1
|
Maleeva G, Nin-Hill A, Wirth U, Rustler K, Ranucci M, Opar E, Rovira C, Bregestovski P, Zeilhofer HU, König B, Alfonso-Prieto M, Gorostiza P. Light-Activated Agonist-Potentiator of GABA A Receptors for Reversible Neuroinhibition in Wildtype Mice. J Am Chem Soc 2024; 146:28822-28831. [PMID: 39383450 PMCID: PMC11503767 DOI: 10.1021/jacs.4c08446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024]
Abstract
Gamma aminobutyric acid type A receptors (GABAARs) play a key role in the mammalian central nervous system (CNS) as drivers of neuroinhibitory circuits, which are commonly targeted for therapeutic purposes with potentiator drugs. However, due to their widespread expression and strong inhibitory action, systemic pharmaceutical potentiation of GABAARs inevitably causes adverse effects regardless of the drug selectivity. Therefore, therapeutic guidelines must often limit or exclude clinically available GABAAR potentiators, despite their high efficacy, good biodistribution, and favorable molecular properties. One solution to this problem is to use drugs with light-dependent activity (photopharmacology) in combination with on-demand, localized illumination. However, a suitable light-activated potentiator of GABAARs has been elusive so far for use in wildtype mammals. We have met this need by developing azocarnil, a diffusible GABAergic agonist-potentiator based on the anxiolytic drug abecarnil that is inactive in the dark and activated by visible violet light. Azocarnil can be rapidly deactivated with green light and by thermal relaxation in the dark. We demonstrate that it selectively inhibits neuronal currents in hippocampal neurons in vitro and in the dorsal horns of the spinal cord of mice, decreasing the mechanical sensitivity as a function of illumination without displaying systemic adverse effects. Azocarnil expands the in vivo photopharmacological toolkit with a novel chemical scaffold and achieves a milestone toward future phototherapeutic applications to safely treat muscle spasms, pain, anxiety, sleep disorders, and epilepsy.
Collapse
Affiliation(s)
- Galyna Maleeva
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona 08028, Spain
- Networking
Biomedical Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), ISCIII, Barcelona 08028, Spain
| | - Alba Nin-Hill
- Departament
de Química Inorgànica i Orgànica (Secció
de Química Orgànica) & Institut de Química
Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona 08020, Spain
| | - Ulrike Wirth
- Institute
of Organic Chemistry, University of Regensburg, Regensburg 93053, Germany
| | - Karin Rustler
- Institute
of Organic Chemistry, University of Regensburg, Regensburg 93053, Germany
| | - Matteo Ranucci
- Institute
of Pharmacology and Toxicology, University
of Zurich, Zürich 8057, Switzerland
| | - Ekin Opar
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona 08028, Spain
- Networking
Biomedical Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), ISCIII, Barcelona 08028, Spain
- Doctorate
program of the University of Barcelona, Barcelona 08020, Spain
| | - Carme Rovira
- Departament
de Química Inorgànica i Orgànica (Secció
de Química Orgànica) & Institut de Química
Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona 08020, Spain
- Catalan
Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| | - Piotr Bregestovski
- Institut
de Neurosciences des Systèmes, UMR INSERM 1106, Aix-Marseille Université, Marseille 13005, France
| | - Hanns Ulrich Zeilhofer
- Institute
of Pharmacology and Toxicology, University
of Zurich, Zürich 8057, Switzerland
- Institute
of Pharmaceutical Sciences, Swiss Federal
Institute of Technology (ETH) Zürich, Zürich 8093, Switzerland
| | - Burkhard König
- Institute
of Organic Chemistry, University of Regensburg, Regensburg 93053, Germany
| | - Mercedes Alfonso-Prieto
- Institute
of Neuroscience and Medicine INM-9 Computational Biomedicine, Forschungszentrum Jülich GmbH, D-52428 Jülich, Germany
| | - Pau Gorostiza
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona 08028, Spain
- Networking
Biomedical Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), ISCIII, Barcelona 08028, Spain
- Catalan
Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| |
Collapse
|
2
|
Shields BC, Yan H, Lim SSX, Burwell SCV, Cammarata CM, Fleming EA, Yousefzadeh SA, Goldenshtein VZ, Kahuno EW, Vagadia PP, Loughran MH, Zhiquan L, McDonnell ME, Scalabrino ML, Thapa M, Hawley TM, Field GD, Hull C, Schiltz GE, Glickfeld LL, Reitz AB, Tadross MR. DART.2: bidirectional synaptic pharmacology with thousandfold cellular specificity. Nat Methods 2024; 21:1288-1297. [PMID: 38877316 PMCID: PMC11569460 DOI: 10.1038/s41592-024-02292-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/25/2024] [Indexed: 06/16/2024]
Abstract
Precision pharmacology aims to manipulate specific cellular interactions within complex tissues. In this pursuit, we introduce DART.2 (drug acutely restricted by tethering), a second-generation cell-specific pharmacology technology. The core advance is optimized cellular specificity-up to 3,000-fold in 15 min-enabling the targeted delivery of even epileptogenic drugs without off-target effects. Additionally, we introduce brain-wide dosing methods as an alternative to local cannulation and tracer reagents for brain-wide dose quantification. We describe four pharmaceuticals-two that antagonize excitatory and inhibitory postsynaptic receptors, and two that allosterically potentiate these receptors. Their versatility is showcased across multiple mouse-brain regions, including cerebellum, striatum, visual cortex and retina. Finally, in the ventral tegmental area, we find that blocking inhibitory inputs to dopamine neurons accelerates locomotion, contrasting with previous optogenetic and pharmacological findings. Beyond enabling the bidirectional perturbation of chemical synapses, these reagents offer intersectional precision-between genetically defined postsynaptic cells and neurotransmitter-defined presynaptic partners.
Collapse
Affiliation(s)
- Brenda C Shields
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Haidun Yan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Shaun S X Lim
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | | | | | | | | | | - Purav P Vagadia
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | | | - Lei Zhiquan
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | | | | | - Mishek Thapa
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Tammy M Hawley
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Greg D Field
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Court Hull
- Department of Neurobiology, Duke University, Durham, NC, USA
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | | | - Allen B Reitz
- Fox Chase Therapeutics Discovery, Inc., Doylestown, PA, USA
| | - Michael R Tadross
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
3
|
McClain SP, Ma X, Johnson DA, Johnson CA, Layden AE, Yung JC, Lubejko ST, Livrizzi G, He XJ, Zhou J, Chang-Weinberg J, Ventriglia E, Rizzo A, Levinstein M, Gomez JL, Bonaventura J, Michaelides M, Banghart MR. In vivo photopharmacology with light-activated opioid drugs. Neuron 2023; 111:3926-3940.e10. [PMID: 37848025 PMCID: PMC11188017 DOI: 10.1016/j.neuron.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 08/02/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023]
Abstract
Traditional methods for site-specific drug delivery in the brain are slow, invasive, and difficult to interface with recordings of neural activity. Here, we demonstrate the feasibility and experimental advantages of in vivo photopharmacology using "caged" opioid drugs that are activated in the brain with light after systemic administration in an inactive form. To enable bidirectional manipulations of endogenous opioid receptors in vivo, we developed photoactivatable oxymorphone (PhOX) and photoactivatable naloxone (PhNX), photoactivatable variants of the mu opioid receptor agonist oxymorphone and the antagonist naloxone. Photoactivation of PhOX in multiple brain areas produced local changes in receptor occupancy, brain metabolic activity, neuronal calcium activity, neurochemical signaling, and multiple pain- and reward-related behaviors. Combining PhOX photoactivation with optical recording of extracellular dopamine revealed adaptations in the opioid sensitivity of mesolimbic dopamine circuitry in response to chronic morphine administration. This work establishes a general experimental framework for using in vivo photopharmacology to study the neural basis of drug action.
