<<

REVIEW ARTICLE published: 23 May 2013 doi: 10.3389/fnint.2013.00040 Modulation of firing and synaptic transmission of serotonergic neurons by intrinsic G -coupled receptors and ion channels

Takashi Maejima, Olivia A. Masseck, Melanie D. Mark and Stefan Herlitze* Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany

Edited by: Serotonergic neurons project to virtually all regions of the central nervous system and Kae Nakamura, Kansai Medical are consequently involved in many critical physiological functions such as mood, sexual University, Japan behavior, feeding, sleep/wake cycle, memory, cognition, blood pressure regulation, Reviewed by: breathing, and reproductive success. Therefore, serotonin release and serotonergic David M. Lovinger, National Institutes of Health, USA neuronal activity have to be precisely controlled and modulated by interacting brain circuits Evan Deneris, Case Western Reserve to adapt to specific emotional and environmental states. We will review the current University, USA knowledge about G protein-coupled receptors and ion channels involved in the regulation of *Correspondence: serotonergic system, how their regulation is modulating the intrinsic activity of serotonergic Stefan Herlitze, Department of neurons and its transmitter release and will discuss the latest methods for controlling the Zoology and Neurobiology, Ruhr-University Bochum, modulation of serotonin release and intracellular signaling in serotonergic neurons in vitro Universitätsstr. 150, ND 7/31, and in vivo. D-44780 Bochum, Germany. e-mail: [email protected] Keywords: 5-HT system, GPCRs, auto-regulation, hetero-regulation, optogenetics

INTRODUCTION densely packed in the midline and some 5-HT neurons are scat- The serotonergic system consists of a small number of neurons tered in the periphery. Within the DRN and MRN 5-HT neurons that are born in the ventral regions of the hindbrain (Deneris and project to defined target area in brain (Adell et al., 2002; Lechin View metadata, citation and similar papers at core.ac.uk brought to you by CORE Wyler, 2012). In the adult nervous system, serotonergic neurons et al., 2006). For example, DRN 5-HT neurons innervate the pre- [5-HT (5-hydroxytryptamine) neurons] are located in the nine frontal cortex, lateral septum, and ventral hippocampus, while provided by Frontiers - Publisher Connector raphe nuclei that are restricted to the basal plate of the mid- MRN 5-HT neurons innervate the temporal cortex, medial sep- brain, pons, and medulla (Dahlstrom and Fuxe, 1964). 5-HT tum, and dorsal hippocampus. Afferent projections to the raphe neurons located in the rostral raphe nuclei, such as the dorsal nuclei are diverse and include acetylcholine (ACh) from the lat- raphe nucleus (DRN) and the median raphe nucleus (MRN), give erodorsal tegmental nucleus, dopamine from the substantia nigra rise to the majority of the serotonergic ascending fibers into the and ventral tegmentum area, histamine from tuberomammil- forebrain including cerebral cortex, limbic system, and basal gan- lary hypothalamic nucleus, noradrenaline (NA) from the locus glia (Jacobs and Azmitia, 1992). The activity of the serotonergic coeruleus, serotonin itself from the raphe nuclei and several system is regulated via transmitter release from local interneurons neuropeptides as well as excitatory glutamatergic and inhibitory and/or afferents to the raphe nuclei (hetero-regulation), via mech- GABAergic inputs. Glutamatergic inputs come from several nuclei anisms arising from 5-HT neurons themselves (auto-regulation), including medial prefrontal cortex and lateral habenula nucleus and potentially via alterations in the extracellular milieu (e.g., (Aghajanian and Wang, 1977; Celada et al., 2001; Adell et al., increase in CO2; Pineyro and Blier, 1999; Richerson, 2004). In this 2002; Bernard and Veh, 2012). Some inputs make direct contacts review, we will discuss G protein-coupled receptors (GPCRs) and with 5-HT neurons, while others project onto local GABAergic ion channels located at somatodendritic and presynaptic regions interneurons that provide feedforward inhibitory input to 5-HT of 5-HT neurons in the DRN and MRN that contribute to the mod- neurons (Sharp et al., 2007). In addition to the local GABAergic ulation of 5-HT neuronal activity and 5-HT release (Figure 1). interneurons located in the raphe nuclei and in the neighboring The DRN and MRN are the primary nuclei of 5-HT projections periaqueductal gray area, extrinsic GABAergic projections have to forebrain and provide the neural substrate to communicate been suggested (Gervasoni et al., 2000). The responsiveness of 5- between global forebrain and other neuromodulatory systems by HT neurons to each of the inputs differs between DRN and MRN, sending a wide range of 5-HT projections and receiving a wide and also within the subnuclei of the DRN (Adell et al.,2002; Lechin variety of afferents (Jacobs and Azmitia, 1992). The DRN is located et al., 2006). The differences in responsiveness most likely depend right beneath the posterior part of cerebella aqueduct and con- on differences in the strength of the afferent inputs for each of tains about half of all 5-HT neurons in the central nervous system the nuclei and subnuclei and also on the expression and types of (CNS), which can be further divided into six regions: rostral, cau- the ionotropic and metabotropic receptors in 5-HT neurons. Fur- dal, dorsomedial, ventromedial, interfascicular, and lateral parts. thermore, the intrinsic membrane excitability of 5-HT neurons The MRN is located at the ventral expansion of the DRN or the has been reported to differ in the distinct raphe nuclei (Beck et al., midline of the pontine tegmentum where many 5-HT neurons are 2004; Crawford et al., 2010). Importantly, beyond the anatomical

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 1

“fnint-07-00040” — 2013/5/21 — 14:57 — page1—#1 Maejima et al. Modulation of 5-HT neurons

and physiological differences, it has been reported that subpop- The predominant 5-HT receptors at the presynaptic termi- ulation of 5-HT neurons have distinct implications in specific nal are the 5-HT1B/1D receptors. 5-HT1B/1D receptorshavebeen physiological function and behavior (Abrams et al., 2004; Lechin shown to inhibit 5-HT release from the axonal varicosities as et al., 2006; Hale and Lowry, 2010). demonstrated with electrophysiological experiments (Sprouse and Aghajanian, 1987; Boeijinga and Boddeke, 1993; Morikawa et al., AUTO-REGULATION 2000), probably due to inhibition of Ca2+ influx through voltage- The midbrain 5-HT neurons elicit spontaneous action potentials gated Ca2+ channels such as P/Q-type and N-type Ca2+ channel (APs), with a regular, slow firing pattern (1–5 APs/s; Aghajanian (Kimura et al., 1995; Harvey et al., 1996). The 5-HT1B receptors and Vandermaelen, 1982; Vandermaelen and Aghajanian, 1983). have been shown to underlie presynaptic autoinhibition of 5- 5-HT released from 5-HT neurons act either on the 5-HT neu- HT release in which 5-HT1A-mediated slow IPSPs are reduced by ron itself or on the target circuits. There are several ways how previous released 5-HT activating presynaptic 5-HT1B receptors 5-HT neurons may receive 5-HT. First, dendrodendritic (Morikawa et al., 2000). In addition, 5-HT1B receptorshavebeen releasing 5-HT have been described in raphe nuclei between 5-HT suggested to up-regulate 5-HT reuptake by serotonin transporters neurons. Second, recurrent axonal collaterals have been suggested (Xie et al., 2008; Hagan et al., 2012) and 5-HT synthesis by itself to back-propagate to the raphe nucleus itself to release 5-HT. might be under the control of 5-HT1B (Hjorth et al., 1995). Thus, Finally, 5-HT neurons between different raphe nuclei such as 5-HT1B autoreceptors may have the ability to control 5-HT release DRN and MRN communicate with each other via 5-HT (for independently from the actual firing rate. review, see Adell et al., 2002; Harsing, 2006; Lechin et al., 2006). 5-HT1F mRNA has also been detected in the raphe Indeed electrical stimulation in DRN slice preparations induces nuclei (Bruinvels et al., 1994). Since 5-HT1F receptorshavea 5-HT1A receptor-mediated slow inhibitory postsynaptic poten- high affinity for sumatriptan, a 5-HT1B/1D agonist, and the tials (IPSPs) in 5-HT neurons (Pan et al., 1989; Morikawa et al., sumatriptan-induced reduction in 5-HT release (monitored by 2000), demonstrating 5-HT release in the proximity of 5-HT voltammetry in brain slices) could not be blocked by 5-HT1A/1B/1D neurons. receptor antagonists, 5-HT1F had been suggested as a possible can- Once 5-HT is released, 5-HT receptors will be activated. Seven didate of a serotonergic autoreceptor in the MRN (Hopwood and subgroups of 5-HT receptors encoding ionotropic as well as Stamford, 2001). metabotropic receptors have been described (5-HT1–5-HT7), with While the expression and function of 5-HT1 receptors has been 15 total variants identified to date (Barnes and Sharp, 1999; Hoyer directly demonstrated in 5-HT neurons, involvement of other 5- et al., 2002; Kroeze et al., 2002). The 5-HT GPCRs can be divided HT receptors such as 5-HT2, 5-HT3, and 5-HT4−7 is less clear and into three major subgroups depending on which G protein sig- may differ among species, the developmental stage of the animal naling pathway they activate. 5-HT1 receptors couple mainly to and 5-HT neuron subtypes. the Gi/o pathway; 5-HT4, 5-HT5, 5-HT6, and 5-HT7 receptors 5-HT2 receptors are functionally expressed in particular on couple to the Gs pathway; and 5-HT2 receptors activate the Gq/11 GABAergic interneurons in the DRN, since activation of 5- pathway. The 5-HT3 receptors are -gated ion channels. HT2A/2C receptors increase fast inhibitory postsynaptic current 5-HT1A, 5-HT1B, and 5-HT1D receptors are found on soma- (IPSC) frequency in 5-HT neurons and reduce 5-HT neuronal fir- todendritic and axonal region of 5-HT neurons (McDevitt and ing as electrophysiologically measured in brain slices (Liu et al., Neumaier, 2011). All three receptors act as negative feedback effec- 2000; Leysen, 2004). 5-HT2 receptor mRNA and have tors for 5-HT neuronal firing and 5-HT release (Andrade, 1998). been identified in the DRN and embryonic 5-HT neurons (Wright Somatodendritically located 5-HT1A receptors down-regulate the et al., 1995; Clemett et al., 2000; Wylie et al., 2010). Additionally, 5- firing rate of 5-HT neurons via activation of G protein-coupled HT2 receptors have been postulated to increase 5-HT1A-mediated inwardly rectifying potassium channels (GIRK) leading to mem- responses in 5-HT neurons (Kidd et al., 1991). However, a direct brane hyperpolarization, and reduction or complete block of modulatory effect of 5-HT2 in 5-HT neurons has not been AP firing (Colino and Halliwell, 1987; Sprouse and Aghajanian, demonstrated. 1987; Blier et al., 1989; Hjorth and Sharp, 1991; Penington et al., The 5-HT3 receptors have also been suggested to act as presy- 1993; Stamford et al., 2000). In addition, 5-HT1A receptors inhibit naptic autoreceptors in serotonergic nerve terminals. Although 2+ voltage-gated Ca channels of the N- and P/Q-type in 5-HT 5-HT3 receptors have been shown to enhance 5-HT release in neurons, as application of a selective 5-HT1A agonist dimin- various brain areas including the raphe nuclei as monitored ishes somatic Ca2+ channel currents (Penington and Kelly, 1990; by [3H]5-HT assays (Bagdy et al., 1998), there is no direct Penington et al., 1992; Bayliss et al., 1997b). The functional conse- immunohistochemical and electrophysiological evidence of the 2+ quence of Ca channel inhibition is an increase in the firing rate presence of 5-HT3 receptors in 5-HT neurons (van Hooft and due to reduction in the afterhyperpolarization, which may involve Vijverberg, 2000). 2+ + Ca activated-K channel (Bayliss et al., 1997b). The physio- For the Gs protein-coupled 5-HT4−7 receptors, mainly indirect logical role of the differential effects of 5-HT1A receptorsonthe evidence exists for an autoregulatory role of these GPCRs in 5-HT AP firing has not been addressed so far, but may involve input neurons. 2+ specificity due to 5-HT1A/GIRK and 5-HT1A/Ca channel colo- 5-HT4 receptors seem to be located somatodendritically and calization in specific subcellular domains and/or differences in the presynaptically. A presynaptic potentiating effect of 5-HT4 recep- regulatory properties of heterogeneous 5-HT neurons within and tor on glutamate release, which can be counteracted by 5- among different raphe nuclei (Calizo et al., 2011). HT1A receptor-mediated inhibitory action, has been described in

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 2

“fnint-07-00040” — 2013/5/21 — 14:57 — page2—#2 Maejima et al. Modulation of 5-HT neurons

FIGURE 1 | Intrinsic somatodendritic and presynaptic modulation of ion channels. Gq/11 pathway activation can lead to the activation of action potential (AP) firing and transmitter release in 5-HT neurons. (A) non-selective cation channels or inhibition of K+ conductance. In addition, the AP firing of 5-HT neurons is modulated by GPCRs and ion channels. AP firing activation of L-type Ca2+ channels via protein kinase C (PKC) can most likely is reduced (red arrows) via activation of GPCRs coupling to the Gi/o pathway increase membrane depolarization and transcription. (B) 5-HT release of 5-HT or activation of inhibitory ligand-gated ion channels. Gi/o pathway activation neurons is also modulated by GPCRs and ion channels. 5-HT release is 2+ can lead to the activation of GIRK channels, inhibition of voltage-gated Ca reduced (red arrow) via activation of GPCRs coupling to the Gi/o pathway. Gi/o 2+ channels or increase in GABAA receptor currents. In addition, Gq/11 activation pathway activation leads to the inhibition of presynaptic Ca channels, can lead to synthesis of endocannabinoids, which inhibit transmitter release reduction in Ca2+ influx and therefore reduction in transmitter release. 5-HT onto 5-HT neurons. AP firing is increased (blue arrows) via activation of release is increased (blue arrows) via activation of GPCRs coupling to the Gs GPCRs coupling to the Gq/11 pathway or activation of excitatory ligand-gated pathway and via opening of excitatory, ligand-gated ion channels.

