The First Structure–Activity Relationship Studies for Designer Receptors Exclusively Activated by Designer DrugsClick to copy article linkArticle link copied!
Abstract
Over the past decade, two independent technologies have emerged and been widely adopted by the neuroscience community for remotely controlling neuronal activity: optogenetics which utilize engineered channelrhodopsin and other opsins, and chemogenetics which utilize engineered G protein-coupled receptors (Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)) and other orthologous ligand–receptor pairs. Using directed molecular evolution, two types of DREADDs derived from human muscarinic acetylcholine receptors have been developed: hM3Dq which activates neuronal firing, and hM4Di which inhibits neuronal firing. Importantly, these DREADDs were not activated by the native ligand acetylcholine (ACh), but selectively activated by clozapine N-oxide (CNO), a pharmacologically inert ligand. CNO has been used extensively in rodent models to activate DREADDs, and although CNO is not subject to significant metabolic transformation in mice, a small fraction of CNO is apparently metabolized to clozapine in humans and guinea pigs, lessening the translational potential of DREADDs. To effectively translate the DREADD technology, the next generation of DREADD agonists are needed and a thorough understanding of structure–activity relationships (SARs) of DREADDs is required for developing such ligands. We therefore conducted the first SAR studies of hM3Dq. We explored multiple regions of the scaffold represented by CNO, identified interesting SAR trends, and discovered several compounds that are very potent hM3Dq agonists but do not activate the native human M3 receptor (hM3). We also discovered that the approved drug perlapine is a novel hM3Dq agonist with >10 000-fold selectivity for hM3Dq over hM3.
Results and Discussion
Design and Synthesis
Biological Evaluation
hM3Dq | hM3 | |||
---|---|---|---|---|
compd | EC50 (nM) | Emax (relative to CNO) | EC50 (nM) | Emax (relative to acetylcholine) |
4a | 1.1 | 95 | 360 | 88 |
4b | 7.0 | 91 | >30 000 | NA |
4c | 13 | 45 | >30 000 | NA |
4d | 71 | 50 | >30 000 | NA |
5a | 6.0 | 100 | >30 000 | NA |
5b | 19 | 50 | >30 000 | NA |
5c | 190 | 45 | >30 000 | NA |
5d | 740 | 79 | >30 000 | NA |
6 | 0.069 | 100 | 9.5 | 92 |
7 | >30 000 | NA | >30 000 | NA |
9 | >30 000 | NA | >30 000 | NA |
10 | >30 000 | NA | >30 000 | NA |
11 | 2.1 | 95 | 490 | 86 |
12 | >30 000 | NA | >30 000 | NA |
13 | 9.6 | 86 | >30 000 | NA |
14 | >30 000 | NA | >30 000 | NA |
21 | 1.7 | 100 | NA | ∼20 |
22 | 1.3 | 81 | >30 000 | NA |
23 | 220 | 59 | >30 000 | NA |
EC50 values are the average of at least two duplicate experiments with standard deviation (SD) values that are 3-fold less than the average. NA: not applicable.
Ki (nM) | ||||
---|---|---|---|---|
compd | 5HT2A | 5HT2C | α1A | H1 |
4a | 5.4 | 9.4 | 1.6 | 1.1 |
4b | 29 | 24 | 46 | 1.9 |
4c | 16 | 17 | 37 | 4.6 |
5b | 1900 | 5100 | >10 000 | 160 |
5c | 5200 | 6700 | 320 | 6200 |
13 | 71 | 280 | 67 | 5.0 |
21 | 66 | 170 | 280 | 6.0 |
Ki values are the average of at least 2 duplicate experiments with standard deviation (SD) values that are 3-fold less than the average.
Conclusion
Methods
Chemistry
General Methods
8-Chloro-11-(4-isopropylpiperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine (4c)
8-Chloro-11-(4-propylpiperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine (4d)
4-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)-1-ethylpiperazine N-oxide (5b)
4-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)-1-isopropylpiperazine N-oxide (5c)
4-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)-1-propylpiperazine N-oxide (5d)
4-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)-1,1-dimethylpiperazin-1-ium iodide (6)
4-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)piperazin-2-one (7)
8-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)-1,3,8-triazaspiro[4.5]decane-2,4-dione (9)
4-Amino-1-(8-chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)piperidine-4-carboxylic acid (10)
1-(4-(8-Chloro-5H-dibenzo[b,e][1,4]diazepin-11-yl)piperazin-1-yl)ethanone (12)
8-Chloro-11-[4-(1,1-dideutrioethyl)piperazin-1-yl]-5H-dibenzo[b,e][1,4]diazepine (13)
8-Chloro-11-(4-(methylsulfonyl)piperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine (14)
11-(Piperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine (21)
11-(4-Ethylpiperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine (22)
4-(5H-Dibenzo[b,e][1,4]diazepin-11-yl)-1-ethylpiperazine N-oxide (23)
Biological Assays
Acknowledgment
We thank Dr. H. Ümit Kaniskan for checking synthetic procedures and NMR spectra of novel compounds.
References
This article references 51 other publications.
- 1Sternson, S. M. and Roth, B. L. (2014) Chemogenetic Tools to Interrogate Brain Functions Annu. Rev. Neurosci. 37, 387– 407Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVKgtL7E&md5=43a0b89232bd0b63e787a9caab7584daChemogenetic tools to interrogate brain functionsSternson, Scott M.; Roth, Bryan L.Annual Review of Neuroscience (2014), 37 (), 387-407CODEN: ARNSD5; ISSN:0147-006X. (Annual Reviews)A review. Elucidating the roles of neuronal cell types for physiol. and behavior is essential for understanding brain functions. Perturbation of neuron elec. activity can be used to probe the causal relationship between neuronal cell types and behavior. New genetically encoded neuron perturbation tools have been developed for remotely controlling neuron function using small mols. that activate engineered receptors that can be targeted to cell types using genetic methods. Here we describe recent progress for approaches using genetically engineered receptors that selectively interact with small mols. Called "chemogenetics," receptors with diverse cellular functions have been developed that facilitate the selective pharmacol. control over a diverse range of cell-signaling processes, including elec. activity, for molecularly defined cell types. These tools have revealed remarkably specific behavioral physiol. influences for molecularly defined cell types that are often intermingled with populations having different or even opposite functions.
- 2Urban, D. J. and Roth, B. L. (2015) DREADDs (Designer Receptors Exclusively Activated by Designer Drugs): Chemogenetic Tools with Therapeutic Utility Annu. Rev. Pharmacol. Toxicol. 55, 399– 417Google ScholarThere is no corresponding record for this reference.
- 3Penfield, W. and Jasper, H. H. (1954) Epilepsy and the Functional Anatomy of the Human Brain (First ed.), Little Brown, Boston.Google ScholarThere is no corresponding record for this reference.
- 4Boyden, E. S., Zhang, F., Bamberg, E., Nagel, G., and Deisseroth, K. (2005) Millisecond-timescale, genetically targeted optical control of neural activity Nat. Neurosci. 8, 1263– 1268Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpt1egtrc%253D&md5=7d71b89559e8825798cbc4b81ccf8c32Millisecond-timescale, genetically targeted optical control of neural activityBoyden, Edward S.; Zhang, Feng; Bamberg, Ernst; Nagel, Georg; Deisseroth, KarlNature Neuroscience (2005), 8 (9), 1263-1268CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Temporally precise, noninvasive control of activity in well-defined neuronal populations is a long-sought goal of systems neuroscience. We adapted for this purpose the naturally occurring algal protein Channelrhodopsin-2, a rapidly gated light-sensitive cation channel, by using lentiviral gene delivery in combination with high-speed optical switching to photostimulate mammalian neurons. We demonstrate reliable, millisecond-timescale control of neuronal spiking, as well as control of excitatory and inhibitory synaptic transmission. This technol. allows the use of light to alter neural processing at the level of single spikes and synaptic events, yielding a widely applicable tool for neuroscientists and biomedical engineers.
- 5Armbruster, B. N. and Roth, B. L. (2005) Mining the receptorome J. Biol. Chem. 280, 5129– 5132Google ScholarThere is no corresponding record for this reference.
- 6Zhang, F., Wang, L. P., Brauner, M., Liewald, J. F., Kay, K., Watzke, N., Wood, P. G., Bamberg, E., Nagel, G., Gottschalk, A., and Deisseroth, K. (2007) Multimodal fast optical interrogation of neural circuitry Nature 446, 633– 639Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjslOltr4%253D&md5=011fc19954bb36af7622fc7b5f5b25f5Multimodal fast optical interrogation of neural circuitryZhang, Feng; Wang, Li-Ping; Brauner, Martin; Liewald, Jana F.; Kay, Kenneth; Watzke, Natalie; Wood, Phillip G.; Bamberg, Ernst; Nagel, Georg; Gottschalk, Alexander; Deisseroth, KarlNature (London, United Kingdom) (2007), 446 (7136), 633-639CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The understanding of the cellular implementation of systems-level neural processes like action, thought and emotion has been limited by the availability of tools to interrogate specific classes of neural cells within intact, living brain tissue. Here the authors identify and develop an archaeal light-driven chloride pump (NpHR) from Natronomonas pharaonis for temporally precise optical inhibition of neural activity. NpHR allows either knockout of single action potentials, or sustained blockade of spiking. NpHR is compatible with ChR2, the previous optical excitation technol. the authors have described, in that the two opposing probes operate at similar light powers but with well-sepd. action spectra. NpHR, like ChR2, functions in mammals without exogenous cofactors, and the two probes can be integrated with calcium imaging in mammalian brain tissue for bidirectional optical modulation and readout of neural activity. Likewise, NpHR and ChR2 can be targeted together to Caenorhabditis elegans muscle and cholinergic motor neurons to control locomotion bidirectionally. NpHR and ChR2 form a complete system for multimodal, high-speed, genetically targeted, all-optical interrogation of living neural circuits.
- 7Airan, R. D., Thompson, K. R., Fenno, L. E., Bernstein, H., and Deisseroth, K. (2009) Temporally precise in vivo control of intracellular signalling Nature 458, 1025– 1029Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXjsVCqur4%253D&md5=7b3f9805ba9e9a5e802247c7a272a41bTemporally precise in vivo control of intracellular signallingAiran, Raag D.; Thompson, Kimberly R.; Fenno, Lief E.; Bernstein, Hannah; Deisseroth, KarlNature (London, United Kingdom) (2009), 458 (7241), 1025-1029CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)In the study of complex mammalian behaviors, technol. limitations have prevented spatiotemporally precise control over intracellular signaling processes. Here we report the development of a versatile family of genetically encoded optical tools ('optoXRs') that leverage common structure-function relationships among G-protein-coupled receptors (GPCRs) to recruit and control, with high spatiotemporal precision, receptor-initiated biochem. signaling pathways. In particular, we have developed and characterized two optoXRs that selectively recruit distinct, targeted signaling pathways in response to light. The two optoXRs exerted opposing effects on spike firing in nucleus accumbens in vivo, and precisely timed optoXR photostimulation in nucleus accumbens by itself sufficed to drive conditioned place preference in freely moving mice. The optoXR approach allows testing of hypotheses regarding the causal impact of biochem. signaling in behaving mammals, in a targetable and temporally precise manner.
- 8Berndt, A., Yizhar, O., Gunaydin, L. A., Hegemann, P., and Deisseroth, K. (2009) Bi-stable neural state switches Nat. Neurosci. 12, 229– 234Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2hsrnI&md5=72cfb3b77c8272c77a7b8a837e37130bBi-stable neural state switchesBerndt, Andre; Yizhar, Ofer; Gunaydin, Lisa A.; Hegemann, Peter; Deisseroth, KarlNature Neuroscience (2009), 12 (2), 229-234CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Here we describe bi-stable channel rhodopsins that convert a brief pulse of light into a stable step in membrane potential. These molecularly engineered probes nevertheless retain millisecond-scale temporal precision. Photocurrents can be precisely initiated and terminated with different colors of light, but operate at vastly longer time scales than conventional channel rhodopsins as a result of modification at the C128 position that extends the lifetime of the open state. Because of their enhanced kinetic stability, these step-function tools are also effectively responsive to light at orders of magnitude lower intensity than wild-type channel rhodopsins. These mols. therefore offer important new capabilities for a broad range of in vivo applications.
- 9Li, X., Gutierrez, D. V., Hanson, M. G., Han, J., Mark, M. D., Chiel, H., Hegemann, P., Landmesser, L. T., and Herlitze, S. (2005) Fast noninvasive activation and inhibition of neural and network activity by vertebrate rhodopsin and green algae channelrhodopsin Proc. Natl. Acad. Sci. U. S. A. 102, 17816– 17821Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlegs7vO&md5=77f01d459514e66d35e9a51994353217Fast noninvasive activation and inhibition of neural and network activity by vertebrate rhodopsin and green algae channelrhodopsinLi, Xiang; Gutierrez, Davina V.; Hanson, M. Gartz; Han, Jing; Mark, Melanie D.; Chiel, Hillel; Hegemann, Peter; Landmesser, Lynn T.; Herlitze, StefanProceedings of the National Academy of Sciences of the United States of America (2005), 102 (49), 17816-17821CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Techniques for fast noninvasive control of neuronal excitability will be of major importance for analyzing and understanding neuronal networks and animal behavior. To develop these tools we demonstrated that two light-activated signaling proteins, vertebrate rat rhodopsin 4 (RO4) and the green algae channelrhodopsin 2 (ChR2), could be used to control neuronal excitability and modulate synaptic transmission. Vertebrate rhodopsin couples to the Gi/o, pertussis toxin-sensitive pathway to allow modulation of G protein-gated inward rectifying potassium channels and voltage-gated Ca2+ channels. Light-mediated activation of RO4 in cultured hippocampal neurons reduces neuronal firing within ms by hyperpolarization of the somato-dendritic membrane, and when activated at presynaptic sites modulates synaptic transmission and paired-pulse facilitation. In contrast, somato-dendritic activation of ChR2 depolarizes neurons sufficiently to induce immediate action potentials, which precisely follow the ChR2 activation up to light stimulation frequencies of 20 Hz. To demonstrate that these constructs are useful for regulating network behavior in intact organisms, embryonic chick spinal cords were electroporated with either construct, allowing the frequency of episodes of spontaneous bursting activity, known to be important for motor circuit formation, to be precisely controlled. Thus light-activated vertebrate RO4 and green algae ChR2 allow the antagonistic control of neuronal function within ms to s in a precise, reversible, and noninvasive manner in cultured neurons and intact vertebrate spinal cords.
- 10Zhang, F., Prigge, M., Beyriere, F., Tsunoda, S. P., Mattis, J., Yizhar, O., Hegemann, P., and Deisseroth, K. (2008) Red-shifted optogenetic excitation: a tool for fast neural control derived from Volvox carteri Nat. Neurosci. 11, 631– 633Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXmtlCisbw%253D&md5=307a16c246c6fd78a9b145bbfe2c0357Red-shifted optogenetic excitation: a tool for fast neural control derived from Volvox carteriZhang, Feng; Prigge, Matthias; Beyriere, Florent; Tsunoda, Satoshi P.; Mattis, Joanna; Yizhar, Ofer; Hegemann, Peter; Deisseroth, KarlNature Neuroscience (2008), 11 (6), 631-633CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)The introduction of two microbial opsin-based tools, channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), to neuroscience has generated interest in fast, multimodal, cell type-specific neural circuit control. Here the authors describe a cation-conducting channelrhodopsin (VChR1) from Volvox carteri that can drive spiking at 589 nm, with excitation max. red-shifted ∼70 nm compared with ChR2. These results demonstrate fast photostimulation with yellow light, thereby defining a functionally distinct third category of microbial rhodopsin proteins.
