Abstract
Decades of research confirm that noradrenergic locus coeruleus (LC) neurons are essential for arousal, attention, motivation, and stress responses. While most studies on LC transmission focused unsurprisingly on norepinephrine (NE), adrenergic signaling cannot account for all the consequences of LC activation. Galanin coexists with NE in the vast majority of LC neurons, yet the precise function of this neuropeptide has proved to be surprisingly elusive given our solid understanding of the LC system. To elucidate the contribution of galanin to LC physiology, here we briefly summarize the nature of stimuli that drive LC activity from a neuroanatomical perspective. We go on to describe the LC pathways in which galanin most likely exerts its effects on behavior, with a focus on addiction, depression, epilepsy, stress, and Alzheimer’s disease. We propose a model in which LC-derived galanin has two distinct functions: as a neuromodulator, primarily acting via the galanin 1 receptor (GAL1), and as a trophic factor, primarily acting via galanin receptor 2 (GAL2). Finally, we discuss how the recent advances in neuropeptide detection, optogenetics and chemical genetics, and galanin receptor pharmacology can be harnessed to identify the roles of LC-derived galanin definitively.
Keywords: locus coeruleus, galanin, norepinephrine, Alzheimer’s disease, depression, addiction, epilepsy, stress
Introduction
The brainstem locus coeruleus (LC) is the major noradrenergic nucleus in the brain and is the sole source of norepinephrine (NE) in many parts of the limbic system and forebrain, such as the hippocampus and frontal cortex. The LC is implicated in many aspects of physiology and behavior, including attention, arousal, motivation, and stress. Although NE is considered the “primary” neurotransmitter employed by the LC, these neurons also synthesize and release a host of neuropeptides, including galanin, neuropeptide Y, cocaine- and amphetamine-related transcript, and brain-derived neurotrophic factor. In particular, galanin is expressed in most LC neurons in rodents and humans. The purpose of this review is to catalog the putative functions and consequences of LC-derived galanin, integrate them into a comprehensive model, and provide a roadmap for the use of new technologies to define how galanin release from LC neurons impacts brain function and disease.
Overview of LC anatomy
Electrophysiological evidence consistently shows that LC neurons respond to sensory stimulation of all modalities with a high degree of fidelity to the intensity and temporal pattern of the stimulation. The response of LC neurons to sensory stimuli appears to be more sensitive than the brainstem cholinergic, serotonergic, or dopaminergic systems (Koyama et al., 1994; Strecker and Jacobs, 1985), suggesting the LC plays a primary role amongst these systems in directing attentional resources to a dynamic environment. The responsiveness of LC neuronal discharge to novel and conditioned stimuli that represent salient features of the immediate environment is well established in a variety species under awake, freely behaving conditions (Aston-Jones et al., 1991; Bouret and Sara, 2005; Rasmussen et al., 1986; Sara and Bouret, 2012). An early study of freely behaving cats is particularly informative and demonstrates the wide range and nature of stimuli influencing LC discharge (Rasmussen et al., 1986). These experiments revealed that the treadmill exercise was a potent driver of LC firing, more so than other stimuli, including the presence of rats or other cats. This entrainment to physical activity presumably reflects the high degree of innervation carrying somatosensory, mechanoreceptive, vestibular, and visceral information to the LC, in addition to other sensory inputs.
LC afferents
The LC is remarkably sensitive to a range of general and special sensory input from somatic and visceral sources; however, early tract-tracing studies of LC afferents revealed surprisingly few structures projecting to the LC and identified the nucleus paragigantocellularis (PGi) and nucleus prepositus hypoglossi (NPH) as the major afferent sources (Aston-Jones et al., 1986). Subsequent work expanded the range of systems innervating the LC, although these connections are comparatively minor.
In addition to direct input from the dorsal horn and spinal trigeminal system (Craig, 1992), the nucleus PGi, a structure in the rostral ventrolateral medulla, can account for much of the somatosensory and mechanoreceptor input from the skeletomotor system (Samuels and Szabadi, 2008; Sara and Bouret, 2012). The PGi also carries auditory input via the inferior colliculus (Van Bockstaele et al., 1993). These pathways excite LC neurons primarily via glutamatergic transmission (Aston-Jones et al., 1991; Ennis et al., 1992; Park et al., 2005), though distinct populations of inhibitory, GABAergic neurons arising from the PGi innervate the LC, as well (Sirieix et al., 2012). Sympathetic afferent input to the LC also arises from the PGi, which itself functions as a sympathoregulatory center. Parasympathetic regulation of the LC occurs via projections from the nucleus tractus solitarius (NTS) (Groves et al., 2005; Van Bockstaele et al., 2001). Though known primarily as a brainstem micturition center, Barrington’s nucleus projects to both the LC and a range of sympathetic and parasympathetic targets (Guo et al., 2013; Valentino et al., 1995) and may thus coordinate autonomic input to the LC. GABAergic projections from the NPH constitute a major pathway to the LC that regulates the inhibition of REM sleep induction (Kaur et al., 2001; Sirieix et al., 2012). The NPH also integrates vestibular (Seo et al., 2004) and visual information (Korp et al., 1989) and thus may coordinate kinetic, positional, and visual input to the LC.
A wide range of neural systems involved in motivational, emotional, and cognitive functions impact the LC. Hypothalamic innervation of the LC is substantial and includes projections from the ventrolateral preoptic area (Lee et al., 2005a), tuberomammillary nucleus, and lateral hypothalamus (Espana et al., 2005; Lee et al., 2005b). All of these pathways are implicated in the control of wakefulness and regulation of sleep architecture, but the orexin-containing neurons arising from the lateral hypothalamus are particularly relevant to arousal (Espana et al., 2005). Projections from the preoptic area to the LC may also coordinate noradrenergic activity with thermoregulatory, osmoregulatory, and reproductive functions (Steininger et al., 2001). Extensive innervation of the LC arising from the paraventricular nucleus represents a secondary, indirect output for sympathetic activation originating from the hypothalamus (Luppi et al., 1995; Samuels and Szabadi, 2008).
In addition to the direct sensory and autonomic inputs arising through the circuits outlined above, the LC receives indirect sensory input via excitatory pathways originating in the central nucleus of the amygdala, which contain corticotropin-releasing factor (Charney et al., 1998; Van Bockstaele et al., 1998). Glutamatergic projections arising from the prefrontal cortex (PFC) also innervate the LC (Jodo et al., 1998). These systems thus provide an important path to convey information about stimuli with emotional salience.
Considering the anatomical relationship of the brainstem networks of biogenic amines that regulate arousal, motivation, and stress, the LC system is especially well situated to command these functions. The LC circuitry thus includes extensive, reciprocal connections to cholinergic, dopaminergic, and serotonergic nuclei. The cholinergic lateral dorsal tegmental nucleus (LDT) and pedunculopontine tegmental nucleus (PPT), which are the major sources of the ascending cortical and thalamic projections of the reticular activating system, also project to and activate the LC via nicotinic receptors (Bitner and Nikkel, 2002; Ganesh et al., 2008; Le Novere et al., 1996; Samuels and Szabadi, 2008). Dopaminergic input to the LC arising from the ventral tegmental area (VTA) regulates the activity of LC neurons in a complex fashion, with both excitatory (Deutch et al., 1986; Ornstein et al., 1987) and inhibitory influences reported (Guiard et al., 2008a). By contrast, serotonergic projections from the dorsal raphe to the LC are predominantly inhibitory and mediated via 5-HT1A receptors (Charlety et al., 1993; Kim et al., 2004), though more recent evidence suggests this influence may involve polysynaptic rather than direct mechanisms (Pudovkina et al., 2002).
LC efferents: focus on prefrontal cortex, hippocampus, and ventral tegmental area
The widespread and highly collateralized nature of LC efferent projections to the forebrain has been characterized thoroughly in the literature (Sara and Bouret, 2012; Szabadi, 2013). To appreciate the scope of the LC efferent network, it helps to recognize that nearly all the connections described above are reciprocal. Also noteworthy is the relatively high degree to which the LC innervates limbic structures. For the purposes of the present review, the following section will focus on the PFC, hippocampus, and mesolimbic dopamine system because of their prominent roles in stress, addiction, epilepsy, and cognition. As detailed below, regions of LC from which cortical and hippocampal projections arise also tend to express the highest levels of galanin.
The preponderance of anatomical evidence indicates that the LC projects to all of the forebrain isocortex and allocortex, and it accounts entirely for the noradrenergic innervation of these targets (Loughlin et al., 1986; Samuels and Szabadi, 2008). Electrophysiological experiments reveal that the innervation of the PFC is significantly more robust than in other isocortical areas (Chandler et al., 2013; Chandler et al., 2014). The LC projection to the hippocampal formation is similarly robust (Melander et al., 1986b). Noradrenergic transmission in the hippocampus regulates LTD and LTP and induces trophic factor expression in several hippocampal subfields, predominantly via mechanisms involving β-adrenergic receptors (Hansen and Manahan-Vaughan, 2015; Tully and Bolshakov, 2010; Yanpallewar et al., 2010). Of the cortical structures to which the LC projects, the olfactory bulb receives by far the most extensive innervation (Shipley et al., 1985), and the major noradrenergic influence on physiology and plasticity in this allocortical structure is well established (Eckmeier and Shea, 2014; Linster et al., 2011). LC pathways thus consistently modulate synaptic plasticity across a range of isocortical and allocortical structures, thereby revealing a prominent role for LC forebrain systems in trophic functions.
LC input to the VTA regulates the firing rate of dopaminergic neurons through α-adrenergic receptors, with both excitatory (Goertz et al., 2015; Grenhoff and Svensson, 1993; Mitrano et al., 2012; Velasquez-Martinez et al., 2012) and inhibitory actions of noradrenergic signaling in the VTA reported (Guiard et al., 2008a; Guiard et al., 2008b; Jimenez-Rivera et al., 2012; Paladini and Williams, 2004; Velasquez-Martinez et al., 2015; White and Wang, 1984). The nature of this regulation is therefore complex and depends on the relative contribution of α1, α2, and somatodendritic D-2 autoreceptor activation (Guiard et al., 2008a; Weinshenker and Schroeder, 2007).
Galanin coexistence in LC neurons and galanin receptors in prefrontal cortex, hippocampus, and ventral tegmental area
Double-labeling immunocytochemical studies reveal that galanin coexists with tyrosine hydroxylase in approximately 80% of LC neurons in rats, with a distribution concentrated in more dorsal regions (Holets et al., 1988; Melander et al., 1986a). This anatomical distribution is relevant to the function of galanin because neurons of the dorsal LC predominantly innervate cortex, hippocampus, and hypothalamus (Loughlin et al., 1986; Sara and Bouret, 2012). Galanin expression is also high in the LC of mice (Perez et al., 2001) and humans (Le Maitre et al., 2013).
The LC provides the sole source of galanin-immunoreactive fibers in the cortex and hippocampus, and most of these fibers express dopamine β-hydroxylase (Hokfelt et al., 1998; Xu et al., 1998a). Galaninergic transmission in the cortex and hippocampus may be mediated by either the GAL1 or GAL2 receptor subtype. GAL1 signaling is predominantly inhibitory, with Gi/o activation leading to inhibition of andenylate cyclase and the opening of G protein-regulated K+ channels, whereas GAL2 is much more complex and signals through Gi/o and Gq/11-type G proteins (Lang et al., 2015), the latter pathway being essential in mediating the trophic actions of galanin (see below). The GAL2 subtype predominates in isocortex, whereas both GAL1 and GAL2 are distributed throughout the hippocampal formation (O’Donnell et al., 1999; Yoshitake et al., 2014). The LC itself expresses high levels of GAL1 and moderate levels of GAL2 (O’Donnell et al., 1999; Yoshitake et al., 2014).
The GAL3 subtype is expressed predominantly in the hypothalamus and brainstem reticular formation in rats (Mennicken et al., 2002). Though the human LC contains high levels of GAL3 (Le Maitre et al., 2013), GAL3 expression in the rodent LC is very low relative to the abundant expression observed in the hypothalamus (Mennicken et al., 2002). Cellular signaling associated with GAL3 is relatively less understood owing to the lack of stable expression of this receptor subtype in commonly used cell lines (Robinson et al., 2013). However, available evidence suggests that signaling through GAL3 is inhibitory and involves interactions with G protein-regulated K+ channels and inhibition of adenylate cyclase, similar to that of GAL1 (Lang et al., 2015).
The well-established projection from the LC to VTA and the high degree of coexistence of galanin and NE suggest that LC-derived galanin regulates mesocorticolimbic dopamine function (see below). Though galaninergic fibers projecting to the VTA have yet to be definitively characterized, both the VTA and substantia nigra express GAL2 receptors (O’Donnell et al., 1999), and evidence discussed below reveals a role for galanin signaling in the regulation of mesolimbic and mesostriatal dopamine function.
In summary, the anatomy of the LC and the distribution of galanin within LC pathways that mediate stress responses and appetitive motivation support the prediction that galanin function would be tightly linked to drug-seeking and stress-related behaviors. Evidence reviewed below implicates galanin in both phenomena, though many such actions are complex and unanticipated. Conditions that alter galanin gene expression in the LC provide critical insight into the function and nature of LC-derived galanin. Surprisingly, galanin expression in the LC is not consistently altered by stress. Unexpectedly, the literature points more to galanin adaptations to chronic conditions that may alter LC physiology over extended periods, rather than responsiveness to acute stressors per se (Holmes et al., 1995a; Holmes et al., 1995b; Kuteeva et al., 2008b; Sweerts et al., 1999). Our laboratories have shown that various modes of chronic exercise elevate LC galanin expression in a “dose-dependent” manner (Murray et al., 2010). Such regulation is consistent with the role the LC plays in integrating the range of sensory and visceral activity associated with dynamic interactions with changing environmental stimuli.
Neuromodulatory effects of galanin
The most well-characterized cellular effect of LC-derived galanin is its ability to inhibit LC activity (Xu et al., 2005). Slice electrophysiology experiments indicate that galanin and GAL1-preferring agonists potently hyperpolarize LC neurons, suppress their spontaneous firing rate, and enhance α2-adrenergic receptor-mediated negative feedback via activation of GAL1 and an increase in K+ conductance, even when synaptic input is blocked (Ma et al., 2001; Pieribone et al., 1995; Seutin et al., 1989). Immunohistochemical evidence suggests that endogenous galanin is likely released from LC dendrites and soma and acts on GAL1 receptors on LC neurons in an autocrine manner (Pieribone et al., 1995; Vila-Porcile et al., 2009). GAL2 and GAL3 receptors are also expressed in LC neurons and may contribute, although GAL2 agonists suppress LC activity only at very high concentrations (Ma et al., 2001). Furthermore, unlike GAL1, which responds to changes in LC activity and intracellular signaling, the levels of GAL2 and GAL3 receptors appear to be static, further implicating GAL1 as the most important neuromodulatory galanin receptor in LC neurons (Hawes et al., 2005). Analysis of galanin-induced suppression of LC activity in GAL1, GAL2, and GAL3 knockout mice would yield more definitive data. It is possible that galanin also acts as a negative feedback signal at the level of noradrenergic nerve terminals, but this putative action has yet to be explored meticulously.
Galanin released from noradrenergic terminals can also likely modulate the activity of neurons innervated by the LC. As discussed in the anatomy section above, the LC projects to a wide range of brain areas that express galanin receptors, each of which has the potential to transduce a galanin signal. Some prime examples are the dorsal raphe, VTA, basal forebrain, hippocampus, and frontal cortex. From a neuromodulatory standpoint, the main effect of galanin is to suppress the excitability of target cells via GAL1, and many of these nuclei are indeed inhibited by galanin. While these brain regions contain galanin-positive fibers originating from the LC and are inhibited by galanin (Counts et al., 2010; Xu et al., 1998a; Xu et al., 1998b), we still lack conclusive evidence that LC-derived galanin per se is responsible. The best circumstantial evidence to date comes from a study showing that high-frequency stimulation of the LC suppresses dopamine neuron activity in the VTA, an effect that cannot be blocked by adrenergic receptor antagonists and has been attributed to galanin (Grenhoff et al., 1993; Weiss et al., 1998).
