REVIEW ARTICLE published: 09 September 2013 doi: 10.3389/fphar.2013.00110 Reduced brain in mood disorders: a common pathophysiological substrate and drug target?

Li-Chun Lin and Etienne Sibille* Department of Psychiatry, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA

Edited by: Our knowledge of the pathophysiology of affect dysregulation has progressively increased, Thibault Renoir, Florey Institute of but the pharmacological treatments remain inadequate. Here, we summarize the cur- Neuroscience and Mental Health, Australia rent literature on deficits in somatostatin, an inhibitory modulatory neuropeptide, in major depression and other neurological disorders that also include mood disturbances. Reviewed by: Jean-Philippe Guilloux,Université We focus on direct evidence in the human postmortem brain, and review rodent Paris Sud, France genetic and pharmacological studies probing the role of the somatostatin system in Jacques Epelbaum, Université Paris relation to mood. We also briefly go over pharmacological developments targeting the Descartes, France Elif Engin, McLean Hospital/Harvard somatostatin system in peripheral organs and discuss the challenges of targeting the Medical School, USA brain somatostatin system. Finally, the fact that somatostatin deficits are frequently *Correspondence: observed across neurological disorders suggests a selective cellular vulnerability of Etienne Sibille, Department of somatostatin-expressing neurons. Potential cell intrinsic factors mediating those changes Psychiatry, Center for Neuroscience, are discussed, including nitric oxide induced oxidative stress, mitochondrial dysfunc- University of Pittsburgh, Bridgeside Point II, 450 Technology Drive, Suite tion, high inflammatory response, high demand for neurotrophic environment, and 231, Pittsburgh, PA 15219, USA overall aging processes. Together, based on the co-localization of somatostatin with e-mail: [email protected] gamma-aminobutyric acid (GABA), its presence in dendritic-targeting GABA neuron sub- types, and its temporal-specific function, we discuss the possibility that deficits in somatostatin play a central role in cortical local inhibitory circuit deficits leading to abnormal corticolimbic network activity and clinical mood symptoms across neurological disorders. View metadata, citation and similar papers at core.ac.uk brought to you by CORE

Keywords: somatostatin, somatostatin-expressing interneurons, SST, SOM, SRIF, depression, mood disorders, provided by Frontiers - Publisher Connector GABA inhibition

INTRODUCTION This article highlights current findings on the functional roles of Mood disturbances are commonly observed in many neuro- somatostatin in local neuronal circuits, and reviews somatostatin logical disorders. The chronic, recurrent and long duration deficits across neurological disorders, including neuropsychi- of mood disturbances not only place an enormous emotional atric disorders [e.g., major depressive disorder (MDD), bipolar and financial burden on patients, but also on their fami- disorder, schizophrenia], and neurodegenerative disorders (e.g., lies and society. Nearly 10% of all primary care office visits Parkinson’s, Alzheimer’s, and Huntington’s diseases; Table 1). are depression-related (Stafford et al., 2000), but only 30% of This raises interesting questions, including first; whether the patients with mood disturbances achieve remission with ini- somatostatin deficits observed in neurological disorders repre- tial treatment (Trivedi et al., 2006). Somatostatin is a pep- sent common, distinct, or partly overlapping mechanisms of tide expressed in multiple organs. In the brain, somatostatin symptoms across disorders and, second, what may be the causes (also known as somatotrophin release inhibiting factor and and biological mechanisms underlying the selective neuronal vul- often abbreviated as SST, SRIF, or SOM) acts as a modulatory nerability of somatostatin-expressing neurons. In addition, we and inhibitory neuropeptide that is co-localized with gamma- review somatostatin findings associated with affect regulation aminobutyric acid (GABA), and that is involved in regulating at the genetic, cellular, and pharmacological levels in animal multiple aspects of physiological and behavioral stress responses, studies. So far, these findings suggest that somatostatin deficits including inhibition of hypothalamic release, amyg- across different brain systems and diseases may play a central dala central nucleus output, and cortical local circuit integra- role in the affective symptom dimension rather than non-specific tion of sensory input. Research advances over the past three signals in neurological disorders (Figure 1). As somatostatin decades suggest a critical role for somatostatin in the patho- itself is not an ideal drug target, including for antidepres- physiology of mood disorders, and potential new therapeutic sant effect, we suggest that further studies characterizing the strategies. Several recent reviews have summarized the role of the intrinsic properties and biological vulnerabilities of somatostatin- somatostatin system, including in receptor subtypes (Patel, 1999; expressing neurons, may identify novel targets with implications Csaba and Dournaud, 2001), pharmacological developments for understanding the function of local cell circuits and brain (Neggers and van der Lely, 2009), and during normal and patho- regions underlying affective symptoms across several neurological logical aging (Patel, 1999; Viollet et al., 2008; Martel et al., 2012). disorders.

www.frontiersin.org September 2013 | Volume 4 | Article 110 | 1

“fphar-04-00110” — 2013/9/5 — 15:44 — page1—#1 Lin and Sibille Somatostatin deficits in mood disorders

Table 1 | Low somatostatin in human neurological disorders.

Neurological disorders Brain region Pathological findings Reference

Major depressive disorder CSF Decreased Agren and Lundqvist (1984), Kling et al. (1993), Molchan et al. (1993) Dorsolateral prefrontal cortex Decreased (RNA expression) Sibille et al. (2011) Anterior cingulate cortex Decreased (RNA expression) Tripp et al. (2011), Tripp et al. (2012) Amygdala Decreased (RNA and protein expression) Guilloux et al. (2012) Schizophrenia CSF Decreased Bissette et al. (1986), Reinikainen et al. (1990) Dorsolateral prefrontal cortex Decreased (RNA expression) Morris et al. (2008), Guillozet-Bongaarts et al. (2013) Hippocampus Decreased (neuron number and density) Konradi et al. (2011a) Caudal entorhinal cortex Decreased (neuron number and density) Wang et al. (2011) Parasubiculum Decreased (neuron number and density) Wang et al. (2011) Bipolar disorder Caudal entorhinal cortex Decreased (neuron density) Wang et al. (2011) Parasubiculum Decreased (neuron density) Wang et al. (2011) Hippocampus Decreased (neuron number and RNA expression) Konradi et al. (2011b) Dorsolateral prefrontal cortex Decreased (RNA expression; trend level) Sibille et al. (2011) Alzheimer’s disease CSF Decreased Bissette et al. (1986); Tamminga et al. (1987) Temporal cortex Decreased (immune-reactivity) Rossor et al. (1980); Candy et al. (1985) Frontal cortex Decreased (immune-reactivity) Davies and Terry (1981); Candy et al. (1985) Hippocampus Decreased (gene expression per cell) Dournaud et al. (1994) Parahippocampal cortex Decreased (neuronal density) Dournaud et al. (1994) Parkinson’s disease CSF Decreased Dupont et al. (1982) Frontal cortex Decreased (radioimmune-reactivity) Epelbaum et al. (1988) Others Temporal cortex Decreased (immune-reactivity) Beal et al. (1986)