- 11Armbruster, B. N., Li, X., Pausch, M. H., Herlitze, S., and Roth, B. L. (2007) Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand Proc. Natl. Acad. Sci. U. S. A. 104, 5163– 5168Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2s7nt1egtQ%253D%253D&md5=935cd23bf341eff865375f7d2e253210Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligandArmbruster Blaine N; Li Xiang; Pausch Mark H; Herlitze Stefan; Roth Bryan LProceedings of the National Academy of Sciences of the United States of America (2007), 104 (12), 5163-8 ISSN:0027-8424.We evolved muscarinic receptors in yeast to generate a family of G protein-coupled receptors (GPCRs) that are activated solely by a pharmacologically inert drug-like and bioavailable compound (clozapine-N-oxide). Subsequent screening in human cell lines facilitated the creation of a family of muscarinic acetylcholine GPCRs suitable for in vitro and in situ studies. We subsequently created lines of telomerase-immortalized human pulmonary artery smooth muscle cells stably expressing all five family members and found that each one faithfully recapitulated the signaling phenotype of the parent receptor. We also expressed a G(i)-coupled designer receptor in hippocampal neurons (hM(4)D) and demonstrated its ability to induce membrane hyperpolarization and neuronal silencing. We have thus devised a facile approach for designing families of GPCRs with engineered ligand specificities. Such reverse-engineered GPCRs will prove to be powerful tools for selectively modulating signal-transduction pathways in vitro and in vivo.
- 12Alexander, G. M., Rogan, S. C., Abbas, A. I., Armbruster, B. N., Pei, Y., Allen, J. A., Nonneman, R. J., Hartmann, J., Moy, S. S., Nicolelis, M. A., McNamara, J. O., and Roth, B. L. (2009) Remote Control of Neuronal Activity in Transgenic Mice Expressing Evolved G Protein-Coupled Receptors Neuron 63, 27– 39Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXpsFOns7g%253D&md5=a7fed676083d592279f5497c6956c781Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptorsAlexander, Georgia M.; Rogan, Sarah C.; Abbas, Atheir I.; Armbruster, Blaine N.; Pei, Ying; Allen, John A.; Nonneman, Randal J.; Hartmann, John; Moy, Sheryl S.; Nicolelis, Miguel A.; McNamara, James O.; Roth, Bryan L.Neuron (2009), 63 (1), 27-39CODEN: NERNET; ISSN:0896-6273. (Cell Press)Examg. the behavioral consequences of selective CNS neuronal activation is a powerful tool for elucidating mammalian brain function in health and disease. Newly developed genetic, pharmacol., and optical tools allow activation of neurons with exquisite spatiotemporal resoln.; however, the inaccessibility to light of widely distributed neuronal populations and the invasiveness required for activation by light or infused ligands limit the utility of these methods. To overcome these barriers, we created transgenic mice expressing an evolved G protein-coupled receptor (hM3Dq) selectively activated by the pharmacol. inert, orally bioavailable drug clozapine-N-oxide (CNO). Here, we expressed hM3Dq in forebrain principal neurons. Local field potential and single-neuron recordings revealed that peripheral administration of CNO activated hippocampal neurons selectively in hM3Dq-expressing mice. Behavioral correlates of neuronal activation included increased locomotion, stereotypy, and limbic seizures. These results demonstrate a powerful chem.-genetic tool for remotely controlling the activity of discrete populations of neurons in vivo.
- 13Rogan, S. C. and Roth, B. L. (2011) Remote control of neuronal signaling Pharmacol. Rev. 63, 291– 315Google ScholarThere is no corresponding record for this reference.
- 14Farrell, M. S. and Roth, B. L. (2012) Pharmacosynthetics: Reimagining the pharmacogenetic approach Brain Res. 1511, 6– 20Google ScholarThere is no corresponding record for this reference.
- 15Jennings, J. H. and Stuber, G. D. (2014) Tools for Resolving Functional Activity and Connectivity within Intact Neural Circuits Curr. Biol. 24, R41– 50Google ScholarThere is no corresponding record for this reference.
- 16Lee, H. M., Giguere, P. M., and Roth, B. L. (2013) DREADDs: Novel tools for drug discovery and development Drug Discovery Today 19, 469– 473Google ScholarThere is no corresponding record for this reference.
- 17Giguere, P. M., Kroeze, W. K., and Roth, B. L. (2014) Tuning up the right signal: Chemical and genetic approaches to study GPCR functions Curr. Opin. Cell Biol. 27, 51– 55Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtVCiu74%253D&md5=9a1e7fdf05facf40734fd6758ac1e698Tuning up the right signal: chemical and genetic approaches to study GPCR functionsGiguere, Patrick M.; Kroeze, Wesley K.; Roth, Bryan L.Current Opinion in Cell Biology (2014), 27 (), 51-55CODEN: COCBE3; ISSN:0955-0674. (Elsevier Ltd.)A review. The G protein-coupled receptor (GPCR) family is among the most druggable families in the human proteome. GPCRs are involved in most physiol. processes, and our ability to modulate their activity is a hallmark of modern pharmacol. The means by which the activity of GPCRs can be modulated have been expanded by emerging data and concepts in pharmacol., which has created new strategies for their control. These new approaches will lead to the generation of more potent, selective, and efficient pharmaceutics, while reducing inappropriate actions and adverse effects. Herein, we review and comment on some recent advances in chem. and genetic approaches to the profiling of GPCR function, as well as the validation of orphan GPCRs as potential therapeutic targets using engineered receptors.
- 18Armbruster, B. N., Li, X., Pausch, M. H., Herlitze, S., and Roth, B. L. (2007) Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand Proc. Natl. Acad. Sci. U. S. A. 104, 5163– 5168Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2s7nt1egtQ%253D%253D&md5=935cd23bf341eff865375f7d2e253210Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligandArmbruster Blaine N; Li Xiang; Pausch Mark H; Herlitze Stefan; Roth Bryan LProceedings of the National Academy of Sciences of the United States of America (2007), 104 (12), 5163-8 ISSN:0027-8424.We evolved muscarinic receptors in yeast to generate a family of G protein-coupled receptors (GPCRs) that are activated solely by a pharmacologically inert drug-like and bioavailable compound (clozapine-N-oxide). Subsequent screening in human cell lines facilitated the creation of a family of muscarinic acetylcholine GPCRs suitable for in vitro and in situ studies. We subsequently created lines of telomerase-immortalized human pulmonary artery smooth muscle cells stably expressing all five family members and found that each one faithfully recapitulated the signaling phenotype of the parent receptor. We also expressed a G(i)-coupled designer receptor in hippocampal neurons (hM(4)D) and demonstrated its ability to induce membrane hyperpolarization and neuronal silencing. We have thus devised a facile approach for designing families of GPCRs with engineered ligand specificities. Such reverse-engineered GPCRs will prove to be powerful tools for selectively modulating signal-transduction pathways in vitro and in vivo.
- 19Ray, R. S., Corcoran, A. E., Brust, R. D., Kim, J. C., Richerson, G. B., Nattie, E., and Dymecki, S. M. (2011) Impaired Respiratory and Body Temperature Control Upon Acute Serotonergic Neuron Inhibition Science 333, 637– 642Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpt1yisb8%253D&md5=62a37269a9b28b710df2c483cc92f7a7Impaired Respiratory and Body Temperature Control Upon Acute Serotonergic Neuron InhibitionRay, Russell S.; Corcoran, Andrea E.; Brust, Rachael D.; Kim, Jun Chul; Richerson, George B.; Nattie, Eugene; Dymecki, Susan M.Science (Washington, DC, United States) (2011), 333 (6042), 637-642CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Physiol. homeostasis is essential for organism survival. Highly responsive neuronal networks are involved, but their constituent neurons are just beginning to be resolved. To query brain serotonergic neurons in homeostasis, the authors used a neuronal silencing tool, mouse RC::FPDi (based on the synthetic G protein-coupled receptor Di), designed for cell type-specific, ligand-inducible, and reversible suppression of action potential firing. In mice harboring Di-expressing serotonergic neurons, administration of the ligand clozapine-N-oxide (CNO) by systemic injection attenuated the chemoreflex that normally increases respiration in response to tissue carbon dioxide (CO2) elevation and acidosis. At the cellular level, CNO suppressed firing rate increases evoked by CO2 acidosis. Body thermoregulation at room temp. was also disrupted after CNO triggering of Di; core temps. plummeted, then recovered. This work establishes that serotonergic neurons regulate life-sustaining respiratory and thermoregulatory networks, and demonstrates a noninvasive tool for mapping neuron function.
- 20Garner, A. R., Rowland, D. C., Hwang, S. Y., Baumgaertel, K., Roth, B. L., Kentros, C., and Mayford, M. (2012) Generation of a synthetic memory trace Science 335, 1513– 1516Google ScholarThere is no corresponding record for this reference.
- 21Atasoy, D., Betley, J. N., Su, H. H., and Sternson, S. M. (2012) Deconstruction of a neural circuit for hunger Nature 488, 172– 177Google ScholarThere is no corresponding record for this reference.
- 22Krashes, M. J., Shah, B. P., Madara, J. C., Olson, D. P., Strochlic, D. E., Garfield, A. S., Vong, L., Pei, H., Watabe-Uchida, M., Uchida, N., Liberles, S. D., and Lowell, B. B. (2014) An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger Nature 507, 238– 242Google ScholarThere is no corresponding record for this reference.
- 23Kuhlman, S. J., Olivas, N. D., Tring, E., Ikrar, T., Xu, X., and Trachtenberg, J. T. (2013) A disinhibitory microcircuit initiates critical-period plasticity in the visual cortex Nature 501, 543– 546Google ScholarThere is no corresponding record for this reference.
- 24Carter, M. E., Soden, M. E., Zweifel, L. S., and Palmiter, R. D. (2013) Genetic identification of a neural circuit that suppresses appetite Nature 503, 111– 114Google ScholarThere is no corresponding record for this reference.
- 25Vrontou, S., Wong, A. M., Rau, K. K., Koerber, H. R., and Anderson, D. J. (2013) Genetic identification of C fibres that detect massage-like stroking of hairy skin in vivo Nature 493, 669– 673Google ScholarThere is no corresponding record for this reference.
- 26Kozorovitskiy, Y., Saunders, A., Johnson, C. A., Lowell, B. B., and Sabatini, B. L. (2012) Recurrent network activity drives striatal synaptogenesis Nature 485, 646– 650Google ScholarThere is no corresponding record for this reference.
- 27Ferguson, S. M., Eskenazi, D., Ishikawa, M., Wanat, M. J., Phillips, P. E., Dong, Y., Roth, B. L., and Neumaier, J. F. (2011) Transient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitization Nat. Neurosci. 14, 22– 24Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFajurfO&md5=7f40c4cd375e4ae2e1c39397ef65990aTransient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitizationFerguson, Susan M.; Eskenazi, Daniel; Ishikawa, Masago; Wanat, Matthew J.; Phillips, Paul E. M.; Dong, Yan; Roth, Bryan L.; Neumaier, John F.Nature Neuroscience (2011), 14 (1), 22-24CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Dorsal striatum is important for the development of drug addiction; however, a precise understanding of the roles of striatopallidal (indirect) and striatonigral (direct) pathway neurons in regulating behaviors remains elusive. Using viral-mediated expression of an engineered G protein-coupled receptor (hM4D), we found that activation of hM4D receptors with clozapine-N-oxide (CNO) potently reduced striatal neuron excitability. When hM4D receptors were selectively expressed in either direct or indirect pathway neurons, CNO did not change acute locomotor responses to amphetamine, but did alter behavioral plasticity assocd. with repeated drug treatment. Specifically, transiently disrupting striatopallidal neuronal activity facilitated behavioral sensitization, whereas decreasing excitability of striatonigral neurons impaired its persistence. These findings suggest that acute drug effects can be parsed from the behavioral adaptations assocd. with repeated drug exposure and highlight the utility of this approach for deconstructing neuronal pathway contributions to behavior.
- 28Silva, B. A., Mattucci, C., Krzywkowski, P., Murana, E., Illarionova, A., Grinevich, V., Canteras, N. S., Ragozzino, D., and Gross, C. T. (2013) Independent hypothalamic circuits for social and predator fear Nat. Neurosci. 16, 1731– 1733Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCntLjI&md5=4cf52a3a86ab6908a7e8009bfe0eb157Independent hypothalamic circuits for social and predator fearSilva, Bianca A.; Mattucci, Camilla; Krzywkowski, Piotr; Murana, Emanuele; Illarionova, Anna; Grinevich, Valery; Canteras, Newton S.; Ragozzino, Davide; Gross, Cornelius T.Nature Neuroscience (2013), 16 (12), 1731-1733CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)The neural circuits mediating fear to naturalistic threats are poorly understood. We found that functionally independent populations of neurons in the ventromedial hypothalamus (VMH), a region that has been implicated in feeding, sex and aggression, are essential for predator and social fear in mice. Our results establish a crit. role for VMH in fear and have implications for selective intervention in pathol. fear in humans.
- 29Bock, R., Shin, J. H., Kaplan, A. R., Dobi, A., Markey, E., Kramer, P. F., Gremel, C. M., Christensen, C. H., Adrover, M. F., and Alvarez, V. A. (2013) Strengthening the accumbal indirect pathway promotes resilience to compulsive cocaine use Nat. Neurosci. 16, 632– 638Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkvFOrt7w%253D&md5=87ce19690e192ac9d9d64cfc56ff9f45Strengthening the accumbal indirect pathway promotes resilience to compulsive cocaine useBock, Roland; Shin, J. Hoon; Kaplan, Alanna R.; Dobi, Alice; Markey, Eric; Kramer, Paul F.; Gremel, Christina M.; Christensen, Christine H.; Adrover, Martin F.; Alvarez, Veronica A.Nature Neuroscience (2013), 16 (5), 632-638CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)A hallmark of addiction is the loss of control over drug intake, which is seen in only a fraction of those exposed to stimulant drugs such as cocaine. The cellular mechanisms underlying vulnerability or resistance to compulsive drug use remain unknown. We found that individual variability in the development of highly motivated and perseverative behavior toward cocaine is assocd. with synaptic plasticity in medium spiny neurons expressing dopamine D2 receptors (D2-MSNs) in the nucleus accumbens (NAc) of mice. Potentiation of glutamatergic inputs onto indirect pathway D2-MSNs was assocd. with resilience toward compulsive cocaine seeking. Inhibition of D2-MSNs using a chemicogenetic approach enhanced the motivation to obtain cocaine, whereas optogenetic activation of D2-MSNs suppressed cocaine self-administration. These results indicate that recruitment of D2-MSNs in NAc functions to restrain cocaine self-administration and serves as a natural protective mechanism in drug-exposed individuals.
- 30Parnaudeau, S., O’Neill, P. K., Bolkan, S. S., Ward, R. D., Abbas, A. I., Roth, B. L., Balsam, P. D., Gordon, J. A., and Kellendonk, C. (2013) Inhibition of mediodorsal thalamus disrupts thalamofrontal connectivity and cognition Neuron 77, 1151– 1162Google ScholarThere is no corresponding record for this reference.
- 31Brancaccio, M., Maywood, E. S., Chesham, J. E., Loudon, A. S., and Hastings, M. H. (2013) A Gq-Ca2+ axis controls circuit-level encoding of circadian time in the suprachiasmatic nucleus Neuron 78, 714– 728Google ScholarThere is no corresponding record for this reference.
- 32Kong, D., Tong, Q., Ye, C., Koda, S., Fuller, P. M., Krashes, M. J., Vong, L., Ray, R. S., Olson, D. P., and Lowell, B. B. (2012) GABAergic RIP-Cre neurons in the arcuate nucleus selectively regulate energy expenditure Cell 151, 645– 657Google ScholarThere is no corresponding record for this reference.
