO) using a single intraperitoneal injection, which is suitable for the acute activation/inhibition of the targeted neuronal population. There are, however, only a few examples of strategies for chronic modulation of DREADD-controlled neurons, the majority of which rely on the use of delivery systems that require surgical intervention. Here, we expand on two non-invasive strategies for delivering the ligand CNO to chronically manipulate neural population in mice. CNO was administered either by using repetitive (daily) eye-drops, or chronically through the animal's drinking water. These non-invasive paradigms result in robust activation of the designer receptors that persisted throughout the CNO treatments. The methods described here offer alternatives for the chronic DREADD-mediated control of neuronal activity and may be useful for experiments designed to evaluate behavior in freely moving animals, focusing on less-invasive CNO delivery methods.
Collapse
Affiliation(s)
- Jesse Zhan
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health
| | - Ruchi Komal
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health
| | - William T Keenan
- Howard Hughes Medical Institute, Department of Neuroscience, Scripps Research Institute
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health
| | - Diego C Fernandez
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health;
| |
Collapse
|
266
|
DePoy LM, Shapiro LP, Kietzman HW, Roman KM, Gourley SL. β1-Integrins in the Developing Orbitofrontal Cortex Are Necessary for Expectancy Updating in Mice. J Neurosci 2019; 39:6644-6655. [PMID: 31253753 PMCID: PMC6703883 DOI: 10.1523/jneurosci.3072-18.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/11/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Navigating a changing environment requires associating stimuli and actions with their likely outcomes and modifying these associations when they change. These processes involve the orbitofrontal cortex (OFC). Although some molecular mediators have been identified, developmental factors are virtually unknown. We hypothesized that the cell adhesion factor β1-integrin is essential to OFC function, anticipating developmental windows during which β1-integrins might be more influential than others. We discovered that OFC-selective β1-integrin silencing before adolescence, but not later, impaired the ability of mice to extinguish conditioned fear and select actions based on their likely outcomes. Early-life knock-down also reduced the densities of dendritic spines, the primary sites of excitatory plasticity in the brain, and weakened sensitivity to cortical inputs. Notwithstanding these defects in male mice, females were resilient to OFC (but not hippocampal) β1-integrin loss. Existing literature suggests that resilience may be explained by estradiol-mediated transactivation of β1-integrins and tropomyosin receptor kinase B (trkB). Accordingly, we discovered that a trkB agonist administered during adolescence corrected reward-related decision making in β1-integrin-deficient males. In sum, developmental β1-integrins are indispensable for OFC function later in life.SIGNIFICANCE STATEMENT The orbitofrontal cortex (OFC) is a subregion of the frontal cortex that allows organisms to link behaviors and stimuli with anticipated outcomes, and to make predictions about the consequences of one's behavior. Aspects of OFC development are particularly prolonged, extending well into adolescence, likely optimizing organisms' abilities to prospectively calculate the consequences of their actions and select behaviors appropriately; these decision making strategies improve as young individuals mature into adulthood. Molecular factors are not, however, well understood. Our experiments reveal that a cell adhesion protein termed "β1-integrin" is necessary for OFC neuronal maturation and function. Importantly, β1-integrins operate during a critical period equivalent to early adolescence in humans to optimize the ability of organisms to update expectancies later in life.
Collapse
Affiliation(s)
- Lauren M DePoy
- Department of Pediatrics
- Department of Psychiatry
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience, and
| | - Lauren P Shapiro
- Department of Pediatrics
- Department of Psychiatry
- Yerkes National Primate Research Center
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| | - Henry W Kietzman
- Department of Pediatrics
- Department of Psychiatry
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience, and
| | - Kaitlyn M Roman
- Department of Pediatrics
- Department of Psychiatry
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience, and
| | - Shannon L Gourley
- Department of Pediatrics,
- Department of Psychiatry
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience, and
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| |
Collapse
|
267
|
Wang D, Stoveken HM, Zucca S, Dao M, Orlandi C, Song C, Masuho I, Johnston C, Opperman KJ, Giles AC, Gill MS, Lundquist EA, Grill B, Martemyanov KA. Genetic behavioral screen identifies an orphan anti-opioid system. Science 2019; 365:1267-1273. [PMID: 31416932 DOI: 10.1126/science.aau2078] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 02/22/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
Opioids target the μ-opioid receptor (MOR) to produce unrivaled pain management, but their addictive properties can lead to severe abuse. We developed a whole-animal behavioral platform for unbiased discovery of genes influencing opioid responsiveness. Using forward genetics in Caenorhabditis elegans, we identified a conserved orphan receptor, GPR139, with anti-opioid activity. GPR139 is coexpressed with MOR in opioid-sensitive brain circuits, binds to MOR, and inhibits signaling to heterotrimeric guanine nucleotide-binding proteins (G proteins). Deletion of GPR139 in mice enhanced opioid-induced inhibition of neuronal firing to modulate morphine-induced analgesia, reward, and withdrawal. Thus, GPR139 could be a useful target for increasing opioid safety. These results also demonstrate the potential of C. elegans as a scalable platform for genetic discovery of G protein-coupled receptor signaling principles.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Hannah M Stoveken
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Stefano Zucca
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Maria Dao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Caitlin Johnston
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Karla J Opperman
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Andrew C Giles
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Matthew S Gill
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Erik A Lundquist
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Brock Grill
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
268
|
V2a Neurons Constrain Extradiaphragmatic Respiratory Muscle Activity at Rest. eNeuro 2019; 6:ENEURO.0492-18.2019. [PMID: 31324674 PMCID: PMC6709210 DOI: 10.1523/eneuro.0492-18.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/28/2019] [Accepted: 06/17/2019] [Indexed: 02/01/2023] Open
Abstract
Breathing requires precise control of respiratory muscles to ensure adequate ventilation. Neurons within discrete regions of the brainstem produce oscillatory activity to control the frequency of breathing. Less is understood about how spinal and pontomedullary networks modulate the activity of respiratory motor neurons to produce different patterns of activity during different behaviors (i.e., during exercise, coughing, swallowing, vocalizing, or at rest) or following disease or injury. Here, we use a chemogenetic approach to inhibit the activity of glutamatergic V2a neurons in the brainstem and spinal cord of neonatal and adult mice to assess their potential roles in respiratory rhythm generation and patterning respiratory muscle activity. Using whole-body plethysmography (WBP), we show that V2a neuron function is required in neonatal mice to maintain the frequency and regularity of respiratory rhythm. However, silencing V2a neurons in adult mice increases respiratory frequency and ventilation, without affecting regularity. Thus, the excitatory drive provided by V2a neurons is less critical for respiratory rhythm generation in adult compared to neonatal mice. In addition, we used simultaneous EMG recordings of the diaphragm and extradiaphragmatic respiratory muscles in conscious adult mice to examine the role of V2a neurons in patterning respiratory muscle activity. We find that silencing V2a neurons activates extradiaphragmatic respiratory muscles at rest, when they are normally inactive, with little impact on diaphragm activity. Thus, our results indicate that V2a neurons participate in a circuit that serves to constrain the activity of extradiaphragmatic respiratory muscles so that they are active only when needed.
Collapse
|
269
|
Grund T, Tang Y, Benusiglio D, Althammer F, Probst S, Oppenländer L, Neumann ID, Grinevich V. Chemogenetic activation of oxytocin neurons: Temporal dynamics, hormonal release, and behavioral consequences. Psychoneuroendocrinology 2019; 106:77-84. [PMID: 30954921 DOI: 10.1016/j.psyneuen.2019.03.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/15/2019] [Accepted: 03/20/2019] [Indexed: 01/17/2023]
Abstract
Chemogenetics provides cell type-specific remote control of neuronal activity. Here, we describe the application of chemogenetics used to specifically activate oxytocin (OT) neurons as representatives of a unique class of neuroendocrine cells. We injected recombinant adeno-associated vectors, driving the stimulatory subunit hM3Dq of a modified human muscarinic receptor into the rat hypothalamus to achieve cell type-specific expression in OT neurons. As chemogenetic activation of OT neurons has not been reported, we provide systematic analysis of the temporal dynamics of OT neuronal responses in vivo by monitoring calcium fluctuations in OT neurons, and intracerebral as well as peripheral release of OT. We further provide evidence for the efficiency of chemogenetic manipulation at behavioral levels, demonstrating that evoked activation of OT neurons leads to social motivation and anxiolysis. Altogether, our results will be profitable for researchers working on the physiology of neuroendocrine systems, peptidergic modulation of behaviors and translational psychiatry.