Neurotrophic actions of galanin
In addition to its neuromodulatory actions, described above, galanin is a potent trophic and neuroprotective factor throughout the nervous system, and much of its known trophic activity occurs in targets of the LC via activation of GAL2. Galanin promotes neurite outgrowth in hippocampal slices and dorsal root ganglion cultures via activation of GAL2 and a mechanism involving the actin-depolymerizing factor cofilin (Hobson et al., 2008; Hobson et al., 2013). GAL2 signaling is associated with Gq/11-mediated inhibition of RhoA GTPase. Inhibition of RhoA subsequently reduces activation of Rho-associated protein kinase (ROCK) signaling, which dephosphorylates and thereby activates cofilin (Hobson et al., 2008; Hobson et al., 2013). Galanin signaling may also contribute to dendritic spine maintenance through GAL2-mediated inhibition of RhoA. The GAL2 agonist AR-M1896 promotes the aggregation of MAP-2 and β-tubulin in hippocampal neurons (Pirondi et al., 2005a), whereas RhoA signaling contributes to MAP-2/β-tubulin disaggregation (Koleske, 2013). The aggregation reflects normal microtubule integrity and is vital for delivery of the microtubule-based cargoes necessary to maintain dendritic spines (Koleske, 2013). Consistent with this molecular action, our laboratories have reported that daily administration of galanin protects against acute stress-induced reductions in dendritic spines in layer V prefrontocortical neurons (Sciolino et al., 2015).
GAL2 signaling protects against a variety of physiological insults in vitro, including excitotoxicity and β-amyloid toxicity (Elliott-Hunt et al., 2007; Elliott-Hunt et al., 2011), and this neuroprotection may involve activation of ERK or Akt (Elliott-Hunt et al., 2007). The prominence of galanin-induced neuroprotection in vivo is particularly obvious when examining the response of endogenous galanin systems to neural injury and neurodegeneration. Galanin expression and galanin-mediated plasticity increases in response to a wide range of pathophysiological insults (Counts et al., 2010; Hobson et al., 2008) and is necessary for neural degeneration following injury (Hobson et al., 2008; Holmes et al., 2000). The widely studied changes in galanin associated with Alzheimer’s disease offer an instructive example. Upregulation of galanin expression and galaninergic hyperinnervation occur in forebrain structures and LC neurons exhibiting Alzheimer’s-related pathology in humans and rodent models (Counts et al., 2010). The fact that galaninergic neurons are relatively spared in late-stage Alzheimer’s patients supports a neuroprotective function and reveals a likely source of galaninergic hyperinnervation of the forebrain (Miller et al., 1999). The neuroprotective actions of galanin described above, particularly against β-amyloid toxicity, argue for the theory that galanin overexpression occurs in response to neurodegeneration and serves to slow or reverse this process (Counts et al., 2010; Hobson et al., 2008). On the other hand, the effects of exogenous administration and galanin overexpression on cognitive performance in rodents support the theory that galaninergic hyperinnervation may contribute to some of the cognitive impairments associated with Alzheimer’s disease (Crawley, 2008), although it is important to note that these two theories are not mutually exclusive. More specifically, the cognitive impairment could be due to the GAL1-mediated neuromodulatory effects, while the neuroprotective effects of galanin may be mediated by GAL2. While the LC may serve as the primary source of galanin to the cortical and hippocampal structures vulnerable to pathological atrophy, galanin derived from the serotonergic raphe and cholinergic basal forebrain nuclei must also be considered in these trophic and neuromodulatory actions (Counts et al., 2010; Perez et al., 2001; Xu et al., 1998b).
Addiction
Although there has been limited work examining the role of galanin in drug addiction, the information available suggests that the effects of this neuropeptide depend on drug class. In general, galanin protects against the rewarding and aversive withdrawal effects of opiates and psychostimulants, while it facilitates responding for nicotine and ethanol in rodents. The effects of galanin on opiate responses are the most comprehensively studied. Transgenic overexpression or centrally administered galanin or the synthetic galanin receptor agonist galnon blocks morphine conditioned place preference and withdrawal signs, while galanin knockout mice show enhanced morphine conditioned place preference and exacerbated morphine withdrawal (Hawes et al., 2008; Holmes et al., 2012; Picciotto et al., 2005; Picciotto, 2008; Zachariou et al., 1999; Zachariou et al., 2003). Similarly, galanin knockout mice are more sensitive to cocaine conditioned place preference, which is reversed by galnon (Narasimhaiah et al., 2009). We and others detected no significant effects of increased galanin signaling on cocaine self-administration, but we found that galnon attenuates cocaine-primed reinstatement of cocaine seeking in rats, an operant model of relapse (Brabant et al., 2010; Ogbonmwan et al., 2014).
In contrast, galanin knockout mice have impaired nicotine conditioned place preference and reduced voluntary ethanol consumption, while galanin-overexpressing mice drink more ethanol than their wild-type counterparts (Karatayev et al., 2009; Karatayev et al., 2010; Neugebauer et al., 2011). Notably, not all studies agree on these general findings. For example, another group has reported that galnon administration decreases nicotine conditioned place preference (Jackson et al., 2011). Galanin, particularly GAL1, has also been implicated in human drug dependence (Gold et al., 2012; Jackson et al., 2011; Lori et al., 2011), although whether the genetic polymorphisms associated with addiction examined in these studies increase or decrease galanin transmission is unknown. By contrast, a variant in a galanin enhancer that appears to impact galanin expression did not significantly affect cannabis, alcohol, or tobacco use (Richardson et al., 2014).
Several lines of converging evidence suggest that LC-derived galanin is at least partially responsible for some of these phenotypes. First, chronic opiate exposure and withdrawal increases galanin and galanin receptor expression in the LC, and withdrawal-induced LC activity is decreased by galanin (Georgescu et al., 2003; Holmes et al., 2012; McClung et al., 2005; Zachariou et al., 1999; Zachariou et al., 2000). Second, given the suppression of LC firing produced by autocrine release of galanin discussed earlier, one might predict that NE depletion would phenocopy increased galanin transmission; indeed, this is the case in many instances. For example, like transgenic galanin overexpression or galanin receptor agonist administration, selective suppression of NE transmission via knockout of α1-adrenergic receptors or the NE biosynthetic enzyme dopamine β-hydroxylase, 6-OHDA lesions, or the administration of adrenergic receptor antagonists can attenuate the rewarding effects of morphine and withdrawal symptoms (Drouin et al., 2002; Maldonado, 1997; Mazei-Robison and Nestler, 2012; Olson et al., 2006; Sahraei et al., 2004; Ventura et al., 2005; Weinshenker and Schroeder, 2007; Zarrindast et al., 2002). Similar to manipulations of galanin itself, suppression of NE transmission has no effect for the most part on operant psychostimulant self-administration, but we and others have shown that psychostimulant conditioned place preference and reinstatement are also reduced upon blockade of NE signaling (Leri et al., 2002; Mantsch et al., 2010; Schroeder et al., 2010; Schroeder et al., 2013; Smith and Aston-Jones, 2011; Ventura et al., 2007; Vranjkovic et al., 2014; Wee et al., 2008; Weinshenker and Schroeder, 2007; Zhang and Kosten, 2005; Zhang and Kosten, 2007) (our unpublished data). Both chronic voluntary exercise and galnon block cocaine-primed reinstatement of cocaine seeking, and exercise is associated with increased galanin mRNA specifically in the LC (Eisenstein and Holmes, 2007; Murray et al., 2010; O’Neal et al., 2001; Ogbonmwan et al., 2015; Sciolino et al., 2012; Sciolino et al., 2015; Van Hoomissen et al., 2004). By contrast, NE depletion and increasing galanin transmission have opposite effects on ethanol and nicotine reward, suggesting that these addiction-related behaviors are mediated by galanin outside of the LC (Forget et al., 2010; Weinshenker et al., 2000). Finally, transgenic mice overexpressing galanin under the control of the noradrenergic-specific dopamine β-hydroxylase promoter, which display a robust increase of galanin in LC neurons, are resistant to morphine withdrawal (Zachariou et al., 2003).
Although the studies described above suggest that LC-derived galanin regulates responses to drugs of abuse, these results should be interpreted with caution. For example, some of the phenotypes associated with blockade of NE transmission probably involve the A2 noradrenergic cell group, rather than the LC (Olson et al., 2006). In addition, not all studies have reported an increase of galanin mRNA in the LC following chronic morphine exposure and withdrawal (Holmes et al., 1995b). Furthermore, the relationship between drug-induced increases of galanin in the LC and subsequent behavioral changes, as well as similarities in the effects of galanin signaling and reduced NE transmission, are evidence of association, not necessarily causation. Although at first blush the attenuation of morphine withdrawal in mice overexpressing galanin in the LC appears to show convincingly that LC-derived galanin is important for this phenotype, it is noteworthy that ectopic overexpression of galanin occurs in several other brain regions in these mice, including the piriform cortex, entorhinal cortex, and subiculum (Crawley et al., 2002; He et al., 2005; Steiner et al., 2001). Combined, these data argue strongly that galanin released from LC neurons modulates some responses to drugs of abuse, but we lack definitive experiments employing manipulations of galanin exclusively in LC neurons.
Both neuromodulatory and trophic actions of galanin probably contribute to its influence on responses to addictive drugs. For example, acute administration of galanin or galnon effectively modulates many of the responses to addictive drugs described above, and GAL1 appears to transduce this signal. On the other hand, GAL2 is the most abundant receptor subtype in the mesocorticolimbic system that controls reward and reinforcement (O’Donnell et al., 1999), and voluntary exercise, which attenuates drug seeking in several different paradigms (Lynch et al., 2013; Ogbonmwan et al., 2015), potently and selectively increases galanin mRNA in the LC in a chronic manner that lends itself to long-term trophic effects (Eisenstein and Holmes, 2007; Murray et al., 2010; O’Neal et al., 2001; Ogbonmwan et al., 2015; Sciolino et al., 2012; Sciolino et al., 2015; Van Hoomissen et al., 2004).
Seizure susceptibility
Galanin has potent endogenous and exogenous anticonvulsant properties. For example, galanin or galnon administration suppresses multiple types of induced seizures, while galanin gene knockout or receptor antagonists exacerbate seizures, and these effects appear to be mediated primarily by GAL1 (Gottsch et al., 2005; Lerner et al., 2008; Mazarati and Lu, 2005; Mitsukawa et al., 2008). The best evidence that LC-derived galanin is anticonvulsant comes from two studies, one employing galanin-overexpressing mice, and the other, exercise. Transgenic mice that overexpress galanin in LC neurons are resistant to seizures produced by perforant path stimulation, pentylenetetrazol, or kainic acid (Mazarati et al., 2000). However, as noted above, these mice also have ectopic galanin expression in seizure-sensitive brain regions that could contribute to the phenotype. We showed that the anticonvulsant effect of chronic running wheel exercise is abolished by the galanin receptor antagonist M-40 (Reiss et al., 2009). Importantly, exercise upregulates galanin mRNA exclusively in LC neurons (Eisenstein and Holmes, 2007; Murray et al., 2010; O’Neal et al., 2001; Ogbonmwan et al., 2015; Van Hoomissen et al., 2004), strongly suggesting that the galanin in question is coming from the LC. In contrast to the influence of galanin on drug addiction-related behaviors, the protective effects of galanin against seizures would presumably not be autocrine in nature since NE itself is also anticonvulsant (Giorgi et al., 2004; Weinshenker and Szot, 2002) and suppression of LC firing by galanin would be predicted to exacerbate epileptic activity. Instead, galanin released by noradrenergic terminals in the hippocampus probably contributes to the anticonvulsant effect of galanin (Holmes et al., 2015; Reiss et al., 2009). Several clues indicate that neuromodulation by galanin, rather than its trophic effects, underlies its anticonvulsant properties: GAL1 appears to have a greater impact on seizures than GAL2, acute administration of galnon attenuates seizures, and acute administration of galanin receptor antagonist blocks the anticonvulsant effect of galanin and exercise.
Depression
A significant body of preclinical evidence implicates galanin in depressive disorders, but this literature is often contradictory and difficult to ascertain. Human genetic evidence strongly implicates the galanin system in the risk of developing stress-related disorders, such as depression. Juhasz and colleagues identified polymorphisms in genes encoding galanin and its receptors that increase the risk for depression in subjects who have experienced significant stress(Juhasz et al., 2014). Surprisingly, these investigators found that variants in galanin genes are more predictive of stress-related depression and anxiety than the canonical 5HTTLPR polymorphism. Several other laboratories report that galanin polymorphisms predict symptom severity in anxiety and depressive disorders in a wide range of different populations (Davidson et al., 2011; Unschuld et al., 2010; Wray et al., 2012). Interpretation of these results is difficult, however, because it is not clear whether the alleles in question increase or decrease galanin transmission. Thus, the human genetic data accumulated so far cannot tell us anything about valence, only that differences in galanin may contribute to depression
Tests of acute administration of non-selective galanin receptor agonists in preclinical bioassays, such as the forced swim and tail suspension tests in rats and mice, respectively, have resulted in decreased immobility (an “antidepressant-like” pattern) (Bartfai et al., 2004; Lu et al., 2005a), increased immobility (Kuteeva et al., 2008a; Kuteeva et al., 2008b), or no effect (Holmes et al., 2005). Much of this confusion comes from interpreting the results of tests that represent pharmacological “screens” or “bioassays” of acute antidepressant actions in rodents, rather than chronic depression models that meet systematic, validating criteria (Holmes, 2003); potentially opposing effects of GAL1 versus GAL2 activation and site of action also contribute to the confusion.
Although the original “Monoamine Hypothesis of Depression” posited that depression was caused by a deficit in NE (and/or serotonin) levels, a more modern hypothesis based on both clinical and preclinical evidence is that LC hyperactivity causes depression-like behaviors. For example, biochemical findings point to markedly elevated NE in the brains of depressed patients (Ehnvall et al., 2003; Wong et al., 2000), and postmortem studies revealed reduced α2 inhibitory autoreceptor function in the LC (Ordway et al., 1994; Ordway et al., 2003), indicating diminished negative feedback that would lead directly to elevated LC activity. A convergence of studies, mostly by Dr. Jay Weiss and colleagues, spanning many years and multiple approaches has established LC hyperactivity as a root cause of depression-like behavior in rodents. A strong uncontrollable stressor (tail shock) induced depression-like behavior (reduced psychomotor activity) and heightened burst firing of LC neurons, and the magnitude of the motor activity suppression was highly and significantly correlated with the degree of LC neuron hyper-responsivity in individual animals (Simson and Weiss, 1988). Intra-LC administration of drugs that activate LC neurons produced depressive-like symptoms in rats and mice, whereas drugs that suppress LC activity attenuated these symptoms (Stone et al., 2009; Weiss et al., 1986). An independent line of corroborating evidence comes from the finding that chronic administration of all known clinically effective antidepressant treatments, including multiple classes of antidepressants with different pharmacological actions (e.g. tricyclics, SSRIs, the α2-antagonist mirtazapine) and electroconvulsive shock, decreased spontaneous and sensory-evoked burst firing of LC neurons in mature adult rats, while drugs that lacked clinical antidepressant efficacy did not (Grant and Weiss, 2001; West et al., 2009). An “exception that proves the rule” can be seen in young, adolescent rats. There is a “black box” warning on antidepressants because of the risk of increased depression/suicide in human adolescents when they are first treated with antidepressants, and the data indicate that this applies exclusively to selective serotonin reuptake inhibitors (SSRIs). In adolescent rats, SSRIs early in treatment had the effect of increasing, rather than decreasing, LC activity and depression-like behavior. By contrast, tricyclics, which are not associated with increased risk of suicidality in adolescents, decreased LC activity in both adult and adolescent rats (West et al., 2010a; West et al., 2010b).