Collapse
Affiliation(s)
- Shannan P McClain
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Neurosciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Xiang Ma
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Desiree A Johnson
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Caroline A Johnson
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Aryanna E Layden
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jean C Yung
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Susan T Lubejko
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Neurosciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Giulia Livrizzi
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - X Jenny He
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Jingjing Zhou
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Janie Chang-Weinberg
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Emilya Ventriglia
- Biobehavioral Imaging and Molecular, Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Arianna Rizzo
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat 08907, Catalonia, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat 08907, Catalonia, Spain
| | - Marjorie Levinstein
- Biobehavioral Imaging and Molecular, Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular, Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Jordi Bonaventura
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat 08907, Catalonia, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat 08907, Catalonia, Spain
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular, Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA
| | - Matthew R Banghart
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
4
|
Hou QQ, Huang QT, Xu Q, Zhou C, Du YY, Ji YF, Xu ZP, Cheng JG, Zhao CQ, Li Z, Shao XS. Synthesis and activity-detection of photoswitchable ligands with fipronil to insect. PEST MANAGEMENT SCIENCE 2023; 79:1086-1093. [PMID: 36334017 DOI: 10.1002/ps.7279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Ionotropic γ-aminobutyric acid (GABA) receptor (GABAR) in an insect is the major inhibitory receptor and is one of the most important targets for insecticides. Due to the high spatiotemporal resolution of GABAR, the photopharmacological ligands acting on it in vertebrates but not insect have been developed. RESULTS In this study, two types of photochromic ligands (PCLs) including DTFIPs (DTFIP1 and DTFIP2) and ABFIPs (p-, m-, and o-ABFIP) were synthesized by incorporating photoswitch azobenzene or dithienylethene into fipronil (FIP), which is the antagonist of insect GABAR. Their photomodulation was measured by mosquito larval behavior, and their potential action mechanism was explored by the two-electrode voltage clamp (TEVC) technique in vitro. DTFIP1 and m-ABFIP exhibited the most significant difference of insecticidal activity by about 90- and 5-fold to mosquito larvae between non-irradiated and irradiated formation, respectively, and allowed for optical control of mosquito swimming activity. TEVC assay results indicated that m-ABFIP and DTFIP1 enable optical control over the homomeric LsRDL-type GABAR, which is achieved by regulating the chloride channel of resistance to dieldrin (RDL)-type GABAR by photoisomerization. CONCLUSION Our results suggested that PCLs synthesized from fipronil provide an alternative and precise tool for studying insect ionotropic GABARs and GABA-dependent behavior. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qing-Qing Hou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Qiu-Tang Huang
- Key Laboratory of Integrated Pest Management on Crops in East China, Ministry of Agriculture, College of Plant Protection, Nanjing Agricultural University, Nanjing, P. R. China
| | - Qi Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Cong Zhou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Yao-Yao Du
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Yun-Fan Ji
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Zhi-Ping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Jia-Gao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| | - Chun-Qing Zhao
- Key Laboratory of Integrated Pest Management on Crops in East China, Ministry of Agriculture, College of Plant Protection, Nanjing Agricultural University, Nanjing, P. R. China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- Key Laboratory of Integrated Pest Management on Crops in East China, Ministry of Agriculture, College of Plant Protection, Nanjing Agricultural University, Nanjing, P. R. China
| | - Xu-Sheng Shao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, P. R. China
| |
Collapse
|
5
|
McClain SP, Ma X, Johnson DA, Johnson CA, Layden AE, Yung JC, Lubejko ST, Livrizzi G, Jenny He X, Zhou J, Ventriglia E, Rizzo A, Levinstein M, Gomez JL, Bonaventura J, Michaelides M, Banghart MR. In vivo photopharmacology with light-activated opioid drugs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526901. [PMID: 36778286 PMCID: PMC9915677 DOI: 10.1101/2023.02.02.526901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traditional methods for site-specific drug delivery in the brain are slow, invasive, and difficult to interface with recordings of neural activity. Here, we demonstrate the feasibility and experimental advantages of in vivo photopharmacology using "caged" opioid drugs that are activated in the brain with light after systemic administration in an inactive form. To enable bidirectional manipulations of endogenous opioid receptors in vivo , we developed PhOX and PhNX, photoactivatable variants of the mu opioid receptor agonist oxymorphone and the antagonist naloxone. Photoactivation of PhOX in multiple brain areas produced local changes in receptor occupancy, brain metabolic activity, neuronal calcium activity, neurochemical signaling, and multiple pain- and reward-related behaviors. Combining PhOX photoactivation with optical recording of extracellular dopamine revealed adaptations in the opioid sensitivity of mesolimbic dopamine circuitry during chronic morphine administration. This work establishes a general experimental framework for using in vivo photopharmacology to study the neural basis of drug action. Highlights A photoactivatable opioid agonist (PhOX) and antagonist (PhNX) for in vivo photopharmacology. Systemic pro-drug delivery followed by local photoactivation in the brain. In vivo photopharmacology produces behavioral changes within seconds of photostimulation. In vivo photopharmacology enables all-optical pharmacology and physiology.
Collapse
|
6
|
Bryson A, Reid C, Petrou S. Fundamental Neurochemistry Review: GABA A receptor neurotransmission and epilepsy: Principles, disease mechanisms and pharmacotherapy. J Neurochem 2023; 165:6-28. [PMID: 36681890 DOI: 10.1111/jnc.15769] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/23/2023]
Abstract
Epilepsy is a common neurological disorder associated with alterations of excitation-inhibition balance within brain neuronal networks. GABAA receptor neurotransmission is the most prevalent form of inhibitory neurotransmission and is strongly implicated in both the pathophysiology and treatment of epilepsy, serving as a primary target for antiseizure medications for over a century. It is now established that GABA exerts a multifaceted influence through an array of GABAA receptor subtypes that extends far beyond simply negating excitatory activity. As the role of GABAA neurotransmission within inhibitory circuits is elaborated, this will enable the development of precision therapies that correct the network dysfunction underlying epileptic pathology.
Collapse
Affiliation(s)
- Alexander Bryson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, Austin Health, Heidelberg, Victoria, Australia
| | - Christopher Reid
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,Praxis Precision Medicines, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
7
|
Liu P, Dai S, Mi T, Tang G, Wang Z, Wang H, Du H, Tang Y, Teng Z, Liu C. Acetate supplementation restores cognitive deficits caused by ARID1A haploinsufficiency in excitatory neurons. EMBO Mol Med 2022; 14:e15795. [PMID: 36385502 PMCID: PMC9728054 DOI: 10.15252/emmm.202215795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Mutations in AT-rich interactive domain-containing protein 1A (ARID1A) cause Coffin-Siris syndrome (CSS), a rare genetic disorder that results in mild to severe intellectual disabilities. However, the biological role of ARID1A in the brain remains unclear. In this study, we report that the haploinsufficiency of ARID1A in excitatory neurons causes cognitive impairment and defects in hippocampal synaptic transmission and dendritic morphology in mice. Similarly, human embryonic stem cell-derived excitatory neurons with deleted ARID1A exhibit fewer dendritic branches and spines, and abnormal electrophysiological activity. Importantly, supplementation of acetate, an epigenetic metabolite, can ameliorate the morphological and electrophysiological deficits observed in mice with Arid1a haploinsufficiency, as well as in ARID1A-null human excitatory neurons. Mechanistically, transcriptomic and ChIP-seq analyses demonstrate that acetate supplementation can increase the levels of H3K27 acetylation at the promoters of key regulatory genes associated with neural development and synaptic transmission. Collectively, these findings support the essential roles of ARID1A in the excitatory neurons and cognition and suggest that acetate supplementation could be a potential therapeutic intervention for CSS.
Collapse
Affiliation(s)
- Pei‐Pei Liu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Shang‐Kun Dai
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,School of Life Sciences and MedicineShandong University of TechnologyZiboChina
| | - Ting‐Wei Mi
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
| | - Gang‐Bin Tang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
| | - Zhuo Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Hui Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Hong‐Zhen Du
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Yi Tang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Zhao‐Qian Teng
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Chang‐Mei Liu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina,Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| |
Collapse
|
8
|
Castagna R, Maleeva G, Pirovano D, Matera C, Gorostiza P. Donor-Acceptor Stenhouse Adduct Displaying Reversible Photoswitching in Water and Neuronal Activity. J Am Chem Soc 2022; 144:15595-15602. [PMID: 35976640 DOI: 10.1021/jacs.2c04920] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The interest in the photochromism and functional applications of donor-acceptor Stenhouse adducts (DASAs) soared in recent years owing to their outstanding advantages and flexible design. However, their low solubility and irreversible conversion in aqueous solutions hampered exploring DASAs for biology and medicine. It is notably unknown whether the barbiturate electron acceptor group retains the pharmacological activity of drugs such as phenobarbital, which targets γ-aminobutyric acid (GABA)-type A receptors (GABAARs) in the brain. Here, we have developed the model compound DASA-barbital based on a scaffold of red-switching second-generation DASAs, and we demonstrate that it is active in GABAARs and alters the neuronal firing rate in a physiological medium at neutral pH. DASA-barbital can also be reversibly photoswitched in acidic aqueous solutions using cyclodextrin, an approved ingredient of drug formulations. These findings clarify the path toward the biological applications of DASAs and to exploit the versatility displayed in polymers and materials science.
Collapse
Affiliation(s)
- Rossella Castagna
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain.,CIBER, Madrid 282029, Spain
| | - Galyna Maleeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain
| | - Deborah Pirovano
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain
| | - Carlo Matera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain.,CIBER, Madrid 282029, Spain
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain.,CIBER, Madrid 282029, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| |
Collapse
|
9
|
Orthogonal Control of Neuronal Circuits and Behavior Using Photopharmacology. J Mol Neurosci 2022; 72:1433-1442. [PMID: 35737209 DOI: 10.1007/s12031-022-02037-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/04/2022] [Indexed: 10/17/2022]
Abstract
Over the last decades, photopharmacology has gone far beyond its proof-of-concept stage to become a bona fide approach to study neural systems in vivo. Indeed, photopharmacological control has expanded over a wide range of endogenous targets, such as receptors, ion channels, transporters, kinases, lipids, and DNA transcription processes. In this review, we provide an overview of the recent progresses in the in vivo photopharmacological control of neuronal circuits and behavior. In particular, the use of small aquatic animals for the in vivo screening of photopharmacological compounds, the recent advances in optical modulation of complex behaviors in mice, and the development of adjacent techniques for light and drug delivery in vivo are described.
Collapse
|
10
|
Ojima K, Kakegawa W, Yamasaki T, Miura Y, Itoh M, Michibata Y, Kubota R, Doura T, Miura E, Nonaka H, Mizuno S, Takahashi S, Yuzaki M, Hamachi I, Kiyonaka S. Coordination chemogenetics for activation of GPCR-type glutamate receptors in brain tissue. Nat Commun 2022; 13:3167. [PMID: 35710788 PMCID: PMC9203742 DOI: 10.1038/s41467-022-30828-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Direct activation of cell-surface receptors is highly desirable for elucidating their physiological roles. A potential approach for cell-type-specific activation of a receptor subtype is chemogenetics, in which both point mutagenesis of the receptors and designed ligands are used. However, ligand-binding properties are affected in most cases. Here, we developed a chemogenetic method for direct activation of metabotropic glutamate receptor 1 (mGlu1), which plays essential roles in cerebellar functions in the brain. Our screening identified a mGlu1 mutant, mGlu1(N264H), that was activated directly by palladium complexes. A palladium complex showing low cytotoxicity successfully activated mGlu1 in mGlu1(N264H) knock-in mice, revealing that activation of endogenous mGlu1 is sufficient to evoke the critical cellular mechanism of synaptic plasticity, a basis of motor learning in the cerebellum. Moreover, cell-type-specific activation of mGlu1 was demonstrated successfully using adeno-associated viruses in mice, which shows the potential utility of this chemogenetics for clarifying the physiological roles of mGlu1 in a cell-type-specific manner. Cell-type-specific activation of receptors is desirable for elucidating their roles in tissues or animals. Here, the authors developed a chemogenetic method for direct activation of mGlu1, a GPCR-type glutamate receptor subtype, and demonstrate its use in mouse brain tissue.