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 3

“fnint-07-00040” — 2013/5/21 — 14:57 — page3—#3 Maejima et al. Modulation of 5-HT neurons

hippocampal neurons (Kobayashi et al., 2008). Since neurotrans- needs to be defined and investigated (for a complete list, see Wylie mitter release of various transmitters including 5-HT is modulated et al., 2010). We will summarize recent findings on the intrinsic by 5-HT4 agonists, a presynaptic localization of 5-HT4 receptors hetero-regulation of the serotonergic transmitter system. on 5-HT neurons seems possible (Mengod et al., 2010). While the function of 5-HT5 receptors in the CNS has not been GABA, GLYCINE, AND GLUTAMATE thoroughly studied, two studies suggest a role of 5-HT5 receptors 5-HT neurons within the raphe nuclei receive in particular for modulating 5-HT neurons. First, 5-HT5B receptor mRNA, a GABAergic but also glutamatergic input. As expected, the receptor which is expressed in rodents but not in humans, is colo- GABAergic input onto 5-HT neurons reduces the neuronal firing, calized with the mRNA of 5-HT transporter in the DRN (Serrats while glutamatergic input increases the firing activity (Pan and et al., 2004). Second, block of 5-HT5A receptors in the DRN atten- Williams, 1989; Levine and Jacobs, 1992; Becquet et al., 1993a,b; uates the 5-carboxamidotryptamine (5-CT; non-selective agonist for review, see Adell et al., 2002; Harsing, 2006). These effects have in particular for 5-HT1A/1B/1D receptors) induced reduction of 5- been mainly attributed to the expression of ionotropic GABA and HT neuronal firing but fail to affect 5-HT release measured using glutamate receptors (GluRs) in 5-HT neurons (Tao and Auerbach, fast cyclic voltammetry in vitro (Thomas et al., 2006). The data 2000; Gartside et al., 2007), which is in agreement with the expres- suggest an autoreceptor modulation of 5-HT neurons via 5-HT5A sion of 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic receptors. acid (AMPA), N-methyl-D-aspartate (NMDA), kainate receptors 5-HT6 and 5-HT7 receptor protein and mRNA have been [GluR 1,2; NMDA-R-2B, kainate receptor 5 (Grik5)] as well as detected in cells in the raphe nuclei including the DRN (Ruat GABAA receptor subunits (GABAA β1–3 and γ2) in embryonic et al., 1993; To et al., 1995; Gustafson et al., 1996; Woolley et al., 5-HT neurons (Wylie et al., 2010). Interestingly, the glycine recep- 2004; see also Gerard et al., 1997; Hamon et al., 1999), but a func- tor α1 and β subunits have also been identified in human brain tional role as autoreceptors for modulating 5-HT neurons could and mice embryonic 5-HT neurons (Baer et al., 2003; Wylie et al., not be demonstrated so far (Bourson et al., 1998; Roberts et al., 2010). Indeed the DRN receives input from glycinergic fibers 2001). (Rampon et al., 1996, 1999) to inhibit 5-HT neuronal firing and 5- Thus, the auto-regulation of 5-HT neuronal firing is in par- HT release (Gallager and Aghajanian, 1976; Wang and Aghajanian, ticular regulated by 5-HT1A receptors via activation of the Gi/o 1977; Becquet et al., 1993a). + 2+ pathway and opening K and closing Ca conductance. At the The modulation of 5-HT neurons by GABAB receptors has presynaptic terminal, 5-HT1B/1D receptor activation reduces 5-HT been addressed in various studies. In general, activation of GABAB release most likely via Gi/o protein-mediated inhibition of presy- receptor by selective agonists decreases 5-HT release (Becquet 2+ naptic Ca channels. In addition, potentiation of 5-HT release et al., 1993b). The decrease of 5-HT release by GABAB receptors by activating 5-HT4 receptors via the Gs pathway seems possible. located within 5-HT neurons is most likely mediated via activa- Since other 5-HT receptor mRNAs have been detected in 5-HT tion of GIRK channels leading to a reduction in AP firing (Innis neurons, other autoregulatory mechanisms may exist in subgroups and Aghajanian, 1987; Williams et al., 1988; Bayliss et al., 1997a; of 5-HT neurons or during different developmental stages of the Cornelisse et al., 2007). Within 5-HT neurons, GABAB receptors serotonergic transmitter system. In particular, animal models for are located extrasynaptically, suggesting that spillover of GABA the selective activation of these GPCRs during development will during high activity of GABAergic neurons would modulate 5- further elucidate the modulatory role of other 5-HT receptors in HT neuronal activity within raphe nuclei (Varga et al., 2002). the auto-regulation of 5-HT neuronal firing and 5-HT release. On the other hand, there is little information about modula- tory effects of metabotropic GluRs (mGluRs) in 5-HT neurons HETERO-REGULATION so far. Although administration of group II mGluR (mGluR2/3) 5-HT modulates various complex behaviors and therefore the antagonist has been reported to increase 5-HT neuronal activity, serotonergic transmitter system receives feedback and feedforward an indirect effect on presynaptic excitatory neurons seems to be information from other brain areas and networks involved in reg- involved (Kawashima et al., 2005). ulating the different behaviors (Adell et al., 2002; Lechin et al., 2006; Sharp et al., 2007). Thus, the hetero-regulation of the 5-HT CORELEASE OF GLUTAMATE OR GABA FROM 5-HT NEURONS neurons involves various transmitter systems. Previous reports have suggested the possibility of glutamate release Forty-nine different GPCRs belonging to all four GPCR sub- from 5-HT neurons based on the presence of vesicular glutamate families were identified in postmitotic embryonic 5-HT neurons transporter type 3 in a subset of 5-HT neurons (Gras et al., 2002; using microarray expression profiling (Wylie et al., 2010). These Amilhon et al., 2010). Using optogenetic techniques, the core- GPCRs include adrenergic, calcitonin, cannabinoid, GABA, his- lease of glutamate and 5-HT from serotonergic terminals could tamine, opioid, and serotonin receptors. For these transmitter be demonstrated in a serotonergic projection from the MRN to systems, a postnatal modulatory role for 5-HT neurons has been hippocampal GABAergic interneurons (Varga et al., 2009). The described (see below). In addition, other GPCRs such as thrombin, serotonergic fibers make direct synaptic contacts to the GABAer- chemokine, prostaglandin E, melanin-concentrating hormone, gic neurons and exert fast synaptic transmission mediated by cadherin, and parathyroid hormone receptors as well as ionotropic GluRs and 5-HT3 receptors. In addition to the glu- (FZD) and (SMO) homolog and the orphan recep- tamate transporters, GABA and its synthesizing enzyme, glutamic tors (GPR 19, 56, 85, 98, 125 135, 162, and 173) were also identified acid decarboxylase (GAD) have also been reported in subsets but a physiological role for their modulation of 5-HT neurons still of 5-HT neurons, suggesting the corelease of GABA and 5-HT

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 4

“fnint-07-00040” — 2013/5/21 — 14:57 — page4—#4 Maejima et al. Modulation of 5-HT neurons

(Nanopoulos et al., 1982; Belin et al., 1983; Fu et al., 2010; Hioki coupling might be explained by the heterodimerization between et al., 2010; Shikanai et al., 2012). However, vesicular inhibitory D1- and D2-like receptors (Rashid et al., 2007; Hasbi et al., 2010). amino acid transporter which is necessary for filling synaptic vesi- It has to be noted that the experiments suggesting the modula- cles with GABA was absent in 5-HT/GAD67 positive neurons and tion of 5-HT neurons by intrinsic D2-like receptors have been their projections, suggesting that GABA may be released by non- performed with relatively high concentrations of quinpirole and vesicular mechanisms such as a reverse transport through GABA sulpiride in in vitro preparations. Therefore, the experiments have transporters (Shikanai et al., 2012). Thus, 5-HT axonal projections to be interpreted carefully. have a potential to modulate the neuronal activity in target areas using at least three different transmitters, i.e., 5-HT, glutamate, NORADRENALINE and GABA. It is intriguing to speculate that the auto-regulation of 5-HT neurons in the raphe nuclei receive noradrenergic input 5-HT neurons itself might be modulated by corelease of glutamate in particular from the locus coeruleus (Adell et al., 2002; Lechin and GABA. et al., 2006). α1 and α2 mRNA and protein have been detected in the DRN and MRN (Unnerstall et al., 1985; ACETYLCHOLINE Rosin et al., 1993; Scheinin et al., 1994; Talley et al., 1996; Day et al., The DRN also receives cholinergic input from the laterodorsal 1997; Strazielle et al., 1999). α1 and α2 adrenergic receptors couple tegmental nucleus (Wang et al., 2000). Modulation of 5-HT neu- to the Gq/11 and Gi/o pathway, respectively. Therefore depending rons by ACh mainly involves nicotinic ACh receptors (nAChRs) on pathway activation, an increase or decrease in 5-HT neuronal and can increase 5-HT neuronal firing (Mihailescu et al., 2002) activity and 5-HT release can be postulated if adrenergic recep- for example, via presynaptic modulation of glutamate release tors are expressed in 5-HT neurons. Early studies revealed that NA (Garduno et al., 2012) or via opening of nAChR expressed in causes an increase in 5-HT neuronal firing in DRN. This effect 5-HT neurons (Galindo-Charles et al., 2008; Chang et al., 2011). has been suggested to be mediated via activation of α1 adreno- Very limited information is available for the expression and func- ceptors (presumably α1B adrenoceptor subtype) located on 5-HT tion of muscarinic ACh receptors (mAChRs) in 5-HT neurons. neurons (Vandermaelen and Aghajanian, 1983; Day et al., 1997) mAChR-M1 receptors (Gq/11) might be expressed on serotoner- and may involve the suppression of a 4-aminopyridine sensitive + gic projections into the hippocampus (Rouse and Levey, 1996) K conductance (IA; Aghajanian, 1985). In vivo experiments sug- and application of mAChR antagonist, atropine into the DRN gest that α1 adrenoceptors are tonically activated by endogenous enhances antidepressant-like 5-HT1A agonist effects (Haddjeri NA (Adell and Artigas, 1999; Pudovkina et al., 2003). In con- 3 et al., 2004), which may involve M2 (Gi/o) but not M1 (Gq/11) trast, 5-HT release detected by voltammetry or [ H]5-HT assay receptors (Haddjeri et al., 2000). Nevertheless a detailed and direct in the DRN slice preparation is inhibited by NA, an effect which demonstration of the involvement of mAChRs in 5-HT neurons has been attributed to α2 adrenoceptors (involving α2A adreno- is essential. ceptor subtype) and also perhaps indirectly to α1 adrenoceptors (Frankhuijzen et al., 1988; Hopwood and Stamford, 2001). Since DOPAMINE α2 receptors couple to the Gi/o pathway, 5-HT release and 5-HT The 5-HT neurons of the DRN reciprocally interact with neuronal firing could be reduced via α2 adrenoceptors located at the dopaminergic mesencephalic transmitter system involving the soma or presynaptic terminal of 5-HT neurons itself (Hop- dopamine receptors. D1-like receptors (D1 and D5) couple to wood and Stamford, 2001), or via inhibition of NA release at the Gs pathway, while dopamine D2-like receptors (D2−4)have noradrenergic synaptic terminals lowering the effective NA con- been described to couple to the Gi/o pathway. In the DRN, D2 centration for α1 adrenoceptors/Gq signaling pathway activation. and D3 receptor expression has been detected so far, with very The direct effect of α2 adrenoceptors in 5-HT neurons is sup- little or no expression of D1-like receptors (Bouthenet et al., 1987; ported by the fact that embryonic 5-HT neurons express α2A Dawson et al., 1988; Cortes et al., 1989; Wamsley et al., 1989; adrenoceptors (Wylie et al., 2010). Yokoyama et al., 1994; Suzuki et al., 1998). Dopamine increases 5-HT neuronal firing in DRN in vivo and in slice electrophysio- HISTAMINE logical recording, and also increases 5-HT release detected by in There are four histamine receptors (H1−4), which couple to dif- vivo microdialysis in DRN and other brain areas (Ferre and Arti- ferent G protein pathways, i.e., H1 (Gq/11), H2 (Gs), H3, and H4 gas, 1993; Ferre et al., 1994; Matsumoto et al., 1996; Mendlin et al., (Gi/o). Early studies suggested that histamine reduces the firing 1998; Haj-Dahmane, 2001; Martin-Ruiz et al., 2001). The modu- of 5-HT neurons in the DRN (Lakoski and Aghajanian, 1983) latory effects on 5-HT neurons have been shown to be mediated by via H2 receptors (Lakoski et al., 1984). Since H2 couples to the D1- and D2-like receptors located outside the DRN (Martin-Ruiz Gq/11 pathway, the results suggest that H2 receptors are localized et al., 2001)orbydirectactivationofD2 receptors expressed in on GABAergic terminals. Later findings showed that histamine 5-HT neurons (Haj-Dahmane, 2001). Activation of D2-like recep- increased 5-HT neuronal firing in the DRN via activation of H1 tors in 5-HT neurons leads to membrane depolarization, involving receptors and the opening of a non-selective cation conductance activation of G proteins, phospholipase C, and a non-selective through Gq/11 signaling pathways (Barbara et al., 2002; Brown cation current, most likely mediated by a transient receptor poten- et al., 2002). Expression profiling in mouse embryos suggested tial (TRP) channel (Aman et al.,2007). The D2-like receptor effects the expression of H3 receptors in 5-HT neurons, which is consis- in 5-HT neurons suggest that D2-like receptors may activate the tent with high mRNA levels in the DRN (Lovenberg et al., 1999; Gq/11 rather than the Gi/o signaling pathway. The Gq/11 protein Drutel et al., 2001; Pillot et al., 2002). However, the low binding