LOW SOMATOSTATIN IN NEUROPSYCHIATRIC AND dorsolateral prefrontal cortex (dlPFC), subgenual anterior cingu- NEURODEGENERATIVE DISORDERS late cortex (sgACC), and amygdala (Sibille et al., 2011; Tripp et al., MAJOR DEPRESSIVE DISORDER 2011, 2012; Guilloux et al., 2012). In addition, two peptides co- Patients with major depressive disorder (MDD) show decreased localized with somatostatin, neuropeptide Y and cortistatin, are somatostatin levels in the cerebrospinal fluid (CSF; Agren and both significantly down-regulated in MDD patients (Tripp et al., Lundqvist, 1984; Molchan et al., 1991; Kling et al., 1993), and tran- 2011, 2012).These three neuropeptides (somatostatin, neuropep- siently decreased CSF somatostatin which normalize with recovery tide Y, and cortistatin) are markers of GABAergic neurons that in MDD (Rubinow et al., 1985; Post et al., 1988). Evidence for low specifically target the dendritic compartment of pyramidal cells levels of CSF somatostatin was found to correlate significantly (de Lecea et al., 1997; Viollet et al., 2008), and that are essential with elevated urinary cortisol in MDD patients (Molchan et al., in gating incoming sensory information (Figure 1). Other types 1993). This is consistent with the altered hypothalamic-pituitary- of GABAergic cell markers, such as parvalbumin and cholecys- adrenal (HPA) axis function described in some depressed patients tokinin, are mostly not affected by MDD (although see Tripp et al., (Holsboer, 2000). The route and characterization, however, from 2012). Interestingly, these somatostatin deficits were systematically CSF somatostatin to MDD pathophysiology is not direct, poten- more robust in female subjects across cohorts and regions (Sibille tially due to a paucity of information on factors regulating CSF et al., 2011; Tripp et al., 2011, 2012; Guilloux et al., 2012), consis- somatostatin, and to inconclusive somatostatin/HPA axis studies tent with the female heightened vulnerability to develop MDD, in MDD patients. Hence, despite these early findings, interest in and suggesting that low somatostatin may represent a molec- somatostatin in mood disorders has declined over time. ular correlate of sexual dimorphism in vulnerability to affect Human post-mortem studies from our group have described dysregulation. Notably, these findings are also consistent with region-specific somatostatin deficits in MDD patients, includ- earlier postmortem studies showing reduced calbindin-positive ing a down-regulation of somatostatin gene expression in the cell numbers in MDD (Rajkowska et al., 2007; Maciag et al.,

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 110 | 2

“fphar-04-00110” — 2013/9/5 — 15:44 — page2—#2 Lin and Sibille Somatostatin deficits in mood disorders

FIGURE 1 | Schematic of somatostatin signaling, pathological regulators expressing interneurons are key conduits for regulating incoming information and biological functions relevant to affect regulation. Somatostatin and pyramidal cell function. In contrast, other GABA neurons subtypes pathway activity is responsive to (left panel), and regulates (right panel), targeting the perisomatic pyramidal cell compartment are mostly not affected several biological events, and molecular and cellular properties that have in major depression. NPY, neuropeptide Y; PYR, pyramidal neuron; PV, been linked to mood disturbances. Somatostatin and somatostatin- parvalbumin.

2010), as somatostatin is mostly expressed in a subgroup of 2011b). In addition, patients with bipolar disorder show elevated calbindin-positive cells (reviewed in Viollet et al., 2008). Converg- CSF somatostatin during manic states (Sharma et al., 1995). ing evidence from down-regulation of somatostatin co-localized GABA markers in MDD across multiple human post-mortem NEURODEGENERATIVE DISORDERS studies suggests that this particular GABA subpopulation in the Alzheimer’s disease is a neurodegenerative disease with neu- forebrain is selectively vulnerable, among other subtypes of GABA ropsychiatric symptoms (Bungener et al., 1996). Decreased CSF neurons. Furthermore, these local cell circuit-based findings intro- somatostatin (Bissette et al., 1986; Tamminga et al., 1987) and duce a new role for somatostatin in depression, which is distinct decreased somatostatin immune-reactivity across cortical and from its previously investigated role in the regulation of the HPA subcortical regions is reported in subjects with Alzheimer’s dis- axis (Rubinow et al., 1983; Molchan et al., 1993; Weckbecker et al. ease, including temporal cortex, frontal cortex, and hippocampus 2003). (Davies et al., 1980; Rossor et al., 1980; Davies and Terry, 1981; Candy et al., 1985; Dournaud et al., 1994). Depression is a com- OTHER NEUROPSYCHIATRIC DISORDERS mon comorbid symptom in Parkinson’s disease and predicts Schizophrenia is a neuropsychiatric disorder characterized by greater disability at any assessment point (Aarsland et al., 1999). positive (e.g., hallucination), negative symptoms (e.g., emo- Decreased CSF somatostatin, decreased somatostatin immuno- tional blunting, apathy) and cognitive symptoms. Somatostatin reactivity, and binding sites are also observed in the temporal deficits in schizophrenia are demonstrated by a reduction of cortex and frontal cortex of patients with Parkinson’s disease (Beal CSF somatostatin (Bissette et al., 1986; Reinikainen et al., 1990), et al., 1986; Epelbaum et al., 1988). Notably, reduced CSF somato- decreased somatostatin gene expression in the dlPFC (Mor- statin in Parkinson’s disease appears to be irreversibly present at ris et al., 2008; Guillozet-Bongaarts et al., 2013), and decreased the onset of symptoms (Dupont et al., 1982). number and density of somatostatin-expressing neurons in the hippocampus (Konradi et al., 2011a), caudal entorhinal cortex REDUCED SOMATOSTATIN AND LOW MOOD? and parasubiculum (Wang et al., 2011). Changes in somatostatin The evidence outlined in this review provides only a glimpse of the are also identified in bipolar disorder, which is clinically char- potential full range of somatostatin deficits across neurological dis- acterized by fluctuating mood. Studies in subjects with bipolar orders, as multiple other brain regions and disease categories await disorder indicate decreases in somatostatin cellular density in the further characterization (Table 1). Taken together, the cumula- caudal entorhinal cortex and parasubiculum (Wang et al., 2011), tive evidence demonstrates that somatostatin deficits are common number of somatostatin-expressing neurons in the hippocampus neurochemical and molecular features in individuals with neuro- (Konradi et al., 2011b), somatostatin gene expression in the dlPFC logical disorders, regardless of their categorical diagnosis. While (trend level; Sibille et al., 2011) and hippocampus (Konradi et al., somatostatin studies of cell number and gene expression in human

www.frontiersin.org September 2013 | Volume 4 | Article 110 | 3

“fphar-04-00110” — 2013/9/5 — 15:44 — page3—#3 Lin and Sibille Somatostatin deficits in mood disorders