Collapse
Affiliation(s)
- Kento Ojima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan.,Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Wataru Kakegawa
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Tokiwa Yamasaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuta Miura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Masayuki Itoh
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yukiko Michibata
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Ryou Kubota
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Eriko Miura
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Michisuke Yuzaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| | - Shigeki Kiyonaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan. .,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya, 464-8603, Japan.
| |
Collapse
|
11
|
Frank JA. Optofluidic neural interfaces for in vivo photopharmacology. Curr Opin Pharmacol 2022; 63:102195. [DOI: 10.1016/j.coph.2022.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 11/03/2022]
|
12
|
Miura Y, Senoo A, Doura T, Kiyonaka S. Chemogenetics of cell surface receptors: beyond genetic and pharmacological approaches. RSC Chem Biol 2022; 3:269-287. [PMID: 35359495 PMCID: PMC8905536 DOI: 10.1039/d1cb00195g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/20/2022] [Indexed: 11/29/2022] Open
Abstract
Cell surface receptors transmit extracellular information into cells. Spatiotemporal regulation of receptor signaling is crucial for cellular functions, and dysregulation of signaling causes various diseases. Thus, it is highly desired to control receptor functions with high spatial and/or temporal resolution. Conventionally, genetic engineering or chemical ligands have been used to control receptor functions in cells. As the alternative, chemogenetics has been proposed, in which target proteins are genetically engineered to interact with a designed chemical partner with high selectivity. The engineered receptor dissects the function of one receptor member among a highly homologous receptor family in a cell-specific manner. Notably, some chemogenetic strategies have been used to reveal the receptor signaling of target cells in living animals. In this review, we summarize the developing chemogenetic methods of transmembrane receptors for cell-specific regulation of receptor signaling. We also discuss the prospects of chemogenetics for clinical applications.
Collapse
Affiliation(s)
- Yuta Miura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Akinobu Senoo
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Shigeki Kiyonaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| |
Collapse
|
13
|
Kramer RH, Rajappa R. Interrogating the function of GABA A receptors in the brain with optogenetic pharmacology. Curr Opin Pharmacol 2022; 63:102198. [PMID: 35276498 DOI: 10.1016/j.coph.2022.102198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/26/2022]
Abstract
To better understand neural circuits and behavior, microbial opsins have been developed as optogenetic tools for stimulating or inhibiting action potentials with high temporal and spatial precision. However, if we seek a more reductionist understanding of how neuronal circuits operate, we also need high-resolution tools for perturbing the function of synapses. By combining photochemical tools and molecular biology, a wide variety of light-regulated neurotransmitter receptors have been developed, enabling photo-control of excitatory, inhibitory, and modulatory synaptic transmission. Here we focus on photo-control of GABAA receptors, ligand-gated Cl- channels that underlie almost all synaptic inhibition in the mammalian brain. By conjugating a photoswitchable tethered ligand onto a genetically-modified subunit of the GABAA receptor, light-sensitivity can be conferred onto specific isoforms of the receptor. Through gene editing, this attachment site can be knocked into the genome, enabling photocontrol of endogenous GABAA receptors. This strategy can be employed to explore the cell biology and neurophysiology of GABAA receptors. This includes investigating how specific isoforms contribute to synaptic and tonic inhibition and understanding the roles they play in brain development, long-term synaptic plasticity, and learning and memory.
Collapse
Affiliation(s)
- Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, United States; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States.
| | - Rajit Rajappa
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, United States; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States
| |
Collapse
|
14
|
Advances in tethered photopharmacology for precise optical control of signaling proteins. Curr Opin Pharmacol 2022; 63:102196. [PMID: 35245800 DOI: 10.1016/j.coph.2022.102196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/25/2022]
Abstract
To overcome the limitations of traditional pharmacology, the field of photopharmacology has developed around the central concept of using light to endow drug action with spatiotemporal precision. Tethered photopharmacology, where a photoswitchable ligand is covalently attached to a target protein, offers a particularly high degree of spatiotemporal control, as well as the ability to genetically target drug action and limit effects to specific protein subtypes. In this review, we describe the core engineering concepts of tethered pharmacology and highlight recent advances in harnessing the power of tethered photopharmacology for an expanded palette of targets and conjugation modes using new, complementary strategies. We also discuss the various applications, including mechanistic studies from the molecular biophysical realm to in vivo studies in behaving animals, that demonstrate the power of tethered pharmacology.
Collapse
|
15
|
Scheiner M, Sink A, Hoffmann M, Vrigneau C, Endres E, Carles A, Sotriffer C, Maurice T, Decker M. Photoswitchable Pseudoirreversible Butyrylcholinesterase Inhibitors Allow Optical Control of Inhibition in Vitro and Enable Restoration of Cognition in an Alzheimer's Disease Mouse Model upon Irradiation. J Am Chem Soc 2022; 144:3279-3284. [PMID: 35138833 DOI: 10.1021/jacs.1c13492] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To develop tools to investigate the biological functions of butyrylcholinesterase (BChE) and the mechanisms by which BChE affects Alzheimer's disease (AD), we synthesized several selective, nanomolar active, pseudoirreversible photoswitchable BChE inhibitors. The compounds were able to specifically influence different kinetic parameters of the inhibition process by light. For one compound, a 10-fold difference in the IC50-values (44.6 nM cis, 424 nM trans) in vitro was translated to an "all or nothing" response with complete recovery in a murine cognition-deficit AD model at dosages as low as 0.3 mg/kg.
Collapse
Affiliation(s)
- Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius-Maximilian-Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Alexandra Sink
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius-Maximilian-Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Matthias Hoffmann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius-Maximilian-Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Cassandre Vrigneau
- MMDN, University of Montpellier, INSERM, EPHE, 34095 Montpellier, France
| | - Erik Endres
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius-Maximilian-Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Allison Carles
- MMDN, University of Montpellier, INSERM, EPHE, 34095 Montpellier, France
| | - Christoph Sotriffer
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius-Maximilian-Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, 34095 Montpellier, France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius-Maximilian-Universität Würzburg, Am Hubland, 97074, Würzburg, Germany
| |
Collapse
|
16
|
Shen Y, Luchetti A, Fernandes G, Do Heo W, Silva AJ. The emergence of molecular systems neuroscience. Mol Brain 2022; 15:7. [PMID: 34983613 PMCID: PMC8728933 DOI: 10.1186/s13041-021-00885-5] [Citation(s) in RCA: 5] [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: 09/07/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Systems neuroscience is focused on how ensemble properties in the brain, such as the activity of neuronal circuits, gives rise to internal brain states and behavior. Many of the studies in this field have traditionally involved electrophysiological recordings and computational approaches that attempt to decode how the brain transforms inputs into functional outputs. More recently, systems neuroscience has received an infusion of approaches and techniques that allow the manipulation (e.g., optogenetics, chemogenetics) and imaging (e.g., two-photon imaging, head mounted fluorescent microscopes) of neurons, neurocircuits, their inputs and outputs. Here, we will review novel approaches that allow the manipulation and imaging of specific molecular mechanisms in specific cells (not just neurons), cell ensembles and brain regions. These molecular approaches, with the specificity and temporal resolution appropriate for systems studies, promise to infuse the field with novel ideas, emphases and directions, and are motivating the emergence of a molecularly oriented systems neuroscience, a new discipline that studies how the spatial and temporal patterns of molecular systems modulate circuits and brain networks, and consequently shape the properties of brain states and behavior.
Collapse
Affiliation(s)
- Yang Shen
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Alessandro Luchetti
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Giselle Fernandes
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Nin-Hill A, Mueller NPF, Molteni C, Rovira C, Alfonso-Prieto M. Photopharmacology of Ion Channels through the Light of the Computational Microscope. Int J Mol Sci 2021; 22:12072. [PMID: 34769504 PMCID: PMC8584574 DOI: 10.3390/ijms222112072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
The optical control and investigation of neuronal activity can be achieved and carried out with photoswitchable ligands. Such compounds are designed in a modular fashion, combining a known ligand of the target protein and a photochromic group, as well as an additional electrophilic group for tethered ligands. Such a design strategy can be optimized by including structural data. In addition to experimental structures, computational methods (such as homology modeling, molecular docking, molecular dynamics and enhanced sampling techniques) can provide structural insights to guide photoswitch design and to understand the observed light-regulated effects. This review discusses the application of such structure-based computational methods to photoswitchable ligands targeting voltage- and ligand-gated ion channels. Structural mapping may help identify residues near the ligand binding pocket amenable for mutagenesis and covalent attachment. Modeling of the target protein in a complex with the photoswitchable ligand can shed light on the different activities of the two photoswitch isomers and the effect of site-directed mutations on photoswitch binding, as well as ion channel subtype selectivity. The examples presented here show how the integration of computational modeling with experimental data can greatly facilitate photoswitchable ligand design and optimization. Recent advances in structural biology, both experimental and computational, are expected to further strengthen this rational photopharmacology approach.
Collapse
Affiliation(s)
- Alba Nin-Hill
- Departament de Química Inorgànica i Orgànica (Secció de Química Orgànica) and Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, 08028 Barcelona, Spain; (A.N.-H.); (C.R.)
| | - Nicolas Pierre Friedrich Mueller
- Institute for Advanced Simulations IAS-5 and Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany;
- Faculty of Mathematics and Natural Sciences, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Carla Molteni
- Physics Department, King’s College London, London WC2R 2LS, UK;
| | - Carme Rovira
- Departament de Química Inorgànica i Orgànica (Secció de Química Orgànica) and Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, 08028 Barcelona, Spain; (A.N.-H.); (C.R.)