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 5

“fnint-07-00040” — 2013/5/21 — 14:57 — page5—#5 Maejima et al. Modulation of 5-HT neurons

of a H3 receptor selective radioligand in the DRN suggests that and corticotropin-releasing factor (CRF). Among these, various H3 receptors are mainly functional at the presynaptic terminal peptide receptors have been identified and functionally described of 5-HT projections (Pillot et al., 2002). Indeed increasing lev- in 5-HT neurons. els of histamine decrease 5-HT release detected by an in vivo electrochemical technique (Hashemi et al., 2011). BOMBESIN To date three types of bombesin receptors have been described, ENDOCANNABINOIDS i.e., the neuromedin B (NMB) receptor (BB1 receptor), the gastrin- The family consists of two subtypes, CB1 and releasing peptide (GRP) receptor (BB2 receptor) and the bombesin CB2. Modulation of neuronal activity is mainly exerted via CB1, receptor subtype 3 (BRS-3 or BB3 receptor; Jensen et al.,2008). The which couples to the Gi/o protein (Howlett et al., 1986) and proba- NMB receptors are functionally expressed in 5-HT neurons in the bly also to the Gs pathway (Glass and Felder, 1997). CB1 receptors DRN (Pinnock et al., 1994; Woodruff et al., 1996). BB1 receptor are localized in particular at presynaptic terminals, where they activation leads to an increase in 5-HT neuronal firing via sup- inhibit presynaptic Ca2+ influx and reduce transmitter release via pression of K+ current, involving most likely the activation of the endocannabinoid (eCB)-mediated retrograde signaling (Maejima Gq/11 pathway (Benya et al., 1992; Woodruff et al., 1996) and as a et al., 2001). CB1 receptors are expressed in serotonergic fibers consequence, an increase in 5-HT release to projection site such (Haring et al., 2007; Ferreira et al., 2012) and their mRNA is found as the hippocampus, which is monitored by in vivo microdialysis early in development (Wylie et al., 2010). 5-HT release in pro- (Merali et al., 2006). jection areas from the DRN is reduced by activation of CB1 as monitored by microdialysis (Egashira et al., 2002) and [3H]5-HT CALCITONIN assay (Nakazi et al., 2000; Egashira et al., 2002), and increased by (CalcR) mRNA has been localized in 5-HT inhibition of CB1 in vivo and in vitro (Darmani et al.,2003; Tzavara neurons in the DRN (Nakamoto et al.,2000), which is in agreement et al., 2003; Aso et al., 2009). Studies in CB1 knock-out animals with the expression profiling studies of postmitotic embryonic 5- and chronic activation of CB1 receptors in vivo suggest that CB1 HT neurons (Wylie et al., 2010). The CalcR couples to the Gs may regulate the function and expression of 5-HT1A receptors pathway (Hay et al., 2005) and Gq/11 pathway (Offermanns et al., (Aso et al., 2009; Moranta et al., 2009; Zavitsanou et al., 2010). 1996). Since high levels of amylin binding sites are detected in Interestingly, 5-HT neurons itself synthesize eCBs in an activity- the DRN (Sexton et al., 1994) and mRNA for CGRP has been dependent manner (Haj-Dahmane and Shen, 2009, 2011). eCB localized in 5-HT positive axon terminals in monkeys (Arvidsson release from 5-HT neurons can be induced by orexin-B leading et al., 1990), it is most likely that CalcR assemble with receptor to the activation of orexin (OX) receptors via the Gq/11 pathway activity-modifying proteins (RAMPs) in 5-HT neurons to respond (Liu et al., 2002b; Haj-Dahmane and Shen, 2005). It has been to the various peptide transmitters (i.e., CGRP, adrenomedullin, therefore speculated that the activity-dependent activation of the and amylin; Tilakaratne et al., 2000). A direct function of CalcR in Gq/11 pathway in general may lead to the production of eCBs in 5-HT neurons has not yet been demonstrated. However, injection 5-HT neurons (Haj-Dahmane and Shen, 2011). Within the DRN of CGRP into rats induces anxiety-like behaviors and increases eCBs mainly act on glutamatergic terminals and probably also on c-Fos expression in the DRN (Sink et al., 2011). GABAergic terminals (Liu et al., 2002b; Haj-Dahmane and Shen, 2009; Mendiguren and Pineda, 2009; Tao and Ma, 2012), lead- CHEMOKINE RECEPTORS ing to a reduction in glutamate and GABA release onto 5-HT Two subtypes of the 18 identified chemokine receptors have been neurons and therefore changes the activity of the 5-HT neurons detected in microarray analyses from embryonic 5-HT neurons, itself. i.e., Duffy antigen/chemokine (C-X-C motif) receptor 4 (CXCR4; Wylie et al., 2010). CXCR4 is expressed in the majority of 5- FRIZZLED RECEPTORS HT neuron outer membranes in the DRN (Heinisch and Kirby, Four frizzled receptors (FZD1–3 and SMO) have been detected in 2010). So far only an indirect action of CXCR4 for modula- postmitotic embryonic 5-HT neurons (Wylie et al., 2010). These tion of 5-HT neuron has been demonstrated, since application receptors mainly couple to the Wnt signaling cascade and are of the CXCR4 ligands and antagonists modulate GABA and glu- most likely involved in the development and maturation of 5- tamate release onto 5-HT neurons (Heinisch and Kirby, 2010), HT neurons (Simon et al., 2005; Song et al., 2012). However, which is in agreement with the described Gi/o protein-mediated the role of frizzled receptors in 5-HT neurons remains to be inhibition of Ca2+ channels via CXCR4 (Oh et al., 2002). In addi- determined. tion, CX3CR1 is also expressed in 5-HT neurons in the DRN and MRN (Heinisch and Kirby, 2009). Here the CX3CR1 spe- NEUROPEPTIDES cific ligand, fractalkine/CX3CL1 increased evoked IPSC amplitude Dr. Hökfelt’s laboratory demonstrated that various peptide trans- on 5-HT neurons (Heinisch and Kirby, 2009), an effect which mitters are expressed in the DRN with species-specific differences is most likely mediated postsynaptically and not presynaptically. between mice and rats (Fu et al., 2010). The identified neu- The effect is surprising, since CX3CR1 has been described to cou- ropeptides are cholecystokinin (CCK), calcitonin -related ple to the Gi/o pathway (Oh et al., 2002), which would inhibit peptide (CGRP), vasoactive intestinal peptide (VIP), somato- synaptic transmitter release and induce paired-pulse facilitation statin, substance P,dynorphin, neurotensin, thyrotropin-releasing (PPF) if activated on GABAergic terminals. Therefore, the CX3CL1 hormone (TRH), enkephalin, galanin, neuropeptide Y (NPY), could increase/modulate GABAA receptor trafficking and GABAA

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 6

“fnint-07-00040” — 2013/5/21 — 14:57 — page6—#6 Maejima et al. Modulation of 5-HT neurons

receptor currents in 5-HT neurons via activation of CX3CR1. A (Mazarati et al., 2005). Therefore GalR2 receptors may activate 5- signaling function for Duffy antigen remains to be determined. HT neurons via reduction in GABAergic input onto 5-HT neurons and/or via Gq/11-mediated increase in 5-HT neuronal firing. CHOLECYSTOKININ Galanin also modulates 5-HT1A autoreceptor responses in vivo.A The expression of CCK receptors in 5-HT neurons in the DRN has possible mechanism of this modulation could be the heterodimer- also been suggested. Application of CCK increases 5-HT neuronal ization of GalR with 5-HT1A which has been observed in het- erologous expression systems (for review, see Kuteeva et al., 2008; firing, which is blocked by the CCK1 antagonist L-364,718 (Boden et al., 1991). In addition, 5-HT release measured as outflow of Borroto-Escuela et al., 2010). [3H]5-HT in cortical slices is increased by CCK-4, which involves HYPOCRETIN–OREXIN CCK2 receptors (Siniscalchi et al., 2001). Both CCK1 and CCK2 The two hypocretin/orexin (OX1 and OX2) receptors are expressed receptors mainly couple to the Gq/11 and Gs pathway (de Weerth et al., 1993; Lee et al., 1993; Ulrich et al., 1993). in tryptophan hydroxylase-positive neurons in the DRN (Brown et al., 2002). Their intracellular signaling targets are rather com- plex involving activation of Gi/o,Gq/11,Gs, and other G proteins CORTICOTROPIN-RELEASING FACTOR (Scammell and Winrow, 2010). Orexin positive fibers project onto Two CRF (CRF1 and CRF2) receptor subtypes have been described GABAergic as well as 5-HT neurons in the DRN (Peyron et al., in brain and both seem to be localized in GABAergic neurons in 1998). Application of the neuropeptides orexin-A and orexin-B the DRN as well as in 5-HT and non-5-HT neurons with differen- causes a Na+/K+ non-selective cation current in 5-HT neurons tial subcellular localizations (for review, see Valentino et al., 2010). (Brown et al., 2002; Liu et al., 2002b; Kohlmeier et al., 2008), sug- CRF1 and CRF2 couple to the Gs pathway (Chang et al., 1993; Per- gesting that activation of OX1 and OX2 leads to the increase rin et al., 1993; Vita et al., 1993; Lovenberg et al., 1995; Liaw et al., of 5-HT neuronal firing. The neuropeptides also induce GABA 1996) and also the Gq/11 pathway in heterologous expression sys- release onto 5-HT neurons at higher peptide concentrations (Liu tems (Dautzenberg et al., 2004), leading to the assumption that et al., 2002b). In addition, orexin increases the somatic L-type stimulation of CRF1 or CRF2 will increase neuronal firing. Indeed, Ca2+ current in 5-HT neurons in a protein kinase C-dependent activation of CRF1 on GABAergic neurons increases GABA release manner (Kohlmeier et al., 2008). It has therefore been suggested onto 5-HT neurons, while activation of CRF2 elicits an inward cur- that modulation of Ca2+ transients by orexin may be involved in rent in 5-HT neurons (Kirby et al., 2008). Based on the differential the transcriptional regulation of long-term processes (Kohlmeier localization of the CRF receptors within the DRN and its receptor et al., 2008). type-specific action on 5-HT neurons, it has been suggested that at low concentrations of CRF,5-HT neuronal activity is decreased, OPIOIDS while at high concentrations, 5-HT neuronal activity is increased Raphe nuclei receive dynorphinergic, enkephalinergic, and β- (Kirby et al., 2008; Valentino et al., 2010). endophinergic innervation (Adell et al., 2002). These transmitters activate μ, κ and δ opioid receptors, which primarily couple to GALANIN the Gi/o pathway. Injection of morphine into the DRN causes The neuropeptide galanin activates three types of galanin recep- an increase in 5-HT release detected in forebrain microdialysis tors (GalR1−3). GalR1 and GalR2 are highly expressed in the DRN (Tao and Auerbach, 1994). The increase in 5-HT release is most (Melander et al., 1988; Larm et al., 2003; Lu et al., 2005; Sharkey likely mediated via Gi/o protein-mediated inhibition of GABAer- et al., 2008). Moreover, GalR1 expression has also been described gic interneurons in the DRN, involving μ opioid receptors located in 5-HT neurons from rats but not in mice (Xu et al., 1998; Larm on GABAergic neurons (Jolas and Aghajanian, 1997). The mod- et al., 2003). GalR activation in the DRN causes a K+ conductance- ulation of 5-HT release in the DRN by κ and δ opioid receptors mediated hyperpolarization in rat brain slices (Xu et al., 1998), has also been described (Tao and Auerbach, 2002). Activation of most likely via GalR3-mediated Gi/o activation of GIRK channels δ receptors increased, while activation of κ receptors decreased 5- (Swanson et al., 2005). These effects are in agreement with in vivo HT release measured with in vivo microdialysis. The κ receptors microdialysis studies showing that injection of galanin into the effects do not involve the modulation of GABAergic or gluta- DRN reduces 5-HT release via GalR activation in the hippocampus matergic inputs in the DRN (Tao and Auerbach, 2002), suggesting (Kehr et al.,2002). In contrast to the inhibitory action of galanin on that κ receptors are expressed in 5-HT neurons. Likewise, opi- 5-HT neuronal activity, a reduction in inhibitory input onto 5-HT oid receptor-like1 (Oprl1) are most likely located and expressed neurons has also been described (Sharkey et al., 2008). Here, the in 5-HT neurons as follows. Oprl1 or nociceptin (NOP) recep- pan GalR1−3 agonist reduced GABA-mediated fast synaptic trans- tors belong to the family but are activated by mission accompanied by increase of PPF,suggesting that GalRs are NOP (orphanin FQ), a neuropeptide derived from prepronoci- expressed on GABAergic terminals and inhibit presynaptic Ca2+ ceptin protein. High levels of NOP receptor binding sites have channels via the Gi/o pathway. On the other hand, GalR2 agonist, been detected in the DRN (Florin et al., 2000) and Oprl1 recep- galanin (2–11) reduced IPSP amplitude but did not cause PPF,sug- tor expression could be detected in embryonic 5-HT neurons gesting a postsynaptic action (Branchek et al., 2000; Sharkey et al., (Wylie et al., 2010). NOP/orphanin FQ inhibits 5-HT release in 2008). Additionally, galanin (2–11) was demonstrated to increase the DRN via Oprl1 (Tao et al., 2007), suggesting a functional role 5-HT release in hippocampal tissue by immunofluorescence and of Oprl1 in 5-HT neurons early in development and in the adult high-performance liquid chromatography (HPLC) measurement brain.