An important question is how LC hyperactivity might cause behavioral depression. Elevated brain NE, a direct consequence of LC hyperactivity, is unlikely to be responsible for depression-related symptoms because many drugs that increase brain NE transmission lack pro-depressant effects, and in fact have antidepressant properties. One clue that points towards galanin as the mediator of LC hyperactivity-induced depression comes from the finding that low-frequency LC stimulation provides excitatory drive onto dopaminergic neurons in the VTA, whereas burst firing of LC neurons (which occurs prominently when LC neurons are hyperactive) inhibits firing of these cells (Grenhoff et al., 1993). Importantly, the excitatory effects of single-pulse LC stimulation are abolished following blockade of adrenergic receptors, whereas the inhibitory effects of phasic LC stimulation are not (Grenhoff et al., 1993). Given the importance of these mesolimbic dopamine neurons for reward and psychomotor activity, it has been suggested that core symptoms of depression, such as anhedonia and depressed activity, are consistent with and could be caused by inhibition of the VTA (Nestler and Carlezon, 2006). An obvious neuropeptide candidate to mediate these effects is galanin, which is expressed in LC neurons that project to the VTA (Gundlach et al., 1990; Melander et al., 1986a; Weiss et al., 1998; Xu et al., 1998a) and is a potent inhibitor of dopamine transmission (de Weille et al., 1989; Gopalan et al., 1993; Nordstrom et al., 1987; Ogbonmwan et al., 2014), and its release probably occurs preferentially during high phasic bursting (Bartfai et al., 1988; Consolo et al., 1994; Karhunen et al., 2001; Verhage et al., 1991). Moreover, galanin overexpression is associated with increased immobility in the forced swim test in mice (Kuteeva et al., 2005a; Kuteeva et al., 2005b; Pirondi et al., 2005b). Based on this evidence, Weiss and colleagues have hypothesized that LC hyperactivity produces depression-related behavioral changes by galanin-mediated inhibition affecting the cell bodies of the VTA dopamine neurons that play a critical role in reward and motivation (Weiss et al., 1996; Weiss et al., 1998), not because LC hyperactivity increases NE transmission. This hypothesis is supported by several key results: (1) chronic antidepressant administration was reported to reduce galanin mRNA in the LC by qRT-PCR (Rovin et al., 2012), (2) effective antidepressant treatments (drugs and electroconvulsive shock) increase VTA dopamine neuronal activity, consistent with their decreasing LC neuronal activity and thereby presumably decreasing galanin release (West and Weiss, 2011), and (3) intra-VTA administration of galanin increases immobility in the forced swim test, while blockade of galanin receptors in the VTA reverses stress-induced behavioral depression that is associated with LC hyperactivity and burst firing (Weiss et al., 2005). Although all of these data are consistent with a role for LC-derived galanin in depression, conclusive evidence must await direct measurements of galanin release in the VTA, coupled with behavioral analysis following specific manipulations of LC firing.
These results indicating a pro-depressant effect of LC-derived galanin must be tempered with conflicting evidence suggesting that galanin has potent antidepressant properties, as well. For example, contrary to the findings of the Weiss group (Rovin et al., 2012), others have reported that experimental manipulations that are clinically effective in treating depression, such chronic administration of a variety of antidepressants (Holmes et al., 2006; Lu et al., 2005a), electroconvulsive therapy (Christiansen, 2011), sleep deprivation (Lu et al., 2005a), and exercise (Ogbonmwan et al., 2015; Sciolino et al., 2012), all similarly induce galanin gene expression in the LC. While GAL1-selective agonists increase immobility in the forced swim and tail suspension tests, GAL2 agonists or GAL2 overexpression consistently produce antidepressant-like effects (Kuteeva et al., 2008a; Kuteeva et al., 2008b; Le Maitre et al., 2013; Saar et al., 2013a; Saar et al., 2013b), and the galanin receptor antagonist M40 reverses the antidepressant-like actions of fluoxetine (Lu et al., 2005a). Finally, rapid, antidepressive actions of galanin have also been reported in humans (Murck et al., 1999; Murck et al., 2004).
How can we reconcile these disparate results? The most obvious possibility is that the discrepancies depend on receptor selectivity. Also noteworthy is that antidepressant therapies in humans require chronic treatment for efficacy, many of which increase galanin in the LC (see above). An appealing explanation is that neuromodulatory LC-derived galanin signaling via GAL1 acutely suppresses mesolimbic dopamine function, thereby reducing behavioral activity in a manner reminiscent of some depression symptoms. On the other hand, trophic galanin actions mediated by GAL2 may exert long-lasting antidepressant actions through neural adaptations in mesocorticolimbic targets of the LC. The use of other tests for depression-like behavior, particularly ones that are only reversed by chronic antidepressant treatment (e.g. novelty-suppressed feeding test, chronic mild stress, or corticolimbic lesion models), will be necessary to test the latter hypothesis. Another potential mechanism that may account for the complexity of galanin-mediated effects could relate to the relative magnitude or patterns of electrophysiological activity in LC-VTA circuits that underlie some depression-like behaviors. Though decreased activity in VTA neurons may produce immobility in the forced swim test, as described above, extremely high firing rates of VTA neurons is associated with a depressive-like phenotype in the social defeat paradigm (Chaudhury et al., 2013; Friedman et al., 2014; Krishnan et al., 2007). Galanin-mediated inhibition of the VTA may therefore produce antidepressant-like effects in this model, either through autoinhibition of LC firing or direct inhibitory influences on the VTA itself. Overall, assessing the role of LC galanin in depression must take into account not only the depression-related behaviors being measured, but also the contribution of a particular LC circuit in those behaviors.
Stress and anxiety
The anatomical capacity for the LC to influence multiple neurotransmitter systems and the direct, neuromodulatory effects of galanin on all such systems support the hypothesis that galanin may potently modulate stress and addictive behaviors via acute neuromodulatory actions. The literature examining the effects of galanin on stress-related behavior consists mostly of experiments that involved acute administration of galanin receptor ligands and subsequent measures in a variety of standard paradigms to detect anxiolytic-like actions. Most of these investigations tested the prediction that galanin would acutely diminish stress and anxiety-related behaviors and are based on a hypothetical mechanism involving acute reductions in noradrenergic LC activity. Centrally administered galanin or the nonselective agonist galnon indeed exerts anxiolytic actions in rats or mice in the conflict test (Bing et al., 1993) and elevated zero maze (Rajarao et al., 2007), and peripherally administered galanin is reported to produce lasting anxiolytic-like effects on open field exploration in rats (Klenerova et al., 2011). By contrast, centrally administered galanin fails to alter behavior in mice in the elevated plus maze, light-dark exploration, and novel open field tests (Karlsson et al., 2005; Karlsson and Holmes, 2006). Reviewing the literature involving galanin/GAL1/GAL2 transgenic mice, the findings are predominantly negative in most standard anxiety models (Karlsson and Holmes, 2006). Galanin-overexpressing mice do not differ from wild-type controls in baseline behavior in the elevated plus maze, light-dark exploration test, or open field test (Holmes et al., 2002). For the most part, neither GAL1 nor GAL2 knockout influences spontaneous behavior in these models, with the exception of an anxiogenic-like profile in GAL1-knockout mice in the elevated plus maze (Holmes et al., 2003).
As described above, much of the inconsistency concerning a role for galanin in stress responses may be due to the difficulties in interpreting acute effects in bioassays that are optimal for detecting anxiolytic-like activity, rather than paradigms involving prior exposure to stress, which may more closely model anxiety-related disorders. Indeed, a clearer picture emerges in experiments that focus on stress resilience or resistance. These studies consistently reveal that galanin may influence the longitudinal impact of stress. Our laboratories have employed a paradigm that involves a single, brief session of footshock that produces an anxiogenic effect when assessed the following day in the elevated plus maze in rats. Daily icv administration of galanin for 20 days prior to the footshock session blocks the anxiogenic effects observed in the elevated plus maze (Sciolino et al., 2015). This finding reveals that galanin promotes stress resilience and suggests this action may occur through long-term neural adaptations mediated by galanin. Similar to chronic administration of galanin, the same period of voluntary exercise increases galanin expression in the LC and induces resilience to stress. This exercise-induced resilience is reversed by chronic but not acute administration of the galanin receptor antagonist M40 (Sciolino et al., 2015). The results therefore reveal a potential role for LC-derived galanin in promoting stress resilience, potentially via a trophic mechanism.
Several other studies support a role for galanin in stress resilience. Although galanin overexpression does not impact spontaneous behavior in anxiety models, galanin-overexpressing mice are insensitive to the anxiogenic effects of yohimbine in the light-dark exploration test (Holmes et al., 2002). This finding directly supports a neuromodulatory hypothesis in which galanin dampens stress-related behavior by regulating noradrenergic function, though it demonstrates that such a mechanism must be relatively minor since it does not generalize to other anxiety paradigms. In a similar vein, GAL2 knockout had no effect on spontaneous behaviors, such as those assessed by the elevated plus maze, light/dark transition, forced swim, or tail suspension tests (Lu et al., 2008). GAL2 knockout mice did, however, exhibit increased susceptibility to the repeated stress employed in a “learned helplessness”-like paradigm compared to wild-type mice. Unlike the other tests, “learned helplessness” paradigms examine the impact of uncontrollable stress exposure on subsequent responding to repeated stressors. Loss of GAL2 led to a susceptible phenotype in this paradigm, suggesting that galanin signaling through GAL2 is necessary for resilience. In further support of a specific role for GAL2 in resilience, transgenic overexpression of GAL2 in several fronto-cortical areas, including mPFC, was found to decrease immobility in a version of the forced swim test that involved pre-exposure to swim stress on the previous day (Le Maitre et al., 2011). By contrast, GAL2 overexpression had no effect on elevated plus-maze or novel open field exploration in mice not previously exposed to stress. The role of galanin in stress resilience was further demonstrated by the ability of the mixed galanin receptor agonist galnon to prevent the development of an “extreme stress responsive” phenotype in a putative rat model of post-traumatic stress disorder, which measures the impact of an acute stress on the development of subsequent anxiety-like behaviors over time (Kozlovsky et al., 2009).
The behavioral literature thus points to functions for galanin beyond neuromodulatory effects. Galanin, and GAL2 in particular, are evidently involved in protecting against the lasting consequences of an acute stress event by diminishing reactivity to subsequent stressors, rather than by modulating reactivity to the acute stress event itself. This long-term protection points to a role for galanin in the plasticity necessary for resilience.
Summary
The evidence reviewed above reveals the complex actions of LC-derived galanin, which depend on target circuits, receptor subtype, presynaptic or postsynaptic influences, and/or involvement of NE. Above all, the LC galanin system owes its complexity to the anatomy of the LC itself, and it is thus not surprising that galanin exerts multiple physiological and behavioral effects depending on the particular target innervated by the LC and the receptor subtype involved. Nonetheless, general principles emerge when considering the influence of galanin on physiology and behavior. The model that follows summarizes these principles, specifically as they relate to LC circuitry. By inhibiting activity of the LC itself through somatodendritic autocrine actions, galanin is poised to regulate all noradrenergic functions throughout the brain. GAL1-mediated inhibition of the LC may thus decrease drug seeking and produce anxiolytic and antidepressant effects. It may also lead to cognitive impairment by suppressing attention/arousal mechanisms. Direct inhibition of the LC may also produce a decrease in VTA tone, which could contribute to pro-depressant effects, though dampening hyperexcitability of the VTA may promote resilience to stress and depression-related behaviors. While yet to be definitively established, LC-derived galanin may also regulate VTA electrophysiological activity directly via the abundant GAL2 receptors present in this structure. LC-derived galanin acting via neuromodulatory GAL1-mediated mechanisms appears to have anticonvulsant properties in models of epilepsy and produces cognitive impairment that has been implicated in Alzheimer’s disease. By contrast, trophic actions of galanin in the PFC, hippocampus, and LC mediated by GAL2 may contribute to beneficial, long-term adaptations that promote stress resilience, elevate mood, and protect against neurodegeneration.
Future directions
There is substantial evidence pointing to a role for LC-derived galanin in drug addiction, seizure susceptibility, depression, Alzheimer’s disease, and stress resilience, although upon close examination, the evidence is all circumstantial. We propose that the following criteria must be met as definitive proof of a functional role for LC-derived galanin: (1) bona fide galanin release must accompany selective LC activity, (2) the effects of LC activity must be blocked by a galanin receptor antagonist or selective depletion of galanin from LC neurons, and (3) selective overexpression of galanin in the LC should mimic the effects of increased LC activity. One reason these criteria have not been met before is that until recently we lacked the necessary tools. Notably, there has been no good way to specifically manipulate LC neurons in vivo with the necessary precision or to measure galanin release in the brain. The lack of galanin receptor antagonist specificity has also held the field back. Today, with the development of optogenetics, advances in brain extracellular peptide measurements, and more selective galanin receptor antagonists, the stage is set to rigorously test the neuromodulatory and trophic consequences of galanin release from LC neurons.
The advent of optogenetics, which uses genetically encoded light-sensitive ion channels to selectively activate neurons of a defined brain region and neurotransmitter phenotype within physiological firing parameters, has overcome the limitations of previous techniques (e.g. electrical stimulation, pharmacological activation), transforming the neuroscience and depression fields (Albert, 2014; Nieh et al., 2013; Roeper, 2013; Stuber et al., 2012; Tye et al., 2013). In these studies, a virus containing the light-activated cation channel, channelrhodopsin2 (ChR2), which is driven by a cell type-selective promoter, is injected into a brain region of interest. A fiber optic ferrule or LED is then implanted just dorsal to the injection site, and a laser delivers light pulses that activate the channel and depolarize the neurons expressing ChR2. Importantly, the laser will only activate neurons that express ChR2, and expression of ChR2 is controlled by the cell type-specific promoter in the virus. Two approaches have been used successfully to drive ChR2 expression selectively in LC neurons. First, Cre-dependent ChR2 viruses can be injected into the LC of transgenic mice or rats expressing Cre recombinase under the control of the tyrosine hydroxylase promoter (Carter et al., 2010; Witten et al., 2011). This approach appears to work well in mice, but despite promising data in the original paper, several labs have failed to observe robust ChR2 expression in the LC of TH-Cre rats (E. Vazey and G. Aston-Jones, personal communication; our unpublished data). To overcome this obstacle, a virus was developed that uses the noradrenergic-specific PRSx8 promoter (Hwang et al., 2001) to drive gene expression in the LC. The initial study describing this method used it to achieve DREADD (Designer Receptors Exclusively Activated by Designer Drugs) expression (Vazey and Aston-Jones, 2014), but it has since been adapted for ChR2-mediated optogenetics (E. Vazey and Gary Aston-Jones, personal communication; our unpublished data). Following implantation of an optic ferrule just dorsal to the viral injection site in the LC, only LC neurons, and not non-LC neurons in the region or fibers of passage/terminals from other brain regions, are activated by laser stimulation. LC neurons fire in two distinct modes: low tonic (typically sustained 0.1–5.0 Hz, but can be as high as 10 Hz) and high phasic (typically 5–15-Hz bursts, with suppression of spontaneous activity in between bursts, but bursts can be as high as 20–25 Hz) (Foote et al., 1983). Although peptide transmission from the LC has been primarily inferred from peripheral nerve data, the release of peptide co-transmitters is thought to be frequency dependent: NE release occurs at both low- and high-frequency discharge, while release of peptide co-transmitters occurs preferentially during high phasic bursting (Bartfai et al., 1988; Consolo et al., 1994; Dutton and Dyball, 1979; Karhunen et al., 2001; Verhage et al., 1991). Moreover, because the activity of the neurons is entrained to the frequency of laser stimulation, we can selectively activate LC neurons in low tonic mode, which preferentially releases NE, or high phasic (bursting) mode, which would be expected to release both NE and galanin, suppress VTA dopamine neuron activity, and induce behavioral depression.