Collapse
Affiliation(s)
- Thomas Grund
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040 Regensburg, Germany
| | - Yan Tang
- Division of Neuropeptides (V078), German Cancer Research Center and Central Institute of Mental Health, University of Heidelberg, 69120 Heidelberg, Germany
| | - Diego Benusiglio
- Division of Neuropeptides (V078), German Cancer Research Center and Central Institute of Mental Health, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ferdinand Althammer
- Division of Neuropeptides (V078), German Cancer Research Center and Central Institute of Mental Health, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sophia Probst
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040 Regensburg, Germany
| | - Lena Oppenländer
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040 Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040 Regensburg, Germany.
| | - Valery Grinevich
- Division of Neuropeptides (V078), German Cancer Research Center and Central Institute of Mental Health, University of Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
270
|
Donthamsetti PC, Broichhagen J, Vyklicky V, Stanley C, Fu Z, Visel M, Levitz JL, Javitch JA, Trauner D, Isacoff EY. Genetically Targeted Optical Control of an Endogenous G Protein-Coupled Receptor. J Am Chem Soc 2019; 141:11522-11530. [PMID: 31291105 PMCID: PMC7271769 DOI: 10.1021/jacs.9b02895] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that play important roles in biology. However, our understanding of their function in complex living systems is limited because we lack tools that can target individual receptors with sufficient precision. State-of-the-art approaches, including DREADDs, optoXRs, and PORTL gated-receptors, control GPCR signaling with molecular, cell type, and temporal specificity. Nonetheless, these tools are based on engineered non-native proteins that may (i) express at nonphysiological levels, (ii) localize and turnover incorrectly, and/or (iii) fail to interact with endogenous partners. Alternatively, membrane-anchored ligands (t-toxins, DARTs) target endogenous receptors with molecular and cell type specificity but cannot be turned on and off. In this study, we used a combination of chemistry, biology, and light to control endogenous metabotropic glutamate receptor 2 (mGluR2), a Family C GPCR, in primary cortical neurons. mGluR2 was rapidly, reversibly, and selectively activated with photoswitchable glutamate tethered to a genetically targeted-plasma membrane anchor (membrane anchored Photoswitchable Orthogonal Remotely Tethered Ligand; maPORTL). Photoactivation was tuned by adjusting the length of the PORTL as well as the expression level and geometry of the membrane anchor. Our findings provide a template for controlling endogenous GPCRs with cell type specificity and high spatiotemporal precision.
Collapse
Affiliation(s)
- Prashant C. Donthamsetti
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Johannes Broichhagen
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Vojtech Vyklicky
- 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
| | - Zhu Fu
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Meike Visel
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Joshua L. Levitz
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10024, United States
| | - Jonathan A. Javitch
- Departments of Psychiatry & Pharmacology, Columbia University, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Dirk Trauner
- Department of Chemistry, New York University, New York, New York 10003, 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
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
271
|
Drozd US, Shaburova EV, Dygalo NN. Genetic approaches to the investigation of serotonergic neuron functions in animals. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The serotonergic system is one of the most important neurotransmitter systems that take part in the regulation of vital CNS functions. The understanding of its mechanisms will help scientists create new therapeutic approaches to the treatment of mental and neurodegenerative diseases and find out how this neurotransmitter system interacts with other parts of the brain and regulates their activity. Since the serotonergic system anatomy and functionality are heterogeneous and complex, the best tools for studying them are based on manipulation of individual types of neurons without affecting neurons of other neurotransmitter systems. The selective cell control is possible due to the genetic determinism of their functions. Proteins that determine the uniqueness of the cell type are expressed under the regulation of cell-specific promoters. By using promoters that are specific for genes of the serotonin system, one can control the expression of a gene of interest in serotonergic neurons. Here we review approaches based on such promoters. The genetic models to be discussed in the article have already shed the light on the role of the serotonergic system in modulating behavior and processing sensory information. In particular, genetic knockouts of serotonin genes sert, pet1, and tph2 promoted the determination of their contribution to the development and functioning of the brain. In addition, the review describes inducible models that allow gene expression to be controlled at various developmental stages. Finally, the application of these genetic approaches in optogenetics and chemogenetics provided a new resource for studying the functions, discharge activity, and signal transduction of serotonergic neurons. Nevertheless, the advantages and limitations of the discussed genetic approaches should be taken into consideration in the course of creating models of pathological conditions and developing pharmacological treatments for their correction.
Collapse
Affiliation(s)
- U. S. Drozd
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| | - E. V. Shaburova
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| | - N. N. Dygalo
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| |
Collapse
|
272
|
Ciriachi C, Svane‐Petersen D, Rickhag M. Genetic tools to study complexity of striatal function. J Neurosci Res 2019; 97:1181-1193. [DOI: 10.1002/jnr.24479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 01/11/2023]
Affiliation(s)
- Chiara Ciriachi
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - David Svane‐Petersen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
273
|
Biane JS, Takashima Y, Scanziani M, Conner JM, Tuszynski MH. Reorganization of Recurrent Layer 5 Corticospinal Networks Following Adult Motor Training. J Neurosci 2019; 39:4684-4693. [PMID: 30948479 PMCID: PMC6561695 DOI: 10.1523/jneurosci.3442-17.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 01/27/2023] Open
Abstract
Recurrent synaptic connections between neighboring neurons are a key feature of mammalian cortex, accounting for the vast majority of cortical inputs. Although computational models indicate that reorganization of recurrent connectivity is a primary driver of experience-dependent cortical tuning, the true biological features of recurrent network plasticity are not well identified. Indeed, whether rewiring of connections between cortical neurons occurs during behavioral training, as is widely predicted, remains unknown. Here, we probe M1 recurrent circuits following motor training in adult male rats and find robust synaptic reorganization among functionally related layer 5 neurons, resulting in a 2.5-fold increase in recurrent connection probability. This reorganization is specific to the neuronal subpopulation most relevant for executing the trained motor skill, and behavioral performance was impaired following targeted molecular inhibition of this subpopulation. In contrast, recurrent connectivity is unaffected among neighboring layer 5 neurons largely unrelated to the trained behavior. Training-related corticospinal cells also express increased excitability following training. These findings establish the presence of selective modifications in recurrent cortical networks in adulthood following training.SIGNIFICANCE STATEMENT Recurrent synaptic connections between neighboring neurons are characteristic of cortical architecture, and modifications to these circuits are thought to underlie in part learning in the adult brain. We now show that there are robust changes in recurrent connections in the rat motor cortex upon training on a novel motor task. Motor training results in a 2.5-fold increase in recurrent connectivity, but only within the neuronal subpopulation most relevant for executing the new motor behavior; recurrent connectivity is unaffected among adjoining neurons that do not execute the trained behavior. These findings demonstrate selective reorganization of recurrent synaptic connections in the adult neocortex following novel motor experience, and illuminate fundamental properties of cortical function and plasticity.
Collapse
Affiliation(s)
| | | | - Massimo Scanziani
- Neurobiology, University of California, San Diego, California 92093
- Howard Hughes Medical Institute, San Diego, California, 92093, and
| | | | - Mark H Tuszynski
- Departments of Neurosciences,
- Veterans Administration Medical Center, San Diego, California 92161
| |
Collapse
|
274
|
Qin L, Ma K, Yan Z. Chemogenetic Activation of Prefrontal Cortex in Shank3-Deficient Mice Ameliorates Social Deficits, NMDAR Hypofunction, and Sgk2 Downregulation. iScience 2019; 17:24-35. [PMID: 31247448 PMCID: PMC6599088 DOI: 10.1016/j.isci.2019.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/21/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Haploinsufficiency of the SHANK3 gene is causally linked to autism spectrum disorders (ASDs) in human genetic studies. Here we found that chemogenetic activation of pyramidal neurons in the prefrontal cortex (PFC) of Shank3-deficient mice with the hM3D (Gq) DREADD restored social preference behaviors and elevated glutamatergic synaptic function in PFC. Moreover, the expression of Sgk2 (serum- and glucocorticoid-inducible kinase 2), a member of the Sgk family, which plays a key role in regulating the membrane trafficking of glutamate receptors, was diminished by Shank3 deficiency and rescued by Gq DREADD activation of PFC. Blocking Sgk function in Shank3-deficient mice prevented Gq DREADD from rescuing social and synaptic deficits, whereas blocking Sgk function in wild-type mice led to the attenuation of PFC glutamatergic signaling and the induction of autism-like social deficits. These results have provided a potential circuit intervention and molecular target for autism treatment.