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 7

“fnint-07-00040” — 2013/5/21 — 14:57 — page7—#7 Maejima et al. Modulation of 5-HT neurons

SUBSTANCE P addition, TRP channels have been described to be modulated by Substance P belongs to the tachykinin family and has a high D2-like receptors (Aman et al., 2007). According to the microar- affinity for the three different neurokinin receptors (NK1−3), ray expression profiling studies, various ion channel targets of in particular to NK1 (Hokfelt et al., 2001). These GPCRs cou- GPCRs are expressed in embryonic 5-HT neurons including ple mainly to the Gq/11 pathway (Stratowa et al., 1995), but TRP (Trpm4 and Trpm7), two-pore channels (TPCN1), cyclic Gs pathway activation has also been reported for NK1 in cell nucleotide gate channels (Hcn3), and KCNQ (Kcnq2; Wylie et al., culture systems (Martini et al., 2002). Various histological stud- 2010). Therefore, more detailed studies have to be performed to ies have revealed extensive expression of NK1 receptorsinthe determine the role of other ion channel targets and in particu- DRN (Maeno et al., 1993; Saffroy et al., 1994; Vigna et al., 1994; lar long-term effects of GPCR modulation for the serotonergic Charara and Parent, 1998; Sergeyev et al., 1999; Froger et al., system. 2001). Most studies suggest that NK1 receptors are not local- ized on 5-HT neurons (Froger et al., 2001; Santarelli et al., 2001), while others revealed NK1 receptor expression in a subpopu- INTEGRATION AND SIGNAL PROCESSING OF MODULATORY lation of 5-HT neurons (Santarelli et al., 2001; Lacoste et al., INFORMATION BY SEROTONERGIC NEURONS: WHY SO 2006, 2009). Interestingly, NK1 receptors are found in the cyto- MANY GPCRs? plasm of the 5-HT neurons and in dendritic membranes of The serotonergic transmitter system modulates many physiologi- GABAergic neurons. After administration of NK1 antagonist or cal functions such as mood, sexual behavior, feeding, sleep/wake deafferentation of substance P releasing projections, the density cycle, memory, cognition, blood pressure regulation, breath- of membrane bound NK1 receptors is increased in the soma- ing, and reproductive success (Mooney et al., 1998; Abrams et al., todendritic region of 5-HT neurons, suggesting that membrane 2004; Lechin et al., 2006; Lerch-Haner et al., 2008; McDevitt and trafficking of NK1 receptors may be regulated by Substance Neumaier, 2011). Because of the complexity and variety of the P input. This mechanism may contribute to the modulation different behaviors modulated by serotonin, it is expected that of 5-HT neuronal firing under certain physiological conditions modulatory signals from other brain areas including sensory (Lacoste et al., 2009). In addition, controversial results exist for information is integrated by GPCR signals in nuclei containing the effect of NK1 on 5-HT release and firing of 5-HT neurons. 5-HT neurons using a high diversity of GPCRs. While GABA Inhibition of NK1 in the DRN using antagonists or knock-out and glutamatergic input into the raphe nuclei will adjust 5- strategies leads to an increase in firing activity of 5-HT neu- HT neurons to the current inhibitory/excitatory state of the rons in vivo (Haddjeri and Blier, 2001; Santarelli et al., 2001). brain, other transmitter systems will inform 5-HT neurons more In contrast, activation of NK1 and NK3 increases spontaneous directly about the serotonin-associated behavior. For example, excitatory postsynaptic currents (EPSCs) in DRN 5-HT neu- dopamine is involved in reward-driven learning; ACh modu- rons resulting in an increased firing of the 5-HT neurons as lates arousal and reward; NA and CRF are involved in stress observed in brain slice recording (Liu et al., 2002a). These effects responses; histamine is involved in sleep regulation and sexual could be blocked by NK1 and NK3 antagonists (Liu et al., 2002a). function; bombesin and CCK regulate eating behavior; eCBs Also, activation of NK1 via intra-raphe injection of substance modulate memory, appetite, stress, social behavior, anxiety, and P in the DRN increases 5-HT release within the DRN, but sleep; galanin has been implicated in the regulation of sleep–wake decreases 5-HT release in frontal cortex as measured with in cycle, cognition, emotion, and blood pressure; hypocretin–orexin vivo microdialysis (Guiard et al., 2007). These effects and also modulate arousal, wakefulness, and appetite; and opioids and the described increase in 5-HT firing in NK1 knock-out mice substance P are involved in pain perception and mood (White involve changes in 5-HT1A autoreceptor levels, suggesting at least and Rumbold, 1988; Woodruff et al., 1996; Greenough et al., 1998; a functional coupling between NK1 and 5-HT1A receptors. Fur- Bear et al., 2001; Merali et al., 2006; Monti, 2010; Haj-Dahmane ther investigations to verify these interactions in 5-HT neurons are and Shen, 2011). Since all different behavioral responses can be required. integrated in nuclei containing 5-HT neurons, regulation of sero- In summary, the various heteroreceptors integrate incoming tonin release will affect similar behaviors as stated above. Thus information via two main pathways, i.e., Gi/o and Gq/11 lead- there is a tight interaction and signaling exchange between the ing to inhibition or activation of 5-HT neuronal firing and 5-HT different transmitter systems to precisely modulate behavioral out- release, respectively. Besides the “classical” Gi/o protein-mediated, put. Consequently, long-term changes in serotonin release can + membrane-delimited modulation of GIRK and presynaptic Ca2 involve changes in the auto-regulation involving 5-HT receptor channels, other ion channel targets have been identified in 5-HT or hetero-regulation involving the above mentioned GPCRs and + neurons. For example, two-pore-domain K channels [TWIK- can cause neuropsychiatric disorders, most notably depression, related acid-sensitive K-1 (TASK-1) and TASK-3] have been anxiety, schizophrenia, and dementia (Lucki, 1998; Davidson described in dorsal and caudal raphe 5-HT neurons (Washburn et al., 2000; Mann et al., 2001; Nelson and Chiavegatto, 2001). et al., 2002). TASK channels are inhibited by GPCRs coupling to The modulation of the different behaviors is even more com- the Gq/11 pathway most likely in a membrane-delimited man- plex since other GPCRs, such as orphan GPCRs, with so far ner involving the direct binding of Gαq subunits (Chen et al., unknown function are also expressed in 5-HT neurons. Therefore, 2006). The existence of voltage-sensitive but not ATP-dependent new strategies and techniques have to be applied and devel- + K channels in DRN neurons including 5-HT neurons have been oped to understand complex behaviors related to the serotonergic proposed based on drug application studies (Harsing, 2006). In system.

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 8

“fnint-07-00040” — 2013/5/21 — 14:57 — page8—#8 Maejima et al. Modulation of 5-HT neurons

NEW APPROACHES TO CONTROL AND UNDERSTAND most likely via activation of GIRK and inhibition of presynap- SEROTONERGIC G PROTEIN-COUPLED RECEPTOR tic Ca2+ channels (Li et al., 2005; Oh et al., 2010; Gutierrez et al., SIGNALING PATHWAYS 2011). Since vRh belongs to class A or like group of Recently, several new approaches to manipulate the activity of GPCRs, like serotonergic autoreceptors 5-HT1A/1B/1D, the idea 5-HT neurons in a cell type-specific manner have been devel- arose to generate light-activated chimeric receptors which couple oped. Various promoter/enhancer sequences have been isolated to intracellular signaling pathways in 5-HT1 receptor domains. and characterized, which allow for the expression of proteins The feasibility of receptor domain swapping has been demon- of choice within at least subsets of 5-HT neurons. These DNA strated by Dr. Khorana’s group (Kim et al., 2005). They replaced sequences include the promoter or enhancer sequences of Pet- the intracellular loops of vRh with that of the β2-adrenergic recep- 1/Fev transcription factor, the serotonin transporter (SLC6A4), tor and turned vRh into a light-activated Gs protein-coupled and the tryptophan hydroxylase 2 (TPH2; Scott et al., 2005). receptor, Opto-β2AR. The approach however was not suitable Using these different promoter/enhancer sequences, different for the exchange of vRh intracellular peptide loops by the 5- mouse lines and virus approaches have been developed to acti- HT1A intracellular receptor domains, since this chimeric receptor vate and/or silence/delete reporter such as green fluorescent revealed altered activation and deactivation kinetics in respect to protein (GFP) or tdTomato, Cre or Flip recombinases, tetracy- GIRK channel modulation (Oh et al., 2010). However, it could cline inducible systems, tetanus toxin light chain, and genes of be shown that the C-terminus (CT) of the 5-HT1A receptor was interest (Scott et al., 2005; Kim et al., 2009; Madisen et al., 2010, sufficient to target vRh into expression domains of 5-HT1A recep- 2012; Richardson-Jones et al., 2010; Zhao et al., 2011). Using the tor and functionally substitute for Gi/o pathway activation. The tetracycline inducible system, for example, the genetic ablation of CTs of GPCRs contain peptide signal domains for subcellular 5-HT1A from the majority of 5-HT neurons could be achieved targeting and G protein interaction. The CT of 5-HT1A had (Richardson-Jones et al., 2010, 2011). been shown to be necessary for trafficking of the receptor to For the investigation of the modulation and function of neu- dendritic domains via interaction with trafficking protein Yif1B ronal circuits in general and for the serotonergic system in (Carrel et al., 2008). Fusion of the 5-HT1A receptor CT onto particular, chemical and optogenetic techniques have been devel- vRh was therefore sufficient to target the chimeric construct Rh- oped in recent years (Herlitze and Landmesser, 2007; Masseck CT5-HT1A into somatodendritic regions of hippocampal neurons et al., 2010). For control of neuronal activity in various neuronal and 5-HT neurons in the DRN and exclude the expression of circuits including the 5-HT system, the light-gated non-selective the chimeric receptor from the axons. The Rh-CT5-HT1A receptors cation channel, ChR2 has been used and allows for the dissection were able to functionally substitute for intracellular 5-HT1A signals of 5-HT-mediated behavioral effects in different raphe nuclei (Li in DRN neurons of 5-HT1A knock-out mice, i.e., light illumi- + et al.,2005; Vargaet al.,2009; Zhao et al.,2011; Madisen et al.,2012; nation induced K conductance (most likely GIRK) reduced the see also Kim et al., 2009). For the investigation of GPCR signals, firing rate of spontaneously active 5-HT neurons. Thus, the adjust- various chemically and light-activated GPCRs have been devel- ment of for example light-activated or chemically activated GPCRs oped (Masseck et al., 2010). For example vertebrate rhodopsin and their coupling and anchoring to and in intracellular signaling (vRh) has been used to regulate Gi/o signaling pathways in neu- domains will allow for the dissection of multiple GPCR pathways rons by light (Li et al., 2005). Exogenously expressed vRh inhibits within the serotonergic system and their interaction in vitro and neuronal firing and neurotransmitter release in vitro and in vivo in vivo.

REFERENCES Aghajanian, G. K., and Vandermaelen, transmission and anxiety. J. Neurosci. human brainstem and spinal cord: an Abrams, J. K., Johnson, P. L., Hol- C. P. (1982). Intracellular record- 30, 2198–2210. immunohistochemical analysis. Neu- lis, J. H., and Lowry, C. A. ings from serotonergic dorsal raphe Andrade, R. (1998). Regulation of roscience 122, 773–784. (2004). Anatomic and functional neurons: pacemaker potentials and membrane excitability in the central Bagdy, E., Solyom, S., and Harsing, L. topography of the dorsal raphe the effect of LSD. Brain Res. 238, nervous system by serotonin receptor G. Jr. (1998). Feedback stimulation nucleus. Ann. N. Y. Acad. Sci. 1018, 463–469. subtypes. Ann. N. Y. Acad. Sci. 861, of somatodendritic serotonin release: 46–57. Aghajanian, G. K., and Wang, R. 190–203. a 5-HT3 receptor-mediated effect in Adell, A., and Artigas, F. (1999). Y. (1977). Habenular and other Arvidsson, U., Schalling, M., Cullheim, the raphe nuclei of the rat. Brain Res. Regulation of the release of 5- midbrain raphe afferents demon- S., Ulfhake, B., Terenius, L., Verhof- Bull. 45, 203–208. hydroxytryptamine in the median strated by a modified retrograde stad, A., et al. (1990). Evidence for Barbara, A., Aceves, J., and Arias- raphe nucleus of the rat by cate- tracing technique. Brain Res. 122, coexistence between calcitonin gene- Montano, J. A. (2002). Histamine cholaminergic afferents. Eur. J. Neu- 229–242. related peptide and serotonin in the H1 receptors in rat dorsal raphe rosci. 11, 2305–2311. Aman, T. K., Shen, R. Y., and bulbospinal pathway in the monkey. nucleus: pharmacological charac- Adell, A., Celada, P., Abellan, M. Haj-Dahmane, S. (2007). D2-like Brain Res. 532, 47–57. terisation and linking to increased T., and Artigas, F. (2002). Origin dopamine receptors depolarize dor- Aso, E., Renoir, T., Mengod, G., Ledent, neuronal activity. Brain Res. 954, and functional role of the extracel- sal raphe serotonin neurons through C., Hamon, M., Maldonado, R., 247–255. lular serotonin in the midbrain raphe the activation of nonselective cationic et al. (2009). Lack of CB1 receptor Barnes, N. M., and Sharp, T. (1999). A nuclei. Brain Res. Brain Res. Rev. 39, conductance. J. Pharmacol. Exp. Ther. activity impairs serotonergic nega- review of central 5-HT receptors and 154–180. 320, 376–385. tive feedback. J. Neurochem. 109, their function. Neuropharmacology Aghajanian, G. K. (1985). Modula- Amilhon, B., Lepicard, E., Renoir, T., 935–944. 38, 1083–1152. tion of a transient outward cur- Mongeau, R., Popa, D., Poirel, O., Baer, K., Waldvogel, H. J., During, M. Bayliss, D. A., Li, Y. W., and Talley, rent in serotonergic neurones by et al. (2010). VGLUT3 (vesicular glu- J., Snell, R. G., Faull, R. L., and E. M. (1997a). Effects of serotonin alpha 1-adrenoceptors. Nature 315, tamate transporter type 3) contribu- Rees, M. I. (2003). Association of on caudal raphe neurons: activation 501–503. tion to the regulation of serotonergic gephyrin and glycine receptors in the of an inwardly rectifying potassium