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08020 Barcelona, Spain
| | - Mercedes Alfonso-Prieto
- Institute for Advanced Simulations IAS-5 and Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany;
- Cécile and Oskar Vogt Institute for Brain Research, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
18
|
Tsai YH, Doura T, Kiyonaka S. Tethering-based chemogenetic approaches for the modulation of protein function in live cells. Chem Soc Rev 2021; 50:7909-7923. [PMID: 34114579 DOI: 10.1039/d1cs00059d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Proteins are the workhorse molecules performing various tasks to sustain life. To investigate the roles of a protein under physiological conditions, the rapid modulation of the protein with high specificity in a living system would be ideal, but achieving this is often challenging. To address this challenge, researchers have developed chemogenetic strategies for the rapid and selective modulation of protein function in live cells. Here, the target protein is modified genetically to become sensitive to a designer molecule that otherwise has no effect on other cellular biomolecules. One powerful chemogenetic strategy is to introduce a tethering point into the target protein, allowing covalent or non-covalent attachment of the designer molecule. In this tutorial review, we focus on tethering-based chemogenetic approaches for modulating protein function in live cells. We first describe genetic, optogenetic and chemical means to study protein function. These means lay the basis for the chemogenetic concept, which is explained in detail. The next section gives an overview, including advantages and limitations, of tethering tactics that have been employed for modulating cellular protein function. The third section provides examples of the modulation of cell-surface proteins using tethering-based chemogenetics through non-covalent tethering and covalent tethering for irreversible modulation or functional switching. The fourth section presents intracellular examples. The last section summarizes key considerations in implementing tethering-based chemogenetics and shows perspectives highlighting future directions and other applications of this burgeoning research field.
Collapse
Affiliation(s)
- Yu-Hsuan Tsai
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | | | | |
Collapse
|
19
|
Bregestovski PD, Ponomareva DN. Photochromic Modulation of Cys-loop
Ligand-gated Ion Channels. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021020162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Gutzeit VA, Acosta-Ruiz A, Munguba H, Häfner S, Landra-Willm A, Mathes B, Mony J, Yarotski D, Börjesson K, Liston C, Sandoz G, Levitz J, Broichhagen J. A fine-tuned azobenzene for enhanced photopharmacology in vivo. Cell Chem Biol 2021; 28:1648-1663.e16. [PMID: 33735619 DOI: 10.1016/j.chembiol.2021.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/23/2020] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
Despite the power of photopharmacology for interrogating signaling proteins, many photopharmacological systems are limited by their efficiency, speed, or spectral properties. Here, we screen a library of azobenzene photoswitches and identify a urea-substituted "azobenzene-400" core that offers bistable switching between cis and trans with improved kinetics, light sensitivity, and a red-shift. We then focus on the metabotropic glutamate receptors (mGluRs), neuromodulatory receptors that are major pharmacological targets. Synthesis of "BGAG12,400," a photoswitchable orthogonal, remotely tethered ligand (PORTL), enables highly efficient, rapid optical agonism following conjugation to SNAP-tagged mGluR2 and permits robust optical control of mGluR1 and mGluR5 signaling. We then produce fluorophore-conjugated branched PORTLs to enable dual imaging and manipulation of mGluRs and highlight their power in ex vivo slice and in vivo behavioral experiments in the mouse prefrontal cortex. Finally, we demonstrate the generalizability of our strategy by developing an improved soluble, photoswitchable pore blocker for potassium channels.
Collapse
Affiliation(s)
- Vanessa A Gutzeit
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Amanda Acosta-Ruiz
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Stephanie Häfner
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Arnaud Landra-Willm
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Bettina Mathes
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Jürgen Mony
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Dzianis Yarotski
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Karl Börjesson
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Conor Liston
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Guillaume Sandoz
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Joshua Levitz
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Johannes Broichhagen
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany; Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany.
| |
Collapse
|
21
|
Borghese CM, Wang HYL, McHardy SF, Messing RO, Trudell JR, Harris RA, Bertaccini EJ. Modulation of α1β3γ2 GABA A receptors expressed in X. laevis oocytes using a propofol photoswitch tethered to the transmembrane helix. Proc Natl Acad Sci U S A 2021; 118:e2008178118. [PMID: 33593898 PMCID: PMC7923644 DOI: 10.1073/pnas.2008178118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tethered photoswitches are molecules with two photo-dependent isomeric forms, each with different actions on their biological targets. They include reactive chemical groups capable of covalently binding to their target. Our aim was to develop a β-subunit-tethered propofol photoswitch (MAP20), as a tool to better study the mechanism of anesthesia through the GABAA α1β3γ2 receptor. We used short spacers between the tether (methanethiosulfonate), the photosensitive moiety (azobenzene), and the ligand (propofol), to allow a precise tethering adjacent to the putative propofol binding site at the β+α- interface of the receptor transmembrane helices (TMs). First, we used molecular modeling to identify possible tethering sites in β3TM3 and α1TM1, and then introduced cysteines in the candidate positions. Two mutant subunits [β3(M283C) and α1(V227C)] showed photomodulation of GABA responses after incubation with MAP20 and illumination with lights at specific wavelengths. The α1β3(M283C)γ2 receptor showed the greatest photomodulation, which decreased as GABA concentration increased. The location of the mutations that produced photomodulation confirmed that the propofol binding site is located in the β+α- interface close to the extracellular side of the transmembrane helices. Tethering the photoswitch to cysteines introduced in the positions homologous to β3M283 in two other subunits (α1W288 and γ2L298) also produced photomodulation, which was not entirely reversible, probably reflecting the different nature of each interface. The results are in agreement with a binding site in the β+α- interface for the anesthetic propofol.
Collapse
Affiliation(s)
- Cecilia M Borghese
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712;
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Hua-Yu L Wang
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX 78249
| | - Stanton F McHardy
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX 78249
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - James R Trudell
- Department of Anesthesia, Stanford University, Palo Alto, CA 94305
- Beckman Program for Molecular and Genetic Medicine, Stanford University, Palo Alto, CA 94305
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Edward J Bertaccini
- Department of Anesthesia, Stanford University, Palo Alto, CA 94305
- Department of Anesthesia, Palo Alto VA Health Care System, Palo Alto Division, Palo Alto, CA 94304
| |
Collapse
|
22
|
Dunning K, Grutter T. Manipulation of ion channel gating with photoswitchable tweezers. Methods Enzymol 2021; 653:349-376. [PMID: 34099179 DOI: 10.1016/bs.mie.2020.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The modulation of ion channel activity is of central importance within the nervous system, and an in-depth understanding of how such activity occurs on the molecular level is of prime importance for enhancing our understanding of neuronal systems in physiological and pathological states. The use of light as a stimulus has presented the unique opportunity to study these dynamic processes with exquisite spatiotemporal control. We have developed the photoswitchable tweezers method, an optogenetic pharmacology-based technique which relies on the use of a photoswitchable crosslinker as "tweezers" to manipulate the molecular movements involved in ion channel functionalities. Not only does this allow optical control of ion channel activity, but also investigation into the molecular motions and inter-residue distances implicated in such activity. In this chapter we discuss the principles behind the photoswitchable tweezers method, its strategic design and the key experimental steps involved in this technique, using purinergic P2X2 receptor as a case study system.
Collapse
Affiliation(s)
- Kate Dunning
- University of Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Thomas Grutter
- University of Strasbourg, Centre National de la Recherche Scientifique, Strasbourg, France; University of Strasbourg Institute for Advanced Studies (USIAS), Strasbourg, France.
| |
Collapse
|
23
|
Subunit-Specific Photocontrol of Glycine Receptors by Azobenzene-Nitrazepam Photoswitcher. eNeuro 2021; 8:ENEURO.0294-20.2020. [PMID: 33298457 PMCID: PMC7877471 DOI: 10.1523/eneuro.0294-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Photopharmacology is a unique approach that through a combination of photochemistry methods and advanced life science techniques allows the study and control of specific biological processes, ranging from intracellular pathways to brain circuits. Recently, a first photochromic channel blocker of anion-selective GABAA receptors, the azobenzene-nitrazepam-based photochromic compound (Azo-NZ1), has been described. In the present study, using patch-clamp technique in heterologous system and in mice brain slices, site-directed mutagenesis and molecular modeling we provide evidence of the interaction of Azo-NZ1 with glycine receptors (GlyRs) and determine the molecular basis of this interaction. Glycinergic synaptic neurotransmission determines an important inhibitory drive in the vertebrate nervous system and plays a crucial role in the control of neuronal circuits in the spinal cord and brain stem. GlyRs are involved in locomotion, pain sensation, breathing, and auditory function, as well as in the development of such disorders as hyperekplexia, epilepsy, and autism. Here, we demonstrate that Azo-NZ1 blocks in a UV-dependent manner the activity of α2 GlyRs (GlyR2), while being barely active on α1 GlyRs (GlyR1). The site of Azo-NZ1 action is in the chloride-selective pore of GlyR at the 2’ position of transmembrane helix 2 and amino acids forming this site determine the difference in Azo-NZ1 blocking activity between GlyR2 and GlyR1. This subunit-specific modulation is also shown on motoneurons of brainstem slices from neonatal mice that switch during development from expressing “fetal” GlyR2 to “adult” GlyR1 receptors.