Collapse
Affiliation(s)
- Luye Qin
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Kaijie Ma
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA.
| |
Collapse
|
275
|
Fournier DI, Todd TP, Bucci DJ. Permanent damage or temporary silencing of retrosplenial cortex impairs the expression of a negative patterning discrimination. Neurobiol Learn Mem 2019; 163:107033. [PMID: 31173918 DOI: 10.1016/j.nlm.2019.107033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/18/2022]
Abstract
The retrosplenial cortex (RSC) is positioned at the interface between cortical sensory regions and the hippocampal/parahippocampal memory system. As such, it has been theorized that RSC may have a fundamental role in linking sensory stimuli together in the service of forming complex representations. To test this, three experiments were carried out to determine the effects of RSC damage or temporary inactivation on learning or performing a negative patterning discrimination. In this procedure, two conditioned stimuli are reinforced when they are presented individually (i.e., stimulus elements) but are non-reinforced when they are presented simultaneously as a compound stimulus. Normal rats successfully discriminate between the two types of trials as evidenced by more responding to the elements compared to the compound stimulus. This is thought to reflect the formation of a configural representation of the compound stimulus; that is, the two cues are linked together in such a fashion that the compound stimulus is a wholly different, unique stimulus. Permanent lesions of RSC made prior to training (Experiment 1) had no effect on learning the discrimination. However, lesions (Experiment 2) or temporary chemogenetic inactivation (Experiment 3) of RSC made after training impaired subsequent performance of the discrimination. We argue that this pattern of results indicates that RSC may normally be involved in forming the configural representations manifested in negative patterning, but that absent the RSC, other brain systems or structures can compensate sufficiently to result in normal behavior.
Collapse
Affiliation(s)
- Danielle I Fournier
- Program in Experimental and Molecular Medicine, Geisel School of Medicine at Dartmouth, USA
| | - Travis P Todd
- Department of Psychological and Brain Sciences, Dartmouth College, USA
| | - David J Bucci
- Program in Experimental and Molecular Medicine, Geisel School of Medicine at Dartmouth, USA; Department of Psychological and Brain Sciences, Dartmouth College, USA.
| |
Collapse
|
276
|
Whyte AJ, Kietzman HW, Swanson AM, Butkovich LM, Barbee BR, Bassell GJ, Gross C, Gourley SL. Reward-Related Expectations Trigger Dendritic Spine Plasticity in the Mouse Ventrolateral Orbitofrontal Cortex. J Neurosci 2019; 39:4595-4605. [PMID: 30940719 PMCID: PMC6554633 DOI: 10.1523/jneurosci.2031-18.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
An essential aspect of goal-directed decision-making is selecting actions based on anticipated consequences, a process that involves the orbitofrontal cortex (OFC) and potentially, the plasticity of dendritic spines in this region. To investigate this possibility, we trained male and female mice to nose poke for food reinforcers, or we delivered the same number of food reinforcers non-contingently to separate mice. We then decreased the likelihood of reinforcement for trained mice, requiring them to modify action-outcome expectations. In a separate experiment, we blocked action-outcome updating via chemogenetic inactivation of the OFC. In both cases, successfully selecting actions based on their likely consequences was associated with fewer immature, thin-shaped dendritic spines and a greater proportion of mature, mushroom-shaped spines in the ventrolateral OFC. This pattern was distinct from spine loss associated with aging, and we identified no effects on hippocampal CA1 neurons. Given that the OFC is involved in prospective calculations of likely outcomes, even when they are not observable, constraining spinogenesis while preserving mature spines may be important for solidifying durable expectations. To investigate causal relationships, we inhibited the RNA-binding protein fragile X mental retardation protein (encoded by Fmr1), which constrains dendritic spine turnover. Ventrolateral OFC-selective Fmr1 knockdown recapitulated the behavioral effects of inducible OFC inactivation (and lesions; also shown here), impairing action-outcome conditioning, and caused dendritic spine excess. Our findings suggest that a proper balance of dendritic spine plasticity within the OFC is necessary for one's ability to select actions based on anticipated consequences.SIGNIFICANCE STATEMENT Navigating a changing environment requires associating actions with their likely outcomes and updating these associations when they change. Dendritic spine plasticity is likely involved, yet relationships are unconfirmed. Using behavioral, chemogenetic, and viral-mediated gene silencing strategies and high-resolution microscopy, we find that modifying action-outcome expectations is associated with fewer immature spines and a greater proportion of mature spines in the ventrolateral orbitofrontal cortex (OFC). Given that the OFC is involved in prospectively calculating the likely outcomes of one's behavior, even when they are not observable, constraining spinogenesis while preserving mature spines may be important for maintaining durable expectations.
Collapse
Affiliation(s)
- Alonzo J Whyte
- Departments of Cell Biology
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
| | - Henry W Kietzman
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
| | - Andrew M Swanson
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
| | - Laura M Butkovich
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
| | - Britton R Barbee
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| | - Gary J Bassell
- Departments of Cell Biology
- Graduate Program in Neuroscience
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, and
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio 45267
| | - Shannon L Gourley
- Pediatrics, Emory School of Medicine,
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| |
Collapse
|
277
|
Denaxa M, Neves G, Rabinowitz A, Kemlo S, Liodis P, Burrone J, Pachnis V. Modulation of Apoptosis Controls Inhibitory Interneuron Number in the Cortex. Cell Rep 2019; 22:1710-1721. [PMID: 29444425 PMCID: PMC6230259 DOI: 10.1016/j.celrep.2018.01.064] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/18/2017] [Accepted: 01/22/2018] [Indexed: 12/20/2022] Open
Abstract
Cortical networks are composed of excitatory projection neurons and inhibitory interneurons. Finding the right balance between the two is important for controlling overall cortical excitation and network dynamics. However, it is unclear how the correct number of cortical interneurons (CIs) is established in the mammalian forebrain. CIs are generated in excess from basal forebrain progenitors, and their final numbers are adjusted via an intrinsically determined program of apoptosis that takes place during an early postnatal window. Here, we provide evidence that the extent of CI apoptosis during this critical period is plastic and cell-type specific and can be reduced in a cell-autonomous manner by acute increases in neuronal activity. We propose that the physiological state of the emerging neural network controls the activity levels of local CIs to modulate their numbers in a homeostatic manner. Lhx6 is required for survival of CIs generated in the MGE MGE-derived CI loss is compensated for by a decrease in CGE-derived interneuron apoptosis Increases in cortical network activity are correlated with improved CI survival Transient, cell-autonomous depolarization improves the survival of grafted CIs
Collapse
Affiliation(s)
- Myrto Denaxa
- Nervous System Development and Homeostasis Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Guilherme Neves
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.
| | - Adam Rabinowitz
- Bioinformatics and Biostatistics Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sarah Kemlo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Petros Liodis
- Molecular Neurobiology, National Institute for Medical Research, the Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.
| | - Vassilis Pachnis
- Nervous System Development and Homeostasis Laboratory, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
278
|
Bone DBJ, Meister J, Knudsen JR, Dattaroy D, Cohen A, Lee R, Lu H, Metzger D, Jensen TE, Wess J. Skeletal Muscle-Specific Activation of G q Signaling Maintains Glucose Homeostasis. Diabetes 2019; 68:1341-1352. [PMID: 30936140 PMCID: PMC6610017 DOI: 10.2337/db18-0796] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 03/22/2019] [Indexed: 12/19/2022]
Abstract
Skeletal muscle (SKM) insulin resistance plays a central role in the pathogenesis of type 2 diabetes. Because G-protein-coupled receptors (GPCRs) represent excellent drug targets, we hypothesized that activation of specific functional classes of SKM GPCRs might lead to improved glucose homeostasis in type 2 diabetes. At present, little is known about the in vivo metabolic roles of the various distinct GPCR signaling pathways operative in SKM. In this study, we tested the hypothesis that selective activation of SKM Gq signaling can improve SKM glucose uptake and whole-body glucose homeostasis under physiological and pathophysiological conditions. Studies with transgenic mice expressing a Gq-linked designer GPCR selectively in SKM cells demonstrated that receptor-mediated activation of SKM Gq signaling greatly promoted glucose uptake into SKM and significantly improved glucose homeostasis in obese, glucose-intolerant mice. These beneficial metabolic effects required the activity of SKM AMPK. In contrast, obese mutant mice that lacked both Gαq and Gα11 selectively in SKM showed severe deficits in glucose homeostasis. Moreover, GPCR-mediated activation of Gq signaling also stimulated glucose uptake in primary human SKM cells. Taken together, these findings strongly suggest that agents capable of enhancing SKM Gq signaling may prove useful as novel antidiabetic drugs.