postmortem brains suggest a specific alteration of somatostatin- activation of inhibitory G protein (Gi) and the following inhi- positive neurons across neurological disorders, it is possible bition of adenylyl cyclase, leading to reduction of cAMP levels, that changes and dys-synchronization of additional components activation of phosphotyrosine phosphatases, and modulation of of local neuronal circuits contribute to a common symptom mitogen-activated protein kinases and phospholipase C (Koch and dimension, which we speculate includes low affect and mood dys- Schonbrunn, 1984; Koch et al., 1988). regulation. Hence, this review is not comprehensive, but rather, Sst1−5 present different patterns of coexpression in the brain highlights the recent findings in brain somatostatin signaling and (Kluxen et al., 1992; Moller et al., 2003; reviewed in Martel et al., the potential role of somatostatin deficits in affect dysregula- 2012). Sst1 is found in retina, basal ganglia and hypothalamus, tion for integrating categorical models of mood symptoms into Sst2 is highly abundant in several telencephalic structures (neo- a dimensional model across neurological disorders. cortex, hippocampus, and amygdala), Sst3 immunoreactivity has only been described in neuronal cilia (Schulz et al., 2000), Sst4 SOMATOSTATIN: GENES, NEURONS AND PHARMACOLOGY is expressed in olfactory bulb, cerebral cortex and CA1 region of SOMATOSTATIN SIGNALING the hippocampus (Schreff et al., 2000), and expression of Sst5 has Somatostatin is a modulatory neuropeptide that synergizes with been detected in cerebral cortex, hippocampus, amygdala, pre- GABA-mediated inhibition, and that specifically targets the optic area, and hypothalamus (Stroh et al., 1999; Strowski et al., distal dendritic compartment of pyramidal neurons in corti- 2003; Olias et al., 2004). Interestingly, when co-expressed in the cal local circuits (Kawaguchi and Kubota, 1997; Gentet et al., same cells, Sst5 influences Sst2 internalization and trafficking and 2012). Somatostatin inhibits release of numerous from modulates cellular desensitization to the effects of somatostatin-14 the hypothalamus, including corticotrophin releasing hormone (Sharif et al., 2007), suggesting that the precise actions of somato- (CRH;Wanget al.,1987; Patel,1999). The somatostatin gene prod- statin depend on the specific interaction of the Sst1−5 receptors uct is composed of 14 or 28 amino-acid residues. Both forms of expressed locally in each brain region. somatostatin, somatostatin-14 and somatostatin-28, are generated by tissue-specific post-translational processing of the 116 amino- GENETIC POLYMORPHISMS IN THE SOMATOSTATIN SYSTEM acid pre-pro-somatostatin peptide (Warren and Shields, 1984; The relatively high degree of amino acid conservation across Tostivint et al.,2008). Somatostatin-14 is predominantly produced species indicates that somatostatin-related genes have been in the central nervous system (CNS) but also in many peripheral highly constrained during evolution (Patel, 1999; Olias et al., organs (Epelbaum, 1986). Somatostatin-28 is mainly synthesized 2004). Accordingly, there are currently very few reports linking along the gastrointestinal tract (Fitz-Patrick and Patel, 1981). The somatostatin gene polymorphisms with neurological disorders. 5-upstream sequence of the somatostatin gene contains cyclic- A primate-specific single nucleotide polymorphism (SNP) in AMP response element (CRE; Montminy et al., 1986), making the human somatostatin gene [C/T polymorphism (rs4988514)] its expression activity-dependent. Thus, somatostatin expression is associated with increased risk in Alzheimer’s disease pro- is preferentially altered by various stressors, such as seizures gression and additive effect with the APOE epsilon4 allele (Vezzani and Hoyer, 1999; Tallent and Qiu, 2008) and electri- (Vepsalainen et al., 2007; Xue et al., 2009), although this was not cal foot shock (Ponomarev et al., 2010). Moreover, mice with confirmed in larger genome-wide association studies (GWAS) conditional homozygous and constitutive heterozygous brain- (Hollingworth et al., 2011; Guerreiro et al., 2013). Leu48Met and derived neurotrophic factor (Bdnf) knockout or disruption of Pro335Leu SNPs in the SST5 gene are of potential significance to exon IV-expressing Bdnf transcripts show decreased somatostatin patients with bipolar disorder (Nyegaard et al., 2002), but no asso- gene expression (Glorioso et al., 2006; Martinowich et al., 2011; ciations of SST5 SNPs are found in patients with autism (Lauritsen Guilloux et al., 2012), demonstrating that somatostatin expression et al., 2003). The paucity of associations with somatostatin gene depends on Bdnf signaling. However, the molecular mechanisms variants is surprising and may reflect either strong negative selec- by which this neurotrophic factor controls somatostatin and tion against genetic variations in this gene, or alternatively, dilution somatostatin-expressing neurons are still unknown. of signal due to heterogeneity of DSM-IV-based cohorts in genetic Somatostatin, cortistatin and their receptors are closely inter- association studies. So, dimensional phenotypes, as defined by twined systems (de Lecea et al., 1996, 1997; reviewed in Spier and clusters of mood symptoms, which are closer to gene functions de Lecea, 2000; de Lecea, 2008). Sharing high structural homology may have implications for future genetic studies of somatostatin with somatostatin, cortistatin binds to all somatostatin receptor and other genes. subtypes and is known to be regulated by exon IV-expressing Bdnf transcripts (Martinowich et al., 2011). However, distinct SOMATOSTATIN-EXPRESSING NEURONS: DIVERSITY AND ROLES from somatostatin, cortistatin binds to additional receptors (e.g., Gamma-aminobutyric acid (GABA) neurons are a diverse group secretagogue receptor 1a and Mas-related gene of inhibitory cells which co-release neuropeptides in order to sup- X2 receptor) (Robas et al., 2003; Siehler et al., 2008) and has port a fine-tuning of neuronal signaling and architecture. The local different physiological properties (e.g., activation of cation selec- inhibitory circuits provide spatiotemporal control of information tive currents not responsive to somatostatin; Spier and de Lecea, processing through at least 20 subtypes of cortical GABA neurons, 2000), suggesting that somatostatin and cortistatin may both con- which are based on their expression of different calcium binding tribute to affect regulation in an integrated, yet differential mode. proteins and neuropeptides, localization, targeting, and differ- The intracellular pathway of somatostatin signaling coupled to ential electrophysiological properties. Recent detailed reviews on all five somatostatin receptors subtypes (Sst1−5) is through the GABA neuron subpopulations have been published (Csaba and

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 110 | 4

“fphar-04-00110” — 2013/9/5 — 15:44 — page4—#4 Lin and Sibille Somatostatin deficits in mood disorders