Collapse
|
24
|
Davenport CM, Rajappa R, Katchan L, Taylor CR, Tsai MC, Smith CM, de Jong JW, Arnold DB, Lammel S, Kramer RH. Relocation of an Extrasynaptic GABA A Receptor to Inhibitory Synapses Freezes Excitatory Synaptic Strength and Preserves Memory. Neuron 2021; 109:123-134.e4. [PMID: 33096025 PMCID: PMC7790995 DOI: 10.1016/j.neuron.2020.09.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/21/2020] [Accepted: 09/25/2020] [Indexed: 11/27/2022]
Abstract
The excitatory synapse between hippocampal CA3 and CA1 pyramidal neurons exhibits long-term potentiation (LTP), a positive feedback process implicated in learning and memory in which postsynaptic depolarization strengthens synapses, promoting further depolarization. Without mechanisms for interrupting positive feedback, excitatory synapses could strengthen inexorably, corrupting memory storage. Here, we reveal a hidden form of inhibitory synaptic plasticity that prevents accumulation of excitatory LTP. We developed a knockin mouse that allows optical control of endogenous α5-subunit-containing γ-aminobutyric acid (GABA)A receptors (α5-GABARs). Induction of excitatory LTP relocates α5-GABARs, which are ordinarily extrasynaptic, to inhibitory synapses, quashing further NMDA receptor activation necessary for inducing more excitatory LTP. Blockade of α5-GABARs accelerates reversal learning, a behavioral test for cognitive flexibility dependent on repeated LTP. Hence, inhibitory synaptic plasticity occurs in parallel with excitatory synaptic plasticity, with the ensuing interruption of the positive feedback cycle of LTP serving as a possible critical early step in preserving memory.
Collapse
Affiliation(s)
- Christopher M Davenport
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rajit Rajappa
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ljudmila Katchan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Charlotte R Taylor
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ming-Chi Tsai
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Caleb M Smith
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Johannes W de Jong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Don B Arnold
- Department of Biology, Section of Molecular and Computational Biology, University of Southern California, Los Angeles, Los Angeles, CA 90089, USA
| | - Stephan Lammel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
25
|
Lemoine D, Mondoloni S, Tange J, Lambolez B, Faure P, Taly A, Tricoire L, Mourot A. Probing the ionotropic activity of glutamate GluD2 receptor in HEK cells with genetically-engineered photopharmacology. eLife 2020; 9:59026. [PMID: 33112237 PMCID: PMC7679134 DOI: 10.7554/elife.59026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Glutamate delta (GluD) receptors belong to the ionotropic glutamate receptor family, yet they don’t bind glutamate and are considered orphan. Progress in defining the ion channel function of GluDs in neurons has been hindered by a lack of pharmacological tools. Here, we used a chemo-genetic approach to engineer specific and photo-reversible pharmacology in GluD2 receptor. We incorporated a cysteine mutation in the cavity located above the putative ion channel pore, for site-specific conjugation with a photoswitchable pore blocker. In the constitutively open GluD2 Lurcher mutant, current could be rapidly and reversibly decreased with light. We then transposed the cysteine mutation to the native receptor, to demonstrate with high pharmacological specificity that metabotropic glutamate receptor signaling triggers opening of GluD2. Our results assess the functional relevance of GluD2 ion channel and introduce an optogenetic tool that will provide a novel and powerful means for probing GluD2 ionotropic contribution to neuronal physiology. Neurotransmitters are chemicals released by the body that trigger activity in neurons. Receptors on the surface of neurons detect these neurotransmitters, providing a link between the inside and the outside of the cell. Glutamate is one of the major neurotransmitters and is involved in virtually all brain functions. Glutamate binds to two different types of receptors in neurons. Ionotropic receptors have pores known as ion channels, which open when glutamate binds. This is a fast-acting response that allows sodium ions to flow into the neuron, triggering an electrical signal. Metabotropic receptors, on the other hand, trigger a series of events inside the cell that lead to a response. Metabotropic receptors take more time than ionotropic receptors to elicit a response in the cell, but their effects last much longer. One type of receptor, known as the GluD family, is very similar to ionotropic glutamate receptors but does not directly respond to glutamate. Instead, the ion channel of GluD receptors opens after being activated by glutamate metabotropic receptors. GluD receptors are produced throughout the brain and play roles in synapse formation and activity, but the way they work remains unclear. An obstacle to understanding how GluD receptors work is the lack of molecules that can specifically block these receptors’ ion channel activity. Lemoine et al. have developed a tool that enables control of the ion channel in GluD receptors using light. Human cells grown in the lab were genetically modified to produce a version of GluD2 (a member of the GluD family) with a light-sensitive molecule attached. In darkness or under green light, the light-sensitive molecule blocks the channel and prevents ions from passing through. Under violet light, the molecule twists, and ions can flow through the channel. With this control over the GluD2 ion channel activity, Lemoine et al. were able to validate previous research showing that the activation of metabotropic glutamate receptors can trigger GluD2 to open. The next step will be to test this approach in neurons. This will help researchers to understand what role GluD ion channels play in neuron to neuron communication.
Collapse
Affiliation(s)
- Damien Lemoine
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Sarah Mondoloni
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Jérome Tange
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Bertrand Lambolez
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Philippe Faure
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Antoine Taly
- CNRS, Université de Paris, UPR 9080, Laboratoire de Biochimie Théorique, Paris, France.,Institut de Biologie Physico-Chimique-Fondation Edmond de Rothschild, PSL Research University, Paris, France
| | - Ludovic Tricoire
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Alexandre Mourot
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| |
Collapse
|
26
|
Vaden RJ, Gonzalez JC, Tsai MC, Niver AJ, Fusilier AR, Griffith CM, Kramer RH, Wadiche JI, Overstreet-Wadiche L. Parvalbumin interneurons provide spillover to newborn and mature dentate granule cells. eLife 2020; 9:54125. [PMID: 32602839 PMCID: PMC7326496 DOI: 10.7554/elife.54125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/19/2020] [Indexed: 01/09/2023] Open
Abstract
Parvalbumin-expressing interneurons (PVs) in the dentate gyrus provide activity-dependent regulation of adult neurogenesis as well as maintain inhibitory control of mature neurons. In mature neurons, PVs evoke GABAA postsynaptic currents (GPSCs) with fast rise and decay phases that allow precise control of spike timing, yet synaptic currents with fast kinetics do not appear in adult-born neurons until several weeks after cell birth. Here we used mouse hippocampal slices to address how PVs signal to newborn neurons prior to the appearance of fast GPSCs. Whereas PV-evoked currents in mature neurons exhibit hallmark fast rise and decay phases, newborn neurons display slow GPSCs with characteristics of spillover signaling. We also unmasked slow spillover currents in mature neurons in the absence of fast GPSCs. Our results suggest that PVs mediate slow spillover signaling in addition to conventional fast synaptic signaling, and that spillover transmission mediates activity-dependent regulation of early events in adult neurogenesis.
Collapse
Affiliation(s)
- Ryan J Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Jose Carlos Gonzalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Ming-Chi Tsai
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, United States
| | - Anastasia J Niver
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Allison R Fusilier
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Chelsea M Griffith
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Richard H Kramer
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, United States
| | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | | |
Collapse
|
27
|
Zhou C, Ji Y, Ren L, Shao X. Photochromic meta-diamides for optical modulation of ligand activity and neuron function†. Photochem Photobiol Sci 2020; 19:854-857. [PMID: 33856679 DOI: 10.1039/d0pp00045k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/28/2020] [Indexed: 05/18/2024]
Abstract
Photopharmacology offers facile solutions for spatiotemporal control over ligand activity and receptor function. The meta-diamide insecticide acts on insect GABA receptors (GABARs) as an antagonist that causes firing of a neuron. We present here photochromic GABAR ligands azobenzene-meta-diamides (ABMDAs) by incorporating photoswitchable azobenzene with meta-diamides. ABMDAs showed good isomerization efficiency and fatigue resistance. Among them, ABMDA7 shows a 1.5-fold insecticidal activity difference towards mosquito larvae (Aedes albopictus) before and after UV illumination. We translated this light-dependent activity difference to the optical modulation of the membrane potential of American cockroach (Periptaneta americana) DUM neurons. This light-responsive meta-diamide-based GABAR ligand allows for optical regulation of insecticidal activity and DUM neurons.
Collapse
Affiliation(s)
- Cuncun Zhou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yunfan Ji
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Liping Ren
- Sinochem Agro Co., Ltd, Sinochem International Plaza, NO. 233 North Changqing Rd., Pudong New Area, Shanghai, 200126, China
| | - Xusheng Shao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
28
|
Mortensen M, Huckvale R, Pandurangan AP, Baker JR, Smart TG. Optopharmacology reveals a differential contribution of native GABA A receptors to dendritic and somatic inhibition using azogabazine. Neuropharmacology 2020; 176:108135. [PMID: 32445639 PMCID: PMC7482436 DOI: 10.1016/j.neuropharm.2020.108135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 05/09/2020] [Indexed: 12/13/2022]
Abstract
γ-aminobutyric acid type-A receptors (GABAARs) are inhibitory ligand-gated ion channels in the brain that are crucial for controlling neuronal excitation. To explore their physiological roles in cellular and neural network activity, it is important to understand why specific GABAAR isoforms are distributed not only to various brain regions and cell types, but also to specific areas of the membrane in individual neurons. To address this aim we have developed a novel photosensitive compound, azogabazine, that targets and reversibly inhibits GABAARs. The receptor selectivity of the compound is based on the competitive antagonist, gabazine, and photosensitivity is conferred by a photoisomerisable azobenzene group. Azogabazine can exist in either cis or trans conformations that are controlled by UV and blue light respectively, to affect receptor inhibition. We report that the trans-isomer preferentially binds and inhibits GABAAR function, whilst promotion of the cis-isomer caused unbinding of azogabazine from GABAARs. Using cultured cerebellar granule cells, azogabazine in conjunction with UV light applied to defined membrane domains, revealed higher densities of GABAARs at somatic inhibitory synapses compared to those populating proximal dendritic zones, even though the latter displayed a higher number of synapses per unit area of membrane. Azogabazine also revealed more pronounced GABA-mediated inhibition of action potential firing in proximal dendrites compared to the soma. Overall, azogabazine is a valuable addition to the photochemical toolkit that can be used to interrogate GABAAR function and inhibition.