Collapse
Affiliation(s)
- Derek B J Bone
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Jonas R Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Diptadip Dattaroy
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Amanda Cohen
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Regina Lee
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Huiyan Lu
- Mouse Transgenic Core Facility, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U1258, Université de Strasbourg, Illkirch, France
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| |
Collapse
|
279
|
Inoue A, Raimondi F, Kadji FMN, Singh G, Kishi T, Uwamizu A, Ono Y, Shinjo Y, Ishida S, Arang N, Kawakami K, Gutkind JS, Aoki J, Russell RB. Illuminating G-Protein-Coupling Selectivity of GPCRs. Cell 2019; 177:1933-1947.e25. [PMID: 31160049 DOI: 10.1016/j.cell.2019.04.044] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/28/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022]
Abstract
Heterotrimetic G proteins consist of four subfamilies (Gs, Gi/o, Gq/11, and G12/13) that mediate signaling via G-protein-coupled receptors (GPCRs), principally by receptors binding Gα C termini. G-protein-coupling profiles govern GPCR-induced cellular responses, yet receptor sequence selectivity determinants remain elusive. Here, we systematically quantified ligand-induced interactions between 148 GPCRs and all 11 unique Gα subunit C termini. For each receptor, we probed chimeric Gα subunit activation via a transforming growth factor-α (TGF-α) shedding response in HEK293 cells lacking endogenous Gq/11 and G12/13 proteins, and complemented G-protein-coupling profiles through a NanoBiT-G-protein dissociation assay. Interrogation of the dataset identified sequence-based coupling specificity features, inside and outside the transmembrane domain, which we used to develop a coupling predictor that outperforms previous methods. We used the predictor to engineer designer GPCRs selectively coupled to G12. This dataset of fine-tuned signaling mechanisms for diverse GPCRs is a valuable resource for research in GPCR signaling.
Collapse
Affiliation(s)
- Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan; Advanced Research & Development Programs for Medical Innovation (PRIME), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo 100-0004, Japan; Advanced Research & Development Programs for Medical Innovation (LEAP), AMED, Chiyoda-ku, Tokyo 100-0004, Japan.
| | - Francesco Raimondi
- CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany; Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany.
| | | | - Gurdeep Singh
- CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany; Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Takayuki Kishi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Akiharu Uwamizu
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Yuki Ono
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Yuji Shinjo
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Satoru Ishida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Nadia Arang
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan; Advanced Research & Development Programs for Medical Innovation (LEAP), AMED, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Robert B Russell
- CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany; Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany.
| |
Collapse
|
280
|
Wang Y, Chen Z. An update for epilepsy research and antiepileptic drug development: Toward precise circuit therapy. Pharmacol Ther 2019; 201:77-93. [PMID: 31128154 DOI: 10.1016/j.pharmthera.2019.05.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Epilepsy involves neuronal dysfunction at molecular, cellular, and circuit levels. The understanding of the mechanism of the epilepsies has advanced greatly in the last three decades, especially in terms of their cellular and molecular basis. However, despite the availability of ~30 anti-epileptic drugs (AEDs) with diverse molecular targets, there are still many challenges (e.g. drug resistance, side effects) in pharmacological treatment of epilepsies today. Because molecular mechanisms are integrated at the level of neuronal circuits, we suggest a shift in epilepsy treatment and research strategies from the "molecular" level to the "circuit" level. Recent technological advances have facilitated circuit mechanistic discovery at each level and have paved the way for many opportunities of novel therapeutic strategies and AED development toward precise circuit therapy.
Collapse
Affiliation(s)
- Yi Wang
- Institute of Pharmacology and Toxicology, Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
281
|
Feng J, Zhang C, Lischinsky JE, Jing M, Zhou J, Wang H, Zhang Y, Dong A, Wu Z, Wu H, Chen W, Zhang P, Zou J, Hires SA, Zhu JJ, Cui G, Lin D, Du J, Li Y. A Genetically Encoded Fluorescent Sensor for Rapid and Specific In Vivo Detection of Norepinephrine. Neuron 2019; 102:745-761.e8. [PMID: 30922875 PMCID: PMC6533151 DOI: 10.1016/j.neuron.2019.02.037] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/28/2019] [Accepted: 02/21/2019] [Indexed: 12/22/2022]
Abstract
Norepinephrine (NE) is a key biogenic monoamine neurotransmitter involved in a wide range of physiological processes. However, its precise dynamics and regulation remain poorly characterized, in part due to limitations of available techniques for measuring NE in vivo. Here, we developed a family of GPCR activation-based NE (GRABNE) sensors with a 230% peak ΔF/F0 response to NE, good photostability, nanomolar-to-micromolar sensitivities, sub-second kinetics, and high specificity. Viral- or transgenic-mediated expression of GRABNE sensors was able to detect electrical-stimulation-evoked NE release in the locus coeruleus (LC) of mouse brain slices, looming-evoked NE release in the midbrain of live zebrafish, as well as optogenetically and behaviorally triggered NE release in the LC and hypothalamus of freely moving mice. Thus, GRABNE sensors are robust tools for rapid and specific monitoring of in vivo NE transmission in both physiological and pathological processes.
Collapse
Affiliation(s)
- Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Changmei Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Julieta E Lischinsky
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Miao Jing
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Chinese Institute for Brain Research, Beijing 100871, China
| | - Jingheng Zhou
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Yajun Zhang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Ao Dong
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Hao Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weiyu Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Zhang
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jing Zou
- Department of Biological Sciences, Neurobiology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - S Andrew Hires
- Department of Biological Sciences, Neurobiology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - J Julius Zhu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; School of Medicine, Ningbo University, Ningbo 315010, China; Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, 6525 Nijmegen, the Netherlands; Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Dayu Lin
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10016, USA
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Chinese Institute for Brain Research, Beijing 100871, China.
| |
Collapse
|
282
|
Zhang CQ, McMahon B, Dong H, Warner T, Shen W, Gallagher M, Macdonald RL, Kang JQ. Molecular basis for and chemogenetic modulation of comorbidities in GABRG2-deficient epilepsies. Epilepsia 2019; 60:1137-1149. [PMID: 31087664 DOI: 10.1111/epi.15160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE γ-Aminobutyric acid type A (GABAA ) receptor subunit gene mutations are significant causes of epilepsy, which are often accompanied by various neuropsychiatric comorbidities, but the underlying mechanisms are unclear. It has been suggested that the comorbidities are caused by seizures, as the comorbidities often present in severe epilepsy syndromes. However, findings from both humans and animal models argue against this conclusion. Mutations in the GABAA receptor γ2 subunit gene GABRG2 have been associated with anxiety alone or with severe epilepsy syndromes and comorbid anxiety, suggesting that a core molecular defect gives rise to the phenotypic spectrum. Here, we determined the pathophysiology of comorbid anxiety in GABRG2 loss-of-function epilepsy syndromes, identified the central nucleus of the amygdala (CeA) as a primary site for epilepsy comorbid anxiety, and demonstrated a potential rescue of comorbid anxiety via neuromodulation of CeA neurons. METHODS We used brain slice recordings, subcellular fractionation with Western blot, immunohistochemistry, confocal microscopy, and a battery of behavior tests in combination with a chemogenetic approach to characterize anxiety and its underlying mechanisms in a Gabrg2+/Q390X knockin mouse and a Gabrg2+/- knockout mouse, each associated with a different epilepsy syndrome. RESULTS We found that impaired GABAergic neurotransmission in CeA underlies anxiety in epilepsy, which is due to reduced GABAA receptor subunit expression resulting from the mutations. Impaired GABAA receptor expression reduced GABAergic neurotransmission in CeA, but not in basolateral amygdala. Activation or inactivation of inhibitory neurons using a chemogenetic approach in CeA alone modulated anxietylike behaviors. Similarly, pharmacological enhancement of GABAergic signaling via γ2 subunit-containing receptors relieved the anxiety. SIGNIFICANCE Together, these data demonstrate the molecular basis for a comorbidity of epilepsy, anxiety, and suggest that impaired GABAA receptor function in CeA due to a loss-of-function mutation could at least contribute to anxiety. Modulation of CeA neurons could cause or suppress anxiety, suggesting a potential use of CeA neurons as therapeutic targets for treatment of anxiety in addition to traditional pharmacological approaches.