Somatodendritic release of NE from LC neurons and its role in mediating auto-inhibition are well established (Huang et al., 2007; Huang et al., 2012). The overflow of NE in the LC measured by in vivo microdialysis corresponds closely to that occurring in the PFC in response to a variety of manipulations, suggesting that somatodendritic release regulates activity in LC targets (Pudovkina et al., 2001). The co-release of NE and galanin in the pancreas is activity dependent and sensitive to α2 autoreceptor modulation, suggesting a similar mechanism may exist in the LC (Scheurink et al., 1992). Ultrastructural analyses reveal that such NE release occurs via large, dense-core vesicles, which is characteristic of peptide co-localization (Huang et al., 2007). This finding may thus provide the anatomical evidence for galanin/NE co-release and suggests a mechanism by which galanin may modulate all LC activity. However, the lack of evidence directly confirming activity-dependent release of galanin from LC terminals/varicosities is a major gap in our understanding of LC-derived galanin function. In general, it is very difficult to measure galanin release directly by in vivo microdialysis because of low endogenous extracellular concentrations, high molecular weight, and poor recovery due to the “sticky” nature of peptides that cause adherence to probe membrane (Shippenberg and Thompson, 2001). A few published studies have reported success with this technique, however. Consolo and coworkers employed in vivo microdialysis to detect galanin release in the ventral hippocampus following local depolarization or stimulation of the cholinergic diagonal band nuclei (Consolo et al., 1994). Though the former manipulation may reflect release from LC projections, the latter only demonstrates a role of cholinergic projections. In vivo microdialysis experiments in the dorsal hippocampus also revealed that 17-β estradiol treatment caused a delayed increase in galanin overflow in anesthetized rats (Hilke et al., 2005), but the source of this release was not determined. Recent advances promise to improve the detection of neuropeptides in brain dialysate. For example, adding antibodies to the perfusion flow can enhance the concentration gradient and probe recovery, and radioimmunoassay, liquid chromatography coupled to mass spectrometry (LC-MS) and capillary liquid chromatography-multistate mass spectrometry (CLC-MS(n)) have facilitated the detection of neuropeptides (Kennedy, 2013; Zhou et al., 2013). We have used enzyme-linked immunosorbent assay (ELISA) successfully to quantify changes in tissue levels of galanin in the brain (Sciolino et al., 2015), and this technique could be adapted for use with dialysate.
By combining optogenetics and new microdialysis techniques, it should be possible now for the first time to directly measure galanin release from noradrenergic terminals in vivo during behavioral testing. It will be critical to test various optogenetic stimulation parameters to confirm that galanin release occurs mainly during high-frequency burst firing of LC neurons. Preliminary studies indicate that high-fidelity activation of LC neurons occurs at ChR2 stimulation frequencies of up to 15 Hz (E. Vazey and G. Aston-Jones, personal communication), which is in the range of the predicted parameters for galanin release (Grenhoff et al., 1993). Newer opsins, such as ChETA (Gunaydin et al., 2010), can sustain higher frequency spike trains, although it has not been tested in LC neurons. The aforementioned DREADDS may also be used to specifically drive the activity of LC neurons (Vazey and Aston-Jones, 2014). DREADDs are derivatives of muscarinic acetylcholine receptors that are engineered to be insensitive to acetylcholine or any other endogenous chemical, but are rapidly activated upon administration of the clozapine derivative, clozapine-N-oxide (CNO), which has no biological activity in the absence of DREADDs (Armbruster et al., 2007; Aston-Jones and Deisseroth, 2013; Wess et al., 2013). Compared to optogenetics, this approach is limited because it cannot precisely control the firing frequency of the neurons: it is more of an all-or-none response and does not appear to be very dose dependent (Vazey and Aston-Jones, 2014). However, because CNO can be administered systemically and a single dose of CNO can modulate neuronal activity for many hours without the need for constant stimulation or connection to a laser (Alexander et al., 2009; Ferguson et al., 2011; Krashes et al., 2011; Rogan and Roth, 2011; Wulff and Arenkiel, 2012), it may be more useful for inducing chronic LC activation and “trophic” galanin release, akin to the ability of chronic voluntary exercise to increase galanin expression in the LC and promote stress resilience. It remains to be seen whether the magnitude of DREADD-mediated LC stimulation is sufficient to induce galanin release.
As mentioned above, the use of DBH-galanin transgenic mice to probe LC-derived galanin function is limited by the ectopic expression of galanin outside the LC. Nevertheless, it should now also be possible to achieve selective galanin overexpression in the LC via intra-LC delivery of PRSx8-galanin virus or Cre-dependent galanin virus in TH-Cre animals. Likewise, these viral vector strategies could be used to reduce LC galanin expression by siRNA or CRISPR. Intra-LC injection of a PRSx8-Cre virus in a “floxed” galanin mouse would presumably achieve galanin ablation in LC neurons, as well. Optogenetic/chemical genetic activation of LC neurons could also be coupled with local infusion of galanin receptor antagonists to establish the contribution of LC-derived galanin to various aspects of neurochemistry and behavior. Finally, optogenetic and chemical genetic tools that suppress, rather than activate, neuronal firing are also available and could be used in attempts to attenuate the effects of presumed LC-derived galanin-mediated processes, such as chronic exercise.
An array of synthetic compounds is currently available to tease apart the relative contributions of GAL1, GAL2, and GAL3 subtypes in the neuromodulatory and trophic functions of galanin. Fragments of the full galanin peptide sequence (29 amino acids in most species, 30 in humans) provide a moderate degree of receptor selectivity. Galanin 2–11 thus exhibits relative selectivity as an agonist for GAL2 and GAL3 (Lang et al., 2007; Webling et al., 2012). Synthetic peptide agonists, such as M1145 and M617, provide good selectivity and nanomolar affinity for GAL2 and GAL1, respectively (Runesson et al., 2009; Webling et al., 2012). A series of novel synthetic peptides with high relative affinity for GAL2 were developed recently (e.g. J18), and these compounds exert potent antidepressant-like effects in the forced swim and tail suspension bioassays (Saar et al., 2013a). There are also several nonpeptide agonists available now, the most widely studied being galnon. Like most other nonpeptide agonists, galnon’s affinity for both GAL1 and GAL2 is in the micromolar range and produces off-target effects on other G protein-coupled receptors (Floren et al., 2005; Webling et al., 2012). Many of the traditional antagonists, such as M35 and M40, are chimeric peptides that bind all receptor subtypes in the nanomolar range (Lang et al., 2007; Webling et al., 2012). Most of the literature reports antagonist activity of these compounds, although partial agonist activity has been reported, as well (Lu et al., 2005b).
Figure 1.
Hypothetical functions of LC-derived galanin. Shown is a model explaining the differential effects of LC-derived galanin depending on site of action and receptor subtype. In general, GAL1-mediated neuromodulatory effects are responsible for cognitive impairment, anticonvulsant activity, decreased addiction-like behavior, and pro-depressant properties, while GAL2-mediated trophic effects underlie stress resil ience, elevated mood, and neuroprotection.
Highlights.
The neuropeptide galanin is expressed in most locus coeruleus (LC) neurons
LC-derived galanin influences neurological and neuropsychiatric phenotypes
Neuromodulatory actions of LC-derived galanin are mediated by GAL1
Neurotrophic actions of LC-derived galanin are mediated by GAL2
New tools and strategies to study LC-derived galanin are proposed
Acknowledgments
We thank C. Strauss for helpful editing of the manuscript. DW and PVH were supported by the National Institutes of Health (DA038453, AG0476670 to DW, DA027535 to DW and PVH).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Albert PR. Light up your life: optogenetics for depression? J Psychiatry Neurosci. 2014;39:3–5. doi: 10.1503/jpn.130267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander GM, Rogan SC, Abbas AI, Armbruster BN, Pei Y, Allen JA, Nonneman RJ, Hartmann J, Moy SS, Nicolelis MA, McNamara JO, Roth BL. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron. 2009;63:27–39. doi: 10.1016/j.neuron.2009.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Armbruster BN, Li X, Pausch MH, Herlitze S, Roth BL. Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A. 2007;104:5163–8. doi: 10.1073/pnas.0700293104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aston-Jones G, Ennis M, Pieribone VA, Nickell WT, Shipley MT. The brain nucleus locus coeruleus: restricted afferent control of a broad efferent network. Science. 1986;234:734–7. doi: 10.1126/science.3775363. [DOI] [PubMed] [Google Scholar]
- Aston-Jones G, Chiang C, Alexinsky T. Discharge of noradrenergic locus coeruleus neurons in behaving rats and monkeys suggests a role in vigilance. Prog Brain Res. 1991;88:501–20. doi: 10.1016/s0079-6123(08)63830-3. [DOI] [PubMed] [Google Scholar]
- Aston-Jones G, Deisseroth K. Recent advances in optogenetics and pharmacogenetics. Brain Res. 2013;1511:1–5. doi: 10.1016/j.brainres.2013.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartfai T, Iverfeldt K, Fisone G, Serfozo P. Regulation of the release of coexisting neurotransmitters. Annu Rev Pharmacol Toxicol. 1988;28:285–310. doi: 10.1146/annurev.pa.28.040188.001441. [DOI] [PubMed] [Google Scholar]
- Bartfai T, Lu X, Badie-Mahdavi H, Barr AM, Mazarati A, Hua XY, Yaksh T, Haberhauer G, Ceide SC, Trembleau L, Somogyi L, Krock L, Rebek J., Jr Galmic, a nonpeptide galanin receptor agonist, affects behaviors in seizure, pain, and forced-swim tests. Proc Natl Acad Sci U S A. 2004;101:10470–5. doi: 10.1073/pnas.0403802101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bing O, Moller C, Engel JA, Soderpalm B, Heilig M. Anxiolytic-like action of centrally administered galanin. Neurosci Lett. 1993;164:17–20. doi: 10.1016/0304-3940(93)90846-d. [DOI] [PubMed] [Google Scholar]
- Bitner RS, Nikkel AL. Alpha-7 nicotinic receptor expression by two distinct cell types in the dorsal raphe nucleus and locus coeruleus of rat. Brain Res. 2002;938:45–54. doi: 10.1016/s0006-8993(02)02485-x. [DOI] [PubMed] [Google Scholar]
- Bouret S, Sara SJ. Network reset: a simplified overarching theory of locus coeruleus noradrenaline function. Trends Neurosci. 2005;28:574–82. doi: 10.1016/j.tins.2005.09.002. [DOI] [PubMed] [Google Scholar]
- Brabant C, Kuschpel AS, Picciotto MR. Locomotion and self-administration induced by cocaine in 129/OlaHsd mice lacking galanin. Behav Neurosci. 2010;124:828–38. doi: 10.1037/a0021221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carter ME, Yizhar O, Chikahisa S, Nguyen H, Adamantidis A, Nishino S, Deisseroth K, de Lecea L. Tuning arousal with optogenetic modulation of locus coeruleus neurons. Nat Neurosci. 2010;13:1526–33. doi: 10.1038/nn.2682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandler DJ, Lamperski CS, Waterhouse BD. Identification and distribution of projections from monoaminergic and cholinergic nuclei to functionally differentiated subregions of prefrontal cortex. Brain Res. 2013;1522:38–58. doi: 10.1016/j.brainres.2013.04.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandler DJ, Gao WJ, Waterhouse BD. Heterogeneous organization of the locus coeruleus projections to prefrontal and motor cortices. Proc Natl Acad Sci U S A. 2014;111:6816–21. doi: 10.1073/pnas.1320827111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charlety PJ, Chergui K, Akaoka H, Saunier CF, Buda M, Aston-Jones G, Chouvet G. Serotonin differentially modulates responses mediated by specific excitatory amino acid receptors in the rat locus coeruleus. Eur J Neurosci. 1993;5:1024–8. doi: 10.1111/j.1460-9568.1993.tb00954.x. [DOI] [PubMed] [Google Scholar]
- Charney DS, Grillon C, Bremner JD. The neurobiological basis of anxiety and fear: circuits, mechanisms, and neurochemical interactions (part 1) Neuroscientist. 1998;4:35–44. [Google Scholar]
- Chaudhury D, Walsh JJ, Friedman AK, Juarez B, Ku SM, Koo JW, Ferguson D, Tsai HC, Pomeranz L, Christoffel DJ, Nectow AR, Ekstrand M, Domingos A, Mazei-Robison MS, Mouzon E, Lobo MK, Neve RL, Friedman JM, Russo SJ, Deisseroth K, Nestler EJ, Han MH. Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature. 2013;493:532–6. doi: 10.1038/nature11713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christiansen SH. Regulation of the galanin system in the brainstem and hypothalamus by electroconvulsive stimulation in mice. Neuropeptides. 2011;45:337–41. doi: 10.1016/j.npep.2011.07.004. [DOI] [PubMed] [Google Scholar]
- Consolo S, Baldi G, Russi G, Civenni G, Bartfai T, Vezzani A. Impulse flow dependency of galanin release in vivo in the rat ventral hippocampus. Proc Natl Acad Sci U S A. 1994;91:8047–51. doi: 10.1073/pnas.91.17.8047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Counts SE, Perez SE, Ginsberg SD, Mufson EJ. Neuroprotective role for galanin in Alzheimer’s disease. EXS. 2010;102:143–62. doi: 10.1007/978-3-0346-0228-0_11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Craig AD. Spinal and trigeminal lamina I input to the locus coeruleus anterogradely labeled with Phaseolus vulgaris leucoagglutinin (PHA-L) in the cat and the monkey. Brain Res. 1992;584:325–8. doi: 10.1016/0006-8993(92)90915-v. [DOI] [PubMed] [Google Scholar]
- Crawley JN, Mufson EJ, Hohmann JG, Teklemichael D, Steiner RA, Holmberg K, Xu ZQ, Blakeman KH, Xu XJ, Wiesenfeld-Hallin Z, Bartfai T, Hokfelt T. Galanin overexpressing transgenic mice. Neuropeptides. 2002;36:145–56. doi: 10.1054/npep.2002.0891. [DOI] [PubMed] [Google Scholar]