- 33Bender, D., Holschbach, M., and Stocklin, G. (1994) Synthesis of n.c.a. carbon-11 labelled clozapine and its major metabolite clozapine-N-oxide and comparison of their biodistribution in mice Nucl. Med. Biol. 21, 921– 925Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXitVajs70%253D&md5=516515513d4ba3696694a396dfbce520Synthesis of n.c.a. carbon-11 labeled clozapine and its major metabolite clozapine-N-oxide and comparison of their biodistribution in miceBender, D.; Holscbach, M.; Stoecklin, G.Nuclear Medicine and Biology (1994), 21 (7), 921-5CODEN: NMBIEO; ISSN:0883-2897.N.c.a. [11C]clozapine, [8-chloro-11-(4-[methyl-11C]-methyl-1-piperazinyl)-5H-dibenzo[b,e]-1,4-diazepine], 1, an atypical neuroleptic was synthesized by N-methylation of the desmethyl compd. norclozapine, 3, using [11C]methyl iodide or [11C]methyl triflate for comparison. Subsequent oxidn. of 1 with m-chloroperoxybenzoic acid yielded clozapine-N-oxide, 2, the major metabolite of 1. Purifn. of both radiolabeled products was carried out using a combined semi-preparative HPLC/solid phase extn. procedure. In preparative scale runs overall radiochem. yields for 1 and 2 were 70 and 65%, resp. The radiochem. purities of both compds. exceeded 98% and the specific activities were in the range of 92-130 GBq/μmol (2.5-3.5 Ci/μmol). Biodistribution of 1 and 2 has been studied in NMRI mice. At ten min post injection clozapine shows a 24-fold higher brain uptake than its major metabolite. At 60 min post injection, however, the cerebral uptake of both compds. is almost identical.
- 34Chang, W. H., Lin, S. K., Lane, H. Y., Wei, F. C., Hu, W. H., Lam, Y. W. F., and Jann, M. W. (1998) Reversible metabolism of clozapine and clozapine N-oxide in schizophrenic patients Prog. Neuro-Psychopharmacol. 22, 723– 739Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXkvVCjtLo%253D&md5=1eb2520fcb3a96e1130b0089cd513bd8Reversible metabolism of clozapine and clozapine N-oxide in schizophrenic patientsChang, Wen-Ho; Lin, Shih-Ku; Lane, Hsien-Yuan; Wei, Fu-Chuan; Hu, Wei-Herng; Lam, Yw Francis; Jann, Michael W.Progress in Neuro-Psychopharmacology & Biological Psychiatry (1998), 22 (5), 723-739CODEN: PNPPD7; ISSN:0278-5846. (Elsevier Science Inc.)Our objective was to characterize the interconversion process between clozapine and its metabolite clozapine N-oxide (CNO), eight healthy male schizophrenics were administered a single dose of clozapine or CNO in a randomized crossover manner. Using a general pharmacokinetic model for the interconversion process, the mean total clearances of clozapine and CNO were 28.45 L/h and 45.30 L/h, resp. These values were similar to the values obtained by the usual model-independent method of pharmacokinetic anal. When administered clozapine, mean CNO plasma concns. of 17.7 ± 16.4 ng/mL were slightly lower than the other clozapine metabolite - desmethylclozapine (DCLOZ) plasma levels of 24.4 ± 8.6 ng/mL at the 12 h time point. When CNO was administered, plasma concns. at the 12 h time point of clozapine were twice the amt. of CNO (28.1 ± 8.9 ng/mL vs 14.4 ± 8.8 ng/mL). DCLOZ plasma concns. were detected in all patients upon clozapine administration. Upon CNO administration, only one patient had detectable plasma DCLOZ levels. The interconversion process of clozapine and CNO could partially account for the wide interpatient variability reported for clozapine plasma concns. in schizophrenic patients.
- 35Massey, C. A., Kim, G., Corcoran, A. E., Haynes, R. L., Paterson, D. S., Cummings, K. J., Dymecki, S. M., Richerson, G. B., Nattie, E. E., Kinney, H. C., and Commons, K. G. (2013) Development of brainstem 5HT(1A) receptor-binding sites in serotonin-deficient mice J. Neurochem. 126, 749– 757Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtl2lt7zK&md5=7ac056dca6ebf8168be5d30db30f8fc7Development of brainstem 5-HT1A receptor-binding sites in serotonin-deficient miceMassey, Caitlin A.; Kim, Gloria; Corcoran, Andrea E.; Haynes, Robin L.; Paterson, David S.; Cummings, Kevin J.; Dymecki, Susan M.; Richerson, George B.; Nattie, Eugene E.; Kinney, Hannah C.; Commons, Kathryn G.Journal of Neurochemistry (2013), 126 (5&6), 749-757CODEN: JONRA9; ISSN:0022-3042. (Wiley-Blackwell)The sudden infant death syndrome is assocd. with a redn. in brainstem 5-hydroxytryptamine (5-HT) and 5-HT1A receptor binding, yet it is unknown if and how these findings are linked. In this study, we used quant. tissue autoradiog. to det. if post-natal development of brainstem 5-HT1A receptors is altered in two mouse models where the development of 5-HT neurons is defective, the Lmx1bf/f/p, and the Pet-1-/- mouse. 5-HT1A receptor agonist-binding sites were examd. in both 5-HT-source nuclei (autoreceptors) and in sites that receive 5-HT innervation (heteroreceptors). In control mice between postnatal day (P) 3 and 10, 5-HT1A receptor binding increased in several brainstem sites; by P25, there were region-specific increases and decreases, refining the overall binding pattern. In the Lmx1bf/f/p and Pet-1-/- mice, 5-HT1A-autoreceptor binding was significantly lower than in control mice at P3, and remained low at P10 and P25. In contrast, 5-HT1A heteroreceptor levels were comparable between control and 5-HT-deficient mice. These data define the postnatal development of 5-HT1A-receptor binding in the mouse brainstem. Furthermore, the data suggest that 5-HT1A-heteroreceptor deficits detected in sudden infant death syndrome are not a direct consequence of a 5-HT neuron dysfunction nor reduced brain 5-HT levels. To elucidate the developmental relationship between serotonin (5-HT) levels and 5-HT1A receptors in the brainstem, we examd. 5-HT1A binding in two 5-HT-deficient mouse models. In nuclei contg. 5-HT neurons, 5-HT1A binding was decreased (autoreceptors), while binding was maintained in projection sites (heteroreceptors). Thus, brainstem 5-HT1A-heteroreceptor-binding sites do not appear developmentally sensitive to reduced brain 5-HT levels.
- 36Jann, M. W., Lam, Y. W., and Chang, W. H. (1994) Rapid formation of clozapine in guinea-pigs and man following clozapine-N-oxide administration Arch. Int. Pharmacodyn. Ther. 328, 243– 250Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXjt1yjsLc%253D&md5=23184a095504f13225b408b843fa61c0Rapid formation of clozapine in guinea pigs and man following clozapine-N-oxide administrationJann, M. W.; Lam, Y. W. F.; Chang, W. H.Archives Internationales de Pharmacodynamie et de Therapie (1994), 328 (2), 243-50CODEN: AIPTAK; ISSN:0003-9780. (Heymans Institute of Pharmacology)Clozapine and its metabolite clozapine-N-oxide (0.5 mg/kg) were administered i.p. to guinea-pigs. Significant amts. of clozapine were detected in plasma, liver, frontal cortex and caudate after clozapine-N-oxide administration. Tissue concns. of clozapine in liver, frontal cortex and caudate were greater than plasma concns. Clozapine concns. were almost equiv. in the liver. Clozapine concns. after N-oxide injection were approx. 40-50% lower compared to clozapine concns. after clozapine administration in the frontal cortex and caudate. A single dose of clozapine-N-oxide was given to a schizophrenic patient. Clozapine plasma concns. were detected after N-oxide administration. This study shows that clozapine is formed from its N-oxide metabolite and that a reversible metabolic pathway exists for clozapine and clozapine-N-oxide.
- 37Loffler, S., Korber, J., Nubbemeyer, U., and Fehsel, K. (2012) Comment on “Impaired Respiratory and Body Temperature Control Upon Acute Serotonergic Neuron Inhibition” Science 337, 646Google ScholarThere is no corresponding record for this reference.
- 38Roth, B. L., Sheffler, D. J., and Kroeze, W. K. (2004) Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia Nat. Rev. Drug Discovery 3, 353– 359Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1Gkt78%253D&md5=77196583acccca65457fec7a67218463Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophreniaRoth, Bryan L.; Sheffler, Douglas J.; Kroeze, Wesley K.Nature Reviews Drug Discovery (2004), 3 (4), 353-359CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Most common central nervous system disorders - such as depression, bipolar disorder, and schizophrenia - seem to be polygenic in origin, and the most effective medications have exceedingly complex pharmacologies. Attempts to develop more effective treatments for diseases such as schizophrenia and depression by discovering drugs selective for single mol. targets (i.e., 'magic bullets') have, not surprisingly, been largely unsuccessful. Here the authors propose that designing selectively non-selective drugs (i.e., 'magic shotguns') that interact with several mol. targets will lead to new and more effective medications for a variety of central nervous system disorders.
- 39Hunziker, F., Kuenzle, F., and Schmutz, J. (1966) Helv. Chim. Acta 49, 1433– 1439Google ScholarThere is no corresponding record for this reference.
- 40Schmutz, J. (1975) Neuroleptic piperazinyl-dibenzo-azepines. Chemistry and structure–activity relationships Arzneimittelforschung 25, 712– 720Google ScholarThere is no corresponding record for this reference.
- 41Steiner, G., Franke, A., Hadicke, E., Lenke, D., Teschendorf, H. J., Hofmann, H. P., Kreiskott, H., and Worstmann, W. (1986) Tricyclic epines. Novel (E)- and (Z)-11H-dibenz[b,e]azepines as potential central nervous system agents. Variation of the basic side chain J. Med. Chem. 29, 1877– 1888Google ScholarThere is no corresponding record for this reference.
- 42Hunziker, F., Fischer, E., and Schmutz, J. (1967) 11-Amino-5H-Dibenzo[b,e]-1,4-diazepine 0.10. Mitteilung Uber Siebengliedrige Heterocyclen Helv. Chim. Acta 50, 1588– 1599Google ScholarThere is no corresponding record for this reference.
- 43Wenthur, C. J. and Lindsley, C. W. (2013) Classics in Chemical Neuroscience: Clozapine ACS Chem. Neurosci. 4, 1018– 1025Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVyjtbbP&md5=75d84aea0d1ce9bccad9fda9a5425d06Classics in Chemical Neuroscience: ClozapineWenthur, Cody J.; Lindsley, Craig W.ACS Chemical Neuroscience (2013), 4 (7), 1018-1025CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)A review. Clozapine was the first true breakthrough in schizophrenia treatment since the discovery of chlorpromazine in 1950, effectively treating pos., neg., and some cognitive symptoms, as well as possessing unprecedented efficacy in treatment-resistant patients. Despite over 30 years of intense study, the precise mol. underpinnings that account for clozapine's unique efficacy remain elusive. In this Viewpoint, we will showcase the history and importance of clozapine to neuroscience in general, as well as for the treatment of schizophrenia, and review the synthesis, pharmacol., drug metab., and adverse events of clozapine.
- 44Kalhapure, R. S., Patil, B. P., Jadhav, M. N., Kawle, L. A., and Wagh, S. B. (2011) Synthesis of 11-(Piperazin-1-yl)-5H-dibenzo[b,e] [1,4]diazepine on Kilo Scale E-J. Chem. 8, 1747– 1749Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVKisrzI&md5=2e5c34ca9850cc28fbc20a9adf88257dSynthesis of 11-(piperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine on kilo scaleKalhapure, Rahul S.; Patil, Bhushan P.; Jadhav, Mahantesh N.; Kawle, Laxmikant A.; Wagh, Sanjay B.E-Journal of Chemistry (2011), 8 (4), 1747-1749CODEN: ECJHAO ISSN:. (WWW Publications)A synthesis of 11-(piperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine on kilo scale without any chromatog. purifn. step was reported. Key steps were Ullmann condensation, catalytic hydrogenation, and catalyzed cyclization.
- 45Ullmann, F. and Bielecki, J. (1901) Synthesis in the Biphenyl series. (I. Announcement) Ber. Dtsch. Chem. Ges. 34, 2174– 2185Google ScholarThere is no corresponding record for this reference.
- 46Davies, M. A., Compton-Toth, B. A., Hufeisen, S. J., Meltzer, H. Y., and Roth, B. L. (2005) The highly efficacious actions of N-desmethylclozapine at muscarinic receptors are unique and not a common property of either typical or atypical antipsychotic drugs: is M1 agonism a pre-requisite for mimicking clozapine’s actions? Psychopharmacology 178, 451– 460Google ScholarThere is no corresponding record for this reference.
- 47Besnard, J., Ruda, G. F., Setola, V., Abecassis, K., Rodriguiz, R. M., Huang, X. P., Norval, S., Sassano, M. F., Shin, A. I., Webster, L. A., Simeons, F. R., Stojanovski, L., Prat, A., Seidah, N. G., Constam, D. B., Bickerton, G. R., Read, K. D., Wetsel, W. C., Gilbert, I. H., Roth, B. L., and Hopkins, A. L. (2012) Automated design of ligands to polypharmacological profiles Nature 492, 215– 220Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVamurfN&md5=ace6cced250cf968d327b482a5bf1798Automated design of ligands to polypharmacological profilesBesnard, Jeremy; Ruda, Gian Filippo; Setola, Vincent; Abecassis, Keren; Rodriguiz, Ramona M.; Huang, Xi-Ping; Norval, Suzanne; Sassano, Maria F.; Shin, Antony I.; Webster, Lauren A.; Simeons, Frederick R. C.; Stojanovski, Laste; Prat, Annik; Seidah, Nabil G.; Constam, Daniel B.; Bickerton, G. Richard; Read, Kevin D.; Wetsel, William C.; Gilbert, Ian H.; Roth, Bryan L.; Hopkins, Andrew L.Nature (London, United Kingdom) (2012), 492 (7428), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The clin. efficacy and safety of a drug is detd. by its activity profile across many proteins in the proteome. However, designing drugs with a specific multi-target profile is both complex and difficult. Therefore methods to design drugs rationally a priori against profiles of several proteins would have immense value in drug discovery. Here is described a new approach for the automated design of ligands against profiles of multiple drug targets. The method is demonstrated by the evolution of an approved acetylcholinesterase inhibitor drug donepezil (I) into brain-penetrable ligands with either specific polypharmacol. or exquisite selectivity profiles for G-protein-coupled receptors. Overall, 800 ligand-target predictions of prospectively designed ligands were tested exptl., of which 75% were confirmed to be correct. Target engagement in vivo is also demonstrated. The approach can be a useful source of drug leads when multi-target profiles are required to achieve either selectivity over other drug targets or a desired polypharmacol.
- 48Wiberg, K. B. (1955) The Deuterium Isotope Effect Chem. Rev. 55, 713– 743Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaG2MXotFaqsA%253D%253D&md5=011c9eb50698d4d3c74971348c9b3ce7The deuterium isotope effectWiberg, Kenneth B.Chemical Reviews (Washington, DC, United States) (1955), 55 (), 713-43CODEN: CHREAY; ISSN:0009-2665.cf. C.A. 49, 13184e. The D isotope effect arises largely from the difference in zero-point energy between a bond to H and the corresponding bond to D. The max. isotope effect is obtained when the bond to H or D is essentially completely cleaved in the activated complex, and the isotope effect decreases with increasing bonding in the activated complex. In general, in any series of related reactions the reaction with the lowest activation energy has the lowest isotope effect. Although the isotope effect is useful in detg. whether a bond to H is cleaved in the rate-detg. step, it must be used with caution; e.g., no isotope effect was noted in the reaction of Grignard reagents with MeOH or H2O, a reaction which probably does involve the cleavage of a bond to H. There is no apparent difference in magnitude for the isotope effect in the removal of H or D as a proton, a hydride ion, or an atom. 241 references.
- 49Elison, C., Rapoport, H., Laursen, R., and Elliott, H. W. (1961) Effect of Deuteration of N-CH3 Group on Potency and Enzymatic N-Demethylation of Morphine Science 134, 1078– 1079Google ScholarThere is no corresponding record for this reference.
- 50Shao, L., Abolin, C., Hewitt, M. C., Koch, P., and Varney, M. (2006) Derivatives of tramadol for increased duration of effect Bioorg. Med. Chem. Lett. 16, 691– 694Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlCiu7bE&md5=6670712406293bbd421e52ac1f693781Derivatives of tramadol for increased duration of effectShao, Liming; Abolin, Craig; Hewitt, Michael C.; Koch, Patrick; Varney, MarkBioorganic & Medicinal Chemistry Letters (2006), 16 (3), 691-694CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Tramadol is a centrally acting opioid analgesic structurally related to codeine and morphine. Analogs of tramadol with deuterium-for-hydrogen replacement at metabolically active sites were prepd. and evaluated in vitro and in vivo.