Dournaud, 2001; Di Cristo et al., 2004; Markram et al., 2004; Tan demonstrated that SstKO mice are healthy, fertile, and show no et al., 2008; Fishell and Rudy, 2011; Gentet et al., 2012; DeFelipe overt behavioral phenotypes, including anxiety-like behavior in et al., 2013; Le Magueresse and Monyer, 2013). Approximately 20– the open-field and fear conditioning tests. Notably, SstKO mice 30% of GABA neurons in the mouse somatosensory cortex express display high basal plasma levels of corticosterone and growth hor- somatostatin (Lee et al., 2010; Rudy et al., 2011), and 40–50% of mone (Zeyda et al., 2001), confirming a somatostatin-mediated GABA neurons contain parvalbumin without overlapping with inhibition of HPA axis function. Similarly, mice lacking indi- somatostatin in the frontal cortex, primary somatosensory cortex vidual Sst1−5 receptors have been tested in numerous biological and visual cortex of mouse (Gonchar et al., 2007; Xu et al., 2010) fields. Of these, Sst2 emerged as the primary receptor of inter- KO and the visual cortex of rat (Gonchar and Burkhalter, 1997). est (Zeyda and Hochgeschwender, 2008), and Sst2 mice display Recent reports focusing on the patterns of cortical neu- increased anxiety-like behavior in the elevated plus maze and open ronal connectivity show that somatostatin-expressing interneu- field, increased immobility in the forced swim test, decreased loco- rons mediate the firing of pyramidal neurons with a fine level motion coupled with an increase of pituitary adrenocorticotropic of specificity among cortical layers. Integrating optogenetic and hormone release instead of growth hormone (Viollet et al., 2000). KO electrophysiology approaches, mouse somatostatin-expressing In line with the observed changes in Sst2 mice, acute predator interneurons in layer 2/3 of the somatosensory cortex provide stress in rats led to up-regulated Sst2 gene expression in the amyg- a tonic inhibition to the distal dendrites of excitatory pyramidal dala and cingulate cortex, shown correlated with Fos expression neurons by sharpening selectivity during periods of quiet wake- in the amygdala (Nanda et al., 2008). As the product of a different fulness, which may contribute to synchronized firing in cortical gene, cortistatin shares a high structural and functional similar- networks and sensorimotor integration (Gentet et al.,2012). Inter- ity with somatostatin-14 (de Lecea et al., 1996, 1997). Notably, estingly, in mouse somatosensory cortex, somatostatin-expressing compared with the weak inhibitory effects of somatostatin on interneurons show a spatially precise connectivity with pyrami- the basal release of CRH from rat hypothalamus and hippocam- dal neurons through direct targeting in layers 2/3 or indirectly pus, cortistatin exhibits strong inhibition of the expression and through inhibition of local parvalbumin interneurons in layer 4 release of basal CRH (Tringali et al., 2012). These findings sug- (Xu et al., 2013). Moreover, in layers 2/3 of the mouse prefrontal gest that Sst2 may regulate affective phenotypes and HPA axis cortex, somatostatin-expressing interneurons compartmentalizes responses both through somatostatin and cortistatin. Given the inhibitions of calcium signaling to spine heads, not shafts, sug- limitations of human studies, SstKO mice provide an opportunity gesting that dendrite-targeting inhibition through somatostatin- to explore the causal role of somatostatin in affect dysregulation expressing interneurons may contribute to downstream cellular and the underlying neural mechanisms. Such insights, however, processes such as synaptic plasticity (Chiu et al., 2013). In mouse will require systematic behavioral characterization with fine spa- visual cortex, somatostatin-expressing interneurons are found to tial and temporal resolution by including female cohorts and mediate response levels of specific subsets of pyramidal neu- region-specific manipulation at different developmental stages. rons whereas parvalbumin-expressing neurons alter response gain Based on the published studies to date, it is still unclear whether (Wilson et al., 2012). Parvalbumin-expressing neurons receive these mutants recapitulate behavioral features of mood disorders. excitatory input from the thalamus and make strong synapses on Knowing the effects of somatostatin signaling on neuroendocrine the soma and axons of their target cells (Kawaguchi and Kubota, regulation, future studies need to assess the molecular and cellu- 1997) to control spike timing of the output neurons. In contrast, lar systems that somatostatin mutations converge upon, and where somatostatin-expressing neurons mostly do not receive input from the exact neural circuits are affected. Moreover, combining genetic thalamus (Beierlein et al., 2003; Cruikshank et al., 2010) and are and environmental factors in animal models is critical to enhance instead activated through feed-forward mechanisms by activated the accuracy of disease modeling and translational efforts. For pyramidal neurons. Somatostatin-expressing interneurons prefer- example, acute or chronic exposure to stress or to stress hormones entially target distal dendrites of pyramidal neurons in layer 2/3 to may capture how such etiological factors determine the vulnera- modulate the processing of incoming sensory information before bility to external insults, in contrast to baseline behavioral testing. it is integrated at the soma level (Di Cristo et al., 2004; Markram In addition, mood disorder-related sex differences are observed et al., 2004; Tan et al., 2008; Murayama et al., 2009; Xu et al., 2013). in community-based epidemiological studies, where the factor of Hence, the distinct GABAergic and prototypical inhibitory pop- seeking treatment is removed (Kornstein et al., 2000; Festinger ulations, expressing either parvalbumin or somatostatin, shape et al., 2008; Leach et al., 2008) and findings of low somatostatin in the spatiotemporal control of multiple post-synaptic potentials in the amygdala appear more robust in postmortem studies of female cortical local circuits, and provide a framework to investigate the MDD subjects (Tripp et al., 2012), suggesting that gender/sex may role of inhibitory circuits in physiology and pathology. represent a biological predisposing factor, or at least a moderating factor, in the intrinsic vulnerability of the somatostatin system. GENETIC APPROACHES TO INVESTIGATE THE SOMATOSTATIN SYSTEM Although many mood disorders emerge during adolescence Mice mutant for somatostatin were created by deleting the cod- (Paus et al., 2008), behavioral abnormalities including affect dys- ing region of the pre-pro-somatostatin (the last ten codons of regulation are often heritable and apparent before diagnostic crite- the first exon; Zeyda et al., 2001). Somatostatin knockout (KO; riaaremet(McGuffin et al., 2003; Geller et al., 2006). It is unclear SstKO ) mice show intact motor coordination and motor learning, when somatostatin deficits occur and potentially begin to con- but have a significant impairment in motor learning as demands tribute to the formation of affective symptoms. Tracking somato- of motor coordination are increased. Overall, a detailed analysis statin system using new anatomic techniques with refined cellular

www.frontiersin.org September 2013 | Volume 4 | Article 110 | 5

“fphar-04-00110” — 2013/9/5 — 15:44 — page5—#5 Lin and Sibille Somatostatin deficits in mood disorders