- Crawley JN. Galanin impairs cognitive abilities in rodents: relevance to Alzheimer’s disease. Cell Mol Life Sci. 2008;65:1836–41. doi: 10.1007/s00018-008-8158-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davidson S, Lear M, Shanley L, Hing B, Baizan-Edge A, Herwig A, Quinn JP, Breen G, McGuffin P, Starkey A, Barrett P, MacKenzie A. Differential activity by polymorphic variants of a remote enhancer that supports galanin expression in the hypothalamus and amygdala: implications for obesity, depression and alcoholism. Neuropsychopharmacology. 2011;36:2211–21. doi: 10.1038/npp.2011.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Weille JR, Fosset M, Schmid-Antomarchi H, Lazdunski M. Galanin inhibits dopamine secretion and activates a potassium channel in pheochromocytoma cells. Brain Res. 1989;485:199–203. doi: 10.1016/0006-8993(89)90685-9. [DOI] [PubMed] [Google Scholar]
- Deutch AY, Goldstein M, Roth RH. Activation of the locus coeruleus induced by selective stimulation of the ventral tegmental area. Brain Res. 1986;363:307–14. doi: 10.1016/0006-8993(86)91016-4. [DOI] [PubMed] [Google Scholar]
- Drouin C, Darracq L, Trovero F, Blanc G, Glowinski J, Cotecchia S, Tassin JP. Alpha1b-adrenergic receptors control locomotor and rewarding effects of psychostimulants and opiates. J Neurosci. 2002;22:2873–84. doi: 10.1523/JNEUROSCI.22-07-02873.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dutton A, Dyball RE. Phasic firing enhances vasopressin release from the rat neurohypophysis. J Physiol. 1979;290:433–40. doi: 10.1113/jphysiol.1979.sp012781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckmeier D, Shea SD. Noradrenergic plasticity of olfactory sensory neuron inputs to the main olfactory bulb. J Neurosci. 2014;34:15234–43. doi: 10.1523/JNEUROSCI.0551-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehnvall A, Sjogren M, Zachrisson OC, Agren H. Lifetime burden of mood swings and activation of brain norepinephrine turnover in patients with treatment-refractory depressive illness. J Affect Disord. 2003;74:185–9. doi: 10.1016/s0165-0327(02)00011-3. [DOI] [PubMed] [Google Scholar]
- Eisenstein SA, Holmes PV. Chronic and voluntary exercise enhances learning of conditioned place preference to morphine in rats. Pharmacol Biochem Behav. 2007;86:607–15. doi: 10.1016/j.pbb.2007.02.002. [DOI] [PubMed] [Google Scholar]
- Elliott-Hunt CR, Pope RJ, Vanderplank P, Wynick D. Activation of the galanin receptor 2 (GalR2) protects the hippocampus from neuronal damage. J Neurochem. 2007;100:780–9. doi: 10.1111/j.1471-4159.2006.04239.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elliott-Hunt CR, Holmes FE, Hartley DM, Perez S, Mufson EJ, Wynick D. Endogenous galanin protects mouse hippocampal neurons against amyloid toxicity in vitro via activation of galanin receptor-2. J Alzheimers Dis. 2011;25:455–62. doi: 10.3233/JAD-2011-110011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ennis M, Aston-Jones G, Shiekhattar R. Activation of locus coeruleus neurons by nucleus paragigantocellularis or noxious sensory stimulation is mediated by intracoerulear excitatory amino acid neurotransmission. Brain Res. 1992;598:185–95. doi: 10.1016/0006-8993(92)90182-9. [DOI] [PubMed] [Google Scholar]
- Espana RA, Reis KM, Valentino RJ, Berridge CW. Organization of hypocretin/orexin efferents to locus coeruleus and basal forebrain arousal-related structures. J Comp Neurol. 2005;481:160–78. doi: 10.1002/cne.20369. [DOI] [PubMed] [Google Scholar]
- Ferguson SM, Eskenazi D, Ishikawa M, Wanat MJ, Phillips PE, Dong Y, Roth BL, Neumaier JF. Transient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitization. Nat Neurosci. 2011;14:22–4. doi: 10.1038/nn.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Floren A, Sollenberg U, Lundstrom L, Zorko M, Stojan J, Budihna M, Wheatley M, Martin NP, Kilk K, Mazarati A, Bartfai T, Lindgren M, Langel U. Multiple interaction sites of galnon trigger its biological effects. Neuropeptides. 2005;39:547–58. doi: 10.1016/j.npep.2005.09.005. [DOI] [PubMed] [Google Scholar]
- Foote SL, Bloom FE, Aston-Jones G. Nucleus locus ceruleus: new evidence of anatomical and physiological specificity. Physiol Rev. 1983;63:844–914. doi: 10.1152/physrev.1983.63.3.844. [DOI] [PubMed] [Google Scholar]
- Forget B, Wertheim C, Mascia P, Pushparaj A, Goldberg SR, Le Foll B. Noradrenergic alpha1 receptors as a novel target for the treatment of nicotine addiction. Neuropsychopharmacology. 2010;35:1751–60. doi: 10.1038/npp.2010.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman AK, Walsh JJ, Juarez B, Ku SM, Chaudhury D, Wang J, Li X, Dietz DM, Pan N, Vialou VF, Neve RL, Yue Z, Han MH. Enhancing depression mechanisms in midbrain dopamine neurons achieves homeostatic resilience. Science. 2014;344:313–9. doi: 10.1126/science.1249240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganesh A, Gonzalez-Sulser A, Chaijale N, Cucchiaro G. Electrophysiologic effects of systemic and locally infused epibatidine on locus coeruleus neurons. Eur J Pharmacol. 2008;584:93–9. doi: 10.1016/j.ejphar.2008.01.034. [DOI] [PubMed] [Google Scholar]
- Georgescu D, Zachariou V, Barrot M, Mieda M, Willie JT, Eisch AJ, Yanagisawa M, Nestler EJ, DiLeone RJ. Involvement of the lateral hypothalamic peptide orexin in morphine dependence and withdrawal. J Neurosci. 2003;23:3106–11. doi: 10.1523/JNEUROSCI.23-08-03106.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giorgi FS, Pizzanelli C, Biagioni F, Murri L, Fornai F. The role of norepinephrine in epilepsy: from the bench to the bedside. Neurosci Biobehav Rev. 2004;28:507–24. doi: 10.1016/j.neubiorev.2004.06.008. [DOI] [PubMed] [Google Scholar]
- Goertz RB, Wanat MJ, Gomez JA, Brown ZJ, Phillips PE, Paladini CA. Cocaine increases dopaminergic neuron and motor activity via midbrain alpha1 adrenergic signaling. Neuropsychopharmacology. 2015;40:1151–62. doi: 10.1038/npp.2014.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gold AB, Wileyto EP, Lori A, Conti D, Cubells JF, Lerman C. Pharmacogenetic association of the galanin receptor (GALR1) SNP rs2717162 with smoking cessation. Neuropsychopharmacology. 2012;37:1683–8. doi: 10.1038/npp.2012.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gopalan C, Tian Y, Moore KE, Lookingland KJ. Neurochemical evidence that the inhibitory effect of galanin on tuberoinfundibular dopamine neurons is activity dependent. Neuroendocrinology. 1993;58:287–93. doi: 10.1159/000126552. [DOI] [PubMed] [Google Scholar]
- Gottsch ML, Zeng H, Hohmann JG, Weinshenker D, Clifton DK, Steiner RA. Phenotypic analysis of mice deficient in the type 2 galanin receptor (GALR2) Mol Cell Biol. 2005;25:4804–11. doi: 10.1128/MCB.25.11.4804-4811.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grant MM, Weiss JM. Effects of chronic antidepressant drug administration and electroconvulsive shock on locus coeruleus electrophysiologic activity. Biol Psychiatry. 2001;49:117–29. doi: 10.1016/s0006-3223(00)00936-7. [DOI] [PubMed] [Google Scholar]
- Grenhoff J, Nisell M, Ferre S, Aston-Jones G, Svensson TH. Noradrenergic modulation of midbrain dopamine cell firing elicited by stimulation of the locus coeruleus in the rat. J Neural Transm Gen Sect. 1993;93:11–25. doi: 10.1007/BF01244934. [DOI] [PubMed] [Google Scholar]
- Grenhoff J, Svensson TH. Prazosin modulates the firing pattern of dopamine neurons in rat ventral tegmental area. Eur J Pharmacol. 1993;233:79–84. doi: 10.1016/0014-2999(93)90351-h. [DOI] [PubMed] [Google Scholar]
- Groves DA, Bowman EM, Brown VJ. Recordings from the rat locus coeruleus during acute vagal nerve stimulation in the anaesthetised rat. Neurosci Lett. 2005;379:174–9. doi: 10.1016/j.neulet.2004.12.055. [DOI] [PubMed] [Google Scholar]
- Guiard BP, El Mansari M, Blier P. Cross-talk between dopaminergic and noradrenergic systems in the rat ventral tegmental area, locus ceruleus, and dorsal hippocampus. Mol Pharmacol. 2008a;74:1463–75. doi: 10.1124/mol.108.048033. [DOI] [PubMed] [Google Scholar]
- Guiard BP, El Mansari M, Merali Z, Blier P. Functional interactions between dopamine, serotonin and norepinephrine neurons: an in-vivo electrophysiological study in rats with monoaminergic lesions. Int J Neuropsychopharmacol. 2008b;11:625–39. doi: 10.1017/S1461145707008383. [DOI] [PubMed] [Google Scholar]
- Gunaydin LA, Yizhar O, Berndt A, Sohal VS, Deisseroth K, Hegemann P. Ultrafast optogenetic control. Nat Neurosci. 2010;13:387–92. doi: 10.1038/nn.2495. [DOI] [PubMed] [Google Scholar]
- Gundlach AL, Wisden W, Morris BJ, Hunt SP. Localization of preprogalanin mRNA in rat brain: in situ hybridization study with a synthetic oligonucleotide probe. Neurosci Lett. 1990;114:241–7. doi: 10.1016/0304-3940(90)90570-y. [DOI] [PubMed] [Google Scholar]
- Guo YX, Li DP, Chen SR, Pan HL. Distinct intrinsic and synaptic properties of pre-sympathetic and pre-parasympathetic output neurons in Barrington’s nucleus. J Neurochem. 2013;126:338–48. doi: 10.1111/jnc.12290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen N, Manahan-Vaughan D. Hippocampal long-term potentiation that is elicited by perforant path stimulation or that occurs in conjunction with spatial learning is tightly controlled by beta-adrenoreceptors and the locus coeruleus. Hippocampus. 2015 doi: 10.1002/hipo.22436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawes JJ, Brunzell DH, Wynick D, Zachariou V, Picciotto MR. GalR1, but not GalR2 or GalR3, levels are regulated by galanin signaling in the locus coeruleus through a cyclic AMP-dependent mechanism. J Neurochem. 2005;93:1168–76. doi: 10.1111/j.1471-4159.2005.03105.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawes JJ, Brunzell DH, Narasimhaiah R, Langel U, Wynick D, Picciotto MR. Galanin protects against behavioral and neurochemical correlates of opiate reward. Neuropsychopharmacology. 2008;33:1864–73. doi: 10.1038/sj.npp.1301579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He B, Counts SE, Perez SE, Hohmann JG, Koprich JB, Lipton JW, Steiner RA, Crawley JN, Mufson EJ. Ectopic galanin expression and normal galanin receptor 2 and galanin receptor 3 mRNA levels in the forebrain of galanin transgenic mice. Neuroscience. 2005;133:371–80. doi: 10.1016/j.neuroscience.2005.01.068. [DOI] [PubMed] [Google Scholar]
- Hilke S, Theodorsson A, Fetissov S, Aman K, Holm L, Hokfelt T, Theodorsson E. Estrogen induces a rapid increase in galanin levels in female rat hippocampal formation--possibly a nongenomic/indirect effect. Eur J Neurosci. 2005;21:2089–99. doi: 10.1111/j.1460-9568.2005.04050.x. [DOI] [PubMed] [Google Scholar]
- Hobson SA, Bacon A, Elliot-Hunt CR, Holmes FE, Kerr NC, Pope R, Vanderplank P, Wynick D. Galanin acts as a trophic factor to the central and peripheral nervous systems. Cell Mol Life Sci. 2008;65:1806–12. doi: 10.1007/s00018-008-8154-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hobson SA, Vanderplank PA, Pope RJ, Kerr NC, Wynick D. Galanin stimulates neurite outgrowth from sensory neurons by inhibition of Cdc42 and Rho GTPases and activation of cofilin. J Neurochem. 2013;127:199–208. doi: 10.1111/jnc.12379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hokfelt T, Xu ZQ, Shi TJ, Holmberg K, Zhang X. Galanin in ascending systems. Focus on coexistence with 5-hydroxytryptamine and noradrenaline. Ann N Y Acad Sci. 1998;863:252–63. doi: 10.1111/j.1749-6632.1998.tb10700.x. [DOI] [PubMed] [Google Scholar]
- Holets VR, Hokfelt T, Rokaeus A, Terenius L, Goldstein M. Locus coeruleus neurons in the rat containing neuropeptide Y, tyrosine hydroxylase or galanin and their efferent projections to the spinal cord, cerebral cortex and hypothalamus. Neuroscience. 1988;24:893–906. doi: 10.1016/0306-4522(88)90076-0. [DOI] [PubMed] [Google Scholar]
- Holmes A, Yang RJ, Crawley JN. Evaluation of an anxiety-related phenotype in galanin overexpressing transgenic mice. J Mol Neurosci. 2002;18:151–65. doi: 10.1385/JMN:18:1-2:151. [DOI] [PubMed] [Google Scholar]
- Holmes A, Kinney JW, Wrenn CC, Li Q, Yang RJ, Ma L, Vishwanath J, Saavedra MC, Innerfield CE, Jacoby AS, Shine J, Iismaa TP, Crawley JN. Galanin GAL-R1 receptor null mutant mice display increased anxiety-like behavior specific to the elevated plus-maze. Neuropsychopharmacology. 2003;28:1031–44. doi: 10.1038/sj.npp.1300164. [DOI] [PubMed] [Google Scholar]
- Holmes A, Li Q, Koenig EA, Gold E, Stephenson D, Yang RJ, Dreiling J, Sullivan T, Crawley JN. Phenotypic assessment of galanin overexpressing and galanin receptor R1 knockout mice in the tail suspension test for depression-related behavior. Psychopharmacology (Berl) 2005;178:276–85. doi: 10.1007/s00213-004-1997-1. [DOI] [PubMed] [Google Scholar]
- Holmes FE, Mahoney S, King VR, Bacon A, Kerr NC, Pachnis V, Curtis R, Priestley JV, Wynick D. Targeted disruption of the galanin gene reduces the number of sensory neurons and their regenerative capacity. Proc Natl Acad Sci U S A. 2000;97:11563–8. doi: 10.1073/pnas.210221897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes FE, Armenaki A, Iismaa TP, Einstein EB, Shine J, Picciotto MR, Wynick D, Zachariou V. Galanin negatively modulates opiate withdrawal via galanin receptor 1. Psychopharmacology (Berl) 2012;220:619–25. doi: 10.1007/s00213-011-2515-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmes PV, Blanchard DC, Blanchard RJ, Brady LS, Crawley JN. Chronic social stress increases levels of preprogalanin mRNA in the rat locus coeruleus. Pharmacol Biochem Behav. 1995a;50:655–60. doi: 10.1016/0091-3057(94)00334-3. [DOI] [PubMed] [Google Scholar]
- Holmes PV, de Bartolomeis A, Koprivica V, Crawley JN. Lack of effect of chronic morphine treatment and naloxone-precipitated withdrawal on tyrosine hydroxylase, galanin, and neuropeptide Y mRNA levels in the rat locus coeruleus. Synapse. 1995b;19:197–205. doi: 10.1002/syn.890190307. [DOI] [PubMed] [Google Scholar]
- Holmes PV. Rodent models of depression: reexamining validity without anthropomorphic inference. Crit Rev Neurobiol. 2003;15:143–74. doi: 10.1615/critrevneurobiol.v15.i2.30. [DOI] [PubMed] [Google Scholar]
- Holmes PV, Yoo HS, Dishman RK. Voluntary exercise and clomipramine treatment elevate prepro-galanin mRNA levels in the locus coeruleus in rats. Neurosci Lett. 2006;408:1–4. doi: 10.1016/j.neulet.2006.04.057. [DOI] [PubMed] [Google Scholar]
- Holmes PV, Reiss JI, Murray PS, Dishman RK, Spradley JM. Chronic exercise dampens hippocampal glutamate overflow induced by kainic acid in rats. Behav Brain Res. 2015;284:19–23. doi: 10.1016/j.bbr.2015.02.002. [DOI] [PubMed] [Google Scholar]