- 51Korber, J., Loffler, S., Schollmeyer, D., and Nubbemeyer, U. (2013) Synthesis and Oxidant Properties of Phase 1 Benzepine N-Oxides of Common Antipsychotic Drugs Synthesis 45, 2875– 2887Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 111 publications.
- Karen J. Thompson, Elham Khajehali, Sophie J. Bradley, Jovana S. Navarrete, Xi Ping Huang, Samuel Slocum, Jian Jin, Jing Liu, Yan Xiong, Reid H. J. Olsen, Jeffrey F. Diberto, Kristen M. Boyt, Melanie M. Pina, Dipanwita Pati, Colin Molloy, Christoffer Bundgaard, Patrick M. Sexton, Thomas L. Kash, Michael J. Krashes, Arthur Christopoulos, Bryan L. Roth, Andrew B. Tobin. DREADD Agonist 21 Is an Effective Agonist for Muscarinic-Based DREADDs in Vitro and in Vivo. ACS Pharmacology & Translational Science 2018, 1
(1)
, 61-72. https://doi.org/10.1021/acsptsci.8b00012
- Laure Konnert, Frédéric Lamaty, Jean Martinez, and Evelina Colacino . Recent Advances in the Synthesis of Hydantoins: The State of the Art of a Valuable Scaffold. Chemical Reviews 2017, 117
(23)
, 13757-13809. https://doi.org/10.1021/acs.chemrev.7b00067
- Jessica Raper, Ryan D. Morrison, J. Scott Daniels, Leonard Howell, Jocelyne Bachevalier, Thomas Wichmann, and Adriana Galvan . Metabolism and Distribution of Clozapine-N-oxide: Implications for Nonhuman Primate Chemogenetics. ACS Chemical Neuroscience 2017, 8
(7)
, 1570-1576. https://doi.org/10.1021/acschemneuro.7b00079
- Craig W. Lindsley, Kyle A. Emmitte, Corey R. Hopkins, Thomas M. Bridges, Karen J. Gregory, Colleen M. Niswender, and P. Jeffrey Conn . Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chemical Reviews 2016, 116
(11)
, 6707-6741. https://doi.org/10.1021/acs.chemrev.5b00656
- Yoshio Iguchi, Ryoji Fukabori, Shigeki Kato, Kazumi Takahashi, Satoshi Eifuku, Yuko Maejima, Kenju Shimomura, Hiroshi Mizuma, Aya Mawatari, Hisashi Doi, Yilong Cui, Hirotaka Onoe, Keigo Hikishima, Makoto Osanai, Takuma Nishijo, Toshihiko Momiyama, Richard Benton, Kazuto Kobayashi. Chemogenetic activation of mammalian brain neurons expressing insect Ionotropic Receptors by systemic ligand precursor administration. Communications Biology 2024, 7
(1)
https://doi.org/10.1038/s42003-024-06223-4
- Julia B. Davenport, Ali D. Güler, Qi Zhang. Methodology for Studying Hypothalamic Regulation of Feeding Behaviors. Methods and Protocols 2024, 7
(6)
, 86. https://doi.org/10.3390/mps7060086
- Yoshio Iguchi, Richard Benton, Kazuto Kobayashi. A chemogenetic technology using insect Ionotropic Receptors to stimulate target cell populations in the mammalian brain. Neuroscience Research 2024, 69 https://doi.org/10.1016/j.neures.2024.11.003
- Takahito Miyake, Kaho Tanaka, Yutsuki Inoue, Yuji Nagai, Reo Nishimura, Takehito Seta, Shumpei Nakagawa, Ken-ichi Inoue, Emi Hasegawa, Takafumi Minamimoto, Masao Doi. Size-reduced DREADD derivatives for AAV-assisted multimodal chemogenetic control of neuronal activity and behavior. Cell Reports Methods 2024, 4
(10)
, 100881. https://doi.org/10.1016/j.crmeth.2024.100881
- Tyler E. Billipp, Connie Fung, Lily M. Webeck, Derek B. Sargent, Matthew B. Gologorsky, Zuojia Chen, Margaret M. McDaniel, Darshan N. Kasal, John W. McGinty, Kaitlyn A. Barrow, Lucille M. Rich, Alessio Barilli, Mark Sabat, Jason S. Debley, Chuan Wu, Richard Myers, Michael R. Howitt, Jakob von Moltke. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024, 57
(6)
, 1243-1259.e8. https://doi.org/10.1016/j.immuni.2024.03.023
- Philip J. Clark, Zachary D. Brodnik, Rodrigo A. España. Chemogenetic Signaling in Space and Time: Considerations for Designing Neuroscience Experiments Using DREADDs. The Neuroscientist 2024, 30
(3)
, 328-346. https://doi.org/10.1177/10738584221134587
- Ana Gonzalez-Ramos, Claudia Puigsasllosas-Pastor, Ainhoa Arcas-Marquez, Daniel Tornero. Updated Toolbox for Assessing Neuronal Network Reconstruction after Cell Therapy. Bioengineering 2024, 11
(5)
, 487. https://doi.org/10.3390/bioengineering11050487
- Valentina I. Cea Salazar, Melvin D. Perez, A.J. Robison, Brian C. Trainor. Impacts of sex differences on optogenetic, chemogenetic, and calcium-imaging tools. Current Opinion in Neurobiology 2024, 84 , 102817. https://doi.org/10.1016/j.conb.2023.102817
- Yoshiatsu Aomine, Yoshinobu Oyama, Koki Sakurai, Tom Macpherson, Takaaki Ozawa, Takatoshi Hikida. Clozapine N-oxide, compound 21, and JHU37160 do not influence effortful reward-seeking behavior in mice. Psychopharmacology 2024, 241
(1)
, 89-96. https://doi.org/10.1007/s00213-023-06465-w
- Andrew Jo, Sercan Deniz, Suraj Cherian, Jian Xu, Daiki Futagi, Steven H. DeVries, Yongling Zhu. Modular interneuron circuits control motion sensitivity in the mouse retina. Nature Communications 2023, 14
(1)
https://doi.org/10.1038/s41467-023-43382-0
- Makiko Shimizu, Mitsuhiro Yoshimura, Kazuhiko Baba, Naofumi Ikeda, Yuki Nonaka, Takashi Maruyama, Tatsushi Onaka, Yoichi Ueta. Deschloroclozapine exhibits an exquisite agonistic effect at lower concentration compared to clozapine-N-oxide in hM3Dq expressing chemogenetically modified rats. Frontiers in Neuroscience 2023, 17 https://doi.org/10.3389/fnins.2023.1301515
- Kate A. Lawson, Christina M. Ruiz, Stephen V. Mahler. A head-to-head comparison of two DREADD agonists for suppressing operant behavior in rats via VTA dopamine neuron inhibition. Psychopharmacology 2023, 240
(10)
, 2101-2110. https://doi.org/10.1007/s00213-023-06429-0
- Jo Bossuyt, Yana Van Den Herrewegen, Liam Nestor, An Buckinx, Dimitri De Bundel, Ilse Smolders. Chemogenetic modulation of astrocytes and microglia: State‐of‐the‐art and implications in neuroscience. Glia 2023, 71
(9)
, 2071-2095. https://doi.org/10.1002/glia.24390
- Maria João Pereira, Rajagopal Ayana, Matthew G. Holt, Lutgarde Arckens. Chemogenetic manipulation of astrocyte activity at the synapse— a gateway to manage brain disease. Frontiers in Cell and Developmental Biology 2023, 11 https://doi.org/10.3389/fcell.2023.1193130
- Janine Traut, Jose Prius Mengual, Elise J Meijer, Laura E McKillop, Hannah Alfonsa, Anna Hoerder-Suabedissen, Seo Ho Song, Kristoffer D Fehér, Dieter Riemann, Zoltan Molnar, Colin J Akerman, Vladyslav V Vyazovskiy, Lukas B Krone. Effects of clozapine-N-oxide and compound 21 on sleep in laboratory mice. eLife 2023, 12 https://doi.org/10.7554/eLife.84740
- Maricela X. Martinez, Mitchell R. Farrell, Stephen V. Mahler. Pathway-Specific Chemogenetic Manipulation by Applying Ligand to Axonally Expressed DREADDs. 2023, 207-220. https://doi.org/10.1007/978-1-0716-2918-5_11
- Adriana K. Cushnie, Daniel N. Bullock, Ana M.G. Manea, Wei Tang, Jan Zimmermann, Sarah R. Heilbronner. The use of chemogenetic actuator ligands in nonhuman primate DREADDs-fMRI. Current Research in Neurobiology 2023, 4 , 100072. https://doi.org/10.1016/j.crneur.2022.100072
- Raphaël Goutaudier, Fanny Joly, David Mallet, Magali Bartolomucci, Denis Guicherd, Carole Carcenac, Frédérique Vossier, Thibault Dufourd, Sabrina Boulet, Colin Deransart, Benoit Chovelon, Sebastien Carnicella. Hypodopaminergic state of the nigrostriatal pathway drives compulsive alcohol use. Molecular Psychiatry 2023, 28
(1)
, 463-474. https://doi.org/10.1038/s41380-022-01848-5
- Shicheng Zhang, Ryan H. Gumpper, Xi-Ping Huang, Yongfeng Liu, Brian E. Krumm, Can Cao, Jonathan F. Fay, Bryan L. Roth. Molecular basis for selective activation of DREADD-based chemogenetics. Nature 2022, 612
(7939)
, 354-362. https://doi.org/10.1038/s41586-022-05489-0
- Stewart S. Cox, Angela M. Kearns, Samuel K. Woods, Brogan J. Brown, Samantha J. Brown, Carmela M. Reichel. The role of the anterior insula during targeted helping behavior in male rats. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-07365-3
- Ornela Kljakic, Aja E. Hogan-Cann, Hunster Yang, Briannee Dover, Mohammed Al-Onaizi, Marco A.M. Prado, Vania F. Prado. Chemogenetic activation of VGLUT3-expressing neurons decreases movement. European Journal of Pharmacology 2022, 935 , 175298. https://doi.org/10.1016/j.ejphar.2022.175298
- Patrick A. Forcelli. Seizing Control of Neuronal Activity: Chemogenetic Applications in Epilepsy. Epilepsy Currents 2022, 22
(5)
, 303-308. https://doi.org/10.1177/15357597221120348
- Muzi Du, Adrienne Santiago, Cenk Akiz, Chiye Aoki. GABAergic interneurons’ feedback inhibition of dorsal raphe-projecting pyramidal neurons of the medial prefrontal cortex suppresses feeding of adolescent female mice undergoing activity-based anorexia. Brain Structure and Function 2022, 227
(6)
, 2127-2151. https://doi.org/10.1007/s00429-022-02507-9
- Jessica Raper, Adriana Galvan. Applications of chemogenetics in non-human primates. Current Opinion in Pharmacology 2022, 64 , 102204. https://doi.org/10.1016/j.coph.2022.102204
- Zachary A. Rodd, Eric A. Engleman, William A. Truitt, Andrew R. Burke, Andrei I. Molosh, Richard L. Bell, Sheketha R. Hauser. CNO Administration Increases Dopamine and Glutamate in the Medial Prefrontal Cortex of Wistar Rats: Further Concerns for the Validity of the CNO-activated DREADD Procedure. Neuroscience 2022, 491 , 176-184. https://doi.org/10.1016/j.neuroscience.2022.03.028
- Jon G. Dean, Christopher W. Fields, Michael A. Brito, Brian H. Silverstein, Chloe Rybicki-Kler, Anna M. Fryzel, Trent Groenhout, Tiecheng Liu, George A. Mashour, Dinesh Pal. Inactivation of Prefrontal Cortex Attenuates Behavioral Arousal Induced by Stimulation of Basal Forebrain During Sevoflurane Anesthesia. Anesthesia & Analgesia 2022, 28 https://doi.org/10.1213/ANE.0000000000006011
- Marie Claes, Lies De Groef, Lieve Moons. The DREADDful Hurdles and Opportunities of the Chronic Chemogenetic Toolbox. Cells 2022, 11
(7)
, 1110. https://doi.org/10.3390/cells11071110
- Yuta Miura, Akinobu Senoo, Tomohiro Doura, Shigeki Kiyonaka. Chemogenetics of cell surface receptors: beyond genetic and pharmacological approaches. RSC Chemical Biology 2022, 3
(3)
, 269-287. https://doi.org/10.1039/D1CB00195G
- Daicia C. Allen, Vanessa A. Jimenez, Timothy L. Carlson, Nicole A. Walter, Kathleen A. Grant, Verginia C. Cuzon Carlson. Characterization of DREADD receptor expression and function in rhesus macaques trained to discriminate ethanol. Neuropsychopharmacology 2022, 47
(4)
, 857-865. https://doi.org/10.1038/s41386-021-01181-5
- Lívea Dornela Godoy, Tamiris Prizon, Matheus Teixeira Rossignoli, João Pereira Leite, José Luiz Liberato. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Frontiers in Integrative Neuroscience 2022, 16 https://doi.org/10.3389/fnint.2022.765324
- Cooper D. Grossman, Bilal A. Bari, Jeremiah Y. Cohen. Serotonin neurons modulate learning rate through uncertainty. Current Biology 2022, 32
(3)
, 586-599.e7. https://doi.org/10.1016/j.cub.2021.12.006
- Andrea de Bartolomeis, Licia Vellucci, Annarita Barone, Mirko Manchia, Vincenzo De Luca, Felice Iasevoli, Christoph Correll. Clozapine's Multiple Cellular Mechanisms: What Do We Know after More than Fifty Years? a Systematic Review and Critical Assessment of Translational Mechanisms Relevant for Innovative Strategies in Treatment-Resistant Schizophrenia. SSRN Electronic Journal 2022, 4 https://doi.org/10.2139/ssrn.4089530
- Jana Desloovere, Paul Boon, Lars Emil Larsen, Marie-Gabrielle Goossens, Jean Delbeke, Evelien Carrette, Wytse Wadman, Kristl Vonck, Robrecht Raedt. Chemogenetic Seizure Control with Clozapine and the Novel Ligand JHU37160 Outperforms the Effects of Levetiracetam in the Intrahippocampal Kainic Acid Mouse Model. Neurotherapeutics 2022, 19
(1)
, 342-351. https://doi.org/10.1007/s13311-021-01160-0
- Csilla Lea Fazekas, Manon Bellardie, Bibiána Török, Eszter Sipos, Blanka Tóth, Mária Baranyi, Beáta Sperlágh, Mihály Dobos-Kovács, Elodie Chaillou, Dóra Zelena. Pharmacogenetic excitation of the median raphe region affects social and depressive-like behavior and core body temperature in male mice. Life Sciences 2021, 286 , 120037. https://doi.org/10.1016/j.lfs.2021.120037
- Vinod Kumar. Designed Synthesis of Diversely Substituted Hydantoins and Hydantoin-Based Hybrid Molecules: A Personal Account. Synlett 2021, 32
(19)
, 1897-1910. https://doi.org/10.1055/a-1480-6474
- Valérie Van Steenbergen, Florence M. Bareyre. Chemogenetic approaches to unravel circuit wiring and related behavior after spinal cord injury. Experimental Neurology 2021, 345 , 113839. https://doi.org/10.1016/j.expneurol.2021.113839
- Wei Guo, Xiayun Wan, Li Ma, Jiancheng Zhang, Kenji Hashimoto. Abnormalities in the composition of the gut microbiota in mice after repeated administration of DREADD ligands. Brain Research Bulletin 2021, 173 , 66-73. https://doi.org/10.1016/j.brainresbull.2021.05.012
- Jaroslawna Meister, Lei Wang, Sai P. Pydi, Jürgen Wess. Chemogenetic approaches to identify metabolically important GPCR signaling pathways: Therapeutic implications. Journal of Neurochemistry 2021, 158
(3)
, 603-620. https://doi.org/10.1111/jnc.15314
- P. Christiaan Klink, Jean-François Aubry, Vincent P. Ferrera, Andrew S. Fox, Sean Froudist-Walsh, Béchir Jarraya, Elisa E. Konofagou, Richard J. Krauzlis, Adam Messinger, Anna S. Mitchell, Michael Ortiz-Rios, Hiroyuki Oya, Angela C. Roberts, Anna Wang Roe, Matthew F.S. Rushworth, Jérôme Sallet, Michael Christoph Schmid, Charles E. Schroeder, Jordy Tasserie, Doris Y. Tsao, Lynn Uhrig, Wim Vanduffel, Melanie Wilke, Igor Kagan, Christopher I. Petkov. Combining brain perturbation and neuroimaging in non-human primates. NeuroImage 2021, 235 , 118017. https://doi.org/10.1016/j.neuroimage.2021.118017
- Chiara Morelli, Laura Castaldi, Sam J. Brown, Lina L. Streich, Alexander Websdale, Francisco J. Taberner, Blanka Cerreti, Alessandro Barenghi, Kevin M. Blum, Julie Sawitzke, Tessa Frank, Laura K. Steffens, Balint Doleschall, Joana Serrao, Denise Ferrarini, Stefan G. Lechner, Robert Prevedel, Paul A. Heppenstall. Identification of a population of peripheral sensory neurons that regulates blood pressure. Cell Reports 2021, 35
(9)
, 109191. https://doi.org/10.1016/j.celrep.2021.109191
- Adrienne N Santiago, Emily A Makowicz, Muzi Du, Chiye Aoki. Food Restriction Engages Prefrontal Corticostriatal Cells and Local Microcircuitry to Drive the Decision to Run versus Conserve Energy. Cerebral Cortex 2021, 31
(6)
, 2868-2885. https://doi.org/10.1093/cercor/bhaa394
- Akihiko Ozawa, Hiroyuki Arakawa. Chemogenetics drives paradigm change in the investigation of behavioral circuits and neural mechanisms underlying drug action. Behavioural Brain Research 2021, 406 , 113234. https://doi.org/10.1016/j.bbr.2021.113234
- Matthew A. Boehm, Jordi Bonaventura, Juan L. Gomez, Oscar Solís, Elliot A. Stein, Charles W. Bradberry, Michael Michaelides. Translational PET applications for brain circuit mapping with transgenic neuromodulation tools. Pharmacology Biochemistry and Behavior 2021, 204 , 173147. https://doi.org/10.1016/j.pbb.2021.173147