Collapse
Affiliation(s)
- Chun-Qing Zhang
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Neurosurgery, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| | - Bryan McMahon
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Huancheng Dong
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy Warner
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wangzhen Shen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Martin Gallagher
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert L Macdonald
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jing-Qiong Kang
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
283
|
Lieb A, Weston M, Kullmann DM. Designer receptor technology for the treatment of epilepsy. EBioMedicine 2019; 43:641-649. [PMID: 31078519 PMCID: PMC6558262 DOI: 10.1016/j.ebiom.2019.04.059] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Epilepsy remains refractory to medical treatment in ~30% of patients despite decades of new drug development. Neurosurgery to remove or disconnect the seizure focus is often curative but frequently contraindicated by risks of irreversible impairment to brain function. Novel therapies are therefore required that better balance seizure suppression against the risks of side effects. Among experimental gene therapies, chemogenetics has the major advantage that the action on the epileptogenic zone can be modulated on demand. Two broad approaches are to use a designer G-protein-coupled receptor or a modified ligand gated ion channel, targeted to specific neurons in the epileptogenic zone using viral vectors and cell-type selective promoters. The receptor can be activated on demand by either an exogenous compound or by pathological levels of extracellular glutamate that occur in epileptogenic tissue. We review the principal designer receptor technologies and their modes of action. We compare the drawbacks and benefits of each designer receptor with particular focus on the drug activators and the potential for clinical translation in epilepsy.
Collapse
Affiliation(s)
- Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
| | - Mikail Weston
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, UK.
| |
Collapse
|
284
|
Bolognini D, Barki N, Butcher AJ, Hudson BD, Sergeev E, Molloy C, Hodge D, Bradley SJ, Le Gouill C, Bouvier M, Tobin AB, Milligan G. Chemogenetics defines receptor-mediated functions of short chain free fatty acids. Nat Chem Biol 2019; 15:489-498. [PMID: 30992568 DOI: 10.1038/s41589-019-0270-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 03/01/2019] [Indexed: 12/23/2022]
Abstract
Differentiating actions of short chain fatty acids (SCFAs) at free fatty acid receptor 2 (FFA2) from other free fatty acid-responsive receptors and from non-receptor-mediated effects has been challenging. Using a novel chemogenetic and knock-in strategy, whereby an engineered variant of FFA2 (FFA2-DREADD) that is unresponsive to natural SCFAs but is instead activated by sorbic acid replaced the wild-type receptor, we determined that activation of FFA2 in differentiated adipocytes and colonic crypt enteroendocrine cells of mouse accounts fully for SCFA-regulated lipolysis and release of the incretin glucagon-like peptide-1 (GLP-1), respectively. In vivo studies confirmed the specific role of FFA2 in GLP-1 release and also demonstrated a direct role for FFA2 in accelerating gut transit. Thereby, we establish the general principle that such a chemogenetic knock-in strategy can successfully define novel G-protein-coupled receptor (GPCR) biology and provide both target validation and establish therapeutic potential of a 'hard to target' GPCR.
Collapse
Affiliation(s)
- Daniele Bolognini
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Natasja Barki
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Adrian J Butcher
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Eugenia Sergeev
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Colin Molloy
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Daryl Hodge
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - 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, UK
| | - Christian Le Gouill
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montreal, Montreal, Quebec, Canada
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montreal, Montreal, Quebec, Canada
| | - 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, UK.
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
285
|
Temporary inactivation of the medial prefrontal cortex impairs the formation, but not the retrieval of social odor recognition memory in rats. Neurobiol Learn Mem 2019; 161:115-121. [DOI: 10.1016/j.nlm.2019.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/13/2019] [Accepted: 04/02/2019] [Indexed: 11/23/2022]
|
286
|
Cholinergic Neurons of the Medial Septum Are Crucial for Sensorimotor Gating. J Neurosci 2019; 39:5234-5242. [PMID: 31028115 DOI: 10.1523/jneurosci.0950-18.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 03/23/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
Hypofunction of NMDA receptors has been considered a possible cause for the pathophysiology of schizophrenia. More recently, indirect ways to regulate NMDA that would be less disruptive have been proposed and metabotropic glutamate receptor subtype 5 (mGluR5) represents one such candidate. To characterize the cell populations involved, we demonstrated here that knock-out (KO) of mGluR5 in cholinergic, but not glutamatergic or parvalbumin (PV)-positive GABAergic, neurons reduced prepulse inhibition of the startle response (PPI) and enhanced sensitivity to MK801-induced locomotor activity. Inhibition of cholinergic neurons in the medial septum by DREADD (designer receptors exclusively activated by designer drugs) resulted in reduced PPI further demonstrating the importance of these neurons in sensorimotor gating. Volume imaging and quantification were used to compare PV and cholinergic cell distribution, density, and total cell counts in the different cell-type-specific KO lines. Electrophysiological studies showed reduced NMDA receptor-mediated currents in cholinergic neurons of the medial septum in mGluR5 KO mice. These results obtained from male and female mice indicate that cholinergic neurons in the medial septum represent a key cell type involved in sensorimotor gating and are relevant to pathologies associated with disrupted sensorimotor gating such as schizophrenia.SIGNIFICANCE STATEMENT The mechanistic complexity underlying psychiatric disorders remains a major challenge that is hindering the drug discovery process. Here, we generated genetically modified mouse lines to better characterize the involvement of the receptor mGluR5 in the fine-tuning of NMDA receptors, specifically in the context of sensorimotor gating. We evaluated the importance of knocking-out mGluR5 in three different cell types in two brain regions and performed different sets of experiments including behavioral testing and electrophysiological recordings. We demonstrated that cholinergic neurons in the medial septum represent a key cell-type involved in sensorimotor gating. We are proposing that pathologies associated with disrupted sensorimotor gating, such as with schizophrenia, could benefit from further evaluating strategies to modulate specifically cholinergic neurons in the medial septum.
Collapse
|
287
|
Zhu L, Dattaroy D, Pham J, Wang L, Barella LF, Cui Y, Wilkins KJ, Roth BL, Hochgeschwender U, Matschinsky FM, Kaestner KH, Doliba NM, Wess J. Intra-islet glucagon signaling is critical for maintaining glucose homeostasis. JCI Insight 2019; 5:127994. [PMID: 31012868 DOI: 10.1172/jci.insight.127994] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glucagon, a hormone released from pancreatic alpha-cells, plays a key role in maintaining proper glucose homeostasis and has been implicated in the pathophysiology of diabetes. In vitro studies suggest that intra-islet glucagon can modulate the function of pancreatic beta-cells. However, because of the lack of suitable experimental tools, the in vivo physiological role of this intra-islet cross-talk has remained elusive. To address this issue, we generated a novel mouse model that selectively expressed an inhibitory designer G protein-coupled receptor (Gi DREADD) in α-cells only. Drug-induced activation of this inhibitory designer receptor almost completely shut off glucagon secretion in vivo, resulting in significantly impaired insulin secretion, hyperglycemia, and glucose intolerance. Additional studies with mouse and human islets indicated that intra-islet glucagon stimulates insulin release primarily by activating β-cell GLP-1 receptors. These new findings strongly suggest that intra-islet glucagon signaling is essential for maintaining proper glucose homeostasis in vivo. Our work may pave the way toward the development of novel classes of antidiabetic drugs that act by modulating intra-islet cross-talk between α- and β-cells.