- Huang HP, Wang SR, Yao W, Zhang C, Zhou Y, Chen XW, Zhang B, Xiong W, Wang LY, Zheng LH, Landry M, Hokfelt T, Xu ZQ, Zhou Z. Long latency of evoked quantal transmitter release from somata of locus coeruleus neurons in rat pontine slices. Proc Natl Acad Sci U S A. 2007;104:1401–6. doi: 10.1073/pnas.0608897104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang HP, Zhu FP, Chen XW, Xu ZQ, Zhang CX, Zhou Z. Physiology of quantal norepinephrine release from somatodendritic sites of neurons in locus coeruleus. Front Mol Neurosci. 2012;5:29. doi: 10.3389/fnmol.2012.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hwang DY, Carlezon WA, Jr, Isacson O, Kim KS. A high-efficiency synthetic promoter that drives transgene expression selectively in noradrenergic neurons. Hum Gene Ther. 2001;12:1731–40. doi: 10.1089/104303401750476230. [DOI] [PubMed] [Google Scholar]
- Jackson KJ, Chen X, Miles MF, Harenza J, Damaj MI. The neuropeptide galanin and variants in the GalR1 gene are associated with nicotine dependence. Neuropsychopharmacology. 2011;36:2339–48. doi: 10.1038/npp.2011.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jimenez-Rivera CA, Figueroa J, Vazquez-Torres R, Velez-Hernandez ME, Schwarz D, Velasquez-Martinez MC, Arencibia-Albite F. Presynaptic inhibition of glutamate transmission by alpha2 receptors in the VTA. Eur J Neurosci. 2012;35:1406–15. doi: 10.1111/j.1460-9568.2012.08029.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jodo E, Chiang C, Aston-Jones G. Potent excitatory influence of prefrontal cortex activity on noradrenergic locus coeruleus neurons. Neuroscience. 1998;83:63–79. doi: 10.1016/s0306-4522(97)00372-2. [DOI] [PubMed] [Google Scholar]
- Juhasz G, Hullam G, Eszlari N, Gonda X, Antal P, Anderson IM, Hokfelt TG, Deakin JF, Bagdy G. Brain galanin system genes interact with life stresses in depression-related phenotypes. Proc Natl Acad Sci U S A. 2014;111:E1666–73. doi: 10.1073/pnas.1403649111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karatayev O, Baylan J, Leibowitz SF. Increased intake of ethanol and dietary fat in galanin overexpressing mice. Alcohol. 2009;43:571–80. doi: 10.1016/j.alcohol.2009.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karatayev O, Baylan J, Weed V, Chang S, Wynick D, Leibowitz SF. Galanin knockout mice show disturbances in ethanol consumption and expression of hypothalamic peptides that stimulate ethanol intake. Alcohol Clin Exp Res. 2010;34:72–80. doi: 10.1111/j.1530-0277.2009.01068.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karhunen T, Vilim FS, Alexeeva V, Weiss KR, Church PJ. Targeting of peptidergic vesicles in cotransmitting terminals. J Neurosci. 2001;21:RC127. doi: 10.1523/JNEUROSCI.21-03-j0005.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karlsson RM, Holmes A, Heilig M, Crawley JN. Anxiolytic-like actions of centrally-administered neuropeptide Y, but not galanin, in C57BL/6J mice. Pharmacol Biochem Behav. 2005;80:427–36. doi: 10.1016/j.pbb.2004.12.009. [DOI] [PubMed] [Google Scholar]
- Karlsson RM, Holmes A. Galanin as a modulator of anxiety and depression and a therapeutic target for affective disease. Amino Acids. 2006;31:231–9. doi: 10.1007/s00726-006-0336-8. [DOI] [PubMed] [Google Scholar]
- Kaur S, Saxena RN, Mallick BN. GABAergic neurons in prepositus hypoglossi regulate REM sleep by its action on locus coeruleus in freely moving rats. Synapse. 2001;42:141–50. doi: 10.1002/syn.1109. [DOI] [PubMed] [Google Scholar]
- Kennedy RT. Emerging trends in in vivo neurochemical monitoring by microdialysis. Curr Opin Chem Biol. 2013;17:860–7. doi: 10.1016/j.cbpa.2013.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim MA, Lee HS, Lee BY, Waterhouse BD. Reciprocal connections between subdivisions of the dorsal raphe and the nuclear core of the locus coeruleus in the rat. Brain Res. 2004;1026:56–67. doi: 10.1016/j.brainres.2004.08.022. [DOI] [PubMed] [Google Scholar]
- Klenerova V, Flegel M, Skopek P, Sida P, Hynie S. Galanin modulating effect on restraint stress-induced short- and long-term behavioral changes in Wistar rats. Neurosci Lett. 2011;502:147–51. doi: 10.1016/j.neulet.2011.06.051. [DOI] [PubMed] [Google Scholar]
- Koleske AJ. Molecular mechanisms of dendrite stability. Nat Rev Neurosci. 2013;14:536–50. doi: 10.1038/nrn3486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korp BG, Blanks RH, Torigoe Y. Projections of the nucleus of the optic tract to the nucleus reticularis tegmenti pontis and prepositus hypoglossi nucleus in the pigmented rat as demonstrated by anterograde and retrograde transport methods. Vis Neurosci. 1989;2:275–86. doi: 10.1017/s095252380000119x. [DOI] [PubMed] [Google Scholar]
- Koyama Y, Jodo E, Kayama Y. Sensory responsiveness of “broad-spike” neurons in the laterodorsal tegmental nucleus, locus coeruleus and dorsal raphe of awake rats: implications for cholinergic and monoaminergic neuron-specific responses. Neuroscience. 1994;63:1021–31. doi: 10.1016/0306-4522(94)90569-x. [DOI] [PubMed] [Google Scholar]
- Kozlovsky N, Matar MA, Kaplan Z, Zohar J, Cohen H. The role of the galaninergic system in modulating stress-related responses in an animal model of posttraumatic stress disorder. Biol Psychiatry. 2009;65:383–91. doi: 10.1016/j.biopsych.2008.10.034. [DOI] [PubMed] [Google Scholar]
- Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, Cusher DS, Maratos-Flier E, Roth BL, Lowell BB. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Invest. 2011;121:1424–8. doi: 10.1172/JCI46229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan V, Han MH, Graham DL, Berton O, Renthal W, Russo SJ, Laplant Q, Graham A, Lutter M, Lagace DC, Ghose S, Reister R, Tannous P, Green TA, Neve RL, Chakravarty S, Kumar A, Eisch AJ, Self DW, Lee FS, Tamminga CA, Cooper DC, Gershenfeld HK, Nestler EJ. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131:391–404. doi: 10.1016/j.cell.2007.09.018. [DOI] [PubMed] [Google Scholar]
- Kuteeva E, Hokfelt T, Ogren SO. Behavioural characterisation of transgenic mice overexpressing galanin under the PDGF-B promoter. Neuropeptides. 2005a;39:299–304. doi: 10.1016/j.npep.2005.01.004. [DOI] [PubMed] [Google Scholar]
- Kuteeva E, Hokfelt T, Ogren SO. Behavioural characterisation of young adult transgenic mice overexpressing galanin under the PDGF-B promoter. Regul Pept. 2005b;125:67–78. doi: 10.1016/j.regpep.2004.07.028. [DOI] [PubMed] [Google Scholar]
- Kuteeva E, Hokfelt T, Wardi T, Ogren SO. Galanin, galanin receptor subtypes and depression-like behaviour. Cell Mol Life Sci. 2008a;65:1854–63. doi: 10.1007/s00018-008-8160-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuteeva E, Wardi T, Lundstrom L, Sollenberg U, Langel U, Hokfelt T, Ogren SO. Differential role of galanin receptors in the regulation of depression-like behavior and monoamine/stress-related genes at the cell body level. Neuropsychopharmacology. 2008b;33:2573–85. doi: 10.1038/sj.npp.1301660. [DOI] [PubMed] [Google Scholar]
- Lang R, Gundlach AL, Kofler B. The galanin peptide family: receptor pharmacology, pleiotropic biological actions, and implications in health and disease. Pharmacol Ther. 2007;115:177–207. doi: 10.1016/j.pharmthera.2007.05.009. [DOI] [PubMed] [Google Scholar]
- Lang R, Gundlach AL, Holmes FE, Hobson SA, Wynick D, Hokfelt T, Kofler B. Physiology, signaling, and pharmacology of galanin peptides and receptors: three decades of emerging diversity. Pharmacol Rev. 2015;67:118–75. doi: 10.1124/pr.112.006536. [DOI] [PubMed] [Google Scholar]
- Le Maitre E, Barde SS, Palkovits M, Diaz-Heijtz R, Hokfelt TG. Distinct features of neurotransmitter systems in the human brain with focus on the galanin system in locus coeruleus and dorsal raphe. Proc Natl Acad Sci U S A. 2013;110:E536–45. doi: 10.1073/pnas.1221378110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Maitre TW, Xia S, Le Maitre E, Dun XP, Lu J, Theodorsson E, Ogren SO, Hokfelt T, Xu ZQ. Galanin receptor 2 overexpressing mice display an antidepressive-like phenotype: possible involvement of the subiculum. Neuroscience. 2011;190:270–88. doi: 10.1016/j.neuroscience.2011.05.015. [DOI] [PubMed] [Google Scholar]
- Le Novere N, Zoli M, Changeux JP. Neuronal nicotinic receptor alpha 6 subunit mRNA is selectively concentrated in catecholaminergic nuclei of the rat brain. Eur J Neurosci. 1996;8:2428–39. doi: 10.1111/j.1460-9568.1996.tb01206.x. [DOI] [PubMed] [Google Scholar]
- Lee HS, Kim MA, Waterhouse BD. Retrograde double-labeling study of common afferent projections to the dorsal raphe and the nuclear core of the locus coeruleus in the rat. J Comp Neurol. 2005a;481:179–93. doi: 10.1002/cne.20365. [DOI] [PubMed] [Google Scholar]
- Lee HS, Lee BY, Waterhouse BD. Retrograde study of projections from the tuberomammillary nucleus to the dorsal raphe and the locus coeruleus in the rat. Brain Res. 2005b;1043:65–75. doi: 10.1016/j.brainres.2005.02.050. [DOI] [PubMed] [Google Scholar]
- Leri F, Flores J, Rodaros D, Stewart J. Blockade of stress-induced but not cocaine-induced reinstatement by infusion of noradrenergic antagonists into the bed nucleus of the stria terminalis or the central nucleus of the amygdala. J Neurosci. 2002;22:5713–8. doi: 10.1523/JNEUROSCI.22-13-05713.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lerner JT, Sankar R, Mazarati AM. Galanin and epilepsy. Cell Mol Life Sci. 2008;65:1864–71. doi: 10.1007/s00018-008-8161-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Linster C, Nai Q, Ennis M. Nonlinear effects of noradrenergic modulation of olfactory bulb function in adult rodents. J Neurophysiol. 2011;105:1432–43. doi: 10.1152/jn.00960.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lori A, Tang Y, O’Malley S, Picciotto MR, Wu R, Conneely KN, Cubells JF. The galanin receptor 1 gene associates with tobacco craving in smokers seeking cessation treatment. Neuropsychopharmacology. 2011;36:1412–20. doi: 10.1038/npp.2011.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loughlin SE, Foote SL, Bloom FE. Efferent projections of nucleus locus coeruleus: topographic organization of cells of origin demonstrated by three-dimensional reconstruction. Neuroscience. 1986;18:291–306. doi: 10.1016/0306-4522(86)90155-7. [DOI] [PubMed] [Google Scholar]
- Lu X, Barr AM, Kinney JW, Sanna P, Conti B, Behrens MM, Bartfai T. A role for galanin in antidepressant actions with a focus on the dorsal raphe nucleus. Proc Natl Acad Sci U S A. 2005a;102:874–9. doi: 10.1073/pnas.0408891102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu X, Lundstrom L, Langel U, Bartfai T. Galanin receptor ligands. Neuropeptides. 2005b;39:143–6. doi: 10.1016/j.npep.2004.12.012. [DOI] [PubMed] [Google Scholar]
- Lu X, Ross B, Sanchez-Alavez M, Zorrilla EP, Bartfai T. Phenotypic analysis of GalR2 knockout mice in anxiety- and depression-related behavioral tests. Neuropeptides. 2008;42:387–97. doi: 10.1016/j.npep.2008.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luppi PH, Aston-Jones G, Akaoka H, Chouvet G, Jouvet M. Afferent projections to the rat locus coeruleus demonstrated by retrograde and anterograde tracing with cholera-toxin B subunit and Phaseolus vulgaris leucoagglutinin. Neuroscience. 1995;65:119–60. doi: 10.1016/0306-4522(94)00481-j. [DOI] [PubMed] [Google Scholar]
- Lynch WJ, Peterson AB, Sanchez V, Abel J, Smith MA. Exercise as a novel treatment for drug addiction: a neurobiological and stage-dependent hypothesis. Neurosci Biobehav Rev. 2013;37:1622–44. doi: 10.1016/j.neubiorev.2013.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma X, Tong YG, Schmidt R, Brown W, Payza K, Hodzic L, Pou C, Godbout C, Hokfelt T, Xu ZQ. Effects of galanin receptor agonists on locus coeruleus neurons. Brain Res. 2001;919:169–74. doi: 10.1016/s0006-8993(01)03033-5. [DOI] [PubMed] [Google Scholar]
- Maldonado R. Participation of noradrenergic pathways in the expression of opiate withdrawal: biochemical and pharmacological evidence. Neurosci Biobehav Rev. 1997;21:91–104. doi: 10.1016/0149-7634(95)00061-5. [DOI] [PubMed] [Google Scholar]
- Mantsch JR, Weyer A, Vranjkovic O, Beyer CE, Baker DA, Caretta H. Involvement of noradrenergic neurotransmission in the stress- but not cocaine-induced reinstatement of extinguished cocaine-induced conditioned place preference in mice: role for beta-2 adrenergic receptors. Neuropsychopharmacology. 2010;35:2165–78. doi: 10.1038/npp.2010.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazarati A, Lu X. Regulation of limbic status epilepticus by hippocampal galanin type 1 and type 2 receptors. Neuropeptides. 2005;39:277–80. doi: 10.1016/j.npep.2004.12.003. [DOI] [PubMed] [Google Scholar]
- Mazarati AM, Hohmann JG, Bacon A, Liu H, Sankar R, Steiner RA, Wynick D, Wasterlain CG. Modulation of hippocampal excitability and seizures by galanin. J Neurosci. 2000;20:6276–81. doi: 10.1523/JNEUROSCI.20-16-06276.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazei-Robison MS, Nestler EJ. Opiate-induced molecular and cellular plasticity of ventral tegmental area and locus coeruleus catecholamine neurons. Cold Spring Harb Perspect Med. 2012;2:a012070. doi: 10.1101/cshperspect.a012070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McClung CA, Nestler EJ, Zachariou V. Regulation of gene expression by chronic morphine and morphine withdrawal in the locus ceruleus and ventral tegmental area. J Neurosci. 2005;25:6005–15. doi: 10.1523/JNEUROSCI.0062-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melander T, Hokfelt T, Rokaeus A, Cuello AC, Oertel WH, Verhofstad A, Goldstein M. Coexistence of galanin-like immunoreactivity with catecholamines, 5-hydroxytryptamine, GABA and neuropeptides in the rat CNS. J Neurosci. 1986a;6:3640–54. doi: 10.1523/JNEUROSCI.06-12-03640.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melander T, Staines WA, Rokaeus A. Galanin-like immunoreactivity in hippocampal afferents in the rat, with special reference to cholinergic and noradrenergic inputs. Neuroscience. 1986b;19:223–40. doi: 10.1016/0306-4522(86)90017-5. [DOI] [PubMed] [Google Scholar]
- Mennicken F, Hoffert C, Pelletier M, Ahmad S, O’Donnell D. Restricted distribution of galanin receptor 3 (GalR3) mRNA in the adult rat central nervous system. J Chem Neuroanat. 2002;24:257–68. doi: 10.1016/s0891-0618(02)00068-6. [DOI] [PubMed] [Google Scholar]
- Miller MA, Kolb PE, Leverenz JB, Peskind ER, Raskind MA. Preservation of noradrenergic neurons in the locus ceruleus that coexpress galanin mRNA in Alzheimer’s disease. J Neurochem. 1999;73:2028–36. [PubMed] [Google Scholar]