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 9

“fnint-07-00040” — 2013/5/21 — 14:57 — page9—#9 Maejima et al. Modulation of 5-HT neurons

conductance. J. Neurophysiol. 77, of a on 5- Chang, B., Daniele, C. A., Gallagher, K., (2004). Cell-type specific calcium sig- 1349–1361. hydroxytryptamine neurones in the Madonia, M., Mitchum, R. D., Bar- naling by corticotropin-releasing fac- Bayliss, D. A., Li, Y. W., and Talley, dorsal raphe of the rat brain. Br. J. rett, L., et al. (2011). Nicotinic excita- tor type 1 (CRF1) and 2a (CRF2(a)) E. M. (1997b). Effects of serotonin Pharmacol. 102, 635–638. tion of serotonergic projections from receptors: phospholipase C-mediated on caudal raphe neurons: inhibition Boeijinga, P. H., and Boddeke, H. dorsal raphe to the nucleus accum- responses in human embryonic kid- of N- and P/Q-type calcium chan- W. (1993). Serotonergic modula- bens. J. Neurophysiol. 106, 801–808. ney 293 but not SK-N-MC neuroblas- nels and the afterhyperpolarization. tion of neurotransmission in the rat Chang, C. P.,Pearse, R. V. II, O’Connell, toma cells. Biochem. Pharmacol. 68, J. Neurophysiol. 77, 1362–1374. subicular cortex in vitro: a role for 5- S., and Rosenfeld, M. G. (1993). Iden- 1833–1844. Bear, M. F., Connors, B. W., and Par- HT1B receptors. Naunyn Schmiede- tification of a seven transmembrane Davidson, R. J., Putnam, K. M., and Lar- adiso, M. A. (2001). Neuroscience: bergs Arch. Pharmacol. 348, 553–557. helix receptor for corticotropin- son, C. L. (2000). Dysfunction in the Exploring the Brain. Baltimore, MD: Borroto-Escuela, D. O., Narvaez, M., releasing factor and sauvagine in neural circuitry of emotion regula- Lippincott Williams & Wilkins. Marcellino, D., Parrado, C., Nar- mammalian brain. Neuron 11, 1187– tion–apossible prelude to violence. Beck, S. G., Pan, Y. Z., Akanwa, A. C., vaez, J. A., Tarakanov, A. O., et al. 1195. Science 289, 591–594. and Kirby, L. G. (2004). Median and (2010). -1 modu- Charara, A., and Parent, A. (1998). Dawson, T. M., Barone, P., Sidhu, A., dorsal raphe neurons are not elec- lates 5-hydroxtryptamine-1A signal- Chemoarchitecture of the primate Wamsley, J. K., and Chase, T. N. trophysiologically identical. J. Neuro- ing via heterodimerization. Biochem. dorsal raphe nucleus. J. Chem. Neu- (1988). The D1 dopamine recep- physiol. 91, 994–1005. Biophys. Res. Commun. 393, 767–772. roanat. 15, 111–127. tor in the rat brain: quantitative Becquet, D., Hery, M., Deprez, P., Bourson, A., Boess, F. G., Bos, M., Chen, X., Talley, E. M., Patel, N., Gomis, autoradiographic localization using Faudon, M., Fache, M. P., Giraud, P., and Sleight, A. J. (1998). Involvement A., McIntire, W. E., Dong, B., et al. an iodinated ligand. Neuroscience 26, et al. (1993a). N-methyl-D-aspartic of 5-HT6 receptors in nigro-striatal (2006). Inhibition of a background 83–100. acid/glycine interactions on the con- function in rodents. Br. J. Pharmacol. potassium channel by Gq protein Day, H. E., Campeau, S., Watson, S. trol of 5-hydroxytryptamine release 125, 1562–1566. alpha-subunits. Proc. Natl. Acad. Sci. J. Jr., and Akil, H. (1997). Distribu- in raphe primary cultures. J. Neu- Bouthenet, M. L., Martres, M. P., U.S.A. 103, 3422–3427. tion of alpha 1a-, alpha 1b- and alpha rochem. 61, 1692–1697. Sales, N., and Schwartz, J. C. (1987). Clemett, D. A., Punhani, T., Duxon, 1d-adrenergic receptor mRNA in the Becquet, D., Hery, M., Francois-Bellan, A detailed mapping of dopamine M. S., Blackburn, T. P., and Fone, rat brain and spinal cord. J. Chem. A. M., Giraud, P., Deprez, P., Faudon, D-2 receptors in rat central ner- K. C. (2000). Immunohistochemical Neuroanat. 13, 115–139. M., et al. (1993b). Glutamate, GABA, vous system by autoradiography with localisation of the 5-HT2C receptor Deneris, E. S., and Wyler, S. C. glycine and taurine modulate sero- [125I]iodosulpride. Neuroscience 20, protein in the rat CNS. Neurophar- (2012). Serotonergic transcriptional tonin synthesis and release in rostral 117–155. macology 39, 123–132. networks and potential importance and caudal rhombencephalic raphe Branchek, T. A., Smith, K. E., Gerald, C., Colino, A., and Halliwell, J. V. (1987). to mental health. Nat. Neurosci. 15, cells in primary cultures. Neurochem. and Walker, M. W. (2000). Galanin Differential modulation of three sep- 519–527. Int. 23, 269–283. receptor subtypes. Trends Pharmacol. arate K-conductances in hippocam- de Weerth, A., Pisegna, J. R., Huppi, Belin, M. F., Nanopoulos, D., Didier, M., Sci. 21, 109–117. pal CA1 neurons by serotonin. Nature K., and Wank, S. A. (1993). Molec- Aguera, M., Steinbusch, H., Verhofs- Brown, R. E., Sergeeva, O. A., Eriks- 328, 73–77. ular cloning, functional expression tad, A., et al. (1983). Immunohisto- son, K. S., and Haas, H. L. (2002). Cornelisse, L. N., Van der Harst, J. E., and chromosomal localization of the chemical evidence for the presence of Convergent excitation of dorsal Lodder, J. C., Baarendse, P. J., Tim- human cholecystokinin type A recep- gamma-aminobutyric acid and sero- raphe serotonin neurons by multiple merman, A. J., Mansvelder, H. D., tor. Biochem. Biophys. Res. Commun. tonin in one nerve cell. A study on the arousal systems (orexin/hypocretin, et al. (2007). Reduced 5-HT1A- and 194, 811–818. raphe nuclei of the rat using antibod- histamine and noradrenaline). J. GABAB receptor function in dorsal Drutel, G., Peitsaro, N., Karlstedt, K., ies to glutamate decarboxylase and Neurosci. 22, 8850–8859. raphe neurons upon chronic fluox- Wieland, K., Smit, M. J., Timmer- serotonin. Brain Res. 275, 329–339. Bruinvels, A. T., Landwehrmeyer, B., etine treatment of socially stressed man, H., et al. (2001). Identification Benya, R. V., Wada, E., Battey, J. F., Gustafson, E. L., Durkin, M. M., rats. J. Neurophysiol. 98, 196–204. of rat H3 receptor isoforms with dif- Fathi, Z., Wang, L. H., Mantey, Mengod, G., Branchek, T. A., et al. Cortes, R., Gueye, B., Pazos, A., ferent brain expression and signaling S. A., et al. (1992). Neuromedin B (1994). Localization of 5-HT1B, 5- Probst, A., and Palacios, J. M. (1989). properties. Mol. Pharmacol. 59, 1–8. receptors retain functional expres- HT1D alpha, 5-HT1E and 5-HT1F Dopamine receptors in human brain: Egashira, N., Mishima, K., Kat- sion when transfected into BALB receptor messenger RNA in rodent autoradiographic distribution of D1 surabayashi, S., Yoshitake, T., 3T3 fibroblasts: analysis of binding, and primate brain. Neuropharmacol- sites. Neuroscience 28, 263–273. Matsumoto,Y., Ishida, J., et al. (2002). kinetics, stoichiometry, modulation ogy 33, 367–386. Crawford, L. K., Craige, C. P., and Involvement of 5-hydroxytryptamine by guanine nucleotide-binding pro- Calizo, L. H., Akanwa, A., Ma, X., Pan, Beck, S. G. (2010). Increased intrin- neuronal system in delta(9)- teins, and and Y. Z., Lemos, J. C., Craige, C., et al. sic excitability of lateral wing sero- tetrahydrocannabinol-induced comparison with natively expressed (2011). Raphe serotonin neurons are tonin neurons of the dorsal raphe: impairment of spatial memory. receptors. Mol. Pharmacol. 42, 1058– not homogenous: electrophysiologi- a mechanism for selective activation Eur. J. Pharmacol. 445, 221–229. 1068. cal, morphological and neurochemi- in stress circuits. J. Neurophysiol. 103, Ferre, S., and Artigas, F. (1993). Bernard, R., andVeh, R. W.(2012). Indi- cal evidence. Neuropharmacology 61, 2652–2663. Dopamine D2 receptor-mediated vidual neurons in the rat lateral habe- 524–543. Dahlstrom, A., and Fuxe, K. (1964). regulation of serotonin extracellu- nular complex project mostly to the Carrel, D., Masson, J., Al Awabdh, S., Localization of monoamines in the lar concentration in the dorsal raphe dopaminergic ventral tegmental area Capra, C. B., Lenkei, Z., Hamon, lower brain stem. Experientia 20, nucleus of freely moving rats. J. Neu- or to the serotonergic raphe nuclei. J. M., et al. (2008). Targeting of the 398–399. rochem. 61, 772–775. Comp. Neurol. 520, 2545–2558. 5-HT1A serotonin receptor to neu- Darmani, N. A., Janoyan, J. J., Kumar, N. Ferre, S., Cortes, R., and Artigas, F. Blier, P., Steinberg, S., Chaput, Y., ronal dendrites is mediated by Yif1B. N., and J. L Crim, N. (2003). Behav- (1994). Dopaminergic regulation of and de Montigny, C. (1989). Electro- J. Neurosci. 28, 8063–8073. iorally active doses of the CB1 recep- the serotonergic raphe-striatal path- physiological assessment of putative Celada, P., Puig, M. V., Casanovas, J. M., tor antagonist SR 141716A increase way: microdialysis studies in freely antagonists of 5-hydroxytryptamine Guillazo, G., and Artigas, F. (2001). brain serotonin and dopamine levels moving rats. J. Neurosci. 14, 4839– receptors: a single-cell study in the rat Control of dorsal raphe serotoner- and turnover. Pharmacol. Biochem. 4846. dorsal raphe nucleus. Can. J. Physiol. gic neurons by the medial prefrontal Behav. 75, 777–787. Ferreira, S. G., Teixeira, F. M., Gar- Pharmacol. 67, 98–105. cortex: involvement of serotonin-1A, Dautzenberg, F. M., Gutknecht, E., Van cao, P., Agostinho, P., Ledent, C., Boden, P. R., Woodruff, G. N., and Pin- GABA(A),and glutamate receptors. J. der Linden, I., Olivares-Reyes, J. A., Cortes, L., et al. (2012). Presynaptic nock, R. D. (1991). Pharmacology Neurosci. 21, 9917–9929. Durrenberger, F., and Hauger, R. L. CB(1) cannabinoid receptors control

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 10

“fnint-07-00040” — 2013/5/21 — 14:57 — page 10 — #10 Maejima et al. Modulation of 5-HT neurons