Collapse
Affiliation(s)
- Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diptadip Dattaroy
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Jonathan Pham
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Lingdi Wang
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Kenneth J Wilkins
- Biostatistics Program, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Ute Hochgeschwender
- Neuroscience Program and College of Medicine, Central Michigan University, Mt. Pleasant, Michigan, USA
| | - Franz M Matschinsky
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Klaus H Kaestner
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicolai M Doliba
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
288
|
Runegaard AH, Fitzpatrick CM, Woldbye DPD, Andreasen JT, Sørensen AT, Gether U. Modulating Dopamine Signaling and Behavior with Chemogenetics: Concepts, Progress, and Challenges. Pharmacol Rev 2019; 71:123-156. [PMID: 30814274 DOI: 10.1124/pr.117.013995] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
For more than 60 years, dopamine (DA) has been known as a critical modulatory neurotransmitter regulating locomotion, reward-based motivation, and endocrine functions. Disturbances in DA signaling have been linked to an array of different neurologic and psychiatric disorders, including Parkinson's disease, schizophrenia, and addiction, but the underlying pathologic mechanisms have never been fully elucidated. One major obstacle limiting interpretation of standard pharmacological and transgenic interventions is the complexity of the DA system, which only appears to widen as research progresses. Nonetheless, development of new genetic tools, such as chemogenetics, has led to an entirely new era for functional studies of neuronal signaling. By exploiting receptors that are engineered to respond selectively to an otherwise inert ligand, so-called Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), chemogenetics enables pharmacological remote control of neuronal activity. Here we review the recent, extensive application of this technique to the DA field and how its use has advanced the study of the DA system and contributed to our general understanding of DA signaling and related behaviors. Moreover, we discuss the challenges and pitfalls associated with the chemogenetic technology, such as the metabolism of the DREADD ligand clozapine N-oxide (CNO) to the D2 receptor antagonist clozapine. We conclude that despite the recent concerns regarding CNO, the chemogenetic toolbox provides an exceptional approach to study neuronal function. The huge potential should promote continued investigations and additional refinements to further expound key mechanisms of DA signaling and circuitries in normal as well as maladaptive behaviors.
Collapse
Affiliation(s)
- Annika Højrup Runegaard
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience (A.H.R., D.P.D.W., A.T.S., U.G.) and Department of Drug Design and Pharmacology (C.M.F., J.T.A.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ciarán Martin Fitzpatrick
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience (A.H.R., D.P.D.W., A.T.S., U.G.) and Department of Drug Design and Pharmacology (C.M.F., J.T.A.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Paul Drucker Woldbye
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience (A.H.R., D.P.D.W., A.T.S., U.G.) and Department of Drug Design and Pharmacology (C.M.F., J.T.A.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Tobias Andreasen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience (A.H.R., D.P.D.W., A.T.S., U.G.) and Department of Drug Design and Pharmacology (C.M.F., J.T.A.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Toft Sørensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience (A.H.R., D.P.D.W., A.T.S., U.G.) and Department of Drug Design and Pharmacology (C.M.F., J.T.A.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience (A.H.R., D.P.D.W., A.T.S., U.G.) and Department of Drug Design and Pharmacology (C.M.F., J.T.A.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
289
|
Abstract
A great deal of experimental evidence suggests that ligands can stabilize different receptor active states that go on to interact with cellular signaling proteins to form a range of different complexes in varying quantities. In pleiotropically linked receptor systems, this leads to selective activation of some signaling pathways at the expense of others (biased signaling). This article summarizes the current knowledge about the complex components of receptor systems, the evidence that biased signaling is used in natural physiology to fine-tune signaling, and the current thoughts on how this mechanism may be applied to the design of better drugs. Although this is a fairly newly discovered phenomenon, theoretical and experimental data suggest that it is a ubiquitous behavior of ligands and receptors and to be expected. Biased signaling is simple to detect in vitro and there are numerous methods to quantify the effect with scales that can be used to optimize this activity in structure-activity medicinal chemistry studies. At present, the major hurdle in the application of this mechanism to therapeutics is the translation of in vitro bias to in vivo effect; this is because of the numerous factors that can modify measures of bias in natural physiologic systems. In spite of this, biased signaling still has the potential to justify revisiting of receptor targets previously thought to be intractable and also furnishes the means to pursue targets previously thought to be forbidden due to deleterious physiology (as these may be eliminated through biased signaling).
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
290
|
Pisa R, Cupido T, Kapoor TM. Designing Allele-Specific Inhibitors of Spastin, a Microtubule-Severing AAA Protein. J Am Chem Soc 2019; 141:5602-5606. [PMID: 30875216 DOI: 10.1021/jacs.8b13257] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The bump-hole approach is a powerful chemical biology strategy to specifically probe the functions of closely related proteins. However, for many protein families, such as the ATPases associated with diverse cellular activities (AAA), we lack structural data for inhibitor-protein complexes to design allele-specific chemical probes. Here we report the X-ray structure of a pyrazolylaminoquinazoline-based inhibitor bound to spastin, a microtubule-severing AAA protein, and characterize the residues involved in inhibitor binding. We show that an inhibitor analogue with a single-atom hydrogen-to-fluorine modification can selectively target a spastin allele with an engineered cysteine mutation in its active site. We also report an X-ray structure of the fluoro analogue bound to the spastin mutant. Furthermore, analyses of other mutant alleles suggest how the stereoelectronics of the fluorine-cysteine interaction, rather than sterics alone, contribute to the inhibitor-allele selectivity. This approach could be used to design allele-specific probes for studying cellular functions of spastin isoforms. Our data also suggest how tuning stereoelectronics can lead to specific inhibitor-allele pairs for the AAA superfamily.
Collapse
Affiliation(s)
- Rudolf Pisa
- Laboratory of Chemistry and Cell Biology , The Rockefeller University , New York , New York 10065 , United States
| | - Tommaso Cupido
- Laboratory of Chemistry and Cell Biology , The Rockefeller University , New York , New York 10065 , United States
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology , The Rockefeller University , New York , New York 10065 , United States
| |
Collapse
|
291
|
Park S, Loke G, Fink Y, Anikeeva P. Flexible fiber-based optoelectronics for neural interfaces. Chem Soc Rev 2019; 48:1826-1852. [PMID: 30815657 DOI: 10.1039/c8cs00710a] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neurological and psychiatric conditions pose an increasing socioeconomic burden on our aging society. Our ability to understand and treat these conditions relies on the development of reliable tools to study the dynamics of the underlying neural circuits. Despite significant progress in approaches and devices to sense and modulate neural activity, further refinement is required on the spatiotemporal resolution, cell-type selectivity, and long-term stability of neural interfaces. Guided by the principles of neural transduction and by the materials properties of the neural tissue, recent advances in neural interrogation approaches rely on flexible and multifunctional devices. Among these approaches, multimaterial fibers have emerged as integrated tools for sensing and delivering of multiple signals to and from the neural tissue. Fiber-based neural probes are produced by thermal drawing process, which is the manufacturing approach used in optical fiber fabrication. This technology allows straightforward incorporation of multiple functional components into microstructured fibers at the level of their macroscale models, preforms, with a wide range of geometries. Here we will introduce the multimaterial fiber technology, its applications in engineering fields, and its adoption for the design of multifunctional and flexible neural interfaces. We will discuss examples of fiber-based neural probes tailored to the electrophysiological recording, optical neuromodulation, and delivery of drugs and genes into the rodent brain and spinal cord, as well as their emerging use for studies of nerve growth and repair.