definition, from Brainbow (Livet et al., 2007) to CLARITY (Chung (Yeung et al., 2011). Moreover, anxiolytic effects in the elevated et al., 2013) and SeeDB (Ke et al., 2013), across different develop- plus-maze test are described after intra-cerebroventricular infu- mental stages may help identify age-dependent neural architecture sions of a selective Sst2 receptor agonist, but not after infusions of and disease mechanisms related to somatostatin function. the other four receptor agonists; antidepressant-like effects in the forced swim test are observed following infusions of either Sst2 or SOMATOSTATIN ANALOG DEVELOPMENT AND PHARMACOLOGICAL Sst3 agonists (Engin and Treit, 2009). Another agent to enhance STUDIES somatostatin functioning, SRA880 (an antagonist of auto-receptor As native somatostatin peptides have a very short half-life time Sst1), synergizes with imipramine in causing antidepressant-like (approximate 1–3 min; Sheppard et al., 1979), long-acting and effects in the tail suspension test and increases Bdnf mRNA highly potent somatostatin analogues are currently available for expression in the mouse cerebral cortex (Nilsson et al., 2012). the treatment of acromegaly and neuroendocrine tumors, includ- ing octreotide (long-acting; LAR-OCT; Bauer et al., 1982) and EFFECTS OF ANTIDEPRESSANTS ON SOMATOSTATIN IN THE CNS Lanreotide (slow release or autogel; Bevan, 2005; Molitch, 2008). Significant efforts have been directed toward the characterization Compared to somatostatin, pharmacological tools of the five of the downstream targets of antidepressant treatment, with a somatostatin receptor subtypes have lagged behind, partly due focus on somatostatin. A recent study demonstrates that chronic to the lack of high-affinity antagonists. imipramine treatment increases somatostatin expression in mouse In addition, several novel somatostatin therapy models are hypothalamus (Nilsson et al., 2012). However, there is incon- available: (1) Universal somatostatin (Schmid and Schoeffter, sistency regarding the effect of chronic citalopram treatment 2004): a somatostatin molecular analog with high binding affin- on somatostatin levels in rats (Kakigi et al., 1992; Prosperini ity to all or most human somatostatin receptors. An exam- et al., 1997; Pallis et al., 2006, 2009). Repeated administration of ple is SOM230, which interacts with Sst1,2,3,5 and particularly imipramine, maprotiline, mianserin, carbamazepine or zotepine potent at Sst5 compared with LAR-OCT; (2) Chimeric somato- has no effect on somatostatin levels in various brain regions of rats statin/dopamine molecule (Saveanu et al., 2002; Pivonello et al., (Weiss et al., 1987; Kakigi et al., 1992). While some somatostatin 2005): a somatostatin and dopamine hybrid agonist, based on receptors seem to exert anxiolytic or antidepressant-like effects, reports that dopamine and somatostatin receptors can hetero- there is no direct evidence supporting somatostatin receptors as oligomerize to enhance functional responses (Rocheville et al., downstream targets of current antidepressants. Together, these 2000). An example is BIM-23A760, which accelerates the suppres- findings suggest that somatostatin levels are mostly unchanged sion of growth hormone and adrenocorticotropic hormone by by antidepressants. It is unclear whether somatostatin, GABA, or the interaction with Sst2 and Drd2 simultaneously; (3) Chimeric- GABA functioning in somatostatin-expressing interneurons may somatostatin vaccinations (Haffer,2012): a fusion protein express- be the real mediators or antidepressant targets. Future studies are ing chloramphenicol acetyl transferase protein and somatostatin. needed to determine the involvement of somatostatin receptors Two somatostatin vaccinations, JH17 and JH18, can effectively and associated intracellular signaling pathways in the therapeu- reduce weight gain and reduce final body weight percentage of tic effects of antidepressants, or whether somatostatin effects are normal, non-obese mice and mice with diet-induced obesity via independent of current antidepressant modalities. the intra-peritoneal route; (4) Non-peptide antagonists, such as SRA880 (Sst1 selective), ACQ090 (Sst3 selective) and Sst4 selective β peptide agonists (Rivier et al., 2003; Hoyer et al., 2004). Despite POTENTIAL MECHANISMS OF SELECTIVE VULNERABILITY this extensive list, the practical use of somatostatin in the brain is OF SOMATOSTATIN-EXPRESSING INTERNEURONS hampered by the multiple effects of the peptide, by the need for It is possible that low somatostatin in diseases acts as a biomarker small molecules targeting specific, high affinity receptors on the for deregulated function of somatostatin-expressing neurons. As target cells in specific brain regions, and by the need for feasible such, it is essential to identify upstream factors responsible for routes of administration that lead to fast delivery into the brain. the dysfunction of somatostatin-expressing interneurons in neu- The potential for using somatostatin analogues as treatment rological disorders. We speculate that intrinsic cellular properties in the CNS is emerging for treatment of epilepsy (Vezzani and in somatostatin-expressing neurons may determine their selec- Hoyer, 1999; Tallent and Qiu, 2008), pain (Mollenholt et al., 1994; tive vulnerability to various insults. Pathways underlying this Taura et al., 1994) and headaches (Sicuteri et al., 1984; Kapicioglu high vulnerability may include high intrinsic oxidative stress et al., 1997); potential use for treatment of mood disorders is sug- related to mitochondria, high sensitivity to inflammation, high gested by reversal of emotion-like behaviors in rodent models. dependence on neurotrophic environment, and cellular devel- Several pharmacological studies support a role of somatostatin in opmental and aging processes. These canonical pathways might affect regulation. Intra-ventricular administration of somatostatin provide novel cell-based perspectives in the treatment of affected in rats produces anxiolytic- and antidepressant-like behaviors somatostatin-expressing cells across neurological disorders. in the elevated plus-maze and forced swim tests, and a neu- rophysiological signature of anxiolytic drugs (e.g., reduction of OXIDATIVE STRESS AND MITOCHONDRIAL DYSFUNCTIONS theta frequency and theta frequency curve slope; Engin et al., Oxidative stress produced by mitochondria during respiration is 2008). Mice with intra-amygdalar and intra-septal microinfusions a common pathogenic mechanism implicated in neurological dis- of somatostatin-14 and somatostatin-28 display reduced anxiety- orders (Sorce and Krause, 2009; Stefanescu and Ciobica, 2012). like behavior in the elevated plus-maze and shock-probe tests Depressed states in mood disorders are associated with decreased

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 110 | 6

“fphar-04-00110” — 2013/9/5 — 15:44 — page6—#6 Lin and Sibille Somatostatin deficits in mood disorders