Collapse
Affiliation(s)
- Martin Mortensen
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Rosemary Huckvale
- The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Arun P Pandurangan
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - James R Baker
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK
| | - Trevor G Smart
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
29
|
Photocontrol of Metabotropic Glutamate Receptors: When One Agonist Is Not Enough, Make It Two. Neuron 2020; 105:395-397. [PMID: 32027826 DOI: 10.1016/j.neuron.2020.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A current major challenge lies in controlling molecularly defined brain receptor and channel populations to investigate their function in vivo. In this issue of Neuron, Acosta-Ruiz et al. (2020) developed a highly versatile molecular toolkit to efficiently manipulate specific metabotropic glutamate receptor subtypes in brain circuits with light.
Collapse
|
30
|
Paoletti P, Ellis-Davies GCR, Mourot A. Optical control of neuronal ion channels and receptors. Nat Rev Neurosci 2020; 20:514-532. [PMID: 31289380 DOI: 10.1038/s41583-019-0197-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Light-controllable tools provide powerful means to manipulate and interrogate brain function with relatively low invasiveness and high spatiotemporal precision. Although optogenetic approaches permit neuronal excitation or inhibition at the network level, other technologies, such as optopharmacology (also known as photopharmacology) have emerged that provide molecular-level control by endowing light sensitivity to endogenous biomolecules. In this Review, we discuss the challenges and opportunities of photocontrolling native neuronal signalling pathways, focusing on ion channels and neurotransmitter receptors. We describe existing strategies for rendering receptors and channels light sensitive and provide an overview of the neuroscientific insights gained from such approaches. At the crossroads of chemistry, protein engineering and neuroscience, optopharmacology offers great potential for understanding the molecular basis of brain function and behaviour, with promises for future therapeutics.
Collapse
Affiliation(s)
- Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France.
| | | | - Alexandre Mourot
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| |
Collapse
|
31
|
Abstract
As the inhibitory γ-aminobutyric acid-ergic (GABAergic) transmission has a pivotal role in the central nervous system (CNS) and defective forms of its synapses are associated with serious neurological disorders, numerous versions of caged GABA and, more recently, photoswitchable ligands have been developed to investigate such transmission. While the complementary nature of these probes is evident, the mechanisms by which the GABA receptors can be photocontrolled have not been fully exploited. In fact, the ultimate need for specificity is critical for the proper synaptic exploration. No caged allosteric modulators of the GABAA receptor have been reported so far; to introduce such an investigational approach, we exploited the structural motifs of the benzodiazepinic scaffold to develop a photocaged version of diazepam (CD) that was tested on basolateral amygdala (BLa) pyramidal cells in mouse brain slices. CD is devoid of any intrinsic activity toward the GABAA receptor before irradiation. Importantly, CD is a photoreleasable GABAA receptor-positive allosteric modulator that offers a different probing mechanism compared to caged GABA and photoswitchable ligands. CD potentiates the inhibitory signaling by prolonging the decay time of postsynaptic GABAergic currents upon photoactivation. Additionally, no effect on presynaptic GABA release was recorded. We developed a photochemical technology to individually study the GABAA receptor, which specifically expands the toolbox available to study GABAergic synapses.
Collapse
|
32
|
Mondoloni S, Durand-de Cuttoli R, Mourot A. Cell-Specific Neuropharmacology. Trends Pharmacol Sci 2019; 40:696-710. [PMID: 31400823 DOI: 10.1016/j.tips.2019.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/04/2019] [Accepted: 07/11/2019] [Indexed: 01/12/2023]
Abstract
Neuronal communication involves a multitude of neurotransmitters and an outstanding diversity of receptors and ion channels. Linking the activity of cell surface receptors and ion channels in defined neural circuits to brain states and behaviors has been a key challenge in neuroscience, since cell targeting is not possible with traditional neuropharmacology. We review here recent technologies that enable the effect of drugs to be restricted to specific cell types, thereby allowing acute manipulation of the brain's own proteins with circuit specificity. We highlight the importance of developing cell-specific neuropharmacology strategies for decoding the nervous system with molecular and circuit precision, and for developing future therapeutics with reduced side effects.
Collapse
Affiliation(s)
- Sarah Mondoloni
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Romain Durand-de Cuttoli
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France; Nash Family Department of Neuroscience, Center for Affective Neuroscience, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandre Mourot
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| |
Collapse
|
33
|
Maleeva G, Wutz D, Rustler K, Nin-Hill A, Rovira C, Petukhova E, Bautista-Barrufet A, Gomila-Juaneda A, Scholze P, Peiretti F, Alfonso-Prieto M, König B, Gorostiza P, Bregestovski P. A photoswitchable GABA receptor channel blocker. Br J Pharmacol 2019; 176:2661-2677. [PMID: 30981211 PMCID: PMC6609548 DOI: 10.1111/bph.14689] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/03/2019] [Accepted: 04/03/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Anion-selective Cys-loop receptors (GABA and glycine receptors) provide the main inhibitory drive in the CNS. Both types of receptor operate via chloride-selective ion channels, though with different kinetics, pharmacological profiles, and localization. Disequilibrium in their function leads to a variety of disorders, which are often treated with allosteric modulators. The few available GABA and glycine receptor channel blockers effectively suppress inhibitory currents in neurons, but their systemic administration is highly toxic. With the aim of developing an efficient light-controllable modulator of GABA receptors, we constructed azobenzene-nitrazepam (Azo-NZ1), which is composed of a nitrazepam moiety merged to an azobenzene photoisomerizable group. EXPERIMENTAL APPROACH The experiments were carried out on cultured cells expressing Cys-loop receptors of known subunit composition and in brain slices using patch-clamp. Site-directed mutagenesis and molecular modelling approaches were applied to evaluate the mechanism of action of Azo-NZ1. KEY RESULTS At visible light, being in trans-configuration, Azo-NZ1 blocked heteromeric α1/β2/γ2 GABAA receptors, ρ2 GABAA (GABAC ), and α2 glycine receptors, whereas switching the compound into cis-state by UV illumination restored the activity. Azo-NZ1 successfully photomodulated GABAergic currents recorded from dentate gyrus neurons. We demonstrated that in trans-configuration, Azo-NZ1 blocks the Cl-selective ion pore of GABA receptors interacting mainly with the 2' level of the TM2 region. CONCLUSIONS AND IMPLICATIONS Azo-NZ1 is a soluble light-driven Cl-channel blocker, which allows photo-modulation of the activity induced by anion-selective Cys-loop receptors. Azo-NZ1 is able to control GABAergic postsynaptic currents and provides new opportunities to study inhibitory neurotransmission using patterned illumination.
Collapse
Affiliation(s)
- Galyna Maleeva
- INSERM, INS, Institut de Neurosciences des Systèmes, Aix-Marseille University, Marseille, France
| | - Daniel Wutz
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Karin Rustler
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Alba Nin-Hill
- Departament de Química Inorgànica i Orgànica (Secció de Química Orgànica) and Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona, Spain
| | - Carme Rovira
- Departament de Química Inorgànica i Orgànica (Secció de Química Orgànica) and Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Elena Petukhova
- Department of Normal Physiology, Kazan State Medical University, Kazan, Russia
| | - Antoni Bautista-Barrufet
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Alexandre Gomila-Juaneda
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Petra Scholze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Franck Peiretti
- INSERM 1263, INRA 1260, C2VN, Aix-Marseille Université, Marseille, France
| | - Mercedes Alfonso-Prieto
- Department of Computational Biomedicine, Institute for Advanced Simulations IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, Jülich, Germany.,Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Burkhard König
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,Network Biomedical Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBER-BBN), Spain
| | - Piotr Bregestovski
- INSERM, INS, Institut de Neurosciences des Systèmes, Aix-Marseille University, Marseille, France.,Department of Normal Physiology, Kazan State Medical University, Kazan, Russia.,Institute of Neurosciences, Kazan State Medical University, Kazan, Russia
| |
Collapse
|
34
|
Leippe P, Frank JA. Designing azobenzene-based tools for controlling neurotransmission. Curr Opin Struct Biol 2019; 57:23-30. [PMID: 30825844 DOI: 10.1016/j.sbi.2019.01.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/13/2019] [Accepted: 01/28/2019] [Indexed: 12/17/2022]
Abstract
Chemical and electrical signaling at the synapse is a dynamic process that is crucial to neurotransmission and pathology. Traditional pharmacotherapy has found countless applications in both academic labs and the clinic; however, diffusible drugs lack spatial and temporal precision when employed in heterogeneous tissues such as the brain. In the field of photopharmacology, chemical attachment of a synthetic photoswitch to a bioactive ligand allows cellular signaling to be controlled with light. Azobenzenes have remained the go-to photoswitch for biological applications due to their tunable photophysical properties, and can be leveraged to achieve reversible optical control of numerous receptors and ion channels. Here, we discuss the most recent advances in photopharmacology which will improve the use of azobenzene-based probes for neuroscience applications.
Collapse
Affiliation(s)
- Philipp Leippe
- Max Planck Institute for Medical Research, Department of Chemical Biology, Jahnstr. 29, 69120 Heidelberg, Germany
| | - James Allen Frank
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| |
Collapse
|
35
|
Carmi I, De Battista M, Maddalena L, Carroll EC, Kienzler MA, Berlin S. Holographic two-photon activation for synthetic optogenetics. Nat Protoc 2019; 14:864-900. [PMID: 30804570 DOI: 10.1038/s41596-018-0118-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022]
Abstract
Optogenetic tools provide users the ability to photocontrol the activity of cells. Commonly, activation is achieved by expression of proteins from photosynthetic organisms, for example, microbial opsins (e.g., ChR2). Alternatively, a sister approach, synthetic optogenetics, enables photocontrol over proteins of mammalian origin by use of photoswitches, visible light (typically), and genetic modification. Thus, synthetic optogenetics facilitates interrogation of native neuronal signaling mechanisms. However, the poor tissue penetration of visible wavelengths impedes the use of the technique in tissue, as two-photon excitation (2PE) is typically required to access the near-infrared window. Here, we describe an alternative technique that uses 2PE-compatible photoswitches (section 1) for photoactivation of genetically modified glutamate receptors (section 2). Furthermore, for fast, multi-region photoactivation, we describe the use of 2P-digital holography (2P-DH) (section 3). We detail how to combine 2P-DH and synthetic optogenetics with electrophysiology, or with red fluorescence Ca2+ recordings, for all-optical neural interrogation. The time required to complete the methods, aside from obtaining the necessary reagents and illumination equipment, is ~3 weeks.