Collapse
Affiliation(s)
- Seongjun Park
- School of Engineering, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | | | |
Collapse
|
292
|
Roet M, Hescham SA, Jahanshahi A, Rutten BPF, Anikeeva PO, Temel Y. Progress in neuromodulation of the brain: A role for magnetic nanoparticles? Prog Neurobiol 2019; 177:1-14. [PMID: 30878723 DOI: 10.1016/j.pneurobio.2019.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/19/2022]
Abstract
The field of neuromodulation is developing rapidly. Current techniques, however, are still limited as they i) either depend on permanent implants, ii) require invasive procedures, iii) are not cell-type specific, iv) involve slow pharmacokinetics or v) have a restricted penetration depth making it difficult to stimulate regions deep within the brain. Refinements into the different fields of neuromodulation are thus needed. In this review, we will provide background information on the different techniques of neuromodulation discussing their latest refinements and future potentials including the implementation of nanoparticles (NPs). In particular we will highlight the usage of magnetic nanoparticles (MNPs) as transducers in advanced neuromodulation. When exposed to an alternating magnetic field (AMF), certain MNPs can generate heat through hysteresis. This MNP heating has been promising in the field of cancer therapy and has recently been introduced as a method for remote and wireless neuromodulation. This indicates that MNPs may aid in the exploration of brain functions via neuromodulation and may eventually be applied for treatment of neuropsychiatric disorders. We will address the materials chemistry of MNPs, their biomedical applications, their delivery into the brain, their mechanisms of stimulation with emphasis on MNP heating and their remote control in living tissue. The final section compares and discusses the parameters used for MNP heating in brain cancer treatment and neuromodulation. Concluding, using MNPs for nanomaterial-mediated neuromodulation seem promising in a variety of techniques and could be applied for different neuropsychiatric disorders when more extensively investigated.
Collapse
Affiliation(s)
- Milaine Roet
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Sarah-Anna Hescham
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Ali Jahanshahi
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Bart P F Rutten
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands
| | - Polina O Anikeeva
- Department of Materials Science and Engineering, Department of Brain and Cognitive Sciences, Research Laboratory of Electronics, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, 02139, MA, United States of America
| | - Yasin Temel
- School for Mental Health and Neuroscience, Department of Neurosurgery, Maastricht University, Maastricht, 6200, MD, The Netherlands; European Graduate School of Neuroscience (EURON), The Netherlands; Department of Neurosurgery, Maastricht University Medical Center, Maastricht, 6202, AZ, The Netherlands.
| |
Collapse
|
293
|
Namkung H, Lee BJ, Sawa A. Causal Inference on Pathophysiological Mediators in Psychiatry. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:17-23. [PMID: 30850434 DOI: 10.1101/sqb.2018.83.037655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Supported by technological advances and collaborative efforts, psychiatric genetics has provided robust genetic findings in the past decade, particularly through genome-wide association studies (GWASs). However, translating these genetic findings into biological mechanisms and new therapies has been enormously challenging because of the complexity of their interpretation. Furthermore, the heterogeneity among patients with the same diagnosis, such as schizophrenia or major depressive disorder, challenges the biological validity of existing categorical approaches in clinical nosology, which is further complicated by the pleiotropic nature of many genetic variants across multiple disorders. Therefore, in the post-GWAS era, the greatest challenge lies in integrating such enriched genetic information with functional dimensions of neurobiological measures and observable behaviors. In this integration, the causal inference from genotypes to phenotypes through intermediate biological processes is of particular importance. In this review, we aim to construct an intellectual framework in which we may obtain causal, mechanistic insights into how multifactorial etiologies-in particular, many genetic variants-affect downstream biological pathways that lead to dimensions of psychiatric relevance.
Collapse
Affiliation(s)
- Ho Namkung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Brian J Lee
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Akira Sawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Maryland 21287, USA
| |
Collapse
|
294
|
Li H, Illenberger JM, Cranston MN, Mactutus CF, McLaurin KA, Harrod SB, Booze RM. Posterior ventral tegmental area-nucleus accumbens shell circuitry modulates response to novelty. PLoS One 2019; 14:e0213088. [PMID: 30835756 PMCID: PMC6400398 DOI: 10.1371/journal.pone.0213088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 02/14/2019] [Indexed: 11/29/2022] Open
Abstract
Dopamine release in the nucleus accumbens from ventral tegmental area (VTA) efferent neurons is critical for orientation and response to novel stimuli in the environment. However, there are considerable differences between neuronal populations of the VTA and it is unclear which specific cell populations modulate behavioral responses to environmental novelty. A retroDREADDs (designer drugs exclusively activated by designer receptors) technique, comprising designer G protein-coupled receptors exclusively activated by designer drugs and modulated by retrograde transported Cre, was used to selectively stimulate neurons of the VTA which project to the nucleus accumbens shell (AcbSh). First, the selectivity and expression of the human M3 muscarinic receptor-based adeno-associated virus (AAV-hM3D) was confirmed in primary neuronal cell cultures. Second, AAV-CMV-GFP/Cre was infused into the AcbSh and AAV-hSyn-DIO-hM3D(Gq)-mCherry (a presynaptic enhancer in the presence of its cognate ligand clozapine-N-oxide) was infused into the VTA of ovariectomized female Fisher 344 rats to elicit hM3D(Gq)-mCherry production specifically in neurons of the VTA which synapse in the AcbSh. Finally, administration of clozapine-N-oxide significantly altered rodents’ response to novelty (e.g. absence of white background noise) by activation of hM3D(Gq) receptors, without altering gross locomotor activity or auditory processing per se. Confocal imaging confirmed production of mCherry in neurons of the posterior aspect of the VTA (pVTA) suggesting these neurons contribute to novelty responses. These results suggest the pVTA-AcbSh circuit is potentially altered in motivational disorders such as apathy, depression, and drug addiction. Targeting the pVTA-AcbSh circuit, therefore, may be an effective target for pharmacological management of such psychopathologies.
Collapse
Affiliation(s)
- Hailong Li
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
| | - Jessica M. Illenberger
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
| | - Michael N. Cranston
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
| | - Charles F. Mactutus
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
| | - Kristen A. McLaurin
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
| | - Steven B. Harrod
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
| | - Rosemarie M. Booze
- Department of Psychology, Program in Behavioral Neuroscience, University of South Carolina, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
295
|
Abstract
Emotions play a central role in human experience. Over time, methods for manipulating emotion have become increasingly refined and techniques for making sense of the underlying neurobiology have become ever more powerful and precise, enabling new insights into the organization of emotions in the brain. Yet recent years have witnessed a remarkably vigorous debate about the nature and origins of emotion, with leading scientists raising compelling concerns about the canon of facts and principles that has inspired and guided the field for the past quarter century. Here, we consider ways in which recent neuroimaging research informs this dialogue. By focusing attention on the most important outstanding questions about the nature of emotion and the architecture of the emotional brain, we hope to stimulate the kinds of work that will be required to move the field forward. Addressing these questions is critical, not just for understanding the mind, but also for elucidating the root causes of many of its disorders.
Collapse
Affiliation(s)
- Alexander J Shackman
- Department of Psychology, University of Maryland, College Park, MD 20742 USA; Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20742 USA; Maryland Neuroimaging Center, University of Maryland, College Park, MD 20742 USA.
| | - Tor D Wager
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO 80309 USA; Institute of Cognitive Science, University of Colorado, Boulder, CO 80309 USA
| |
Collapse
|
296
|
Allen DC, Carlson TL, Xiong Y, Jin J, Grant KA, Cuzon Carlson VC. A Comparative Study of the Pharmacokinetics of Clozapine N-Oxide and Clozapine N-Oxide Hydrochloride Salt in Rhesus Macaques. J Pharmacol Exp Ther 2019; 368:199-207. [PMID: 30523062 PMCID: PMC6337003 DOI: 10.1124/jpet.118.252031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/04/2018] [Indexed: 11/22/2022] Open
Abstract
Translating chemogenetic techniques from nonhuman primates to potential clinical applications has been complicated in part due to in vivo conversion of the chemogenetic actuator, clozapine N-oxide (CNO), to its pharmacologically active parent compound, clozapine, a ligand with known side effects, including five boxed warnings from the Food and Drug Administration. Additionally, the limited solubility of CNO requires high concentrations of potentially toxic detergents such as dimethylsulfoxide (DMSO). To address these concerns, pharmacokinetic profiling of commercially available CNO in DMSO (CNO-DMSO, 10% v/v DMSO in saline) and a water-soluble salt preparation (CNO-HCl, saline) was conducted in rhesus macaques. A time course of blood plasma and cerebrospinal fluid (CSF) concentrations of CNO and clozapine was conducted (30-240 minutes post-administration) following a range of doses (3-10 mg/kg, i.m. and/or i.v.) of CNO-DMSO or CNO-HCl. CNO-HCl resulted in 6- to 7-fold higher plasma concentrations of CNO compared to CNO-DMSO, and relatively less clozapine (3%-5% clozapine/CNO in the CNO-DMSO group and 0.5%-1.5% clozapine/CNO in the CNO-HCl group). Both groups had large between-subjects variability, pointing to the necessity of performing individual CNO pharmacokinetic studies prior to further experimentation. The ratio of CNO measured in the CSF was between 2% and 6% of that measured in the plasma and did not differ across drug preparation, indicating that CSF concentrations may be approximated from plasma samples. In conclusion, CNO-HCl demonstrated improved bioavailability compared with CNO-DMSO with less conversion to clozapine. Further investigation is needed to determine if brain concentrations of clozapine following CNO-HCl administration are pharmacologically active at off-target monoaminergic receptor systems in the primate brain.