- Weida Shen, Shishuo Chen, Yining Liu, Pufan Han, Tianyu Ma, Ling-Hui Zeng. Chemogenetic manipulation of astrocytic activity: Is it possible to reveal the roles of astrocytes?. Biochemical Pharmacology 2021, 186 , 114457. https://doi.org/10.1016/j.bcp.2021.114457
- Feng Hu, Patrick J. Morris, Jordi Bonaventura, Hong Fan, William B. Mathews, Daniel P. Holt, Sherry Lam, Matthew Boehm, Robert F. Dannals, Martin G. Pomper, Michael Michaelides, Andrew G. Horti. 18F-labeled radiotracers for in vivo imaging of DREADD with positron emission tomography. European Journal of Medicinal Chemistry 2021, 213 , 113047. https://doi.org/10.1016/j.ejmech.2020.113047
- R. Maldonado, P. Calvé, A. García-Blanco, L. Domingo-Rodriguez, E. Senabre, E. Martín-García. Vulnerability to addiction. Neuropharmacology 2021, 186 , 108466. https://doi.org/10.1016/j.neuropharm.2021.108466
- Marie‐Gabrielle Goossens, Paul Boon, Wytse Wadman, Chris Van den Haute, Veerle Baekelandt, Alain G. Verstraete, Kristl Vonck, Lars E. Larsen, Mathieu Sprengers, Evelien Carrette, Jana Desloovere, Alfred Meurs, Jean Delbeke, Christian Vanhove, Robrecht Raedt. Long‐term chemogenetic suppression of seizures in a multifocal rat model of temporal lobe epilepsy. Epilepsia 2021, 62
(3)
, 659-670. https://doi.org/10.1111/epi.16840
- Jongwook Cho, Seungjun Ryu, Sunwoo Lee, Junsoo Kim, Hyoung-Ihl Kim. Optimizing clozapine for chemogenetic neuromodulation of somatosensory cortex. Scientific Reports 2020, 10
(1)
https://doi.org/10.1038/s41598-020-62923-x
- Julia B. Kahn, Russell G. Port, Stewart A. Anderson, Douglas A. Coulter. Modular, Circuit-Based Interventions Rescue Hippocampal-Dependent Social and Spatial Memory in a 22q11.2 Deletion Syndrome Mouse Model. Biological Psychiatry 2020, 88
(9)
, 710-718. https://doi.org/10.1016/j.biopsych.2020.04.028
- Nicholas A. Upright, Mark G. Baxter. Effect of chemogenetic actuator drugs on prefrontal cortex-dependent working memory in nonhuman primates. Neuropsychopharmacology 2020, 45
(11)
, 1793-1798. https://doi.org/10.1038/s41386-020-0660-9
- Raphaël Goutaudier, Véronique Coizet, Carole Carcenac, Sebastien Carnicella, . Compound 21, a two-edged sword with both DREADD-selective and off-target outcomes in rats. PLOS ONE 2020, 15
(9)
, e0238156. https://doi.org/10.1371/journal.pone.0238156
- Yuji Nagai, Naohisa Miyakawa, Hiroyuki Takuwa, Yukiko Hori, Kei Oyama, Bin Ji, Manami Takahashi, Xi-Ping Huang, Samuel T. Slocum, Jeffrey F. DiBerto, Yan Xiong, Takuya Urushihata, Toshiyuki Hirabayashi, Atsushi Fujimoto, Koki Mimura, Justin G. English, Jing Liu, Ken-ichi Inoue, Katsushi Kumata, Chie Seki, Maiko Ono, Masafumi Shimojo, Ming-Rong Zhang, Yutaka Tomita, Jin Nakahara, Tetsuya Suhara, Masahiko Takada, Makoto Higuchi, Jian Jin, Bryan L. Roth, Takafumi Minamimoto. Deschloroclozapine, a potent and selective chemogenetic actuator enables rapid neuronal and behavioral modulations in mice and monkeys. Nature Neuroscience 2020, 23
(9)
, 1157-1167. https://doi.org/10.1038/s41593-020-0661-3
- Seven E. Tomek, Gabriela M. Stegmann, Jonna M. Leyrer-Jackson, Jose Piña, M. Foster Olive. Restoration of prosocial behavior in rats after heroin self-administration via chemogenetic activation of the anterior insular cortex. Social Neuroscience 2020, 15
(4)
, 408-419. https://doi.org/10.1080/17470919.2020.1746394
- Michael D. Sunshine, Tommy W. Sutor, Emily J. Fox, David D. Fuller. Targeted activation of spinal respiratory neural circuits. Experimental Neurology 2020, 328 , 113256. https://doi.org/10.1016/j.expneurol.2020.113256
- Rifka C. Derman, Caroline E. Bass, Carrie R. Ferrario. Effects of hM4Di activation in CamKII basolateral amygdala neurons and CNO treatment on sensory-specific vs. general PIT: refining PIT circuits and considerations for using CNO. Psychopharmacology 2020, 237
(5)
, 1249-1266. https://doi.org/10.1007/s00213-020-05453-8
- O. Keifer, K. Kambara, A. Lau, S. Makinson, D. Bertrand. Chemogenetics a robust approach to pharmacology and gene therapy. Biochemical Pharmacology 2020, 175 , 113889. https://doi.org/10.1016/j.bcp.2020.113889
- Xinzhu Yu, Jun Nagai, Baljit S. Khakh. Improved tools to study astrocytes. Nature Reviews Neuroscience 2020, 21
(3)
, 121-138. https://doi.org/10.1038/s41583-020-0264-8
- Lore M. Peeters, Stephan Missault, Aneta J. Keliris, Georgios A. Keliris. Combining designer receptors exclusively activated by designer drugs and neuroimaging in experimental models: A powerful approach towards neurotheranostic applications. British Journal of Pharmacology 2020, 177
(5)
, 992-1002. https://doi.org/10.1111/bph.14885
- Nhan C. Huynh, Baher A. Ibrahim, Christopher M. Lee, Mickeal N. Key, Daniel A. Llano. Injection of Adeno-Associated Virus Containing Optogenetic and Chemogenetic Probes into the Neonatal Mouse Brain. 2020, 19-43. https://doi.org/10.1007/978-1-4939-9944-6_2
- Wilder Doucette, Elizabeth B. Smedley. Emerging Translational Treatments to Target the Neural Networks of Binge Eating. 2020, 103-118. https://doi.org/10.1007/978-3-030-43562-2_8
- Jordi Bonaventura, Mark A. G. Eldridge, Feng Hu, Juan L. Gomez, Marta Sanchez-Soto, Ara M. Abramyan, Sherry Lam, Matthew A. Boehm, Christina Ruiz, Mitchell R. Farrell, Andrea Moreno, Islam Mustafa Galal Faress, Niels Andersen, John Y. Lin, Ruin Moaddel, Patrick J. Morris, Lei Shi, David R. Sibley, Stephen V. Mahler, Sadegh Nabavi, Martin G. Pomper, Antonello Bonci, Andrew G. Horti, Barry J. Richmond, Michael Michaelides. High-potency ligands for DREADD imaging and activation in rodents and monkeys. Nature Communications 2019, 10
(1)
https://doi.org/10.1038/s41467-019-12236-z
- Martin Jendryka, Monika Palchaudhuri, Daniel Ursu, Bastiaan van der Veen, Birgit Liss, Dennis Kätzel, Wiebke Nissen, Anton Pekcec. Pharmacokinetic and pharmacodynamic actions of clozapine-N-oxide, clozapine, and compound 21 in DREADD-based chemogenetics in mice. Scientific Reports 2019, 9
(1)
https://doi.org/10.1038/s41598-019-41088-2
- Sascha R. A. Alles, Anne-Marie Malfait, Richard J. Miller. Chemo- and Optogenetic Strategies for the Elucidation of Pain Pathways. 2019, 817-832. https://doi.org/10.1093/oxfordhb/9780190860509.013.33
- Dong-oh Seo, Laura E. Motard, Michael R. Bruchas. Contemporary strategies for dissecting the neuronal basis of neurodevelopmental disorders. Neurobiology of Learning and Memory 2019, 165 , 106835. https://doi.org/10.1016/j.nlm.2018.03.015
- Sarah Mondoloni, Romain Durand-de Cuttoli, Alexandre Mourot. Cell-Specific Neuropharmacology. Trends in Pharmacological Sciences 2019, 40
(9)
, 696-710. https://doi.org/10.1016/j.tips.2019.07.007
- Julia B Kahn, Russell G Port, Cuiyong Yue, Hajime Takano, Douglas A Coulter. Circuit-based interventions in the dentate gyrus rescue epilepsy-associated cognitive dysfunction. Brain 2019, 142
(9)
, 2705-2721. https://doi.org/10.1093/brain/awz209
- Raphaël Goutaudier, Véronique Coizet, Carole Carcenac, Sebastien Carnicella. DREADDs: The Power of the Lock, the Weakness of the Key. Favoring the Pursuit of Specific Conditions Rather than Specific Ligands. eneuro 2019, 6
(5)
, ENEURO.0171-19.2019. https://doi.org/10.1523/ENEURO.0171-19.2019
- Bryan L Roth. How structure informs and transforms chemogenetics. Current Opinion in Structural Biology 2019, 57 , 9-16. https://doi.org/10.1016/j.sbi.2019.01.016
- Ofer Yizhar, J. Simon Wiegert. Designer Drugs for Designer Receptors: Unlocking the Translational Potential of Chemogenetics. Trends in Pharmacological Sciences 2019, 40
(6)
, 362-364. https://doi.org/10.1016/j.tips.2019.04.010
- Andreas Lieb, Mikail Weston, Dimitri M. Kullmann. Designer receptor technology for the treatment of epilepsy. EBioMedicine 2019, 43 , 641-649. https://doi.org/10.1016/j.ebiom.2019.04.059
- Christopher J. Magnus, Peter H. Lee, Jordi Bonaventura, Roland Zemla, Juan L. Gomez, Melissa H. Ramirez, Xing Hu, Adriana Galvan, Jayeeta Basu, Michael Michaelides, Scott M. Sternson. Ultrapotent chemogenetics for research and potential clinical applications. Science 2019, 364
(6436)
https://doi.org/10.1126/science.aav5282
- Mikail Weston, Teresa Kaserer, Angela Wu, Alexandre Mouravlev, Jenna C. Carpenter, Albert Snowball, Samuel Knauss, Melanie von Schimmelmann, Matthew J. During, Gabriele Lignani, Stephanie Schorge, Deborah Young, Dimitri M. Kullmann, Andreas Lieb. Olanzapine: A potent agonist at the hM4D(Gi) DREADD amenable to clinical translation of chemogenetics. Science Advances 2019, 5
(4)
https://doi.org/10.1126/sciadv.aaw1567
- Simone Bærentzen, Agata Casado-Sainz, Denise Lange, Vladimir Shalgunov, Isabel Martinez Tejada, Mengfei Xiong, Elina T. L’Estrade, Fraser G. Edgar, Hedok Lee, Matthias M. Herth, Mikael Palner. The Chemogenetic Receptor Ligand Clozapine N-Oxide Induces in vivo Neuroreceptor Occupancy and Reduces Striatal Glutamate Levels. Frontiers in Neuroscience 2019, 13 https://doi.org/10.3389/fnins.2019.00187
- Tanvi Paretkar, Eugene Dimitrov. Activation of enkephalinergic (Enk) interneurons in the central amygdala (CeA) buffers the behavioral effects of persistent pain. Neurobiology of Disease 2019, 124 , 364-372. https://doi.org/10.1016/j.nbd.2018.12.005
- Annika Højrup Runegaard, Ciarán Martin Fitzpatrick, David Paul Drucker Woldbye, Jesper Tobias Andreasen, Andreas Toft Sørensen, Ulrik Gether, Lynette C. Daws. Modulating Dopamine Signaling and Behavior with Chemogenetics: Concepts, Progress, and Challenges. Pharmacological Reviews 2019, 71
(2)
, 123-156. https://doi.org/10.1124/pr.117.013995
- Yifeng Cheng, Jun Wang. The use of chemogenetic approaches in alcohol use disorder research and treatment. Alcohol 2019, 74 , 39-45. https://doi.org/10.1016/j.alcohol.2018.05.012
- Michael V. Baratta, Steven F. Maier. New tools for understanding coping and resilience. Neuroscience Letters 2019, 693 , 54-57. https://doi.org/10.1016/j.neulet.2017.09.049
- Patrick Aldrin-Kirk, Tomas Björklund. Practical Considerations for the Use of DREADD and Other Chemogenetic Receptors to Regulate Neuronal Activity in the Mammalian Brain. 2019, 59-87. https://doi.org/10.1007/978-1-4939-9065-8_4
- Stephanie M. Perez, Daniel J. Lodge. Convergent Inputs from the Hippocampus and Thalamus to the Nucleus Accumbens Regulate Dopamine Neuron Activity. The Journal of Neuroscience 2018, 38
(50)
, 10607-10618. https://doi.org/10.1523/JNEUROSCI.2629-16.2018
- Daniel F. Manvich, Kevin A. Webster, Stephanie L. Foster, Martilias S. Farrell, James C. Ritchie, Joseph H. Porter, David Weinshenker. The DREADD agonist clozapine N-oxide (CNO) is reverse-metabolized to clozapine and produces clozapine-like interoceptive stimulus effects in rats and mice. Scientific Reports 2018, 8
(1)
https://doi.org/10.1038/s41598-018-22116-z
- Jayme R. McReynolds, John P. Christianson, Jordan M. Blacktop, John R. Mantsch. What does the Fos say? Using Fos-based approaches to understand the contribution of stress to substance use disorders. Neurobiology of Stress 2018, 9 , 271-285. https://doi.org/10.1016/j.ynstr.2018.05.004
- Ying Wang, Jiao Liang, Liying Chen, Yating Shen, Junli Zhao, Cenglin Xu, Xiaohua Wu, Heming Cheng, Xiaoying Ying, Yi Guo, Shuang Wang, Yudong Zhou, Yi Wang, Zhong Chen. Pharmaco-genetic therapeutics targeting parvalbumin neurons attenuate temporal lobe epilepsy. Neurobiology of Disease 2018, 117 , 149-160. https://doi.org/10.1016/j.nbd.2018.06.006
- Ann-Kathrin Ilg, Thomas Enkel, Dusan Bartsch, Florian Bähner. Behavioral Effects of Acute Systemic Low-Dose Clozapine in Wild-Type Rats: Implications for the Use of DREADDs in Behavioral Neuroscience. Frontiers in Behavioral Neuroscience 2018, 12 https://doi.org/10.3389/fnbeh.2018.00173
- Sophie J. Bradley, Andrew B. Tobin, Rudi Prihandoko. The use of chemogenetic approaches to study the physiological roles of muscarinic acetylcholine receptors in the central nervous system. Neuropharmacology 2018, 136 , 421-426. https://doi.org/10.1016/j.neuropharm.2017.11.043
- Yasmin Padovan-Hernandez, Lori A. Knackstedt. Dose-dependent reduction in cocaine-induced locomotion by Clozapine-N-Oxide in rats with a history of cocaine self-administration. Neuroscience Letters 2018, 674 , 132-135. https://doi.org/10.1016/j.neulet.2018.03.045
- Stephen V Mahler, Gary Aston-Jones. CNO Evil? Considerations for the Use of DREADDs in Behavioral Neuroscience. Neuropsychopharmacology 2018, 43
(5)
, 934-936. https://doi.org/10.1038/npp.2017.299
- Erin J Campbell, Nathan J Marchant. The use of chemogenetics in behavioural neuroscience: receptor variants, targeting approaches and caveats. British Journal of Pharmacology 2018, 175
(7)
, 994-1003. https://doi.org/10.1111/bph.14146
- Adriana Galvan, Michael J. Caiola, Daniel L. Albaugh. Advances in optogenetic and chemogenetic methods to study brain circuits in non-human primates. Journal of Neural Transmission 2018, 125
(3)
, 547-563. https://doi.org/10.1007/s00702-017-1697-8
- Christophe Varin, Patricia Bonnavion. Pharmacosynthetic Deconstruction of Sleep-Wake Circuits in the Brain. 2018, 153-206. https://doi.org/10.1007/164_2018_183
- Puneet Sharma, Ilse S. Pienaar. The Use of DREADDs for Dissecting the Contribution of Cellular and Neural Circuit Mechanisms in Models of Neurodegenerative Disease. 2018, 565-596. https://doi.org/10.1016/B978-0-12-804078-2.00024-6
- Patrick Aldrin-Kirk, Andreas Heuer, Daniella Rylander Ottosson, Marcus Davidsson, Bengt Mattsson, Tomas Björklund. Chemogenetic modulation of cholinergic interneurons reveals their regulating role on the direct and indirect output pathways from the striatum. Neurobiology of Disease 2018, 109 , 148-162. https://doi.org/10.1016/j.nbd.2017.10.010
- Deniz Atasoy, Scott M. Sternson. Chemogenetic Tools for Causal Cellular and Neuronal Biology. Physiological Reviews 2018, 98
(1)
, 391-418. https://doi.org/10.1152/physrev.00009.2017
- Yevgen Yudin, Tibor Rohacs. Inhibitory G
i/O
-coupled receptors in somatosensory neurons: Potential therapeutic targets for novel analgesics. Molecular Pain 2018, 14 https://doi.org/10.1177/1744806918763646
- Patrick A. Forcelli. Applications of optogenetic and chemogenetic methods to seizure circuits: Where to go next?. Journal of Neuroscience Research 2017, 95
(12)
, 2345-2356. https://doi.org/10.1002/jnr.24135
- Jingwei Jiang, Huxing Cui, Kamal Rahmouni. Optogenetics and pharmacogenetics: principles and applications. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology 2017, 313
(6)
, R633-R645. https://doi.org/10.1152/ajpregu.00091.2017
- C. James Howell, Michael P. Sceniak, Min Lang, Wenceslas Krakowiecki, Fatimah E. Abouelsoud, Saloni U. Lad, Heping Yu, David M. Katz. Activation of the Medial Prefrontal Cortex Reverses Cognitive and Respiratory Symptoms in a Mouse Model of Rett Syndrome. eneuro 2017, 4
(6)
, ENEURO.0277-17.2017. https://doi.org/10.1523/ENEURO.0277-17.2017
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 51 other publications.