frontocortical serotonin and gluta- N., et al. (2000). Role and origin of activity in synaptosomes. 66, Hashemi, P., Dankoski, E. C., Wood, mate release – species differences. the GABAergic innervation of dor- 1024–1034. K. M., Ambrose, R. E., and Wight- Neurochem. Int. 61, 219–226. sal raphe serotonergic neurons. J. Haj-Dahmane, S. (2001). D2-like man, R. M. (2011). In vivo elec- Florin, S., Meunier, J., and Costentin, Neurosci. 20, 4217–4225. activation excites trochemical evidence for simultane- J. (2000). Autoradiographic localiza- Glass, M., and Felder, C. C. (1997). Con- rat dorsal raphe 5-HT neurons ous 5-HT and histamine release in tion of [3H]nociceptin binding sites current stimulation of cannabinoid in vitro. Eur. J. Neurosci. 14, the rat substantia nigra pars reticu- in the rat brain. Brain Res. 880, 11–16. CB1 and dopamine D2 receptors aug- 125–134. lata following medial forebrain bun- Frankhuijzen, A. L., Wardeh, G., ments cAMP accumulation in striatal Haj-Dahmane, S., and Shen, R. Y. dle stimulation. J. Neurochem. 118, Hogenboom, F., and Mulder, A. H. neurons: evidence for a Gs linkage (2005). The wake-promoting pep- 749–759. (1988). Alpha 2-adrenoceptor medi- to the CB1 receptor. J. Neurosci. 17, tide orexin-B inhibits glutamatergic Hay, D. L., Christopoulos, G., ated inhibition of the release of radi- 5327–5333. transmission to dorsal raphe nucleus Christopoulos, A., Poyner, D. R., olabelled 5-hydroxytryptamine and Gras, C., Herzog, E., Bellenchi, G. serotonin neurons through retro- and Sexton, P. M. (2005). Pharma- noradrenaline from slices of the dor- C., Bernard, V., Ravassard, P., Pohl, grade endocannabinoid signaling. J. cological discrimination of calcitonin sal region of the rat brain. Naunyn M., et al. (2002). A third vesicular Neurosci. 25, 896–905. receptor: receptor activity-modifying Schmiedebergs Arch. Pharmacol. 337, glutamate transporter expressed by Haj-Dahmane, S., and Shen, R. Y. protein complexes. Mol. Pharmacol. 255–260. cholinergic and serotoninergic neu- (2009). Endocannabinoids suppress 67, 1655–1665. Froger, N., Gardier, A. M., Moratalla, rons. J. Neurosci. 22, 5442–5451. excitatory synaptic transmission to Heinisch, S., and Kirby, L. G. (2009). R., Alberti, I., Lena, I., Boni, C., Greenough, A., Cole, G., Lewis, J., dorsal raphe serotonin neurons Fractalkine/CX3CL1 enhances et al. (2001). 5-hydroxytryptamine Lockton, A., and Blundell, J. (1998). through the activation of presynap- GABA synaptic activity at sero- (5-HT)1A autoreceptor adaptive Untangling the effects of hunger, anx- tic CB1 receptors. J. Pharmacol. Exp. tonin neurons in the rat dorsal changes in substance P (neu- iety, and nausea on energy intake Ther. 331, 186–196. raphe nucleus. Neuroscience 164, rokinin 1) receptor knock-out during intravenous cholecystokinin Haj-Dahmane, S., and Shen, R. Y. 1210–1223. mice mimic antidepressant-induced octapeptide (CCK-8) infusion. Phys- (2011). Modulation of the sero- Heinisch, S., and Kirby, L. G. desensitization. J. Neurosci. 21, iol. Behav. 65, 303–310. tonin system by endocannabinoid (2010). SDF-1alpha/CXCL12 8188–8197. Guiard, B. P., Guilloux, J. P., Reperant, signaling. Neuropharmacology 61, enhances GABA and glutamate Fu, W., Le Maitre, E., Fabre, V., Bernard, C., Hunt, S. P., Toth, M., and Gardier, 414–420. synaptic activity at serotonin J. F., David Xu, Z. Q., and Hokfelt, A. M. (2007). Substance P neurokinin Hale, M. W., and Lowry, C. A. (2010). neurons in the rat dorsal raphe T. (2010). Chemical neuroanatomy 1 receptor activation within the dor- Functional topography of midbrain nucleus. Neuropharmacology 58, of the dorsal raphe nucleus and adja- sal raphe nucleus controls serotonin and pontine serotonergic systems: 501–514. cent structures of the mouse brain. J. release in the mouse frontal cortex. implications for synaptic regulation Herlitze, S., and Landmesser, L. T. Comp. Neurol. 518, 3464–3494. Mol. Pharmacol. 72, 1411–1418. of serotonergic circuits. Psychophar- (2007). New optical tools for con- Galindo-Charles, L., Hernandez-Lopez, Gustafson, E. L., Durkin, M. M., Bard, J. macology (Berl.) 213, 243–264. trolling neuronal activity. Curr. Opin. S., Galarraga, E., Tapia, D., Bar- A., Zgombick, J., and Branchek, T. A. Hamon, M., Doucet, E., Lefevre, K., Neurobiol. 17, 87–94. gas, J., Garduno, J., et al. (2008). (1996). A receptor autoradiographic Miquel, M. C., Lanfumey, L., Insausti, Hioki, H., Nakamura, H., Ma, Y. F., Serotoninergic dorsal raphe neurons and in situ hybridization analysis of R., et al. (1999). Antibodies and anti- Konno, M., Hayakawa, T., Naka- possess functional postsynaptic nico- the distribution of the 5-ht7 recep- sense oligonucleotide for probing the mura, K. C., et al. (2010). Vesicular tinic acetylcholine receptors. Synapse tor in rat brain. Br. J. Pharmacol. 117, distribution and putative functions of glutamate transporter 3-expressing 62, 601–615. 657–666. central 5-HT6 receptors. Neuropsy- nonserotonergic projection neurons Gallager, D. W., and Aghajanian, G. K. Gutierrez, D. V., Mark, M. D., chopharmacology 21, 68S–76S. constitute a subregion in the rat mid- (1976). Effect of antipsychotic drugs Masseck, O., Maejima, T., Kuckels- Haring, M., Marsicano, G., Lutz, B., brain raphe nuclei. J. Comp. Neurol. on the firing of dorsal raphe cells. berg, D., Hyde, R. A., et al. (2011). and Monory, K. (2007). Identifi- 518, 668–686. II. Reversal by picrotoxin. Eur. J. Optogenetic control of motor coor- cation of the cannabinoid receptor Hjorth, S., and Sharp, T. (1991). Effect Pharmacol. 39, 357–364. dination by Gi/o protein-coupled type 1 in serotonergic cells of raphe of the 5-HT1A receptor agonist 8- Garduno, J., Galindo-Charles, L., vertebrate rhodopsin in cerebellar nuclei in mice. Neuroscience 146, OH-DPAT on the release of 5-HT in Jimenez-Rodriguez, J., Galarraga, E., Purkinje cells. J. Biol. Chem. 286, 1212–1219. dorsal and median raphe-innervated Tapia, D., Mihailescu, S., et al. (2012). 25848–25858. Harsing, L. G. Jr. (2006). The pharma- rat brain regions as measured by Presynaptic alpha4beta2 nicotinic Haddjeri, N., and Blier, P. (2001). cology of the neurochemical trans- in vivo microdialysis. Life Sci. 48, acetylcholine receptors increase glu- Sustained blockade of neurokinin-1 mission in the midbrain raphe nuclei 1779–1786. tamate release and serotonin neu- receptors enhances serotonin neu- of the rat. Curr. Neuropharmacol. 4, Hjorth, S., Suchowski, C. S., and Gal- ron excitability in the dorsal raphe rotransmission. Biol. Psychiatry 50, 313–339. loway, M. P. (1995). Evidence for nucleus. J. Neurosci. 32, 15148– 191–199. Harvey, J., Wedley, S., Findlay, J. 5-HT autoreceptor-mediated, nerve 15157. Haddjeri, N., Faure, C., Lucas, G., D., Sidell, M. R., and Pullar, I. impulse-independent, control of 5- Gartside, S. E., Cole, A. J., Williams, Mnie-Filali, O., Chouvet, G., Astier, A. (1996). omega-Agatoxin IVA HT synthesis in the rat brain. Synapse A. P., McQuade, R., and Judge, S. J. B., et al. (2004). In-vivo modulation identifies a single calcium chan- 19, 170–176. (2007). AMPA and NMDA receptor of central 5-hydroxytryptamine nel subtype which contributes to Hokfelt, T., Pernow, B., and Wahren, regulation of firing activity in 5-HT (5-HT1A) receptor-mediated the potassium-induced release of J. (2001). Substance P: a pioneer neurons of the dorsal and median responses by the cholinergic sys- acetylcholine, 5-hydroxytryptamine, amongst neuropeptides. J. Intern. raphe nuclei. Eur. J. Neurosci. 25, tem. Int. J. Neuropsychopharmacol. 7, dopamine, gamma-aminobutyric Med. 249, 27–40. 3001–3008. 391–399. acid and glutamate from rat brain Hopwood, S. E., and Stamford, J. A. Gerard, C., Martres, M. P., Lefevre, Haddjeri, N., Lucas, G., and Blier, slices. Neuropharmacology 35, (2001). Multiple 5-HT(1) autorecep- K., Miquel, M. C., Verge, D., Lan- P. (2000). Role of cholinergic and 385–392. tor subtypes govern serotonin release fumey, L., et al. (1997). Immuno- GABAergic systems in the feedback Hasbi, A., O’Dowd, B. F., and in dorsal and median raphe nuclei. localization of serotonin 5-HT6 inhibition of dorsal raphe 5-HT neu- George, S. R. (2010). Heteromeriza- Neuropharmacology 40, 508–519. receptor-like material in the rat cen- rons. Neuroreport 11, 3397–3401. tion of dopamine D2 receptors with Howlett, A. C., Qualy, J. M., and tral nervous system. Brain Res. 746, Hagan, C. E., McDevitt, R. A., Liu, dopamine D1 or D5 receptors gen- Khachatrian, L. L. (1986). Involve- 207–219. Y., Furay, A. R., and Neumaier, J. erates intracellular calcium signaling ment of Gi in the inhibition of adeny- Gervasoni, D., Peyron, C., Rampon, C., F. (2012). 5-HT(1B) autoreceptor by different mechanisms. Curr. Opin. late cyclase by cannabimimetic drugs. Barbagli, B., Chouvet, G., Urbain, regulation of serotonin transporter Pharmacol. 10, 93–99. Mol. Pharmacol. 29, 307–313.

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 11

“fnint-07-00040” — 2013/5/21 — 14:57 — page 11 — #11 Maejima et al. Modulation of 5-HT neurons

Hoyer, D., Hannon, J. P., and Martin, calcium channels in high potassium- by 5-HT neurons in dorsal raphe Liu, R., Jolas, T., and Aghajanian, G. G. R. (2002). Molecular, pharma- elicited release of neurotransmitters nucleus of rat and mouse: evidence (2000). Serotonin 5-HT(2) receptors cological and functional diversity of from rat brain slices. Neuroscience 66, for species-dependent modulation of activate local GABA inhibitory inputs 5-HT receptors. Pharmacol. Biochem. 609–615. serotonin transmission. Eur. J. Neu- to serotonergic neurons of the dor- Behav. 71, 533–554. Kirby, L. G., Freeman-Daniels, E., rosci. 17, 481–493. sal raphe nucleus. Brain Res. 873, Innis, R. B., and Aghajanian, G. Lemos, J. C., Nunan, J. D., Lechin, F., van der Dijs, B., and 34–45. K. (1987). Pertussis toxin blocks Lamy, C., Akanwa, A., et al. Hernandez-Adrian, G. (2006). Dor- Lovenberg, T. W., Liaw, C. W., Grigori- 5-HT1A and GABAB receptor- (2008). Corticotropin-releasing fac- sal raphe vs. median raphe serotoner- adis, D. E., Clevenger, W., Chalmers, mediated inhibition of serotonergic tor increases GABA synaptic activ- gic antagonism. Anatomical, physio- D. T., De Souza, E. B., et al. (1995). neurons. Eur. J. Pharmacol. 143, ity and induces inward current in logical, behavioral, neuroendocrino- Cloning and characterization of a 195–204. 5-hydroxytryptamine dorsal raphe logical, neuropharmacological and functionally distinct corticotropin- Jacobs, B. L., and Azmitia, E. C. (1992). neurons. J. Neurosci. 28, 12927– clinical evidences: relevance for neu- releasing factor receptor subtype Structure and function of the brain 12937. ropharmacological therapy. Prog. from rat brain. Proc. Natl. Acad. Sci. serotonin system. Physiol. Rev. 72, Kobayashi, K., Ikeda, Y., Haneda, E., and Neuropsychopharmacol. Biol. Psychi- U.S.A. 92, 836–840. 165–229. Suzuki, H. (2008). Chronic fluoxe- atry 30, 565–585. Lovenberg, T. W., Roland, B. L., Wilson, Jensen, R. T., Battey, J. F., Spindel, tine bidirectionally modulates poten- Lee, Y. M., Beinborn, M., McBride, S. J., Jiang, X., Pyati, J., Huvar, A., E. R., and Benya, R. V. (2008). tiating effects of serotonin on the E. W., Lu, M., Kolakowski, L. et al. (1999). Cloning and functional International Union of Pharmacol- hippocampal mossy fiber synaptic F. Jr., and Kopin, A. S. (1993). expression of the human histamine ogy. LXVIII. Mammalian bombesin transmission. J. Neurosci. 28, 6272– The human brain cholecystokinin- H3 receptor. Mol. Pharmacol. 55, receptors: nomenclature, distribu- 6280. B/gastrin receptor. Cloning and char- 1101–1107. tion, pharmacology, signaling, and Kohlmeier, K. A., Watanabe, S., Tyler, acterization. J. Biol. Chem. 268, Lu, X., Barr, A. M., Kinney, J. W., functions in normal and disease C. J., Burlet, S., and Leonard, C. S. 8164–8169. Sanna, P., Conti, B., Behrens, M. states. Pharmacol. Rev. 60, 1–42. (2008). Dual orexin actions on dorsal Lerch-Haner, J. K., Frierson, D., Craw- M., et al. (2005). A role for galanin Jolas, T., and Aghajanian, G. K. (1997). raphe and laterodorsal tegmentum ford, L. K., Beck, S. G., and in antidepressant actions with a Opioids suppress spontaneous and neurons: noisy cation current acti- Deneris, E. S. (2008). Serotonergic focus on the dorsal raphe nucleus. NMDA-induced inhibitory postsy- vation and selective enhancement of transcriptional programming deter- Proc. Natl. Acad. Sci. U.S.A. 102, naptic currents in the dorsal raphe Ca2+ transients mediated by L-type mines maternal behavior and off- 874–879. nucleusoftheratinvitro.Brain Res. calcium channels. J. Neurophysiol. spring survival. Nat. Neurosci. 11, Lucki, I. (1998). The spectrum of behav- 755, 229–245. 100, 2265–2281. 1001–1003. iors influenced by serotonin. Biol. Kawashima, N., Karasawa, J., Shimazaki, Kroeze, W. K., Kristiansen, K., and Levine, E. S., and Jacobs, B. L. Psychiatry 44, 151–162. T., Chaki, S., Okuyama, S., Yasuhara, Roth, B. L. (2002). Molecular biology (1992). Neurochemical afferents con- Madisen, L., Mao, T., Koch, H., A., et al. (2005). Neuropharmacolog- of serotonin receptors structure and trolling the activity of serotoner- Zhuo, J. M., Berenyi, A., Fuji- ical profiles of antagonists of group function at the molecular level. Curr. gic neurons in the dorsal raphe sawa, S., et al. (2012). A toolbox II metabotropic glutamate receptors. Top. Med. Chem. 2, 507–528. nucleus: microiontophoretic studies of Cre-dependent optogenetic trans- Neurosci. Lett. 378, 131–134. Kuteeva, E., Hokfelt, T., Wardi, T., and in the awake cat. J. Neurosci. 12, genic mice for light-induced activa- Kehr, J., Yoshitake, T., Wang, F. H., Ogren, S. O. (2008). Galanin, galanin 4037–4044. tion and silencing. Nat. Neurosci. 15, Razani, H., Gimenez-Llort, L., Jans- receptor subtypes and depression- Leysen, J. E. (2004). 5-HT2 recep- 793–802. son, A., et al. (2002). Galanin is a like behaviour. Cell.Mol.LifeSci.65, tors. Curr. Drug Targets CNS Neurol. Madisen, L., Zwingman, T. A., Sunkin, potent in vivo modulator of mesen- 1854–1863. Disord. 3, 11–26. S. M., Oh, S. W., Zariwala, H. cephalic serotonergic neurotransmis- Lacoste, B., Riad, M., and Descarries, Li, X., Gutierrez, D. V., Hanson, M. G., A., Gu, H., et al. (2010). A robust sion. Neuropsychopharmacology 27, L. (2006). Immunocytochemical evi- Han, J., Mark, M. D., Chiel, H., et al. and high-throughput Cre reporting 341–356. dence for the existence of substance (2005). Fast noninvasive activation and characterization system for the Kidd, E. J., Garratt, J. C., and Mars- P receptor (NK1) in serotonin neu- and inhibition of neural and net- whole mouse brain. Nat. Neurosci. 13, den, C. A. (1991). Effects of repeated rons of rat and mouse dorsal raphe work activity by vertebrate rhodopsin 133–140. treatment with 1-(2,5-dimethoxy-4- nucleus. Eur. J. Neurosci. 23, 2947– and green algae channelrhodopsin. Maejima, T., Hashimoto, K., Yoshida, iodophenyl)-2-aminopropane (DOI) 2958. Proc. Natl. Acad. Sci. U.S.A. 102, T., Aiba, A., and Kano, M. (2001). on the autoregulatory control of dor- Lacoste, B., Riad, M., Ratte, M. O., Boye, 17816–17821. Presynaptic inhibition caused by ret- sal raphe 5-HT neuronal firing and S. M., Levesque, D., and Descarries, L. Liaw, C. W., Lovenberg, T. W., rograde signal from metabotropic cortical 5-HT release. Eur. J. Pharma- (2009). Trafficking of neurokinin-1 Barry, G., Oltersdorf, T., Grigori- glutamate to cannabinoid receptors. col. 200, 131–139. receptors in serotonin neurons is con- adis, D. E., and de Souza, E. B. Neuron 31, 463–475. Kim, J. C., Cook, M. N., Carey, trolled by substance P within the rat (1996). Cloning and characterization Maeno, H., Kiyama, H., and Tohyama, M. R., Shen, C., Regehr, W. G., dorsal raphe nucleus. Eur. J. Neurosci. of the human corticotropin-releasing M. (1993). Distribution of the sub- and Dymecki, S. M. (2009). Link- 29, 2303–2314. factor-2 receptor complementary stance P receptor (NK-1 receptor) in ing genetically defined neurons to Lakoski, J. M., and Aghajanian, G. K. deoxyribonucleic acid. Endocrinology the central nervous system. Brain Res. behavior through a broadly appli- (1983). Effects of histamine, H1- and 137, 72–77. Mol. Brain Res. 18, 43–58. cable silencing allele. Neuron 63, H2-receptor antagonists on the activ- Liu, R., Ding, Y., and Aghajanian, G. K. Mann, J. J., Brent, D. A., and Arango, 305–315. ity of serotonergic neurons in the (2002a). Neurokinins activate local V. (2001). The neurobiology and Kim, J. M., Hwa, J., Garriga, P.,Reeves, P. dorsal raphe nucleus. J. Pharmacol. glutamatergic inputs to serotonergic genetics of suicide and attempted sui- J., RajBhandary, U. L., and Khorana, Exp. Ther. 227, 517–523. neurons of the dorsal raphe nucleus. cide: a focus on the serotonergic sys- H. G. (2005). Light-driven activation Lakoski, J. M., Gallager, D. W., and Neuropsychopharmacology 27, tem. Neuropsychopharmacology 24, of beta 2-adrenergic receptor signal- Aghajanian, G. K. (1984). Histamine- 329–340. 467–477. ing by a chimeric rhodopsin con- induced depression of serotoninergic Liu, R. J., van den Pol, A. N., and Martini, L., Hastrup, H., Holst, B., taining the beta 2-adrenergic receptor dorsal raphe neurons: antagonism by Aghajanian, G. K. (2002b). Hypocre- Fraile-Ramos, A., Marsh, M., and cytoplasmic loops. Biochemistry 44, cimetidine, a reevaluation. Eur. J. tins (orexins) regulate serotonin neu- Schwartz, T. W. (2002). NK1 recep- 2284–2292. Pharmacol. 103, 153–156. rons in the dorsal raphe nucleus by tor fused to beta-arrestin displays Kimura, M., Yamanishi, Y., Hanada, T., Larm, J. A., Shen, P. J., and Gund- excitatory direct and inhibitory indi- a single-component, high-affinity Kagaya, T.,Kuwada, M.,Watanabe,T., lach, A. L. (2003). Differential galanin rect actions. J. Neurosci. 22, 9453– molecular phenotype. Mol. Pharma- et al. (1995). Involvement of P-type receptor-1 and galanin expression 9464. col. 62, 30–37.