Collapse
Affiliation(s)
- Daicia C Allen
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon (D.C.A., K.A.G., V.C.C.C.); Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon (T.L.C., K.A.G., V.C.C.C.); and Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (Y.X., J.J.)
| | - Timothy L Carlson
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon (D.C.A., K.A.G., V.C.C.C.); Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon (T.L.C., K.A.G., V.C.C.C.); and Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (Y.X., J.J.)
| | - Yan Xiong
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon (D.C.A., K.A.G., V.C.C.C.); Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon (T.L.C., K.A.G., V.C.C.C.); and Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (Y.X., J.J.)
| | - Jian Jin
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon (D.C.A., K.A.G., V.C.C.C.); Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon (T.L.C., K.A.G., V.C.C.C.); and Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (Y.X., J.J.)
| | - Kathleen A Grant
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon (D.C.A., K.A.G., V.C.C.C.); Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon (T.L.C., K.A.G., V.C.C.C.); and Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (Y.X., J.J.)
| | - Verginia C Cuzon Carlson
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon (D.C.A., K.A.G., V.C.C.C.); Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon (T.L.C., K.A.G., V.C.C.C.); and Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (Y.X., J.J.)
| |
Collapse
|
297
|
Leffler KE, Abdel-Rahman AA. Estrogen-Dependent Disruption of Adiponectin-Connexin43 Signaling Underlies Exacerbated Myocardial Dysfunction in Diabetic Female Rats. J Pharmacol Exp Ther 2019; 368:208-217. [PMID: 30523063 PMCID: PMC6337006 DOI: 10.1124/jpet.118.254029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022] Open
Abstract
The reasons for the higher severity of type 2 diabetes (T2DM)-associated cardiomyopathy in women, despite their inherent estrogen (E2)-dependent cardioprotection, remain unknown. We hypothesized that the reliance of the healthy females' hearts on augmented adiponectin (APN)-connexin 43 (Cx43) signaling becomes paradoxically detrimental when disrupted by T2DM in an E2-dependent manner. We tested this hypothesis in high-fat, low- dose streptozotocin diabetic rats and their controls with the following designations: 1) sham-operated (SO), 2) ovariectomized (OVX), 3) ovariectomized with E2 supplementation (OVX + E2), and 4) male. E2-replete (SO or OVX + E2) diabetic rats exhibited higher mortality and greater increases in left ventricular (LV) mass and reduced LV developed pressure, LV contractility, and fractional shortening but preserved ejection fraction. Further, compared with respective nondiabetic counterparts, the hearts of these E2-replete diabetic rats exhibited greater upregulation of cardiac estrogen receptor α and reductions in Cx43 expression and in the phosphorylation levels of the survival molecules extracellular regulating kinases 1/2 and phosphorylated AKT (pAKT). Whereas serum APN was reduced, independent of sex and ovarian hormone status in all DM rats, cardiac APN was most drastically reduced in DM SO rats. The present translational findings are the first to implicate ovarian hormones/E2 in the exacerbated myocardial dysfunction in female diabetic subjects and to suggest a pivotal role for malfunctioning cardiac APN-Cx43 signaling in this sex/E2-specific clinical problem.
Collapse
Affiliation(s)
- Korin E Leffler
- Department of Pharmacology and Toxicology, East Carolina University, Brody School of Medicine, Greenville, North Carolina
| | - Abdel A Abdel-Rahman
- Department of Pharmacology and Toxicology, East Carolina University, Brody School of Medicine, Greenville, North Carolina
| |
Collapse
|
298
|
Cheng Y, Wang J. The use of chemogenetic approaches in alcohol use disorder research and treatment. Alcohol 2019; 74:39-45. [PMID: 30442535 DOI: 10.1016/j.alcohol.2018.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/15/2018] [Accepted: 05/28/2018] [Indexed: 02/09/2023]
Abstract
Several novel techniques were developed recently to explore neural circuit mechanisms of neuropsychiatric disorders. These techniques include the Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-based chemogenetic tools, which represent valuable platforms for selective and non-invasive control of neural activity with a high degree of spatial resolution. Among all variants, Gq- and Gi-DREADDs are widely used by neuroscientists to dissect out the circuitry and cellular signals. This review is focused on strategies to access a specific neuronal population or circuit using the DREADD technique and summarizes the current knowledge of the DREADDs' application in alcohol use disorder research and therapeutics.
Collapse
|
299
|
Coyote-Maestas W, He Y, Myers CL, Schmidt D. Domain insertion permissibility-guided engineering of allostery in ion channels. Nat Commun 2019; 10:290. [PMID: 30655517 PMCID: PMC6336875 DOI: 10.1038/s41467-018-08171-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023] Open
Abstract
Allostery is a fundamental principle of protein regulation that remains hard to engineer, particularly in membrane proteins such as ion channels. Here we use human Inward Rectifier K+ Channel Kir2.1 to map site-specific permissibility to the insertion of domains with different biophysical properties. We find that permissibility is best explained by dynamic protein properties, such as conformational flexibility. Several regions in Kir2.1 that are equivalent to those regulated in homologs, such as G-protein-gated inward rectifier K+ channels (GIRK), have differential permissibility; that is, for these sites permissibility depends on the structural properties of the inserted domain. Our data and the well-established link between protein dynamics and allostery led us to propose that differential permissibility is a metric of latent allosteric capacity in Kir2.1. In support of this notion, inserting light-switchable domains into sites with predicted latent allosteric capacity renders Kir2.1 activity sensitive to light. Allostery is a fundamental principle of protein regulation that remains challenging to engineer. Here authors screen human Inward Rectifier K + Channel Kir2.1 for permissibility to domain insertions and propose that differential permissibility is a metric of latent allosteric capacity in Kir2.1.
Collapse
Affiliation(s)
- Willow Coyote-Maestas
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Yungui He
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, 55455, MN, USA
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, 55455, MN, USA.
| |
Collapse
|
300
|
cAMP-producing chemogenetic and adenosine A2a receptor activation inhibits the inwardly rectifying potassium current in striatal projection neurons. Neuropharmacology 2019; 148:229-243. [PMID: 30659840 DOI: 10.1016/j.neuropharm.2019.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/29/2022]
Abstract
Adenosine A2a receptors (A2aRs) are highly and selectively expressed in D2-medium spiny neurons (D2-MSNs) that also express a high level of dopamine D2 receptors (D2Rs). However, it was not established how A2aR activity affects D2-MSN excitability, let alone the ion channels involved. We have performed two sets of experiments to determine the potential A2aR agonistic effects on D2-MSN intrinsic excitability and the underlying ion channel mechanism. First, we have used the cAMP-producing, Gαs/olf coupled designer receptors exclusively activated by designer drug (Gs-DREADDs) to phenocopy cAMP-stimulating A2aR activation. We found that activation of Gs-DREADD inhibited the inwardly rectifying potassium current (Kir)-a key regulator of MSN excitability, caused a depolarization, increased input resistance, and substantially increased the intrinsic excitability of MSNs such that depolarizing inputs evoked many more action potentials. Second, we have determined that A2aR agonism produced these same excitatory effects on D2-MSN intrinsic excitability and spike firing, although at lower magnitudes than those induced by Gs-DREADD activation; furthermore, these A2aR-triggered excitatory effects were intact in the presence of a D2R antagonist. Taken together, these results clearly establish that in striatal D2-MSNs, A2aR activation can independently inhibit Kir and increase intrinsic excitability and spike and neurotransmitter output; our results also indicate that Gs-DREADD can serve as a broadly useful positive control for neurotransmitter receptors that increase intracellular cAMP levels and hence facilitate the determination of the cellular effects of these neurotransmitter receptors.
Collapse
|