- Mitrano DA, Schroeder JP, Smith Y, Cortright JJ, Bubula N, Vezina P, Weinshenker D. alpha-1 Adrenergic receptors are localized on presynaptic elements in the nucleus accumbens and regulate mesolimbic dopamine transmission. Neuropsychopharmacology. 2012;37:2161–72. doi: 10.1038/npp.2012.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitsukawa K, Lu X, Bartfai T. Galanin, galanin receptors and drug targets. Cell Mol Life Sci. 2008;65:1796–805. doi: 10.1007/s00018-008-8153-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murck H, Antonijevic IA, Frieboes RM, Maier P, Schier T, Steiger A. Galanin has REM-sleep deprivation-like effects on the sleep EEG in healthy young men. J Psychiatr Res. 1999;33:225–32. doi: 10.1016/s0022-3956(98)00059-4. [DOI] [PubMed] [Google Scholar]
- Murck H, Held K, Ziegenbein M, Kunzel H, Holsboer F, Steiger A. Intravenous administration of the neuropeptide galanin has fast antidepressant efficacy and affects the sleep EEG. Psychoneuroendocrinology. 2004;29:1205–11. doi: 10.1016/j.psyneuen.2004.02.006. [DOI] [PubMed] [Google Scholar]
- Murray PS, Groves JL, Pettett BJ, Britton SL, Koch LG, Dishman RK, Holmes PV. Locus coeruleus galanin expression is enhanced after exercise in rats selectively bred for high capacity for aerobic activity. Peptides. 2010;31:2264–8. doi: 10.1016/j.peptides.2010.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narasimhaiah R, Kamens HM, Picciotto MR. Effects of galanin on cocaine-mediated conditioned place preference and ERK signaling in mice. Psychopharmacology (Berl) 2009;204:95–102. doi: 10.1007/s00213-008-1438-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nestler EJ, Carlezon WA., Jr The mesolimbic dopamine reward circuit in depression. Biol Psychiatry. 2006;59:1151–9. doi: 10.1016/j.biopsych.2005.09.018. [DOI] [PubMed] [Google Scholar]
- Neugebauer NM, Henehan RM, Hales CA, Picciotto MR. Mice lacking the galanin gene show decreased sensitivity to nicotine conditioned place preference. Pharmacol Biochem Behav. 2011;98:87–93. doi: 10.1016/j.pbb.2010.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nieh EH, Kim SY, Namburi P, Tye KM. Optogenetic dissection of neural circuits underlying emotional valence and motivated behaviors. Brain Res. 2013;1511:73–92. doi: 10.1016/j.brainres.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nordstrom O, Melander T, Hokfelt T, Bartfai T, Goldstein M. Evidence for an inhibitory effect of the peptide galanin on dopamine release from the rat median eminence. Neurosci Lett. 1987;73:21–6. doi: 10.1016/0304-3940(87)90024-3. [DOI] [PubMed] [Google Scholar]
- O’Donnell D, Ahmad S, Wahlestedt C, Walker P. Expression of the novel galanin receptor subtype GALR2 in the adult rat CNS: distinct distribution from GALR1. J Comp Neurol. 1999;409:469–81. [PubMed] [Google Scholar]
- O’Neal HA, Van Hoomissen JD, Holmes PV, Dishman RK. Prepro-galanin messenger RNA levels are increased in rat locus coeruleus after treadmill exercise training. Neurosci Lett. 2001;299:69–72. doi: 10.1016/s0304-3940(00)01780-8. [DOI] [PubMed] [Google Scholar]
- Ogbonmwan YE, Sciolino NR, Groves-Chapman JL, Freeman KG, Schroeder JP, Edwards GL, Holmes PV, Weinshenker D. The galanin receptor agonist, galnon, attenuates cocaine-induced reinstatement and dopamine overflow in the frontal cortex. Addict Biol. 2014 doi: 10.1111/adb.12166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogbonmwan YE, Schroeder JP, Holmes PV, Weinshenker D. The effects of post-extinction exercise on cocaine-primed and stress-induced reinstatement of cocaine seeking in rats. Psychopharmacology (Berl) 2015;232:1395–403. doi: 10.1007/s00213-014-3778-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olson VG, Heusner CL, Bland RJ, During MJ, Weinshenker D, Palmiter RD. Role of noradrenergic signaling by the nucleus tractus solitarius in mediating opiate reward. Science. 2006;311:1017–20. doi: 10.1126/science.1119311. [DOI] [PubMed] [Google Scholar]
- Ordway GA, Widdowson PS, Smith KS, Halaris A. Agonist binding to alpha 2-adrenoceptors is elevated in the locus coeruleus from victims of suicide. J Neurochem. 1994;63:617–24. doi: 10.1046/j.1471-4159.1994.63020617.x. [DOI] [PubMed] [Google Scholar]
- Ordway GA, Schenk J, Stockmeier CA, May W, Klimek V. Elevated agonist binding to alpha2-adrenoceptors in the locus coeruleus in major depression. Biol Psychiatry. 2003;53:315–23. doi: 10.1016/s0006-3223(02)01728-6. [DOI] [PubMed] [Google Scholar]
- Ornstein K, Milon H, McRae-Degueurce A, Alvarez C, Berger B, Wurzner HP. Biochemical and radioautographic evidence for dopaminergic afferents of the locus coeruleus originating in the ventral tegmental area. J Neural Transm. 1987;70:183–91. doi: 10.1007/BF01253597. [DOI] [PubMed] [Google Scholar]
- Paladini CA, Williams JT. Noradrenergic inhibition of midbrain dopamine neurons. J Neurosci. 2004;24:4568–75. doi: 10.1523/JNEUROSCI.5735-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park HB, Koh M, Cho SH, Hutchinson B, Lee B. Mapping the rat somatosensory pathway from the anterior cruciate ligament nerve endings to the cerebrum. J Orthop Res. 2005;23:1419–24. doi: 10.1016/j.orthres.2005.03.017.1100230626. [DOI] [PubMed] [Google Scholar]
- Perez SE, Wynick D, Steiner RA, Mufson EJ. Distribution of galaninergic immunoreactivity in the brain of the mouse. J Comp Neurol. 2001;434:158–85. doi: 10.1002/cne.1171. [DOI] [PubMed] [Google Scholar]
- Picciotto MR, Hawes JJ, Brunzell DH, Zachariou V. Galanin can attenuate opiate reinforcement and withdrawal. Neuropeptides. 2005;39:313–5. doi: 10.1016/j.npep.2004.12.001. [DOI] [PubMed] [Google Scholar]
- Picciotto MR. Galanin and addiction. Cell Mol Life Sci. 2008;65:1872–9. doi: 10.1007/s00018-008-8151-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pieribone VA, Xu ZQ, Zhang X, Grillner S, Bartfai T, Hokfelt T. Galanin induces a hyperpolarization of norepinephrine-containing locus coeruleus neurons in the brainstem slice. Neuroscience. 1995;64:861–74. doi: 10.1016/0306-4522(94)00450-j. [DOI] [PubMed] [Google Scholar]
- Pirondi S, Fernandez M, Schmidt R, Hokfelt T, Giardino L, Calza L. The galanin-R2 agonist AR-M1896 reduces glutamate toxicity in primary neural hippocampal cells. J Neurochem. 2005a;95:821–33. doi: 10.1111/j.1471-4159.2005.03437.x. [DOI] [PubMed] [Google Scholar]
- Pirondi S, Kuteeva E, Giardino L, Ferraro L, Antonelli T, Bartfai T, Ogren SO, Hokfelt T, Calza L. Behavioral and neurochemical studies on brain aging in galanin overexpressing mice. Neuropeptides. 2005b;39:305–12. doi: 10.1016/j.npep.2005.02.006. [DOI] [PubMed] [Google Scholar]
- Pudovkina OL, Kawahara Y, de Vries J, Westerink BH. The release of noradrenaline in the locus coeruleus and prefrontal cortex studied with dual-probe microdialysis. Brain Res. 2001;906:38–45. doi: 10.1016/s0006-8993(01)02553-7. [DOI] [PubMed] [Google Scholar]
- Pudovkina OL, Cremers TI, Westerink BH. The interaction between the locus coeruleus and dorsal raphe nucleus studied with dual-probe microdialysis. Eur J Pharmacol. 2002;445:37–42. doi: 10.1016/s0014-2999(02)01663-1. [DOI] [PubMed] [Google Scholar]
- Rajarao SJ, Platt B, Sukoff SJ, Lin Q, Bender CN, Nieuwenhuijsen BW, Ring RH, Schechter LE, Rosenzweig-Lipson S, Beyer CE. Anxiolytic-like activity of the non-selective galanin receptor agonist, galnon. Neuropeptides. 2007;41:307–20. doi: 10.1016/j.npep.2007.05.001. [DOI] [PubMed] [Google Scholar]
- Rasmussen K, Morilak DA, Jacobs BL. Single unit activity of locus coeruleus neurons in the freely moving cat. I. During naturalistic behaviors and in response to simple and complex stimuli. Brain Res. 1986;371:324–34. doi: 10.1016/0006-8993(86)90370-7. [DOI] [PubMed] [Google Scholar]
- Reiss JI, Dishman RK, Boyd HE, Robinson JK, Holmes PV. Chronic activity wheel running reduces the severity of kainic acid-induced seizures in the rat: possible role of galanin. Brain Res. 2009;1266:54–63. doi: 10.1016/j.brainres.2009.02.030. [DOI] [PubMed] [Google Scholar]
- Richardson TG, Minica C, Heron J, Tavare J, MacKenzie A, Day I, Lewis G, Hickman M, Vink JM, Gelernter J, Kranzler HR, Farrer LA, Munafo M, Wynick D. Evaluating the role of a galanin enhancer genotype on a range of metabolic, depressive and addictive phenotypes. Am J Med Genet B Neuropsychiatr Genet. 2014;165B:654–64. doi: 10.1002/ajmg.b.32270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson J, Smith A, Sturchler E, Tabrizifard S, Kamenecka T, McDonald P. Development of a high-throughput screening-compatible cell-based functional assay to identify small molecule probes of the galanin 3 receptor (GalR3) Assay Drug Dev Technol. 2013;11:468–77. doi: 10.1089/adt.2013.526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roeper J. Dissecting the diversity of midbrain dopamine neurons. Trends Neurosci. 2013;36:336–42. doi: 10.1016/j.tins.2013.03.003. [DOI] [PubMed] [Google Scholar]
- Rogan SC, Roth BL. Remote control of neuronal signaling. Pharmacol Rev. 2011;63:291–315. doi: 10.1124/pr.110.003020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rovin ML, Boss-Williams KA, Alisch RS, Ritchie JC, Weinshenker D, West CH, Weiss JM. Influence of chronic administration of antidepressant drugs on mRNA for galanin, galanin receptors, and tyrosine hydroxylase in catecholaminergic and serotonergic cell-body regions in rat brain. Neuropeptides. 2012;46:81–91. doi: 10.1016/j.npep.2012.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Runesson J, Saar I, Lundstrom L, Jarv J, Langel U. A novel GalR2-specific peptide agonist. Neuropeptides. 2009;43:187–92. doi: 10.1016/j.npep.2009.04.004. [DOI] [PubMed] [Google Scholar]
- Saar I, Lahe J, Langel K, Runesson J, Webling K, Jarv J, Rytkonen J, Narvanen A, Bartfai T, Kurrikoff K, Langel U. Novel systemically active galanin receptor 2 ligands in depression-like behavior. J Neurochem. 2013a;127:114–23. doi: 10.1111/jnc.12274. [DOI] [PubMed] [Google Scholar]
- Saar I, Runesson J, Jarv J, Kurrikoff K, Langel U. Novel galanin receptor subtype specific ligand in depression like behavior. Neurochem Res. 2013b;38:398–404. doi: 10.1007/s11064-012-0933-8. [DOI] [PubMed] [Google Scholar]
- Sahraei H, Ghazzaghi H, Zarrindast MR, Ghoshooni H, Sepehri H, Haeri-Rohan A. The role of alpha-adrenoceptor mechanism(s) in morphine-induced conditioned place preference in female mice. Pharmacol Biochem Behav. 2004;78:135–41. doi: 10.1016/j.pbb.2004.03.004. [DOI] [PubMed] [Google Scholar]
- Samuels ER, Szabadi E. Functional neuroanatomy of the noradrenergic locus coeruleus: its roles in the regulation of arousal and autonomic function part I: principles of functional organisation. Curr Neuropharmacol. 2008;6:235–53. doi: 10.2174/157015908785777229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sara SJ, Bouret S. Orienting and reorienting: the locus coeruleus mediates cognition through arousal. Neuron. 2012;76:130–41. doi: 10.1016/j.neuron.2012.09.011. [DOI] [PubMed] [Google Scholar]
- Scheurink AJ, Mundinger TO, Dunning BE, Veith RC, Taborsky GJ., Jr Alpha 2-adrenergic regulation of galanin and norepinephrine release from canine pancreas. Am J Physiol. 1992;262:R819–25. doi: 10.1152/ajpregu.1992.262.5.R819. [DOI] [PubMed] [Google Scholar]
- Schroeder JP, Cooper DA, Schank JR, Lyle MA, Gaval-Cruz M, Ogbonmwan YE, Pozdeyev N, Freeman KG, Iuvone PM, Edwards GL, Holmes PV, Weinshenker D. Disulfiram attenuates drug-primed reinstatement of cocaine seeking via inhibition of dopamine beta-hydroxylase. Neuropsychopharmacology. 2010;35:2440–9. doi: 10.1038/npp.2010.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schroeder JP, Epps SA, Grice TW, Weinshenker D. The selective dopamine beta-hydroxylase inhibitor nepicastat attenuates multiple aspects of cocaine-seeking behavior. Neuropsychopharmacology. 2013;38:1032–8. doi: 10.1038/npp.2012.267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sciolino NR, Dishman RK, Holmes PV. Voluntary exercise offers anxiolytic potential and amplifies galanin gene expression in the locus coeruleus of the rat. Behav Brain Res. 2012;233:191–200. doi: 10.1016/j.bbr.2012.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sciolino NR, Smith JM, Stranahan AM, Freeman KG, Edwards GL, Weinshenker D, Holmes PV. Galanin mediates features of neural and behavioral stress resilience afforded by exercise. Neuropharmacology. 2015;89:255–64. doi: 10.1016/j.neuropharm.2014.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo SW, Shin HY, Kim SH, Han SW, Lee KY, Kim SM, Heo JH. Vestibular imbalance associated with a lesion in the nucleus prepositus hypoglossi area. Arch Neurol. 2004;61:1440–3. doi: 10.1001/archneur.61.9.1440. [DOI] [PubMed] [Google Scholar]
- Seutin V, Verbanck P, Massotte L, Dresse A. Galanin decreases the activity of locus coeruleus neurons in vitro. Eur J Pharmacol. 1989;164:373–6. doi: 10.1016/0014-2999(89)90481-0. [DOI] [PubMed] [Google Scholar]
- Shipley MT, Halloran FJ, de la Torre J. Surprisingly rich projection from locus coeruleus to the olfactory bulb in the rat. Brain Res. 1985;329:294–9. doi: 10.1016/0006-8993(85)90537-2. [DOI] [PubMed] [Google Scholar]
- Shippenberg TS, Thompson AC. Overview of microdialysis. Curr Protoc Neurosci. 2001;Chapter 7(Unit 7):1. doi: 10.1002/0471142301.ns0701s00. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simson PE, Weiss JM. Altered activity of the locus coeruleus in an animal model of depression. Neuropsychopharmacology. 1988;1:287–95. [PubMed] [Google Scholar]
- Sirieix C, Gervasoni D, Luppi PH, Leger L. Role of the lateral paragigantocellular nucleus in the network of paradoxical (REM) sleep: an electrophysiological and anatomical study in the rat. PLoS One. 2012;7:e28724. doi: 10.1371/journal.pone.0028724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith RJ, Aston-Jones G. alpha(2) Adrenergic and imidazoline receptor agonists prevent cue-induced cocaine seeking. Biol Psychiatry. 2011;70:712–9. doi: 10.1016/j.biopsych.2011.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steiner RA, Hohmann JG, Holmes A, Wrenn CC, Cadd G, Jureus A, Clifton DK, Luo M, Gutshall M, Ma SY, Mufson EJ, Crawley JN. Galanin transgenic mice display cognitive and neurochemical deficits characteristic of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2001;98:4184–9. doi: 10.1073/pnas.061445598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steininger TL, Gong H, McGinty D, Szymusiak R. Subregional organization of preoptic area/anterior hypothalamic projections to arousal-related monoaminergic cell groups. J Comp Neurol. 2001;429:638–53. [PubMed] [Google Scholar]
- Stone EA, Lin Y, Sarfraz Y, Quartermain D. Marked behavioral activation from inhibitory stimulation of locus coeruleus alpha1-adrenoceptors by a full agonist. Brain Res. 2009;1291:21–31. doi: 10.1016/j.brainres.2009.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strecker RE, Jacobs BL. Substantia nigra dopaminergic unit activity in behaving cats: effect of arousal on spontaneous discharge and sensory evoked activity. Brain Res. 1985;361:339–50. doi: 10.1016/0006-8993(85)91304-6. [DOI] [PubMed] [Google Scholar]