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 12

“fnint-07-00040” — 2013/5/21 — 14:57 — page 12 — #12 Maejima et al. Modulation of 5-HT neurons

Martin-Ruiz, R., Ugedo, L., Honrubia, Mihailescu, S., Guzman-Marin, R., Oh, E., Maejima, T., Liu, C., Deneris, and the rostral hypothalamus in rat M. A., Mengod, G., and Artigas, Dominguez Mdel, C., and Drucker- E., and Herlitze, S. (2010). Substitu- brain slices in vitro. Brain Res. 653, F. (2001). Control of serotonergic Colin, R. (2002). Mechanisms of tion of 5-HT1A receptor signaling by 119–124. neurons in rat brain by dopamin- nicotine actions on dorsal raphe sero- a light-activated G protein-coupled Pudovkina, O. L., Cremers, T. I., and ergic receptors outside the dorsal toninergic neurons. Eur. J. Pharma- receptor. J. Biol. Chem. 285, 30825– Westerink, B. H. (2003). Regulation raphe nucleus. J. Neurochem. 77, col. 452, 77–82. 30836. of the release of serotonin in the 762–775. Monti, J. M. (2010). The role of dorsal Oh, S. B., Endoh, T., Simen, A. A., dorsal raphe nucleus by alpha1 and Masseck, O. A., Rubelowski, J. M., raphe nucleus serotonergic and non- Ren, D., and Miller, R. J. (2002). alpha2 adrenoceptors. Synapse 50, Spoida, K., and Herlitze, S. (2010). serotonergic neurons, and of their Regulation of calcium currents by 77–82. Light- and drug-activated G-protein- receptors, in regulating waking and chemokines and their receptors. J. Rampon, C., Luppi, P. H., Fort, P., Pey- coupled receptors to control intra- rapid eye movement (REM) sleep. Neuroimmunol. 123, 66–75. ron, C., and Jouvet, M. (1996). Dis- cellular signalling. Exp. Physiol. 96, SleepMed.Rev.14, 319–327. Pan, Z. Z., Colmers, W. F., and Williams, tribution of glycine-immunoreactive 51–56. Mooney, R. D., Crnko-Hoppenjans, T. J. T. (1989). 5-HT-mediated synaptic cell bodies and fibers in the rat brain. Matsumoto, M., Yoshioka, M., Togashi, A., Ke, M., Bennett-Clarke, C. A., potentials in the dorsal raphe nucleus: Neuroscience 75, 737–755. H., Ikeda, T., and Saito, H. Lane, R. D., Chiaia, N. L., et al. interactions with excitatory amino Rampon, C., Peyron, C., Gervasoni, D., (1996). Functional regulation by (1998). Augmentation of serotonin acid and GABA neurotransmission. J. Pow, D. V., Luppi, P. H., and Fort, dopamine receptors of serotonin in the developing superior colliculus Neurophysiol. 62, 481–486. P. (1999). Origins of the glycinergic release from the rat hippocampus: alters the normal development of the Pan, Z. Z., and Williams, J. T. inputs to the rat locus coeruleus and in vivo microdialysis study. Naunyn uncrossed retinotectal projection. J. (1989). GABA- and glutamate- dorsal raphe nuclei: a study combin- Schmiedebergs Arch. Pharmacol. 353, Comp. Neurol. 393, 84–92. mediated synaptic potentials in rat ing retrograde tracing with glycine 621–629. Moranta, D., Esteban, S., and Garcia- dorsal raphe neurons in vitro. J. immunohistochemistry. Eur. J. Neu- Mazarati, A. M., Baldwin, R. A., Shin- Sevilla, J. A. (2009). Chronic Neurophysiol. 61, 719–726. rosci. 11, 1058–1066. mei, S., and Sankar, R. (2005). In treatment and withdrawal of the Penington, N. J., and Kelly, J. S. (1990). Rashid, A. J., So, C. H., Kong, M. vivo interaction between serotonin cannabinoid agonist WIN 55,212-2 Serotonin receptor activation reduces M., Furtak, T., El-Ghundi, M., and galanin receptors types 1 and 2 modulate the sensitivity of presynap- calcium current in an acutely dissoci- Cheng, R., et al. (2007). D1-D2 in the dorsal raphe: implication for tic receptors involved in the regula- ated adult central neuron. Neuron 4, dopamine receptor heterooligomers limbic seizures. J. Neurochem. 95, tion of monoamine syntheses in rat 751–758. with unique pharmacology are cou- 1495–503. brain. Naunyn Schmiedebergs Arch. Penington, N. J., Kelly, J. S., and Fox, pled to rapid activation of Gq/11 in McDevitt, R. A., and Neumaier, J. Pharmacol. 379, 61–72. A. P. (1992). Action potential wave- the striatum. Proc. Natl. Acad. Sci. F. (2011). Regulation of dorsal Morikawa, H., Manzoni, O. J., Crabbe, J. forms reveal simultaneous changes in U.S.A. 104, 654–659. raphe nucleus function by sero- C., and Williams, J. T. (2000). Regula- ICa and IK produced by 5-HT in rat Richardson-Jones, J. W., Craige, C. P., tonin autoreceptors: a behavioral per- tion of central synaptic transmission dorsal raphe neurons. Proc. Biol. Sci. Guiard, B. P., Stephen, A., Metzger, spective. J. Chem. Neuroanat. 41, by 5-HT(1B) auto- and heterorecep- 248, 171–179. K. L., Kung, H. F., et al. (2010). 5- 234–246. tors. Mol. Pharmacol. 58, 1271– Penington, N. J., Kelly, J. S., and Fox, HT1A autoreceptor levels determine Melander, T., Kohler, C., Nilsson, S., 1278. A. P.(1993). Whole-cell recordings of vulnerability to stress and response to Hokfelt, T., Brodin, E., Theodorsson, Nakamoto, H., Soeda, Y., Takami, S., inwardly rectifying K+ currents acti- antidepressants. Neuron 65, 40–52. E., et al. (1988). Autoradiographic Minami, M., and Satoh, M. (2000). vated by 5-HT1A receptors on dorsal Richardson-Jones, J. W., Craige, C. quantitation and anatomical map- Localization of calcitonin receptor raphe neurones of the adult rat. J. P., Nguyen, T. H., Kung, H. F., ping of 125I-galanin binding sites mRNA in the mouse brain: coex- Physiol. 469, 387–405. Gardier, A. M., Dranovsky, A., et al. in the rat central nervous system. J. istence with serotonin transporter Perrin, M. H., Donaldson, C. J., (2011). Serotonin-1A autoreceptors Chem. Neuroanat. 1, 213–233. mRNA. Brain Res. Mol. Brain Res. 76, Chen, R., Lewis, K. A., and Vale, are necessary and sufficient for the Mendiguren, A., and Pineda, J. (2009). 93–102. W. W. (1993). Cloning and func- normal formation of circuits under- Effect of the CB(1) receptor antago- Nakazi, M., Bauer, U., Nickel, T., Kath- tional expression of a rat brain lying innate anxiety. J. Neurosci. 31, nists rimonabant and AM251 on the mann, M., and Schlicker, E. (2000). corticotropin releasing factor (CRF) 6008–6018. firing rate of dorsal raphe nucleus Inhibition of serotonin release in receptor. Endocrinology 133, 3058– Richerson, G. B. (2004). Serotonergic neurons in rat brain slices. Br. J. the mouse brain via presynaptic 3061. neurons as carbon dioxide sensors Pharmacol. 158, 1579–1587. cannabinoid CB1 receptors. Naunyn Peyron, C., Tighe, D. K., van den Pol, that maintain pH homeostasis. Nat. Mendlin, A., Martin, F. J., and Schmiedebergs Arch. Pharmacol. 361, A. N., de Lecea, L., Heller, H. C., Rev. Neurosci. 5, 449–461. Jacobs, B. L. (1998). Involve- 19–24. Sutcliffe, J. G., et al. (1998). Neu- Roberts, C., Allen, L., Langmead, C. ment of dopamine D2 receptors in Nanopoulos, D., Belin, M. F., Maitre, rons containing hypocretin (orexin) J., Hagan, J. J., Middlemiss, D. N., apomorphine-induced facilitation of M., Vincendon, G., and Pujol, J. project to multiple neuronal systems. and Price, G. W. (2001). The effect forebrain serotonin output. Eur. J. F. (1982). Immunocytochemical evi- J. Neurosci. 18, 9996–10015. of SB-269970, a 5-HT(7) receptor Pharmacol. 351, 291–298. dence for the existence of GABAergic Pillot, C., Heron, A., Cochois, V., antagonist, on 5-HT release from Mengod, G., Cortes, R., Vilaro, M. T., neurons in the nucleus raphe dorsalis. Tardivel-Lacombe, J., Ligneau, X., serotonergic terminals and cell bod- and Hoyer, D. (2010). “Distribution Possible existence of neurons con- Schwartz, J. C., et al. (2002). A ies. Br. J. Pharmacol. 132, 1574– of 5-HT receptors in the central ner- taining serotonin and GABA. Brain detailed mapping of the histamine 1580. vous system,” in Handbook of Behav- Res. 232, 375–389. H(3) receptor and its gene tran- Rosin, D. L., Zeng, D., Stornetta, R. L., ioral Neurobiology of Serotonin,eds Nelson, R. J., and Chiavegatto, S. (2001). scripts in rat brain. Neuroscience 114, Norton, F. R., Riley, T., Okusa, M. D., C. P. Muller and B. Jacobs (London: Molecular basis of aggression. Trends 173–193. et al. (1993). Immunohistochemical Elsevier B.V.), 123–138. Neurosci. 24, 713–719. Pineyro, G., and Blier, P. (1999). localization of alpha 2A-adrenergic Merali, Z., Bedard, T., Andrews, N., Offermanns, S., Iida-Klein, A., Segre, Autoregulation of serotonin neu- receptors in catecholaminergic and Davis, B., McKnight, A. T., Gonza- G. V., and Simon, M. I. (1996). rons: role in antidepressant other brainstem neurons in the rat. lez, M. I., et al. (2006). Bombesin G alpha q family members couple drug action. Pharmacol. Rev. 51, Neuroscience 56, 139–155. receptors as a novel anti-anxiety ther- parathyroid hormone (PTH)/PTH- 533–591. Rouse, S. T., and Levey, A. I. (1996). apeutic target: BB1 receptor actions related peptide and calcitonin recep- Pinnock, R. D., Reynolds, T., and Expression of m1-m4 muscarinic on anxiety through alterations of tors to phospholipase C in COS- Woodruff, G. N. (1994). Different immunoreac- serotonin activity. J. Neurosci. 26, 7 cells. Mol. Endocrinol. 10, types of bombesin receptors on neu- tivity in septohippocampal neurons 10387–10396. 566–574. rons in the dorsal raphe nucleus and other identified hippocampal