- 1Sternson, S. M. and Roth, B. L. (2014) Chemogenetic Tools to Interrogate Brain Functions Annu. Rev. Neurosci. 37, 387– 4071https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVKgtL7E&md5=43a0b89232bd0b63e787a9caab7584daChemogenetic tools to interrogate brain functionsSternson, Scott M.; Roth, Bryan L.Annual Review of Neuroscience (2014), 37 (), 387-407CODEN: ARNSD5; ISSN:0147-006X. (Annual Reviews)A review. Elucidating the roles of neuronal cell types for physiol. and behavior is essential for understanding brain functions. Perturbation of neuron elec. activity can be used to probe the causal relationship between neuronal cell types and behavior. New genetically encoded neuron perturbation tools have been developed for remotely controlling neuron function using small mols. that activate engineered receptors that can be targeted to cell types using genetic methods. Here we describe recent progress for approaches using genetically engineered receptors that selectively interact with small mols. Called "chemogenetics," receptors with diverse cellular functions have been developed that facilitate the selective pharmacol. control over a diverse range of cell-signaling processes, including elec. activity, for molecularly defined cell types. These tools have revealed remarkably specific behavioral physiol. influences for molecularly defined cell types that are often intermingled with populations having different or even opposite functions.
- 2Urban, D. J. and Roth, B. L. (2015) DREADDs (Designer Receptors Exclusively Activated by Designer Drugs): Chemogenetic Tools with Therapeutic Utility Annu. Rev. Pharmacol. Toxicol. 55, 399– 417There is no corresponding record for this reference.
- 3Penfield, W. and Jasper, H. H. (1954) Epilepsy and the Functional Anatomy of the Human Brain (First ed.), Little Brown, Boston.There is no corresponding record for this reference.
- 4Boyden, E. S., Zhang, F., Bamberg, E., Nagel, G., and Deisseroth, K. (2005) Millisecond-timescale, genetically targeted optical control of neural activity Nat. Neurosci. 8, 1263– 12684https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpt1egtrc%253D&md5=7d71b89559e8825798cbc4b81ccf8c32Millisecond-timescale, genetically targeted optical control of neural activityBoyden, Edward S.; Zhang, Feng; Bamberg, Ernst; Nagel, Georg; Deisseroth, KarlNature Neuroscience (2005), 8 (9), 1263-1268CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Temporally precise, noninvasive control of activity in well-defined neuronal populations is a long-sought goal of systems neuroscience. We adapted for this purpose the naturally occurring algal protein Channelrhodopsin-2, a rapidly gated light-sensitive cation channel, by using lentiviral gene delivery in combination with high-speed optical switching to photostimulate mammalian neurons. We demonstrate reliable, millisecond-timescale control of neuronal spiking, as well as control of excitatory and inhibitory synaptic transmission. This technol. allows the use of light to alter neural processing at the level of single spikes and synaptic events, yielding a widely applicable tool for neuroscientists and biomedical engineers.
- 5Armbruster, B. N. and Roth, B. L. (2005) Mining the receptorome J. Biol. Chem. 280, 5129– 5132There is no corresponding record for this reference.
- 6Zhang, F., Wang, L. P., Brauner, M., Liewald, J. F., Kay, K., Watzke, N., Wood, P. G., Bamberg, E., Nagel, G., Gottschalk, A., and Deisseroth, K. (2007) Multimodal fast optical interrogation of neural circuitry Nature 446, 633– 6396https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjslOltr4%253D&md5=011fc19954bb36af7622fc7b5f5b25f5Multimodal fast optical interrogation of neural circuitryZhang, Feng; Wang, Li-Ping; Brauner, Martin; Liewald, Jana F.; Kay, Kenneth; Watzke, Natalie; Wood, Phillip G.; Bamberg, Ernst; Nagel, Georg; Gottschalk, Alexander; Deisseroth, KarlNature (London, United Kingdom) (2007), 446 (7136), 633-639CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The understanding of the cellular implementation of systems-level neural processes like action, thought and emotion has been limited by the availability of tools to interrogate specific classes of neural cells within intact, living brain tissue. Here the authors identify and develop an archaeal light-driven chloride pump (NpHR) from Natronomonas pharaonis for temporally precise optical inhibition of neural activity. NpHR allows either knockout of single action potentials, or sustained blockade of spiking. NpHR is compatible with ChR2, the previous optical excitation technol. the authors have described, in that the two opposing probes operate at similar light powers but with well-sepd. action spectra. NpHR, like ChR2, functions in mammals without exogenous cofactors, and the two probes can be integrated with calcium imaging in mammalian brain tissue for bidirectional optical modulation and readout of neural activity. Likewise, NpHR and ChR2 can be targeted together to Caenorhabditis elegans muscle and cholinergic motor neurons to control locomotion bidirectionally. NpHR and ChR2 form a complete system for multimodal, high-speed, genetically targeted, all-optical interrogation of living neural circuits.
- 7Airan, R. D., Thompson, K. R., Fenno, L. E., Bernstein, H., and Deisseroth, K. (2009) Temporally precise in vivo control of intracellular signalling Nature 458, 1025– 10297https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXjsVCqur4%253D&md5=7b3f9805ba9e9a5e802247c7a272a41bTemporally precise in vivo control of intracellular signallingAiran, Raag D.; Thompson, Kimberly R.; Fenno, Lief E.; Bernstein, Hannah; Deisseroth, KarlNature (London, United Kingdom) (2009), 458 (7241), 1025-1029CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)In the study of complex mammalian behaviors, technol. limitations have prevented spatiotemporally precise control over intracellular signaling processes. Here we report the development of a versatile family of genetically encoded optical tools ('optoXRs') that leverage common structure-function relationships among G-protein-coupled receptors (GPCRs) to recruit and control, with high spatiotemporal precision, receptor-initiated biochem. signaling pathways. In particular, we have developed and characterized two optoXRs that selectively recruit distinct, targeted signaling pathways in response to light. The two optoXRs exerted opposing effects on spike firing in nucleus accumbens in vivo, and precisely timed optoXR photostimulation in nucleus accumbens by itself sufficed to drive conditioned place preference in freely moving mice. The optoXR approach allows testing of hypotheses regarding the causal impact of biochem. signaling in behaving mammals, in a targetable and temporally precise manner.
- 8Berndt, A., Yizhar, O., Gunaydin, L. A., Hegemann, P., and Deisseroth, K. (2009) Bi-stable neural state switches Nat. Neurosci. 12, 229– 2348https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2hsrnI&md5=72cfb3b77c8272c77a7b8a837e37130bBi-stable neural state switchesBerndt, Andre; Yizhar, Ofer; Gunaydin, Lisa A.; Hegemann, Peter; Deisseroth, KarlNature Neuroscience (2009), 12 (2), 229-234CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Here we describe bi-stable channel rhodopsins that convert a brief pulse of light into a stable step in membrane potential. These molecularly engineered probes nevertheless retain millisecond-scale temporal precision. Photocurrents can be precisely initiated and terminated with different colors of light, but operate at vastly longer time scales than conventional channel rhodopsins as a result of modification at the C128 position that extends the lifetime of the open state. Because of their enhanced kinetic stability, these step-function tools are also effectively responsive to light at orders of magnitude lower intensity than wild-type channel rhodopsins. These mols. therefore offer important new capabilities for a broad range of in vivo applications.
- 9Li, X., Gutierrez, D. V., Hanson, M. G., Han, J., Mark, M. D., Chiel, H., Hegemann, P., Landmesser, L. T., and Herlitze, S. (2005) Fast noninvasive activation and inhibition of neural and network activity by vertebrate rhodopsin and green algae channelrhodopsin Proc. Natl. Acad. Sci. U. S. A. 102, 17816– 178219https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlegs7vO&md5=77f01d459514e66d35e9a51994353217Fast noninvasive activation and inhibition of neural and network activity by vertebrate rhodopsin and green algae channelrhodopsinLi, Xiang; Gutierrez, Davina V.; Hanson, M. Gartz; Han, Jing; Mark, Melanie D.; Chiel, Hillel; Hegemann, Peter; Landmesser, Lynn T.; Herlitze, StefanProceedings of the National Academy of Sciences of the United States of America (2005), 102 (49), 17816-17821CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Techniques for fast noninvasive control of neuronal excitability will be of major importance for analyzing and understanding neuronal networks and animal behavior. To develop these tools we demonstrated that two light-activated signaling proteins, vertebrate rat rhodopsin 4 (RO4) and the green algae channelrhodopsin 2 (ChR2), could be used to control neuronal excitability and modulate synaptic transmission. Vertebrate rhodopsin couples to the Gi/o, pertussis toxin-sensitive pathway to allow modulation of G protein-gated inward rectifying potassium channels and voltage-gated Ca2+ channels. Light-mediated activation of RO4 in cultured hippocampal neurons reduces neuronal firing within ms by hyperpolarization of the somato-dendritic membrane, and when activated at presynaptic sites modulates synaptic transmission and paired-pulse facilitation. In contrast, somato-dendritic activation of ChR2 depolarizes neurons sufficiently to induce immediate action potentials, which precisely follow the ChR2 activation up to light stimulation frequencies of 20 Hz. To demonstrate that these constructs are useful for regulating network behavior in intact organisms, embryonic chick spinal cords were electroporated with either construct, allowing the frequency of episodes of spontaneous bursting activity, known to be important for motor circuit formation, to be precisely controlled. Thus light-activated vertebrate RO4 and green algae ChR2 allow the antagonistic control of neuronal function within ms to s in a precise, reversible, and noninvasive manner in cultured neurons and intact vertebrate spinal cords.
- 10Zhang, F., Prigge, M., Beyriere, F., Tsunoda, S. P., Mattis, J., Yizhar, O., Hegemann, P., and Deisseroth, K. (2008) Red-shifted optogenetic excitation: a tool for fast neural control derived from Volvox carteri Nat. Neurosci. 11, 631– 63310https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXmtlCisbw%253D&md5=307a16c246c6fd78a9b145bbfe2c0357Red-shifted optogenetic excitation: a tool for fast neural control derived from Volvox carteriZhang, Feng; Prigge, Matthias; Beyriere, Florent; Tsunoda, Satoshi P.; Mattis, Joanna; Yizhar, Ofer; Hegemann, Peter; Deisseroth, KarlNature Neuroscience (2008), 11 (6), 631-633CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)The introduction of two microbial opsin-based tools, channelrhodopsin-2 (ChR2) and halorhodopsin (NpHR), to neuroscience has generated interest in fast, multimodal, cell type-specific neural circuit control. Here the authors describe a cation-conducting channelrhodopsin (VChR1) from Volvox carteri that can drive spiking at 589 nm, with excitation max. red-shifted ∼70 nm compared with ChR2. These results demonstrate fast photostimulation with yellow light, thereby defining a functionally distinct third category of microbial rhodopsin proteins.