Collapse
Affiliation(s)
- Ido Carmi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Marco De Battista
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Laura Maddalena
- Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
| | - Elizabeth C Carroll
- Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
| | | | - Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
36
|
Abstract
γ-aminobutyric acid has become one of the most widely known neurotransmitter molecules in the brain over the last 50 years, recognised for its pivotal role in inhibiting neural excitability. It emerged from studies of crustacean muscle and neurons before its significance to the mammalian nervous system was appreciated. Now, after five decades of investigation, we know that most neurons are γ-aminobutyric-acid-sensitive, it is a cornerstone of neural physiology and dysfunction to γ-aminobutyric acid signalling is increasingly documented in a range of neurological diseases. In this review, we briefly chart the neurodevelopment of γ-aminobutyric acid and its two major receptor subtypes: the γ-aminobutyric acidA and γ-aminobutyric acidB receptors, starting from the humble invertebrate origins of being an 'interesting molecule' acting at a single γ-aminobutyric acid receptor type, to one of the brain's most important neurochemical components and vital drug targets for major therapeutic classes of drugs. We document the period of molecular cloning and the explosive influence this had on the field of neuroscience and pharmacology up to the present day and the production of atomic γ-aminobutyric acidA and γ-aminobutyric acidB receptor structures. γ-Aminobutyric acid is no longer a humble molecule but the instigator of rich and powerful signalling processes that are absolutely vital for healthy brain function.
Collapse
Affiliation(s)
- Trevor G Smart
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | | |
Collapse
|
37
|
Luchkina NV, Bolshakov VY. Diminishing fear: Optogenetic approach toward understanding neural circuits of fear control. Pharmacol Biochem Behav 2018; 174:64-79. [PMID: 28502746 PMCID: PMC5681900 DOI: 10.1016/j.pbb.2017.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/13/2017] [Accepted: 05/10/2017] [Indexed: 02/05/2023]
Abstract
Understanding complex behavioral processes, both learned and innate, requires detailed characterization of the principles governing signal flow in corresponding neural circuits. Previous studies were hampered by the lack of appropriate tools needed to address the complexities of behavior-driving micro- and macrocircuits. The development and implementation of optogenetic methodologies revolutionized the field of behavioral neuroscience, allowing precise spatiotemporal control of specific, genetically defined neuronal populations and their functional connectivity both in vivo and ex vivo, thus providing unprecedented insights into the cellular and network-level mechanisms contributing to behavior. Here, we review recent pioneering advances in behavioral studies with optogenetic tools, focusing on mechanisms of fear-related behavioral processes with an emphasis on approaches which could be used to suppress fear when it is pathologically expressed. We also discuss limitations of these methodologies as well as review new technological developments which could be used in future mechanistic studies of fear behavior.
Collapse
Affiliation(s)
- Natalia V Luchkina
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
| | - Vadim Y Bolshakov
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
| |
Collapse
|
38
|
Durand-de Cuttoli R, Mondoloni S, Marti F, Lemoine D, Nguyen C, Naudé J, d'Izarny-Gargas T, Pons S, Maskos U, Trauner D, Kramer RH, Faure P, Mourot A. Manipulating midbrain dopamine neurons and reward-related behaviors with light-controllable nicotinic acetylcholine receptors. eLife 2018; 7:37487. [PMID: 30176987 PMCID: PMC6122951 DOI: 10.7554/elife.37487] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
Dopamine (DA) neurons of the ventral tegmental area (VTA) integrate cholinergic inputs to regulate key functions such as motivation and goal-directed behaviors. Yet the temporal dynamic range and mechanism of action of acetylcholine (ACh) on the modulation of VTA circuits and reward-related behaviors are not known. Here, we used a chemical-genetic approach for rapid and precise optical manipulation of nicotinic neurotransmission in VTA neurons in living mice. We provide direct evidence that the ACh tone fine-tunes the firing properties of VTA DA neurons through β2-containing (β2*) nicotinic ACh receptors (nAChRs). Furthermore, locally photo-antagonizing these receptors in the VTA was sufficient to reversibly switch nicotine reinforcement on and off. By enabling control of nicotinic transmission in targeted brain circuits, this technology will help unravel the various physiological functions of nAChRs and may assist in the design of novel therapies relevant to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Romain Durand-de Cuttoli
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Sarah Mondoloni
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Fabio Marti
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Damien Lemoine
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Claire Nguyen
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Jérémie Naudé
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Thibaut d'Izarny-Gargas
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Stéphanie Pons
- Unité de Neurobiologie Intégrative des Systèmes Cholinergiques, Department of Neuroscience, Institut Pasteur, Paris, France
| | - Uwe Maskos
- Unité de Neurobiologie Intégrative des Systèmes Cholinergiques, Department of Neuroscience, Institut Pasteur, Paris, France
| | - Dirk Trauner
- Department of Chemistry, New York University, New York, United States
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, United States
| | - Philippe Faure
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| | - Alexandre Mourot
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
39
|
Lorenz-Guertin JM, Bambino MJ, Jacob TC. γ2 GABA AR Trafficking and the Consequences of Human Genetic Variation. Front Cell Neurosci 2018; 12:265. [PMID: 30190672 PMCID: PMC6116786 DOI: 10.3389/fncel.2018.00265] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/02/2018] [Indexed: 11/13/2022] Open
Abstract
GABA type A receptors (GABAARs) mediate the majority of fast inhibitory neurotransmission in the central nervous system (CNS). Most prevalent as heteropentamers composed of two α, two β, and a γ2 subunit, these ligand-gated ionotropic chloride channels are capable of extensive genetic diversity (α1-6, β1-3, γ1-3, δ, 𝜀, 𝜃, π, ρ1-3). Part of this selective GABAAR assembly arises from the critical role for γ2 in maintaining synaptic receptor localization and function. Accordingly, mutations in this subunit account for over half of the known epilepsy-associated genetic anomalies identified in GABAARs. Fundamental structure-function studies and cellular pathology investigations have revealed dynamic GABAAR trafficking and synaptic scaffolding as critical regulators of GABAergic inhibition. Here, we introduce in vitro and in vivo findings regarding the specific role of the γ2 subunit in receptor trafficking. We then examine γ2 subunit human genetic variation and assess disease related phenotypes and the potential role of altered GABAAR trafficking. Finally, we discuss new-age imaging techniques and their potential to provide novel insight into critical regulatory mechanisms of GABAAR function.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew J Bambino
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
40
|
Affiliation(s)
- Katharina Hüll
- Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003-6699, United States
| | - Johannes Morstein
- Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003-6699, United States
| | - Dirk Trauner
- Department of Chemistry, New York University, 100 Washington Square East, New York, New York 10003-6699, United States
| |
Collapse
|
41
|
The Benzodiazepine Binding Sites of GABAA Receptors. Trends Pharmacol Sci 2018; 39:659-671. [DOI: 10.1016/j.tips.2018.03.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/15/2018] [Accepted: 03/22/2018] [Indexed: 11/24/2022]
|
42
|
Lin WC, Tsai MC, Rajappa R, Kramer RH. Design of a Highly Bistable Photoswitchable Tethered Ligand for Rapid and Sustained Manipulation of Neurotransmission. J Am Chem Soc 2018; 140:7445-7448. [PMID: 29874068 PMCID: PMC6422952 DOI: 10.1021/jacs.8b03942] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Photoswitchable neurotransmitter receptors are powerful tools for precise manipulation of neural signaling. However, their applications for slow or long-lasting biological events are constrained by fast thermal relaxation of cis-azobenzene. We address this issue by modifying the ortho positions of azobenzene used in the tethered ligand. In cultured cells and intact brain tissue, conjugating inhibitory neurotransmitter receptors with one of the derivatives, dMPC1, allows bidirectional receptor control with 380 and 500 nm light. Moreover, the receptors can be locked in either an active or an inactive state in darkness after a brief pulse of light. This strategy thus enables both rapid and sustained manipulation of neurotransmission, allowing optogenetic interrogation of neural functions over a broad range of time scales.
Collapse
Affiliation(s)
- Wan-Chen Lin
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Ming-Chi Tsai
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Rajit Rajappa
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Richard H. Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| |
Collapse
|
43
|
The State of the NIH BRAIN Initiative. J Neurosci 2018; 38:6427-6438. [PMID: 29921715 DOI: 10.1523/jneurosci.3174-17.2018] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022] Open
Abstract
The BRAIN Initiative arose from a grand challenge to "accelerate the development and application of new technologies that will enable researchers to produce dynamic pictures of the brain that show how individual brain cells and complex neural circuits interact at the speed of thought." The BRAIN Initiative is a public-private effort focused on the development and use of powerful tools for acquiring fundamental insights about how information processing occurs in the central nervous system (CNS). As the Initiative enters its fifth year, NIH has supported >500 principal investigators, who have answered the Initiative's challenge via hundreds of publications describing novel tools, methods, and discoveries that address the Initiative's seven scientific priorities. We describe scientific advances produced by individual laboratories, multi-investigator teams, and entire consortia that, over the coming decades, will produce more comprehensive and dynamic maps of the brain, deepen our understanding of how circuit activity can produce a rich tapestry of behaviors, and lay the foundation for understanding how its circuitry is disrupted in brain disorders. Much more work remains to bring this vision to fruition, and the National Institutes of Health continues to look to the diverse scientific community, from mathematics, to physics, chemistry, engineering, neuroethics, and neuroscience, to ensure that the greatest scientific benefit arises from this unique research Initiative.