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 13

“fnint-07-00040” — 2013/5/21 — 14:57 — page 13 — #13 Maejima et al. Modulation of 5-HT neurons

afferents. J. Comp. Neurol. 375, Shikanai, H., Yoshida, T., Konno, K., Swanson, C. J., Blackburn, T. P., Zhang, To, Z. P., Bonhaus, D. W., Eglen, R. M., 406–416. Yamasaki, M., Izumi, T., Ohmura, Y., X., Zheng, K., Xu, Z. Q., Hok- and Jakeman, L. B. (1995). Character- Ruat, M., Traiffort, E., Leurs, R., et al. (2012). Distinct neurochemical felt, T., et al. (2005). Anxiolytic- ization and distribution of putative Tardivel-Lacombe, J., Diaz, J., Arrang, and functional properties of GAD67- and antidepressant-like profiles of the 5-ht7 receptors in guinea-pig brain. J. M., et al. (1993). Molecular cloning, containing 5-HT neurons in the rat galanin-3 receptor (Gal3) antagonists Br. J. Pharmacol. 115, 107–116. characterization, and localization of dorsal raphe nucleus. J. Neurosci. 32, SNAP 37889 and SNAP 398299. Proc. Tzavara, E. T., Davis, R. J., Perry, a high-affinity serotonin receptor (5- 14415–14426. Natl. Acad. Sci. U.S.A. 102, 17489– K. W., Li, X., Salhoff, C., Bymas- HT7) activating cAMP formation. Simon, H. H., Scholz, C., and O’Leary, 17494. ter, F. P., et al. (2003). The Proc. Natl. Acad. Sci. U.S.A. 90, D. D. (2005). Engrailed genes con- Talley, E. M., Rosin, D. L., Lee, A., CB1 receptor antagonist SR141716A 8547–8551. trol developmental fate of seroton- Guyenet, P. G., and Lynch, K. R. selectively increases monoaminergic Saffroy, M., Beaujouan, J. C., Petitet, ergic and noradrenergic neurons in (1996). Distribution of alpha 2A- neurotransmission in the medial pre- F., Torrens, Y., and Glowinski, J. mid- and hindbrain in a gene dose- adrenergic receptor-like immunore- frontal cortex: implications for thera- (1994). Differential localization of dependent manner. Mol. Cell. Neu- activity in the rat central nervous peutic actions. Br. J. Pharmacol. 138, 3H-[Pro9]SP binding sites in the rosci. 28, 96–105. system. J. Comp. Neurol. 372, 544–553. guinea pig and rat brain. Brain Res. Siniscalchi, A., Rodi, D., Cavallini, S., 111–134. Ulrich, C. D., Ferber, I., Holicky, E., 633, 317–325. Marino, S., Beani, L., and Bianchi, Tao, R., and Auerbach, S. B. (1994). Hadac, E., Buell, G., and Miller, Santarelli, L., Gobbi, G., Debs, P. C. (2001). Effects of cholecystokinin Increased extracellular serotonin in L. J. (1993). Molecular cloning and C., Sibille, E. T., Blier, P., Hen, tetrapeptide (CCK(4)) and anxiolytic rat brain after systemic or intrara- functional expression of the human R., and Heath, M. J. (2001). drugs on the electrically evoked phe administration of morphine. J. gallbladder cholecystokinin A recep- Genetic and pharmacological dis- [(3)H]5-hydroxytryptamine outflow Neurochem. 63, 517–524. tor. Biochem. Biophys. Res. Commun. ruption of neurokinin 1 receptor from rat cortical slices. Brain Res. 922, Tao, R., and Auerbach, S. B. 193, 204–211. function decreases anxiety-related 104–111. (2000). Regulation of serotonin Unnerstall, J. R., Fernandez, I., behaviors and increases serotonergic Sink, K. S., Walker, D. L., Yang, Y., and release by GABA and excitatory and Orensanz, L. M. (1985). The function. Proc. Natl. Acad. Sci. U.S.A. Davis, M. (2011). Calcitonin gene- amino acids. J. Psychopharmacol. 14, alpha-adrenergic receptor: radiohis- 98, 1912–1917. related peptide in the bed nucleus 100–113. tochemical analysis of functional Scammell, T. E., and Winrow, C. J. of the stria terminalis produces an Tao, R., and Auerbach, S. B. (2002). Opi- characteristics and biochemical dif- (2010). Orexin receptors: pharmacol- anxiety-like pattern of behavior and oid receptor subtypes differentially ferences. Pharmacol. Biochem. Behav. ogy and therapeutic opportunities. increases neural activation in anxiety- modulate serotonin efflux in the rat 22, 859–874. Annu. Rev. Pharmacol. Toxicol. 51, related structures. J. Neurosci. 31, central nervous system. J. Pharmacol. Valentino, R. J., Lucki, I., and Van 243–266. 1802–1810. Exp. Ther. 303, 549–556. Bockstaele, E. (2010). Corticotropin- Scheinin, M., Lomasney, J. W., Hayden- Song, L., Liu,Y.,Yu,Y., Duan, X., and Qi, Tao, R., and Ma, Z. (2012). Neu- releasing factor in the dorsal raphe Hixson, D. M., Schambra, U. B., S. (2012). Shh signaling guides spa- ral circuit in the dorsal raphe nucleus: linking stress coping and Caron, M. G., Lefkowitz, R. J., tial pathfinding of raphespinal tract nucleus responsible for cannabinoid- addiction. Brain Res. 1314, 29–37. et al. (1994). Distribution of alpha axons by multidirectional repulsion. mediated increases in 5-HT efflux Vandermaelen, C. P.,and Aghajanian, G. 2-adrenergic receptor subtype gene Cell Res. 22, 697–716. in the nucleus accumbens of the K. (1983). Electrophysiological and expression in rat brain. Brain Res. Sprouse, J. S., and Aghajanian, rat brain. ISRN Pharmacol. 2012, pharmacological characterization of Mol. Brain Res. 21, 133–149. G. K. (1987). Electrophysiological 276902. serotonergic dorsal raphe neurons Scott, M. M., Wylie, C. J., Lerch, J. K., responses of serotoninergic dorsal Tao, R., Ma, Z., Thakkar, M. M., McCar- recorded extracellularly and intracel- Murphy, R., Lobur, K., Herlitze, S., raphe neurons to 5-HT1A and 5- ley, R. W., and Auerbach, S. B. (2007). lularly in rat brain slices. Brain Res. et al. (2005). A genetic approach to HT1B agonists. Synapse 1, 3–9. Nociceptin/orphanin FQ decreases 289, 109–119. access serotonin neurons for in vivo Stamford, J. A., Davidson, C., McLaugh- serotonin efflux in the rat brain but van Hooft, J. A., and Vijverberg, H. P. and in vitro studies. Proc. Natl. Acad. lin, D. P., and Hopwood, S. E. in contrast to a kappa-opioid has (2000). 5-HT(3) receptors and neu- Sci. U.S.A. 102, 16472–16477. (2000). Control of dorsal raphe 5-HT no antagonistic effect on mu-opioid- rotransmitter release in the CNS: a Sergeyev, V., Hokfelt, T., and Hurd, function by multiple 5-HT(1) autore- induced increases in serotonin efflux. nerve ending story? Trends Neurosci. Y. (1999). Serotonin and substance ceptors: parallel purposes or point- Neuroscience 147, 106–116. 23, 605–610. P co-exist in dorsal raphe neurons less plurality? Trends Neurosci. 23, Thomas, D. R., Soffin, E. M., Roberts, Varga,V., Losonczy, A., Zemelman, B.V., of the human brain. Neuroreport 10, 459–465. C., Kew, J. N., de la Flor, R. Borhegyi, Z., Nyiri, G., Domonkos, 3967–3970. Stratowa, C., Machat, H., Burger, M., Dawson, L. A., et al. (2006). A., et al. (2009). Fast synaptic subcor- Serrats, J., Raurich, A., Vilaro, M. T., E., Himmler, A., Schafer, R., Spe- SB-699551-A (3-cyclopentyl-N- tical control of hippocampal circuits. Mengod, G., and Cortes, R. (2004). vak, W., et al. (1995). Functional [2-(dimethylamino)ethyl]-N-[(4- Science 326, 449–453. 5-ht5B receptor mRNA in the raphe characterization of the human neu- {[(2-phenylethyl)amino]methyl}-4- Varga, V., Sik, A., Freund, T. F., and nuclei: coexpression with serotonin rokinin receptors NK1, NK2, and biphenylyl)methyl]propanamide Kocsis, B. (2002). GABA(B) recep- transporter. Synapse 51, 102–111. NK3 based on a cellular assay system. dihydrochloride), a novel 5- tors in the median raphe nucleus: Sexton, P. M., Paxinos, G., Kenney, M. J. Recept. Signal Transduct. Res. 15, ht5A receptor-selective antagonist, distribution and role in the seroton- A., Wookey, P. J., and Beaumont, 617–630. enhances 5-HT neuronal function: ergic control of hippocampal activity. K. (1994). In vitro autoradiographic Strazielle, C., Lalonde, R., Hebert, C., Evidence for an autoreceptor role Neuroscience 109, 119–132. localization of amylin binding sites in and Reader, T. A. (1999). Regional for the 5-ht5A receptor in guinea Vigna, S. R., Bowden, J. J., McDonald, rat brain. Neuroscience 62, 553–567. brain distribution of noradrenaline pig brain. Neuropharmacology 51, D. M., Fisher, J., Okamoto, A., McVey, Sharkey, L. M., Madamba, S. G., Siggins, uptake sites, and of alpha1-alpha2- 566–577. D. C., et al. (1994). Characterization G. R., and Bartfai, T. (2008). Galanin and beta-adrenergic receptors in Tilakaratne, N., Christopoulos, G., of antibodies to the rat substance P alters GABAergic neurotransmission PCD mutant mice: a quantitative Zumpe, E. T., Foord, S. M., and Sex- (NK-1) receptor and to a chimeric in the dorsal raphe nucleus. Neu- autoradiographic study. Neuroscience ton, P. M. (2000). Amylin receptor substance P receptor expressed in rochem. Res. 33, 285–291. 94, 287–304. phenotypes derived from human cal- mammalian cells. J. Neurosci. 14, Sharp, T., Boothman, L., Raley, J., and Suzuki, M., Hurd, Y. L., Sokoloff, citonin receptor/RAMP coexpression 834–845. Queree, P. (2007). Important mes- P., Schwartz, J. C., and Sedvall, exhibit pharmacological differences Vita, N., Laurent, P., Lefort, S., sages in the ‘post’: recent discoveries G. (1998). D3 dopamine receptor dependent on receptor isoform and Chalon, P., Lelias, J. M., Kaghad, in 5-HT neurone feedback control. mRNA is widely expressed in the host cell environment. J. Pharmacol. M., et al. (1993). Primary structure Trends Pharmacol. Sci. 28, 629–636. human brain. Brain Res. 779, 58–74. Exp. Ther. 294, 61–72. and functional expression of mouse

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 14

“fnint-07-00040” — 2013/5/21 — 14:57 — page 14 — #14 Maejima et al. Modulation of 5-HT neurons

pituitary and human brain corti- Psychopharmacology (Berl.) 95, monoamine autoreceptors in human for optogenetic dissection of neural cotrophin releasing factor receptors. 1–14. embryonic kidney 293 cells and brain circuitry function. Nat. Methods 8, FEBS Lett. 335, 1–5. Williams, J. T., Colmers, W. F., and Pan, synaptosomes. J. Pharmacol. Exp. 745–752. Wamsley, J. K., Gehlert, D. R., Filloux, Z. Z. (1988). Voltage- and ligand- Ther. 325, 629–640. F. M., and Dawson, T. M. (1989). activated inwardly rectifying currents Xu, Z. Q., Zhang, X., Pieribone, V. A., Comparison of the distribution of in dorsal raphe neurons in vitro. J. Grillner, S., and Hokfelt, T. (1998). Conflict of Interest Statement: The D-1 and D-2 dopamine receptors in Neurosci. 8, 3499–3506. Galanin-5-hydroxytryptamine authors declare that the research was the rat brain. J. Chem. Neuroanat. 2, Woodruff, G. N., Hall, M. D., interactions: electrophysiological, conducted in the absence of any com- 119–137. Reynolds, T., and Pinnock, R. D. immunohistochemical and in situ mercial or financial relationships that Wang, Q. P., Guan, J. L., and (1996). Bombesin receptors in the hybridization studies on rat dorsal could be construed as a potential con- Shioda, S. (2000). Synaptic contacts brain. Ann. N. Y. Acad. Sci. 780, raphe neurons with a note on galanin flict of interest. between serotonergic and cholin- 223–243. R1 and R2 receptors. Neuroscience ergic neurons in the rat dor- Woolley, M. L., Marsden, C. A., and 87, 79–94. Received: 11 February 2013; accepted: sal raphe nucleus and laterodorsal Fone, K. C. (2004). 5-ht6 recep- Yokoyama, C., Okamura, H., Nakajima, 03 May 2013; published online: 23 May tegmental nucleus. Neuroscience 97, tors. Curr. Drug Targets CNS Neurol. T., Taguchi, J., and Ibata, Y. (1994). 2013. 553–563. Disord. 3, 59–79. Autoradiographic distribution of Citation: Maejima T, Masseck OA, Mark Wang, R. Y., and Aghajanian, G. K. Wright, D. E., Seroogy, K. B., Lundgren, [3H]YM-09151-2, a high-affinity MD and Herlitze S (2013) Modula- (1977). Physiological evidence for K. H., Davis, B. M., and Jennes, L. and selective antagonist ligand for tion of firing and synaptic transmission habenula as major link between fore- (1995). Comparative localization of the dopamine D2 receptor group, of serotonergic neurons by intrinsic G brain and midbrain raphe. Science serotonin1A, 1C, and 2 receptor sub- in the rat brain and spinal cord. J. protein-coupled receptors and ion chan- 197, 89–91. type mRNAs in rat brain. J. Comp. Comp. Neurol. 344, 121–136. nels. Front. Integr. Neurosci. 7:40. doi: Washburn, C. P., Sirois, J. E., Tal- Neurol. 351, 357–373. Zavitsanou, K., Wang, H., Dal- 10.3389/fnint.2013.00040 ley, E. M., Guyenet, P. G., and Wylie, C. J., Hendricks, T. J., Zhang, ton, V. S., and Nguyen, V. Copyright © 2013 Maejima, Masseck, Bayliss, D. A. (2002). Serotoner- B., Wang, L., Lu, P., Leahy, P., (2010). Cannabinoid administration Mark and Herlitze. This is an open- gic raphe neurons express TASK et al. (2010). Distinct transcriptomes increases 5HT1A receptor binding access article distributed under the terms channel transcripts and a TASK- define rostral and caudal serotonin and mRNA expression in the hip- of the Creative Commons Attribution like pH- and halothane-sensitive neurons. J. Neurosci. 30, 670–684. pocampus of adult but not adolescent License, which permits use, distribution K+ conductance. J. Neurosci. 22, Xie, Z., Westmoreland, S. V., and rats. Neuroscience 169, 315–324. and reproduction in other forums, pro- 1256–1265. Miller, G. M. (2008). Modula- Zhao, S., Ting, J. T., Atallah, H. vided the original authors and source White, J. M., and Rumbold, G. R. tion of monoamine transporters E., Qiu, L., Tan, J., Gloss, are credited and subject to any copy- (1988). Behavioural effects of his- by common biogenic amines via B., et al. (2011). Cell type-specific right notices concerning any third-party tamine and its antagonists: a review. trace amine-associated receptor 1 and channelrhodopsin-2 transgenic mice graphics etc.

Frontiers in Integrative Neuroscience www.frontiersin.org May 2013 | Volume 7 | Article 40 | 15

“fnint-07-00040” — 2013/5/21 — 14:57 — page 15 — #15