brain energy generation (Baxter et al., 1985, 1989). Mitochon- episodes (Celik et al., 2010; Maes et al., 2012), suggesting that drial dysfunction together with the oxidative stress accumulation prior depression may sensitize inflammatory responses. Patients has been proposed to synergistically contribute to the neuro- treated with inflammatory cytokines, such as interferon-α,are endangerment processes underlying depression (Gardner et al., at greater risk of developing depressive episodes (Castera et al., 2003; Burnett et al.,2005) and neurodegenerative diseases (Lin and 2006; Lotrich et al., 2007). Somatostatin released from sensory Beal, 2006; Mancuso et al., 2007; Petrozzi et al., 2007). Similarly, nerves and somatostatin receptors on peripheral blood mononu- high baseline oxidative stress could be an intrinsic characteris- clear cells play a crucial role in anti-inflammation through tic of vulnerable neuronal populations. Notably, neuronal nitric inhibition of pro-inflammatory peptide release (Szolcsanyi et al., oxide synthase (nNOS) and NADPH diaphorase (NADPHd), two 1998; Kurnatowska and Pawlikowski, 2000; Helyes et al., 2004). enzymes that produce reactive oxidative species, are extensively Rats with chronic inflammation induced by lipopolysaccharide and almost exclusively co-localized with somatostatin and neu- show decreased hippocampal somatostatin expression (Gavilan ropeptide Y (Dun et al., 1994; Figueredo-Cardenas et al., 1996; et al., 2007). It is possible that there is crosstalk among periph- Jaglin et al., 2012), hence providing a neurochemical basis for eral inflammation, somatostatin function, and central effects high susceptibility of somatostatin-expressing neurons to generate of somatostatin-expressing neurons. Hence, decreasing somato- oxidative stress in response to pathophysiological insults. statin expression due to cellular impairment in the progress of neurological diseases may further enhance inflammation in HIGH DEPENDENCE ON NEUROTROPHIC ENVIRONMENT a vicious cycle, leading to exacerbated cellular vulnerability of Brain-derived neurotrophic factor (BDNF) and its receptor neu- somatostatin-expressing neurons. rotrophic tyrosine kinase receptor type 2 (TrkB) have been Aging is associated with a considerable increase in implicated in mood disorders (Guilloux et al., 2012; Tripp et al., an activated, pro-inflammatory state (Wei et al., 1992; 2012). BDNF-TrkB signaling is one of the key mediators for Bruunsgaard and Pedersen, 2003), a decline in circulating levels maintaining normal somatostatin gene expression (Glorioso et al., of Bdnf (Erickson et al., 2010), and increased oxidative dam- 2006; Martinowich et al., 2011). Progressively impairing BDNF- age (Sohal and Weindruch, 1996). Somatostatin expression is TrkB signaling in patients with mood disturbances may directly significantly decreased with age in human cortical regions, but impact the biology of somatostatin-expressing neurons, resulting parvalbumin expression is not altered by age (Erraji-Benchekroun in somatostatin deficits. In addition, Bdnf-TrkB signaling itself et al., 2005; Glorioso et al., 2011). Similarly, the number of is vulnerable to increased inflammation (Goshen et al., 2008; Koo hippocampal somatostatin-expressing interneurons decreases in and Duman,2008; Song and Wang,2011) and high glucocorticoids aged rats, but the number of parvalbumin-expressing neurons insults (Hodes et al., 2012). Mild oxidative stress inhibits tyrosine remains the same (Vela et al., 2003). Somatostatin and IL-1β phosphatases activity (Barrett et al., 2005), potentially leading to mRNA expression are negatively correlated in aged hippocampus impaired TrkB downstream signaling. Cortistatin and neuropep- of rats (Gavilan et al., 2007). Comparing the effects of aging on tide Y expression partly overlaps with the somatostatin neuron somatostatin expression in the sgACC, an accelerated reduction is population in rodents (Figueredo-Cardenas et al., 1996; de Lecea found in patients with MDD compared to normal aging subjects et al., 1997; Xu et al., 2010). Comparing the profile of gene changes (Tripp et al., 2012), suggesting a pattern resulting in an early aging between subjects with MDD and mice with genetically-altered phenomenon which we have speculated may be synergistically Bdnf signaling suggest that the reduced somatostatin, neuropep- induced by normal age-related changes and depression-related tide Y and cortistatin are partly downstream from a combination pathological change (Douillard-Guilloux et al., 2013). of reduced constitutive and activity-dependent Bdnf signaling (Guilloux et al., 2012). In contrast, markers for other GABA neu- CONCLUSION ron subtypes targeting the perisomatic area region cell body and Here we have focused on somatostatin, a GABA marker, axon initial segment of pyramidal neurons (i.e., cholecystokinin down-regulated in MDD, schizophrenia, bipolar disorder, and and calretinin), appear to be independent of BDNF signaling and neurodegenerative diseases. Exploring cross-disease molecular unaffected in MDD patients (Guilloux et al., 2012; Tripp et al., (somatostatin) and cellular (somatostatin-expressing interneu- 2012). Hence, it is possible that the somatostatin-specific cellu- rons) pathological findings suggests a dimensional pathological lar function and vulnerability are partly mediated by BDNF-TrkB phenotype that is specific to the somatostatin gene/cell biolog- signaling during both physiological and pathological processes of ical entity rather than to categorical brain disorders. Based on affect regulation. these results we speculate that common risk factors affecting somatostatin and somatostatin-expressing neurons may impact INFLAMMATION AND CELLULAR AGING information processing in the cortical local circuits (Figure 1). Inflammation has been implicated as a contributing factor in Clarifying the role of somatostatin and its regulation of GABA the onset and progression of many neurological disorders (Di inhibition in affect regulation could provide new strategies for Filippo et al.,2008). Mood disturbances are associated with an acti- predicting, delaying, and treating neurological diseases with vated inflammatory response system (Padmos et al., 2008; Miller mood disturbances. A number of questions remain. For exam- et al., 2009), including increased levels of peripheral interleukins ple, are the prevalent somatostatin deficits seen in multiple and tumor necrosis factor-alpha in MDD patients (Kaestner diseases reflected in a common symptom dimension, such as et al., 2005; Howren et al., 2009; Dowlati et al., 2010; Maes, low mood, across neurological diseases? What are the critical 2011). Inflammatory illnesses are associated with more depressive events that determine the vulnerability of somatostatin-expressing

www.frontiersin.org September 2013 | Volume 4 | Article 110 | 7

“fphar-04-00110” — 2013/9/5 — 15:44 — page7—#7 Lin and Sibille Somatostatin deficits in mood disorders

neurons? And what are the pathogenic mechanisms that mediate for several neuropsychiatric disorders and neurodegenerative the observed disease-related molecular and cellular phenotypes? diseases. One possibility is that inflammation, oxidative stress, aging, and reduced neurotrophic support may all converge to affect ACKNOWLEDGMENTS somatostatin-expressing neurons. Targeting these pathways may This work was supported by National Institute of Mental Health exert neuro-protective effects on somatostatin-expressing neu- (NIMH-MH093723, MH084060 and MH077159). We thank rons, as a potential therapeutic approach with implications Beverly French for careful review of the manuscript.