- Stuber GD, Britt JP, Bonci A. Optogenetic modulation of neural circuits that underlie reward seeking. Biol Psychiatry. 2012;71:1061–7. doi: 10.1016/j.biopsych.2011.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sweerts BW, Jarrott B, Lawrence AJ. Expression of preprogalanin mRNA following acute and chronic restraint stress in brains of normotensive and hypertensive rats. Brain Res Mol Brain Res. 1999;69:113–23. doi: 10.1016/s0169-328x(99)00095-9. [DOI] [PubMed] [Google Scholar]
- Szabadi E. Functional neuroanatomy of the central noradrenergic system. J Psychopharmacol. 2013;27:659–93. doi: 10.1177/0269881113490326. [DOI] [PubMed] [Google Scholar]
- Tully K, Bolshakov VY. Emotional enhancement of memory: how norepinephrine enables synaptic plasticity. Mol Brain. 2010;3:15. doi: 10.1186/1756-6606-3-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tye KM, Mirzabekov JJ, Warden MR, Ferenczi EA, Tsai HC, Finkelstein J, Kim SY, Adhikari A, Thompson KR, Andalman AS, Gunaydin LA, Witten IB, Deisseroth K. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature. 2013;493:537–41. doi: 10.1038/nature11740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Unschuld PG, Ising M, Roeske D, Erhardt A, Specht M, Kloiber S, Uhr M, Muller-Myhsok B, Holsboer F, Binder EB. Gender-specific association of galanin polymorphisms with HPA-axis dysregulation, symptom severity, and antidepressant treatment response. Neuropsychopharmacology. 2010;35:1583–92. doi: 10.1038/npp.2010.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valentino RJ, Pavcovich LA, Hirata H. Evidence for corticotropin-releasing hormone projections from Barrington’s nucleus to the periaqueductal gray and dorsal motor nucleus of the vagus in the rat. J Comp Neurol. 1995;363:402–22. doi: 10.1002/cne.903630306. [DOI] [PubMed] [Google Scholar]
- Van Bockstaele EJ, Akaoka H, Aston-Jones G. Brainstem afferents to the rostral (juxtafacial) nucleus paragigantocellularis: integration of exteroceptive and interoceptive sensory inputs in the ventral tegmentum. Brain Res. 1993;603:1–18. doi: 10.1016/0006-8993(93)91293-2. [DOI] [PubMed] [Google Scholar]
- Van Bockstaele EJ, Colago EE, Valentino RJ. Amygdaloid corticotropin-releasing factor targets locus coeruleus dendrites: substrate for the coordination of emotional and cognitive limbs of the stress response. J Neuroendocrinol. 1998;10:743–57. doi: 10.1046/j.1365-2826.1998.00254.x. [DOI] [PubMed] [Google Scholar]
- Van Bockstaele EJ, Menko AS, Drolet G. Neuroadaptive responses in brainstem noradrenergic nuclei following chronic morphine exposure. Mol Neurobiol. 2001;23:155–71. doi: 10.1385/mn:23:2-3:155. [DOI] [PubMed] [Google Scholar]
- Van Hoomissen JD, Holmes PV, Zellner AS, Poudevigne A, Dishman RK. Effects of beta-adrenoreceptor blockade during chronic exercise on contextual fear conditioning and mRNA for galanin and brain-derived neurotrophic factor. Behav Neurosci. 2004;118:1378–90. doi: 10.1037/0735-7044.118.6.1378. [DOI] [PubMed] [Google Scholar]
- Vazey EM, Aston-Jones G. Designer receptor manipulations reveal a role of the locus coeruleus noradrenergic system in isoflurane general anesthesia. Proc Natl Acad Sci U S A. 2014;111:3859–64. doi: 10.1073/pnas.1310025111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Velasquez-Martinez MC, Vazquez-Torres R, Jimenez-Rivera CA. Activation of alpha1-adrenoceptors enhances glutamate release onto ventral tegmental area dopamine cells. Neuroscience. 2012;216:18–30. doi: 10.1016/j.neuroscience.2012.03.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Velasquez-Martinez MC, Vazquez-Torres R, Rojas LV, Sanabria P, Jimenez-Rivera CA. Alpha-1 adrenoreceptors modulate GABA release onto ventral tegmental area dopamine neurons. Neuropharmacology. 2015;88:110–21. doi: 10.1016/j.neuropharm.2014.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ventura R, Alcaro A, Puglisi-Allegra S. Prefrontal cortical norepinephrine release is critical for morphine-induced reward, reinstatement and dopamine release in the nucleus accumbens. Cereb Cortex. 2005;15:1877–86. doi: 10.1093/cercor/bhi066. [DOI] [PubMed] [Google Scholar]
- Ventura R, Morrone C, Puglisi-Allegra S. Prefrontal/accumbal catecholamine system determines motivational salience attribution to both reward- and aversion-related stimuli. Proc Natl Acad Sci U S A. 2007;104:5181–6. doi: 10.1073/pnas.0610178104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verhage M, McMahon HT, Ghijsen WE, Boomsma F, Scholten G, Wiegant VM, Nicholls DG. Differential release of amino acids, neuropeptides, and catecholamines from isolated nerve terminals. Neuron. 1991;6:517–24. doi: 10.1016/0896-6273(91)90054-4. [DOI] [PubMed] [Google Scholar]
- Vila-Porcile E, Xu ZQ, Mailly P, Nagy F, Calas A, Hokfelt T, Landry M. Dendritic synthesis and release of the neuropeptide galanin: morphological evidence from studies on rat locus coeruleus neurons. J Comp Neurol. 2009;516:199–212. doi: 10.1002/cne.22105. [DOI] [PubMed] [Google Scholar]
- Vranjkovic O, Gasser PJ, Gerndt CH, Baker DA, Mantsch JR. Stress-induced cocaine seeking requires a beta-2 adrenergic receptor-regulated pathway from the ventral bed nucleus of the stria terminalis that regulates CRF actions in the ventral tegmental area. J Neurosci. 2014;34:12504–14. doi: 10.1523/JNEUROSCI.0680-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Webling KE, Runesson J, Bartfai T, Langel U. Galanin receptors and ligands. Front Endocrinol (Lausanne) 2012;3:146. doi: 10.3389/fendo.2012.00146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wee S, Mandyam CD, Lekic DM, Koob GF. Alpha 1-noradrenergic system role in increased motivation for cocaine intake in rats with prolonged access. Eur Neuropsychopharmacol. 2008;18:303–11. doi: 10.1016/j.euroneuro.2007.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinshenker D, Rust NC, Miller NS, Palmiter RD. Ethanol-associated behaviors of mice lacking norepinephrine. J Neurosci. 2000;20:3157–64. doi: 10.1523/JNEUROSCI.20-09-03157.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weinshenker D, Szot P. The role of catecholamines in seizure susceptibility: new results using genetically engineered mice. Pharmacol Ther. 2002;94:213–33. doi: 10.1016/s0163-7258(02)00218-8. [DOI] [PubMed] [Google Scholar]
- Weinshenker D, Schroeder JP. There and back again: a tale of norepinephrine and drug addiction. Neuropsychopharmacology. 2007;32:1433–51. doi: 10.1038/sj.npp.1301263. [DOI] [PubMed] [Google Scholar]
- Weiss JM, Simson PG, Hoffman LJ, Ambrose MJ, Cooper S, Webster A. Infusion of adrenergic receptor agonists and antagonists into the locus coeruleus and ventricular system of the brain. Effects on swim-motivated and spontaneous motor activity. Neuropharmacology. 1986;25:367–84. doi: 10.1016/0028-3908(86)90231-5. [DOI] [PubMed] [Google Scholar]
- Weiss JM, Demetrikopoulos MK, West CHK, Bonsall RW. Hypothesis linking the noradrenergic and dopaminergic systems in depression. Depression. 1996;3:225–245. [Google Scholar]
- Weiss JM, Bonsall RW, Demetrikopoulos MK, Emery MS, West CH. Galanin: a significant role in depression? Ann N Y Acad Sci. 1998;863:364–82. doi: 10.1111/j.1749-6632.1998.tb10707.x. [DOI] [PubMed] [Google Scholar]
- Weiss JM, Boss-Williams KA, Moore JP, Demetrikopoulos MK, Ritchie JC, West CH. Testing the hypothesis that locus coeruleus hyperactivity produces depression-related changes via galanin. Neuropeptides. 2005;39:281–7. doi: 10.1016/j.npep.2004.12.028. [DOI] [PubMed] [Google Scholar]
- Wess J, Nakajima K, Jain S. Novel designer receptors to probe GPCR signaling and physiology. Trends Pharmacol Sci. 2013;34:385–92. doi: 10.1016/j.tips.2013.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West CH, Ritchie JC, Boss-Williams KA, Weiss JM. Antidepressant drugs with differing pharmacological actions decrease activity of locus coeruleus neurons. Int J Neuropsychopharmacol. 2009;12:627–41. doi: 10.1017/S1461145708009474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West CH, Ritchie JC, Weiss JM. Addendum: Paroxetine-induced increase in activity of locus coeruleus neurons in adolescent rats: implication of a countertherapeutic effect of an antidepressant. Neuropsychopharmacology. 2010a;35:1836–7. doi: 10.1038/npp.2010.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West CH, Ritchie JC, Weiss JM. Paroxetine-induced increase in activity of locus coeruleus neurons in adolescent rats: implication of a countertherapeutic effect of an antidepressant. Neuropsychopharmacology. 2010b;35:1653–63. doi: 10.1038/npp.2010.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- West CH, Weiss JM. Effects of chronic antidepressant drug administration and electroconvulsive shock on activity of dopaminergic neurons in the ventral tegmentum. Int J Neuropsychopharmacol. 2011;14:201–10. doi: 10.1017/S1461145710000489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White FJ, Wang RY. Pharmacological characterization of dopamine autoreceptors in the rat ventral tegmental area: microiontophoretic studies. J Pharmacol Exp Ther. 1984;231:275–80. [PubMed] [Google Scholar]
- Witten IB, Steinberg EE, Lee SY, Davidson TJ, Zalocusky KA, Brodsky M, Yizhar O, Cho SL, Gong S, Ramakrishnan C, Stuber GD, Tye KM, Janak PH, Deisseroth K. Recombinase-driver rat lines: tools, techniques, and optogenetic application to dopamine-mediated reinforcement. Neuron. 2011;72:721–33. doi: 10.1016/j.neuron.2011.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong ML, Kling MA, Munson PJ, Listwak S, Licinio J, Prolo P, Karp B, McCutcheon IE, Geracioti TD, Jr, DeBellis MD, Rice KC, Goldstein DS, Veldhuis JD, Chrousos GP, Oldfield EH, McCann SM, Gold PW. Pronounced and sustained central hypernoradrenergic function in major depression with melancholic features: relation to hypercortisolism and corticotropin-releasing hormone. Proc Natl Acad Sci U S A. 2000;97:325–30. doi: 10.1073/pnas.97.1.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wray NR, Pergadia ML, Blackwood DH, Penninx BW, Gordon SD, Nyholt DR, Ripke S, MacIntyre DJ, McGhee KA, Maclean AW, Smit JH, Hottenga JJ, Willemsen G, Middeldorp CM, de Geus EJ, Lewis CM, McGuffin P, Hickie IB, van den Oord EJ, Liu JZ, Macgregor S, McEvoy BP, Byrne EM, Medland SE, Statham DJ, Henders AK, Heath AC, Montgomery GW, Martin NG, Boomsma DI, Madden PA, Sullivan PF. Genome-wide association study of major depressive disorder: new results, meta-analysis, and lessons learned. Mol Psychiatry. 2012;17:36–48. doi: 10.1038/mp.2010.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wulff P, Arenkiel BR. Chemical genetics: receptor-ligand pairs for rapid manipulation of neuronal activity. Curr Opin Neurobiol. 2012;22:54–60. doi: 10.1016/j.conb.2011.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu ZQ, Shi TJ, Hokfelt T. Galanin/GMAP- and NPY-like immunoreactivities in locus coeruleus and noradrenergic nerve terminals in the hippocampal formation and cortex with notes on the galanin-R1 and -R2 receptors. J Comp Neurol. 1998a;392:227–51. doi: 10.1002/(sici)1096-9861(19980309)392:2<227::aid-cne6>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
- Xu ZQ, Zhang X, Pieribone VA, Grillner S, Hokfelt T. Galanin-5-hydroxytryptamine interactions: electrophysiological, immunohistochemical and in situ hybridization studies on rat dorsal raphe neurons with a note on galanin R1 and R2 receptors. Neuroscience. 1998b;87:79–94. doi: 10.1016/s0306-4522(98)00151-1. [DOI] [PubMed] [Google Scholar]
- Xu ZQ, Zheng K, Hokfelt T. Electrophysiological studies on galanin effects in brain--progress during the last six years. Neuropeptides. 2005;39:269–75. doi: 10.1016/j.npep.2005.02.003. [DOI] [PubMed] [Google Scholar]
- Yanpallewar SU, Fernandes K, Marathe SV, Vadodaria KC, Jhaveri D, Rommelfanger K, Ladiwala U, Jha S, Muthig V, Hein L, Bartlett P, Weinshenker D, Vaidya VA. Alpha2-adrenoceptor blockade accelerates the neurogenic, neurotrophic, and behavioral effects of chronic antidepressant treatment. J Neurosci. 2010;30:1096–109. doi: 10.1523/JNEUROSCI.2309-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshitake S, Kuteeva E, Hokfelt T, Mennicken F, Theodorsson E, Yamaguchi M, Kehr J, Yoshitake T. Correlation between the effects of local and intracerebroventricular infusions of galanin on 5-HT release studied by microdialysis, and distribution of galanin and galanin receptors in prefrontal cortex, ventral hippocampus, amygdala, hypothalamus, and striatum of awake rats. Synapse. 2014;68:179–93. doi: 10.1002/syn.21730. [DOI] [PubMed] [Google Scholar]
- Zachariou V, Parikh K, Picciotto MR. Centrally administered galanin blocks morphine place preference in the mouse. Brain Res. 1999;831:33–42. doi: 10.1016/s0006-8993(99)01476-6. [DOI] [PubMed] [Google Scholar]
- Zachariou V, Thome J, Parikh K, Picciotto MR. Upregulation of galanin binding sites and GalR1 mRNA levels in the mouse locus coeruleus following chronic morphine treatments and precipitated morphine withdrawal. Neuropsychopharmacology. 2000;23:127–37. doi: 10.1016/S0893-133X(00)00094-4. [DOI] [PubMed] [Google Scholar]
- Zachariou V, Brunzell DH, Hawes J, Stedman DR, Bartfai T, Steiner RA, Wynick D, Langel U, Picciotto MR. The neuropeptide galanin modulates behavioral and neurochemical signs of opiate withdrawal. Proc Natl Acad Sci U S A. 2003;100:9028–33. doi: 10.1073/pnas.1533224100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zarrindast MR, Bahreini T, Adl M. Effect of imipramine on the expression and acquisition of morphine-induced conditioned place preference in mice. Pharmacol Biochem Behav. 2002;73:941–9. doi: 10.1016/s0091-3057(02)00951-6. [DOI] [PubMed] [Google Scholar]
- Zhang XY, Kosten TA. Prazosin, an alpha-1 adrenergic antagonist, reduces cocaine-induced reinstatement of drug-seeking. Biol Psychiatry. 2005;57:1202–4. doi: 10.1016/j.biopsych.2005.02.003. [DOI] [PubMed] [Google Scholar]
- Zhang XY, Kosten TA. Previous exposure to cocaine enhances cocaine self-administration in an alpha 1-adrenergic receptor dependent manner. Neuropsychopharmacology. 2007;32:638–45. doi: 10.1038/sj.npp.1301120. [DOI] [PubMed] [Google Scholar]
- Zhou Y, Mabrouk OS, Kennedy RT. Rapid preconcentration for liquid chromatography-mass spectrometry assay of trace level neuropeptides. J Am Soc Mass Spectrom. 2013;24:1700–9. doi: 10.1007/s13361-013-0605-1. [DOI] [PMC free article] [PubMed] [Google Scholar]