Collapse
|
44
|
Christenson Wick Z, Krook-Magnuson E. Specificity, Versatility, and Continual Development: The Power of Optogenetics for Epilepsy Research. Front Cell Neurosci 2018; 12:151. [PMID: 29962936 PMCID: PMC6010559 DOI: 10.3389/fncel.2018.00151] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Optogenetics is a powerful and rapidly expanding set of techniques that use genetically encoded light sensitive proteins such as opsins. Through the selective expression of these exogenous light-sensitive proteins, researchers gain the ability to modulate neuronal activity, intracellular signaling pathways, or gene expression with spatial, directional, temporal, and cell-type specificity. Optogenetics provides a versatile toolbox and has significantly advanced a variety of neuroscience fields. In this review, using recent epilepsy research as a focal point, we highlight how the specificity, versatility, and continual development of new optogenetic related tools advances our understanding of neuronal circuits and neurological disorders. We additionally provide a brief overview of some currently available optogenetic tools including for the selective expression of opsins.
Collapse
Affiliation(s)
- Zoé Christenson Wick
- Graduate Program in Neuroscience and Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
45
|
An Emerging Circuit Pharmacology of GABA A Receptors. Trends Pharmacol Sci 2018; 39:710-732. [PMID: 29903580 DOI: 10.1016/j.tips.2018.04.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/23/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022]
Abstract
In the past 20 years we have learned a great deal about GABAA receptor (GABAAR) subtypes, and which behaviors are regulated or which drug effects are mediated by each subtype. However, the question of where GABAARs involved in specific drug effects and behaviors are located in the brain remains largely unanswered. We review here recent studies taking a circuit pharmacology approach to investigate the functions of GABAAR subtypes in specific brain circuits controlling fear, anxiety, learning, memory, reward, addiction, and stress-related behaviors. The findings of these studies highlight the complexity of brain inhibitory systems and the importance of taking a subtype-, circuit-, and neuronal population-specific approach to develop future therapeutic strategies using cell type-specific drug delivery.
Collapse
|
46
|
Tochitsky I, Kienzler MA, Isacoff E, Kramer RH. Restoring Vision to the Blind with Chemical Photoswitches. Chem Rev 2018; 118:10748-10773. [PMID: 29874052 DOI: 10.1021/acs.chemrev.7b00723] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Degenerative retinal diseases such as retinitis pigmentosa (RP) and age-related macular degeneration (AMD) affect millions of people around the world and lead to irreversible vision loss if left untreated. A number of therapeutic strategies have been developed over the years to treat these diseases or restore vision to already blind patients. In this Review, we describe the development and translational application of light-sensitive chemical photoswitches to restore visual function to the blind retina and compare the translational potential of photoswitches with other vision-restoring therapies. This therapeutic strategy is enabled by an efficient fusion of chemical synthesis, chemical biology, and molecular biology and is broadly applicable to other biological systems. We hope this Review will be of interest to chemists as well as neuroscientists and clinicians.
Collapse
Affiliation(s)
- Ivan Tochitsky
- F.M. Kirby Neurobiology Center , Boston Children's Hospital , Boston , Massachusetts 02115 , United States.,Department of Neurobiology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Michael A Kienzler
- Department of Chemistry , University of Maine , Orono , Maine 04469 , United States
| | - Ehud Isacoff
- Department of Molecular and Cell Biology , University of California , Berkeley , California 94720 , United States.,Helen Wills Neuroscience Institute , University of California , Berkeley , California 94720 , United States.,Bioscience Division , Lawrence Berkeley National Laboratory , Berkeley , California 94720 , United States
| | - Richard H Kramer
- 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
| |
Collapse
|
47
|
Bregestovski P, Maleeva G, Gorostiza P. Light-induced regulation of ligand-gated channel activity. Br J Pharmacol 2018; 175:1892-1902. [PMID: 28859250 PMCID: PMC5979632 DOI: 10.1111/bph.14022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/21/2017] [Accepted: 07/03/2017] [Indexed: 12/25/2022] Open
Abstract
The control of ligand-gated receptors with light using photochromic compounds has evolved from the first handcrafted examples to accurate, engineered receptors, whose development is supported by rational design, high-resolution protein structures, comparative pharmacology and molecular biology manipulations. Photoswitchable regulators have been designed and characterized for a large number of ligand-gated receptors in the mammalian nervous system, including nicotinic acetylcholine, glutamate and GABA receptors. They provide a well-equipped toolbox to investigate synaptic and neuronal circuits in all-optical experiments. This focused review discusses the design and properties of these photoswitches, their applications and shortcomings and future perspectives in the field. LINKED ARTICLES This article is part of a themed section on Nicotinic Acetylcholine Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.11/issuetoc.
Collapse
Affiliation(s)
- Piotr Bregestovski
- Aix Marseille Université, INSERM 1106 Institut de Neurosciences des SystèmesMarseilleFrance
- Department of PhysiologyKazan Medical State UniversityKazanRussia
| | - Galyna Maleeva
- Aix Marseille Université, INSERM 1106 Institut de Neurosciences des SystèmesMarseilleFrance
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and TechnologyBarcelonaSpain
- ICREABarcelonaSpain
- CIBER‐BBNMadridSpain
| |
Collapse
|
48
|
Klippenstein V, Mony L, Paoletti P. Probing Ion Channel Structure and Function Using Light-Sensitive Amino Acids. Trends Biochem Sci 2018; 43:436-451. [PMID: 29650383 DOI: 10.1016/j.tibs.2018.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 02/25/2018] [Accepted: 02/27/2018] [Indexed: 12/13/2022]
Abstract
Approaches to remotely control and monitor ion channel operation with light are expanding rapidly in the biophysics and neuroscience fields. A recent development directly introduces light sensitivity into proteins by utilizing photosensitive unnatural amino acids (UAAs) incorporated using the genetic code expansion technique. The introduction of UAAs results in unique molecular level control and, when combined with the maximal spatiotemporal resolution and poor invasiveness of light, enables direct manipulation and interrogation of ion channel functionality. Here, we review the diverse applications of light-sensitive UAAs in two superfamilies of ion channels (voltage- and ligand-gated ion channels; VGICs and LGICs) and summarize existing UAA tools, their mode of action, potential, caveats, and technical considerations to their use in illuminating ion channel structure and function.
Collapse
Affiliation(s)
- Viktoria Klippenstein
- Institut de Biologie de I'ENS (IBENS), CNRS UMR8197, INSERM U1024, Ecole Normale Supérieure, Université PSL, 46 rue d'Ulm, 75005 Paris, France; These authors contributed equally to this work
| | - Laetitia Mony
- Institut de Biologie de I'ENS (IBENS), CNRS UMR8197, INSERM U1024, Ecole Normale Supérieure, Université PSL, 46 rue d'Ulm, 75005 Paris, France; These authors contributed equally to this work
| | - Pierre Paoletti
- Institut de Biologie de I'ENS (IBENS), CNRS UMR8197, INSERM U1024, Ecole Normale Supérieure, Université PSL, 46 rue d'Ulm, 75005 Paris, France.
| |
Collapse
|
49
|
Lorenz-Guertin JM, Jacob TC. GABA type a receptor trafficking and the architecture of synaptic inhibition. Dev Neurobiol 2018; 78:238-270. [PMID: 28901728 PMCID: PMC6589839 DOI: 10.1002/dneu.22536] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
Ubiquitous expression of GABA type A receptors (GABAA R) in the central nervous system establishes their central role in coordinating most aspects of neural function and development. Dysregulation of GABAergic neurotransmission manifests in a number of human health disorders and conditions that in certain cases can be alleviated by drugs targeting these receptors. Precise changes in the quantity or activity of GABAA Rs localized at the cell surface and at GABAergic postsynaptic sites directly impact the strength of inhibition. The molecular mechanisms constituting receptor trafficking to and from these compartments therefore dictate the efficacy of GABAA R function. Here we review the current understanding of how GABAA Rs traffic through biogenesis, plasma membrane transport, and degradation. Emphasis is placed on discussing novel GABAergic synaptic proteins, receptor and scaffolding post-translational modifications, activity-dependent changes in GABAA R confinement, and neuropeptide and neurosteroid mediated changes. We further highlight modern techniques currently advancing the knowledge of GABAA R trafficking and clinically relevant neurodevelopmental diseases connected to GABAergic dysfunction. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 238-270, 2018.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| |
Collapse
|
50
|
Lin WC, Kramer RH. Light-Switchable Ion Channels and Receptors for Optogenetic Interrogation of Neuronal Signaling. Bioconjug Chem 2018; 29:861-869. [PMID: 29465988 DOI: 10.1021/acs.bioconjchem.7b00803] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Optogenetics is an emerging technique that enables precise and specific control of biological activities in defined space and time. This technique employs naturally occurring or engineered light-responsive proteins to manipulate the physiological processes of the target cells. To better elucidate the molecular bases of neural functions, substantial efforts have been made to confer light sensitivity onto ion channels and neurotransmitter receptors that mediate signaling events within and between neurons. The chemical strategies for engineering light-switchable channels/receptors and the neuronal implementation of these tools are discussed.
Collapse
Affiliation(s)
- Wan-Chen Lin
- Department of Molecular and Cell Biology , University of California, Berkeley , 121 Life Sciences Addition , Berkeley , California 94720 , United States
| | - Richard H Kramer
- Department of Molecular and Cell Biology , University of California, Berkeley , 121 Life Sciences Addition , Berkeley , California 94720 , United States
| |
Collapse
|