REFERENCES Bevan, J. S. (2005). The antitu- GABAergic inhibition by dendritic Di Cristo, G., Wu, C., Chattopadhyaya, Aarsland, D., Larsen, J. P., Lim, N. G., moral effects of somatostatin ana- spines. Science 340, 759–762. doi: B., Ango, F., Knott, G., Welker, E., Janvin, C., Karlsen, K., Tandberg, log therapy in acromegaly. J. Clin. 10.1126/science.1234274 et al. (2004). Subcellular domain- E., et al. (1999). Range of neuropsy- Endocrinol. Metab. 90, 1856–1863. Chung, K., Wallace, J., Kim, S. Y., restricted GABAergic innervation in chiatric disturbances in patients with doi: 10.1210/jc.2004-1093 Kalyanasundaram, S., Andalman, A. primary visual cortex in the absence Parkinson’s disease. J. Neurol. Neu- Bissette, G., Widerlov, E., Walleus, S., Davidson, T. J., et al. (2013). Struc- of sensory and thalamic inputs. rosurg. Psychiatry 67, 492–496. doi: H., Karlsson, I., Eklund, K., tural and molecular interrogation of Nat. Neurosci. 7, 1184–1186. doi: 10.1136/jnnp.67.4.492 Forsman, A., et al. (1986). Alter- intact biological systems. Nature 497, 10.1038/nn1334 Agren, H., and Lundqvist, G. (1984). ations in cerebrospinal fluid 332–337. doi: 10.1038/nature12107 Di Filippo, M., Sarchielli, P., Picconi, Low levels of somatostatin in human concentrations of somatostatinlike Cruikshank, S. J., Urabe, H., Nur- B., and Calabresi, P. (2008). Neuroin- CSF mark depressive episodes. immunoreactivity in neuropsychi- mikko, A. V., and Connors, B. W. flammation and synaptic plasticity: Psychoneuroendocrinology 9, 233– atric disorders. Arch. Gen. Psychiatry (2010). Pathway-specific feedforward theoretical basis for a novel, immune- 248. doi: 10.1016/0306-4530(84) 43, 1148–1151. doi: 10.1001/arch- circuits between thalamus and neo- centred, therapeutic approach to 90003-9 psyc.1986.01800120034008 cortex revealed by selective optical neurological disorders. Trends Phar- Barrett, D. M., Black, S. M., Todor, Bruunsgaard, H., and Pedersen, B. stimulation of axons. Neuron 65, macol. Sci. 29, 402–412. doi: H., Schmidt-Ullrich, R. K., Dawson, K. (2003). Age-related inflamma- 230–245. doi: 10.1016/j.neuron.2009. 10.1016/j.tips.2008.06.005 K. S., and Mikkelsen, R. B. (2005). tory cytokines and disease. Immunol. 12.025 Douillard-Guilloux, G., Guilloux, J. P., Inhibition of protein-tyrosine phos- Allergy Clin. North Am. 23, 15– Csaba, Z., and Dournaud, P. (2001). Lewis, D. A., and Sibille, E. (2013). phatases by mild oxidative stresses 39. doi: 10.1016/S0889-8561(02) Cellular biology of somatostatin Anticipated brain molecular aging is dependent on S-nitrosylation. J. 00056-5 receptors. Neuropeptides 35, 1–23. in major depression. Am. J. Geri- Biol. Chem. 280, 14453–14461. doi: Bungener, C., Jouvent, R., and Der- doi: 10.1054/npep.2001.0848 atr. Psychiatry 21, 450–460. doi: 10.1074/jbc.M411523200 ouesne, C. (1996). Affective distur- Davies, P., Katzman, R., and Terry, R. 10.1016/j.jagp.2013.01.040 Bauer, W., Briner, U., Doepfner, W., bances in Alzheimer’s disease. J. Am. D. (1980). Reduced somatostatin-like Dournaud, P., Cervera-Pierot, P., Haller, R., Huguenin, R., Marbach. Geriatr. Soc. 44, 1066–1071. immunoreactivity in cerebral cor- Hirsch, E., Javoy-Agid, F., Kordon, C., P., et al. (1982). SMS 201-995: a very Burnett, B. B., Gardner, A., and Boles, tex from cases of Alzheimer dis- Agid, Y., et al. (1994). Somatostatin potent and selective octapeptide ana- R. G. (2005). Mitochondrial inheri- ease and Alzheimer senile dementa. messenger RNA-containing neurons logue of somatostatin with prolonged tance in depression, dysmotility and Nature 288, 279–280. doi: 10.1038/ in Alzheimer’s disease: an in situ action. Life Sci. 31, 1133–1140. doi: migraine? J. Affect Disord. 88, 109– 288279a0 hybridization study in hippocampus, 10.1016/0024-3205(82)90087-X 116. doi: 10.1016/j.jad.2005.05.009 Davies, P., and Terry, R. D. (1981). Cor- parahippocampal cortex and frontal Baxter, L. R. Jr., Phelps, M. E., Mazz- Candy, J. M., Gascoigne, A. D., Big- tical somatostatin-like immunoreac- cortex. Neuroscience 61, 755–764. iotta, J. C., Schwartz, J. M., Gerner, gins, J. A., Smith, A. I., Perry, tivity in cases of Alzheimer’s disease doi: 10.1016/0306-4522(94)90399-9 R. H., Selin, C. E. et al. (1985). R. H., Perry, E. K., et al. (1985). and senile dementia of the Alzheimer Dowlati, Y., Herrmann, N., Swardfager, Cerebral metabolic rates for glu- Somatostatin immunoreactivity in type. Neurobiol. Aging 2, 9–14. doi: W., Liu, H., Sham, L., Reim, E. cose in mood disorders. Studies with cortical and some subcortical regions 10.1016/0197-4580(81)90053-1 K., et al. (2010). A meta-analysis positron emission tomography and in Alzheimer’s disease. J. Neurol. DeFelipe, J., Lopez-Cruz, P. L., of cytokines in major depression. fluorodeoxyglucose F 18. Arch. Gen. Sci. 71, 315–323. doi: 10.1016/0022- Benavides-Piccione, R., Bielza, C., Biol. Psychiatry 67, 446–457. doi: Psychiatry 42, 441–447. doi: 10.1001/ 510X(85)90070-X Larranaga, P., Anderson, S., et al. 10.1016/j.biopsych.2009.09.033 archpsyc.1985.01790280019002 Castera, L., Constant, A., Henry, C., (2013). New insights into the clas- Dun, N. J., Dun, S. L., Wong, R. K., Baxter, L. R. Jr., Schwartz, J. M., Phelps, Champbenoit, P., Bernard, P. H., De sification and nomenclature of cor- and Forstermann, U. (1994). Colo- M. E., Mazziotta, J. C., Guze, B. Ledinghen, V., et al. (2006). Impact tical GABAergic interneurons. Nat. calization of nitric oxide synthase and H., Selin, C. E. et al. (1989). Reduc- on adherence and sustained viro- Rev. Neurosci. 14, 202–216. doi: somatostatin immunoreactivity in rat tion of prefrontal cortex glucose logical response of psychiatric side 10.1038/nrn3444 dentate hilar neurons. Proc. Natl. metabolism common to three types effects during peginterferon and rib- de Lecea, L. (2008). Cortistatin – func- Acad. Sci. U.S.A. 91, 2955–2959. doi: of depression. Arch. Gen. Psychia- avirin therapy for chronic hepati- tions in the central nervous system. 10.1073/pnas.91.8.2955 try 46, 243–250. doi: 10.1001/arch- tis C. Aliment. Pharmacol. Ther. Mol. Cell. Endocrinol. 286, 88–95. doi: Dupont, E., Christensen, S. E., psyc.1989.01810030049007 24, 1223–1230. doi: 10.1111/j.1365- 10.1016/j.mce.2007.12.014 Hansen, A. P., de Fine Olivarius, Beal, M. F., Mazurek, M. F., and 2036.2006.03107.x de Lecea, L., Criado, J. R., Prospero- B., and Orskov, H. (1982). Low Martin, J. B. (1986). Somato- Celik, C., Erdem, M., Cayci, T., Ozdemir, Garcia, O., Gautvik, K. M., cerebrospinal fluid somatostatin in statin immunoreactivity is reduced B., Ozgur Akgul, E., Kurt, Y. G., Schweitzer, P., Danielson, P. E., Parkinson disease: an irreversible in Parkinson’s disease dementia with et al. (2010). The association between et al. (1996). A cortical neuropeptide abnormality. Neurology 32, 312–314. Alzheimer’s changes. Brain Res. serum levels of neopterin and num- with neuronal depressant and sleep- doi: 10.1212/WNL.32.3.312 397, 386–388. doi: 10.1016/0006- ber of depressive episodes of major modulating properties. Nature 381, Engin, E., Stellbrink, J., Treit, D., 8993(86)90644-X depression. Prog. Neuropsychophar- 242–245. doi: 10.1038/381242a0 and Dickson, C. T. (2008). Anxi- Beierlein, M., Gibson, J. R., and Con- macol. Biol. Psychiatry 34, 372–375. de Lecea, L., del Rio, J. A., Criado, J. R., olytic and antidepressant effects of nors, B. W. (2003). Two dynam- doi: 10.1016/j.pnpbp.2010.01.002 Alcantara, S., Morales, M., Daniel- intracerebroventricularly admin- ically distinct inhibitory networks Chiu, C. Q., Lur, G., Morse, son, P. E., et al. (1997). Cortistatin istered somatostatin: behavioral in layer 4 of the neocortex. J. T. M., Carnevale, N. T., Ellis- is expressed in a distinct subset of and neurophysiological evidence. Neurophysiol. 90, 2987–3000. doi: Davies, G. C., and Higley, M. cortical interneurons. J. Neurosci. 17, Neuroscience 157, 666–676. doi: 10.1152/jn.00283.2003 J. (2013). Compartmentalization of 5868–5880. 10.1016/j.neuroscience.2008.09.037