- 11Armbruster, B. N., Li, X., Pausch, M. H., Herlitze, S., and Roth, B. L. (2007) Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand Proc. Natl. Acad. Sci. U. S. A. 104, 5163– 516811https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2s7nt1egtQ%253D%253D&md5=935cd23bf341eff865375f7d2e253210Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligandArmbruster Blaine N; Li Xiang; Pausch Mark H; Herlitze Stefan; Roth Bryan LProceedings of the National Academy of Sciences of the United States of America (2007), 104 (12), 5163-8 ISSN:0027-8424.We evolved muscarinic receptors in yeast to generate a family of G protein-coupled receptors (GPCRs) that are activated solely by a pharmacologically inert drug-like and bioavailable compound (clozapine-N-oxide). Subsequent screening in human cell lines facilitated the creation of a family of muscarinic acetylcholine GPCRs suitable for in vitro and in situ studies. We subsequently created lines of telomerase-immortalized human pulmonary artery smooth muscle cells stably expressing all five family members and found that each one faithfully recapitulated the signaling phenotype of the parent receptor. We also expressed a G(i)-coupled designer receptor in hippocampal neurons (hM(4)D) and demonstrated its ability to induce membrane hyperpolarization and neuronal silencing. We have thus devised a facile approach for designing families of GPCRs with engineered ligand specificities. Such reverse-engineered GPCRs will prove to be powerful tools for selectively modulating signal-transduction pathways in vitro and in vivo.
- 12Alexander, G. M., Rogan, S. C., Abbas, A. I., Armbruster, B. N., Pei, Y., Allen, J. A., Nonneman, R. J., Hartmann, J., Moy, S. S., Nicolelis, M. A., McNamara, J. O., and Roth, B. L. (2009) Remote Control of Neuronal Activity in Transgenic Mice Expressing Evolved G Protein-Coupled Receptors Neuron 63, 27– 3912https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXpsFOns7g%253D&md5=a7fed676083d592279f5497c6956c781Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptorsAlexander, Georgia M.; Rogan, Sarah C.; Abbas, Atheir I.; Armbruster, Blaine N.; Pei, Ying; Allen, John A.; Nonneman, Randal J.; Hartmann, John; Moy, Sheryl S.; Nicolelis, Miguel A.; McNamara, James O.; Roth, Bryan L.Neuron (2009), 63 (1), 27-39CODEN: NERNET; ISSN:0896-6273. (Cell Press)Examg. the behavioral consequences of selective CNS neuronal activation is a powerful tool for elucidating mammalian brain function in health and disease. Newly developed genetic, pharmacol., and optical tools allow activation of neurons with exquisite spatiotemporal resoln.; however, the inaccessibility to light of widely distributed neuronal populations and the invasiveness required for activation by light or infused ligands limit the utility of these methods. To overcome these barriers, we created transgenic mice expressing an evolved G protein-coupled receptor (hM3Dq) selectively activated by the pharmacol. inert, orally bioavailable drug clozapine-N-oxide (CNO). Here, we expressed hM3Dq in forebrain principal neurons. Local field potential and single-neuron recordings revealed that peripheral administration of CNO activated hippocampal neurons selectively in hM3Dq-expressing mice. Behavioral correlates of neuronal activation included increased locomotion, stereotypy, and limbic seizures. These results demonstrate a powerful chem.-genetic tool for remotely controlling the activity of discrete populations of neurons in vivo.
- 13Rogan, S. C. and Roth, B. L. (2011) Remote control of neuronal signaling Pharmacol. Rev. 63, 291– 315There is no corresponding record for this reference.
- 14Farrell, M. S. and Roth, B. L. (2012) Pharmacosynthetics: Reimagining the pharmacogenetic approach Brain Res. 1511, 6– 20There is no corresponding record for this reference.
- 15Jennings, J. H. and Stuber, G. D. (2014) Tools for Resolving Functional Activity and Connectivity within Intact Neural Circuits Curr. Biol. 24, R41– 50There is no corresponding record for this reference.
- 16Lee, H. M., Giguere, P. M., and Roth, B. L. (2013) DREADDs: Novel tools for drug discovery and development Drug Discovery Today 19, 469– 473There is no corresponding record for this reference.
- 17Giguere, P. M., Kroeze, W. K., and Roth, B. L. (2014) Tuning up the right signal: Chemical and genetic approaches to study GPCR functions Curr. Opin. Cell Biol. 27, 51– 5517https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtVCiu74%253D&md5=9a1e7fdf05facf40734fd6758ac1e698Tuning up the right signal: chemical and genetic approaches to study GPCR functionsGiguere, Patrick M.; Kroeze, Wesley K.; Roth, Bryan L.Current Opinion in Cell Biology (2014), 27 (), 51-55CODEN: COCBE3; ISSN:0955-0674. (Elsevier Ltd.)A review. The G protein-coupled receptor (GPCR) family is among the most druggable families in the human proteome. GPCRs are involved in most physiol. processes, and our ability to modulate their activity is a hallmark of modern pharmacol. The means by which the activity of GPCRs can be modulated have been expanded by emerging data and concepts in pharmacol., which has created new strategies for their control. These new approaches will lead to the generation of more potent, selective, and efficient pharmaceutics, while reducing inappropriate actions and adverse effects. Herein, we review and comment on some recent advances in chem. and genetic approaches to the profiling of GPCR function, as well as the validation of orphan GPCRs as potential therapeutic targets using engineered receptors.
- 18Armbruster, B. N., Li, X., Pausch, M. H., Herlitze, S., and Roth, B. L. (2007) Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand Proc. Natl. Acad. Sci. U. S. A. 104, 5163– 516818https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2s7nt1egtQ%253D%253D&md5=935cd23bf341eff865375f7d2e253210Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligandArmbruster Blaine N; Li Xiang; Pausch Mark H; Herlitze Stefan; Roth Bryan LProceedings of the National Academy of Sciences of the United States of America (2007), 104 (12), 5163-8 ISSN:0027-8424.We evolved muscarinic receptors in yeast to generate a family of G protein-coupled receptors (GPCRs) that are activated solely by a pharmacologically inert drug-like and bioavailable compound (clozapine-N-oxide). Subsequent screening in human cell lines facilitated the creation of a family of muscarinic acetylcholine GPCRs suitable for in vitro and in situ studies. We subsequently created lines of telomerase-immortalized human pulmonary artery smooth muscle cells stably expressing all five family members and found that each one faithfully recapitulated the signaling phenotype of the parent receptor. We also expressed a G(i)-coupled designer receptor in hippocampal neurons (hM(4)D) and demonstrated its ability to induce membrane hyperpolarization and neuronal silencing. We have thus devised a facile approach for designing families of GPCRs with engineered ligand specificities. Such reverse-engineered GPCRs will prove to be powerful tools for selectively modulating signal-transduction pathways in vitro and in vivo.
- 19Ray, R. S., Corcoran, A. E., Brust, R. D., Kim, J. C., Richerson, G. B., Nattie, E., and Dymecki, S. M. (2011) Impaired Respiratory and Body Temperature Control Upon Acute Serotonergic Neuron Inhibition Science 333, 637– 64219https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpt1yisb8%253D&md5=62a37269a9b28b710df2c483cc92f7a7Impaired Respiratory and Body Temperature Control Upon Acute Serotonergic Neuron InhibitionRay, Russell S.; Corcoran, Andrea E.; Brust, Rachael D.; Kim, Jun Chul; Richerson, George B.; Nattie, Eugene; Dymecki, Susan M.Science (Washington, DC, United States) (2011), 333 (6042), 637-642CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Physiol. homeostasis is essential for organism survival. Highly responsive neuronal networks are involved, but their constituent neurons are just beginning to be resolved. To query brain serotonergic neurons in homeostasis, the authors used a neuronal silencing tool, mouse RC::FPDi (based on the synthetic G protein-coupled receptor Di), designed for cell type-specific, ligand-inducible, and reversible suppression of action potential firing. In mice harboring Di-expressing serotonergic neurons, administration of the ligand clozapine-N-oxide (CNO) by systemic injection attenuated the chemoreflex that normally increases respiration in response to tissue carbon dioxide (CO2) elevation and acidosis. At the cellular level, CNO suppressed firing rate increases evoked by CO2 acidosis. Body thermoregulation at room temp. was also disrupted after CNO triggering of Di; core temps. plummeted, then recovered. This work establishes that serotonergic neurons regulate life-sustaining respiratory and thermoregulatory networks, and demonstrates a noninvasive tool for mapping neuron function.
- 20Garner, A. R., Rowland, D. C., Hwang, S. Y., Baumgaertel, K., Roth, B. L., Kentros, C., and Mayford, M. (2012) Generation of a synthetic memory trace Science 335, 1513– 1516There is no corresponding record for this reference.
- 21Atasoy, D., Betley, J. N., Su, H. H., and Sternson, S. M. (2012) Deconstruction of a neural circuit for hunger Nature 488, 172– 177There is no corresponding record for this reference.
- 22Krashes, M. J., Shah, B. P., Madara, J. C., Olson, D. P., Strochlic, D. E., Garfield, A. S., Vong, L., Pei, H., Watabe-Uchida, M., Uchida, N., Liberles, S. D., and Lowell, B. B. (2014) An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger Nature 507, 238– 242There is no corresponding record for this reference.
- 23Kuhlman, S. J., Olivas, N. D., Tring, E., Ikrar, T., Xu, X., and Trachtenberg, J. T. (2013) A disinhibitory microcircuit initiates critical-period plasticity in the visual cortex Nature 501, 543– 546There is no corresponding record for this reference.
- 24Carter, M. E., Soden, M. E., Zweifel, L. S., and Palmiter, R. D. (2013) Genetic identification of a neural circuit that suppresses appetite Nature 503, 111– 114There is no corresponding record for this reference.
- 25Vrontou, S., Wong, A. M., Rau, K. K., Koerber, H. R., and Anderson, D. J. (2013) Genetic identification of C fibres that detect massage-like stroking of hairy skin in vivo Nature 493, 669– 673There is no corresponding record for this reference.
- 26Kozorovitskiy, Y., Saunders, A., Johnson, C. A., Lowell, B. B., and Sabatini, B. L. (2012) Recurrent network activity drives striatal synaptogenesis Nature 485, 646– 650There is no corresponding record for this reference.
- 27Ferguson, S. M., Eskenazi, D., Ishikawa, M., Wanat, M. J., Phillips, P. E., Dong, Y., Roth, B. L., and Neumaier, J. F. (2011) Transient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitization Nat. Neurosci. 14, 22– 2427https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFajurfO&md5=7f40c4cd375e4ae2e1c39397ef65990aTransient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitizationFerguson, Susan M.; Eskenazi, Daniel; Ishikawa, Masago; Wanat, Matthew J.; Phillips, Paul E. M.; Dong, Yan; Roth, Bryan L.; Neumaier, John F.Nature Neuroscience (2011), 14 (1), 22-24CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)Dorsal striatum is important for the development of drug addiction; however, a precise understanding of the roles of striatopallidal (indirect) and striatonigral (direct) pathway neurons in regulating behaviors remains elusive. Using viral-mediated expression of an engineered G protein-coupled receptor (hM4D), we found that activation of hM4D receptors with clozapine-N-oxide (CNO) potently reduced striatal neuron excitability. When hM4D receptors were selectively expressed in either direct or indirect pathway neurons, CNO did not change acute locomotor responses to amphetamine, but did alter behavioral plasticity assocd. with repeated drug treatment. Specifically, transiently disrupting striatopallidal neuronal activity facilitated behavioral sensitization, whereas decreasing excitability of striatonigral neurons impaired its persistence. These findings suggest that acute drug effects can be parsed from the behavioral adaptations assocd. with repeated drug exposure and highlight the utility of this approach for deconstructing neuronal pathway contributions to behavior.
- 28Silva, B. A., Mattucci, C., Krzywkowski, P., Murana, E., Illarionova, A., Grinevich, V., Canteras, N. S., Ragozzino, D., and Gross, C. T. (2013) Independent hypothalamic circuits for social and predator fear Nat. Neurosci. 16, 1731– 173328https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCntLjI&md5=4cf52a3a86ab6908a7e8009bfe0eb157Independent hypothalamic circuits for social and predator fearSilva, Bianca A.; Mattucci, Camilla; Krzywkowski, Piotr; Murana, Emanuele; Illarionova, Anna; Grinevich, Valery; Canteras, Newton S.; Ragozzino, Davide; Gross, Cornelius T.Nature Neuroscience (2013), 16 (12), 1731-1733CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)The neural circuits mediating fear to naturalistic threats are poorly understood. We found that functionally independent populations of neurons in the ventromedial hypothalamus (VMH), a region that has been implicated in feeding, sex and aggression, are essential for predator and social fear in mice. Our results establish a crit. role for VMH in fear and have implications for selective intervention in pathol. fear in humans.
- 29Bock, R., Shin, J. H., Kaplan, A. R., Dobi, A., Markey, E., Kramer, P. F., Gremel, C. M., Christensen, C. H., Adrover, M. F., and Alvarez, V. A. (2013) Strengthening the accumbal indirect pathway promotes resilience to compulsive cocaine use Nat. Neurosci. 16, 632– 63829https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXkvFOrt7w%253D&md5=87ce19690e192ac9d9d64cfc56ff9f45Strengthening the accumbal indirect pathway promotes resilience to compulsive cocaine useBock, Roland; Shin, J. Hoon; Kaplan, Alanna R.; Dobi, Alice; Markey, Eric; Kramer, Paul F.; Gremel, Christina M.; Christensen, Christine H.; Adrover, Martin F.; Alvarez, Veronica A.Nature Neuroscience (2013), 16 (5), 632-638CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)A hallmark of addiction is the loss of control over drug intake, which is seen in only a fraction of those exposed to stimulant drugs such as cocaine. The cellular mechanisms underlying vulnerability or resistance to compulsive drug use remain unknown. We found that individual variability in the development of highly motivated and perseverative behavior toward cocaine is assocd. with synaptic plasticity in medium spiny neurons expressing dopamine D2 receptors (D2-MSNs) in the nucleus accumbens (NAc) of mice. Potentiation of glutamatergic inputs onto indirect pathway D2-MSNs was assocd. with resilience toward compulsive cocaine seeking. Inhibition of D2-MSNs using a chemicogenetic approach enhanced the motivation to obtain cocaine, whereas optogenetic activation of D2-MSNs suppressed cocaine self-administration. These results indicate that recruitment of D2-MSNs in NAc functions to restrain cocaine self-administration and serves as a natural protective mechanism in drug-exposed individuals.
- 30Parnaudeau, S., O’Neill, P. K., Bolkan, S. S., Ward, R. D., Abbas, A. I., Roth, B. L., Balsam, P. D., Gordon, J. A., and Kellendonk, C. (2013) Inhibition of mediodorsal thalamus disrupts thalamofrontal connectivity and cognition Neuron 77, 1151– 1162There is no corresponding record for this reference.
- 31Brancaccio, M., Maywood, E. S., Chesham, J. E., Loudon, A. S., and Hastings, M. H. (2013) A Gq-Ca2+ axis controls circuit-level encoding of circadian time in the suprachiasmatic nucleus Neuron 78, 714– 728There is no corresponding record for this reference.
- 32Kong, D., Tong, Q., Ye, C., Koda, S., Fuller, P. M., Krashes, M. J., Vong, L., Ray, R. S., Olson, D. P., and Lowell, B. B. (2012) GABAergic RIP-Cre neurons in the arcuate nucleus selectively regulate energy expenditure Cell 151, 645– 657There is no corresponding record for this reference.
- 33Bender, D., Holschbach, M., and Stocklin, G. (1994) Synthesis of n.c.a. carbon-11 labelled clozapine and its major metabolite clozapine-N-oxide and comparison of their biodistribution in mice Nucl. Med. Biol. 21, 921– 92533https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXitVajs70%253D&md5=516515513d4ba3696694a396dfbce520Synthesis of n.c.a. carbon-11 labeled clozapine and its major metabolite clozapine-N-oxide and comparison of their biodistribution in miceBender, D.; Holscbach, M.; Stoecklin, G.Nuclear Medicine and Biology (1994), 21 (7), 921-5CODEN: NMBIEO; ISSN:0883-2897.N.c.a. [11C]clozapine, [8-chloro-11-(4-[methyl-11C]-methyl-1-piperazinyl)-5H-dibenzo[b,e]-1,4-diazepine], 1, an atypical neuroleptic was synthesized by N-methylation of the desmethyl compd. norclozapine, 3, using [11C]methyl iodide or [11C]methyl triflate for comparison. Subsequent oxidn. of 1 with m-chloroperoxybenzoic acid yielded clozapine-N-oxide, 2, the major metabolite of 1. Purifn. of both radiolabeled products was carried out using a combined semi-preparative HPLC/solid phase extn. procedure. In preparative scale runs overall radiochem. yields for 1 and 2 were 70 and 65%, resp. The radiochem. purities of both compds. exceeded 98% and the specific activities were in the range of 92-130 GBq/μmol (2.5-3.5 Ci/μmol). Biodistribution of 1 and 2 has been studied in NMRI mice. At ten min post injection clozapine shows a 24-fold higher brain uptake than its major metabolite. At 60 min post injection, however, the cerebral uptake of both compds. is almost identical.
- 34Chang, W. H., Lin, S. K., Lane, H. Y., Wei, F. C., Hu, W. H., Lam, Y. W. F., and Jann, M. W. (1998) Reversible metabolism of clozapine and clozapine N-oxide in schizophrenic patients Prog. Neuro-Psychopharmacol. 22, 723– 73934https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXkvVCjtLo%253D&md5=1eb2520fcb3a96e1130b0089cd513bd8Reversible metabolism of clozapine and clozapine N-oxide in schizophrenic patientsChang, Wen-Ho; Lin, Shih-Ku; Lane, Hsien-Yuan; Wei, Fu-Chuan; Hu, Wei-Herng; Lam, Yw Francis; Jann, Michael W.Progress in Neuro-Psychopharmacology & Biological Psychiatry (1998), 22 (5), 723-739CODEN: PNPPD7; ISSN:0278-5846. (Elsevier Science Inc.)Our objective was to characterize the interconversion process between clozapine and its metabolite clozapine N-oxide (CNO), eight healthy male schizophrenics were administered a single dose of clozapine or CNO in a randomized crossover manner. Using a general pharmacokinetic model for the interconversion process, the mean total clearances of clozapine and CNO were 28.45 L/h and 45.30 L/h, resp. These values were similar to the values obtained by the usual model-independent method of pharmacokinetic anal. When administered clozapine, mean CNO plasma concns. of 17.7 ± 16.4 ng/mL were slightly lower than the other clozapine metabolite - desmethylclozapine (DCLOZ) plasma levels of 24.4 ± 8.6 ng/mL at the 12 h time point. When CNO was administered, plasma concns. at the 12 h time point of clozapine were twice the amt. of CNO (28.1 ± 8.9 ng/mL vs 14.4 ± 8.8 ng/mL). DCLOZ plasma concns. were detected in all patients upon clozapine administration. Upon CNO administration, only one patient had detectable plasma DCLOZ levels. The interconversion process of clozapine and CNO could partially account for the wide interpatient variability reported for clozapine plasma concns. in schizophrenic patients.
- 35Massey, C. A., Kim, G., Corcoran, A. E., Haynes, R. L., Paterson, D. S., Cummings, K. J., Dymecki, S. M., Richerson, G. B., Nattie, E. E., Kinney, H. C., and Commons, K. G. (2013) Development of brainstem 5HT(1A) receptor-binding sites in serotonin-deficient mice J. Neurochem. 126, 749– 75735https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtl2lt7zK&md5=7ac056dca6ebf8168be5d30db30f8fc7Development of brainstem 5-HT1A receptor-binding sites in serotonin-deficient miceMassey, Caitlin A.; Kim, Gloria; Corcoran, Andrea E.; Haynes, Robin L.; Paterson, David S.; Cummings, Kevin J.; Dymecki, Susan M.; Richerson, George B.; Nattie, Eugene E.; Kinney, Hannah C.; Commons, Kathryn G.Journal of Neurochemistry (2013), 126 (5&6), 749-757CODEN: JONRA9; ISSN:0022-3042. (Wiley-Blackwell)The sudden infant death syndrome is assocd. with a redn. in brainstem 5-hydroxytryptamine (5-HT) and 5-HT1A receptor binding, yet it is unknown if and how these findings are linked. In this study, we used quant. tissue autoradiog. to det. if post-natal development of brainstem 5-HT1A receptors is altered in two mouse models where the development of 5-HT neurons is defective, the Lmx1bf/f/p, and the Pet-1-/- mouse. 5-HT1A receptor agonist-binding sites were examd. in both 5-HT-source nuclei (autoreceptors) and in sites that receive 5-HT innervation (heteroreceptors). In control mice between postnatal day (P) 3 and 10, 5-HT1A receptor binding increased in several brainstem sites; by P25, there were region-specific increases and decreases, refining the overall binding pattern. In the Lmx1bf/f/p and Pet-1-/- mice, 5-HT1A-autoreceptor binding was significantly lower than in control mice at P3, and remained low at P10 and P25. In contrast, 5-HT1A heteroreceptor levels were comparable between control and 5-HT-deficient mice. These data define the postnatal development of 5-HT1A-receptor binding in the mouse brainstem. Furthermore, the data suggest that 5-HT1A-heteroreceptor deficits detected in sudden infant death syndrome are not a direct consequence of a 5-HT neuron dysfunction nor reduced brain 5-HT levels. To elucidate the developmental relationship between serotonin (5-HT) levels and 5-HT1A receptors in the brainstem, we examd. 5-HT1A binding in two 5-HT-deficient mouse models. In nuclei contg. 5-HT neurons, 5-HT1A binding was decreased (autoreceptors), while binding was maintained in projection sites (heteroreceptors). Thus, brainstem 5-HT1A-heteroreceptor-binding sites do not appear developmentally sensitive to reduced brain 5-HT levels.
- 36Jann, M. W., Lam, Y. W., and Chang, W. H. (1994) Rapid formation of clozapine in guinea-pigs and man following clozapine-N-oxide administration Arch. Int. Pharmacodyn. Ther. 328, 243– 25036https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXjt1yjsLc%253D&md5=23184a095504f13225b408b843fa61c0Rapid formation of clozapine in guinea pigs and man following clozapine-N-oxide administrationJann, M. W.; Lam, Y. W. F.; Chang, W. H.Archives Internationales de Pharmacodynamie et de Therapie (1994), 328 (2), 243-50CODEN: AIPTAK; ISSN:0003-9780. (Heymans Institute of Pharmacology)Clozapine and its metabolite clozapine-N-oxide (0.5 mg/kg) were administered i.p. to guinea-pigs. Significant amts. of clozapine were detected in plasma, liver, frontal cortex and caudate after clozapine-N-oxide administration. Tissue concns. of clozapine in liver, frontal cortex and caudate were greater than plasma concns. Clozapine concns. were almost equiv. in the liver. Clozapine concns. after N-oxide injection were approx. 40-50% lower compared to clozapine concns. after clozapine administration in the frontal cortex and caudate. A single dose of clozapine-N-oxide was given to a schizophrenic patient. Clozapine plasma concns. were detected after N-oxide administration. This study shows that clozapine is formed from its N-oxide metabolite and that a reversible metabolic pathway exists for clozapine and clozapine-N-oxide.
- 37Loffler, S., Korber, J., Nubbemeyer, U., and Fehsel, K. (2012) Comment on “Impaired Respiratory and Body Temperature Control Upon Acute Serotonergic Neuron Inhibition” Science 337, 646There is no corresponding record for this reference.
- 38Roth, B. L., Sheffler, D. J., and Kroeze, W. K. (2004) Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia Nat. Rev. Drug Discovery 3, 353– 35938https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1Gkt78%253D&md5=77196583acccca65457fec7a67218463Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophreniaRoth, Bryan L.; Sheffler, Douglas J.; Kroeze, Wesley K.Nature Reviews Drug Discovery (2004), 3 (4), 353-359CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Most common central nervous system disorders - such as depression, bipolar disorder, and schizophrenia - seem to be polygenic in origin, and the most effective medications have exceedingly complex pharmacologies. Attempts to develop more effective treatments for diseases such as schizophrenia and depression by discovering drugs selective for single mol. targets (i.e., 'magic bullets') have, not surprisingly, been largely unsuccessful. Here the authors propose that designing selectively non-selective drugs (i.e., 'magic shotguns') that interact with several mol. targets will lead to new and more effective medications for a variety of central nervous system disorders.
- 39Hunziker, F., Kuenzle, F., and Schmutz, J. (1966) Helv. Chim. Acta 49, 1433– 1439There is no corresponding record for this reference.
- 40Schmutz, J. (1975) Neuroleptic piperazinyl-dibenzo-azepines. Chemistry and structure–activity relationships Arzneimittelforschung 25, 712– 720There is no corresponding record for this reference.
- 41Steiner, G., Franke, A., Hadicke, E., Lenke, D., Teschendorf, H. J., Hofmann, H. P., Kreiskott, H., and Worstmann, W. (1986) Tricyclic epines. Novel (E)- and (Z)-11H-dibenz[b,e]azepines as potential central nervous system agents. Variation of the basic side chain J. Med. Chem. 29, 1877– 1888There is no corresponding record for this reference.
- 42Hunziker, F., Fischer, E., and Schmutz, J. (1967) 11-Amino-5H-Dibenzo[b,e]-1,4-diazepine 0.10. Mitteilung Uber Siebengliedrige Heterocyclen Helv. Chim. Acta 50, 1588– 1599There is no corresponding record for this reference.
- 43Wenthur, C. J. and Lindsley, C. W. (2013) Classics in Chemical Neuroscience: Clozapine ACS Chem. Neurosci. 4, 1018– 102543https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVyjtbbP&md5=75d84aea0d1ce9bccad9fda9a5425d06Classics in Chemical Neuroscience: ClozapineWenthur, Cody J.; Lindsley, Craig W.ACS Chemical Neuroscience (2013), 4 (7), 1018-1025CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)A review. Clozapine was the first true breakthrough in schizophrenia treatment since the discovery of chlorpromazine in 1950, effectively treating pos., neg., and some cognitive symptoms, as well as possessing unprecedented efficacy in treatment-resistant patients. Despite over 30 years of intense study, the precise mol. underpinnings that account for clozapine's unique efficacy remain elusive. In this Viewpoint, we will showcase the history and importance of clozapine to neuroscience in general, as well as for the treatment of schizophrenia, and review the synthesis, pharmacol., drug metab., and adverse events of clozapine.
- 44Kalhapure, R. S., Patil, B. P., Jadhav, M. N., Kawle, L. A., and Wagh, S. B. (2011) Synthesis of 11-(Piperazin-1-yl)-5H-dibenzo[b,e] [1,4]diazepine on Kilo Scale E-J. Chem. 8, 1747– 174944https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVKisrzI&md5=2e5c34ca9850cc28fbc20a9adf88257dSynthesis of 11-(piperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine on kilo scaleKalhapure, Rahul S.; Patil, Bhushan P.; Jadhav, Mahantesh N.; Kawle, Laxmikant A.; Wagh, Sanjay B.E-Journal of Chemistry (2011), 8 (4), 1747-1749CODEN: ECJHAO ISSN:. (WWW Publications)A synthesis of 11-(piperazin-1-yl)-5H-dibenzo[b,e][1,4]diazepine on kilo scale without any chromatog. purifn. step was reported. Key steps were Ullmann condensation, catalytic hydrogenation, and catalyzed cyclization.
- 45Ullmann, F. and Bielecki, J. (1901) Synthesis in the Biphenyl series. (I. Announcement) Ber. Dtsch. Chem. Ges. 34, 2174– 2185There is no corresponding record for this reference.
- 46Davies, M. A., Compton-Toth, B. A., Hufeisen, S. J., Meltzer, H. Y., and Roth, B. L. (2005) The highly efficacious actions of N-desmethylclozapine at muscarinic receptors are unique and not a common property of either typical or atypical antipsychotic drugs: is M1 agonism a pre-requisite for mimicking clozapine’s actions? Psychopharmacology 178, 451– 460There is no corresponding record for this reference.
- 47Besnard, J., Ruda, G. F., Setola, V., Abecassis, K., Rodriguiz, R. M., Huang, X. P., Norval, S., Sassano, M. F., Shin, A. I., Webster, L. A., Simeons, F. R., Stojanovski, L., Prat, A., Seidah, N. G., Constam, D. B., Bickerton, G. R., Read, K. D., Wetsel, W. C., Gilbert, I. H., Roth, B. L., and Hopkins, A. L. (2012) Automated design of ligands to polypharmacological profiles Nature 492, 215– 22047https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhvVamurfN&md5=ace6cced250cf968d327b482a5bf1798Automated design of ligands to polypharmacological profilesBesnard, Jeremy; Ruda, Gian Filippo; Setola, Vincent; Abecassis, Keren; Rodriguiz, Ramona M.; Huang, Xi-Ping; Norval, Suzanne; Sassano, Maria F.; Shin, Antony I.; Webster, Lauren A.; Simeons, Frederick R. C.; Stojanovski, Laste; Prat, Annik; Seidah, Nabil G.; Constam, Daniel B.; Bickerton, G. Richard; Read, Kevin D.; Wetsel, William C.; Gilbert, Ian H.; Roth, Bryan L.; Hopkins, Andrew L.Nature (London, United Kingdom) (2012), 492 (7428), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The clin. efficacy and safety of a drug is detd. by its activity profile across many proteins in the proteome. However, designing drugs with a specific multi-target profile is both complex and difficult. Therefore methods to design drugs rationally a priori against profiles of several proteins would have immense value in drug discovery. Here is described a new approach for the automated design of ligands against profiles of multiple drug targets. The method is demonstrated by the evolution of an approved acetylcholinesterase inhibitor drug donepezil (I) into brain-penetrable ligands with either specific polypharmacol. or exquisite selectivity profiles for G-protein-coupled receptors. Overall, 800 ligand-target predictions of prospectively designed ligands were tested exptl., of which 75% were confirmed to be correct. Target engagement in vivo is also demonstrated. The approach can be a useful source of drug leads when multi-target profiles are required to achieve either selectivity over other drug targets or a desired polypharmacol.
- 48Wiberg, K. B. (1955) The Deuterium Isotope Effect Chem. Rev. 55, 713– 74348https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaG2MXotFaqsA%253D%253D&md5=011c9eb50698d4d3c74971348c9b3ce7The deuterium isotope effectWiberg, Kenneth B.Chemical Reviews (Washington, DC, United States) (1955), 55 (), 713-43CODEN: CHREAY; ISSN:0009-2665.cf. C.A. 49, 13184e. The D isotope effect arises largely from the difference in zero-point energy between a bond to H and the corresponding bond to D. The max. isotope effect is obtained when the bond to H or D is essentially completely cleaved in the activated complex, and the isotope effect decreases with increasing bonding in the activated complex. In general, in any series of related reactions the reaction with the lowest activation energy has the lowest isotope effect. Although the isotope effect is useful in detg. whether a bond to H is cleaved in the rate-detg. step, it must be used with caution; e.g., no isotope effect was noted in the reaction of Grignard reagents with MeOH or H2O, a reaction which probably does involve the cleavage of a bond to H. There is no apparent difference in magnitude for the isotope effect in the removal of H or D as a proton, a hydride ion, or an atom. 241 references.
- 49Elison, C., Rapoport, H., Laursen, R., and Elliott, H. W. (1961) Effect of Deuteration of N-CH3 Group on Potency and Enzymatic N-Demethylation of Morphine Science 134, 1078– 1079There is no corresponding record for this reference.
- 50Shao, L., Abolin, C., Hewitt, M. C., Koch, P., and Varney, M. (2006) Derivatives of tramadol for increased duration of effect Bioorg. Med. Chem. Lett. 16, 691– 69450https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlCiu7bE&md5=6670712406293bbd421e52ac1f693781Derivatives of tramadol for increased duration of effectShao, Liming; Abolin, Craig; Hewitt, Michael C.; Koch, Patrick; Varney, MarkBioorganic & Medicinal Chemistry Letters (2006), 16 (3), 691-694CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Tramadol is a centrally acting opioid analgesic structurally related to codeine and morphine. Analogs of tramadol with deuterium-for-hydrogen replacement at metabolically active sites were prepd. and evaluated in vitro and in vivo.
- 51Korber, J., Loffler, S., Schollmeyer, D., and Nubbemeyer, U. (2013) Synthesis and Oxidant Properties of Phase 1 Benzepine N-Oxides of Common Antipsychotic Drugs Synthesis 45, 2875– 2887There is no corresponding record for this reference.