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 110 | 8

“fphar-04-00110” — 2013/9/5 — 15:44 — page8—#8 Lin and Sibille Somatostatin deficits in mood disorders

Engin, E., and Treit, D. (2009). Anx- Hagenfeldt, L., et al. (2003). Alter- Mol. Psychiatry 13, 717–728. doi: Schuepbach, E., et al. (2004). iolytic and antidepressant actions ations of mitochondrial function and 10.1038/sj.mp.4002055 SRA880, in vitro characterization of somatostatin: the role of sst2 correlations with personality traits Guerreiro, R., Wojtas, A., Bras, J., Car- of the first non-peptide somato- and sst3 receptors. Psychopharma- in selected major depressive disor- rasquillo, M., Rogaeva, E., Majounie, statin sst(1) receptor antagonist. cology (Berl.) 206, 281–289. doi: der patients. J. Affect. Disord. 76, E., et al. (2013). TREM2 variants in Neurosci. Lett. 361, 132–135. doi: 10.1007/s00213-009-1605-5 55–68. doi: 10.1016/S0165-0327(02) Alzheimer’s disease. N. Engl. J. Med. 10.1016/j.neulet.2004.02.017 Epelbaum, J. (1986). Somatostatin in 00067-8 368, 117–127. doi: 10.1056/NEJ- Jaglin, X. H., Hjerling-Leffler, J., Fishell, the central nervous system: physiol- Gavilan, M. P., Revilla, E., Pin- Moa1211851 G., and Batista-Brito, R. (2012). ogy and pathological modifications. tado, C., Castano, A., Vizuete, Guilloux, J. P., Douillard-Guilloux, The origin of neocortical nitric oxide Prog. Neurobiol. 27, 63–100. doi: M. L., Moreno-González, I., et al. G., Kota, R., Wang, X., Gardier, synthase-expressing inhibitory neu- 10.1016/0301-0082(86)90012-2 (2007). Molecular and cellular A. M., Martinowich, K., et al. rons. Front. Neural Circuits 6:44. doi: Epelbaum, J., Javoy-Agid, F., Enjal- characterization of the age-related (2012). Molecular evidence for 10.3389/fncir.2012.00044 bert, A., Krantic, S., Kordon, C., neuroinflammatory processes occur- BDNF- and GABA-related dysfunc- Kaestner, F., Hettich, M., Peters, M., and Agid, Y. (1988). Somatostatin ring in normal rat hippocampus: tions in the amygdala of female sub- Sibrowski, W., Hetzel, G., Ponath, concentrations and binding sites in potential relation with the loss of jects with major depression. Mol. G., et al. (2005). Different acti- human frontal cortex are differen- somatostatin GABAergic neurons. Psychiatry 17, 1130–1142. doi: vation patterns of proinflammatory tially affected in Parkinson’s disease J. Neurochem. 103, 984–996. doi: 10.1038/mp.2011.113 cytokines in melancholic and non- associated dementia and in progres- 10.1111/j.1471-4159.2007.04787.x Guillozet-Bongaarts, A. L., Hyde, T. M., melancholic major depression are sive supranuclear palsy. J. Neurol. Geller, B., Tillman, R., Bolhofner, K., Dalley, R. A., Hawrylycz, M. J., Henry, associated with HPA axis activity. Sci. 87, 167–174. doi: 10.1016/0022- Zimerman, B., Strauss, N. A., and A., Hof, P. R., et al. (2013). Altered J. Affect. Disord. 87, 305–311. doi: 510X(88)90242-0 Kaufmann, P. (2006). Controlled, gene expression in the dorsolateral 10.1016/j.jad.2005.03.012 Erickson, K. I., Prakash, R. S., blindly rated, direct-interview family prefrontal cortex of individuals with Kakigi, T., Maeda, K., Kaneda, H., and Voss, M. W., Chaddock, L., study of a prepubertal and early- schizophrenia. Mol. Psychiatry. doi: Chihara, K. (1992). Repeated admin- Heo, S., McLaren, M., et al. adolescent bipolar I disorder pheno- 10.1038/mp.2013.30 [Epub ahead of istration of antidepressant drugs (2010). Brain-derived neurotrophic type: morbid risk, age at onset, and print]. reduces regional somatostatin con- factor is associated with age-related comorbidity. Arch. Gen. Psychiatry Haffer, K. N. (2012). Effects of centrations in rat brain. J. Affect. Dis- decline in hippocampal volume. 63, 1130–1138. doi: 10.1001/arch- novel vaccines on weight loss in ord. 25, 215–220. doi: 10.1016/0165- J. Neurosci. 30, 5368–5375. doi: psyc.63.10.1130 diet-induced-obese (DIO) mice. J. 0327(92)90078-K 10.1523/JNEUROSCI.6251-09.2010 Gentet, L. J., Kremer, Y., Taniguchi, Anim. Sci. Biotechnol. 3, 21. doi: Kapicioglu, S., Gokce, E., Kapicioglu, Erraji-Benchekroun, L., Underwood, H., Huang, Z. J., Staiger, J. F., and 10.1186/2049-1891-3-21 Z., and Ovali, E. (1997). Treat- M. D., Arango, V., Galfalvy, H., Petersen, C. C. (2012). Unique func- Helyes, Z., Szabo, A., Nemeth, J., ment of migraine attacks with a Pavlidis, P., Smyrniotopoulos, P., tional properties of somatostatin- Jakab, B., Pinter, E., Banvolgyi, long-acting somatostatin analogue et al. (2005). Molecular aging in expressing GABAergic neurons in A., et al. (2004). Antiinflammatory (octreotide, SMS 201–995). Cepha- human prefrontal cortex is selec- mouse barrel cortex. Nat. Neurosci. and analgesic effects of somatostatin lalgia 17, 27–30. doi: 10.1046/j.1468- tive and continuous throughout adult 15, 607–612. doi: 10.1038/nn.3051 released from capsaicin-sensitive sen- 2982.1997.1701027.x life. Biol. Psychiatry 57, 549–558. doi: Glorioso, C., Oh, S., Douillard, G. sory nerve terminals in a Freund’s Kawaguchi, Y., and Kubota, Y. (1997). 10.1016/j.biopsych.2004.10.034 G., and Sibille, E. (2011). Brain adjuvant-induced chronic arthritis GABAergic cell subtypes and their Festinger, D. S., Marlowe, D. B., molecular aging, promotion of neu- model in the rat. Arthritis Rheum. 50, synaptic connections in rat frontal Dugosh, K. L., Croft, J. R., and rological disease and modulation by 1677–1685. doi: 10.1002/art.20184 cortex. Cereb. Cortex 7, 476–486. doi: Arabia, P. L. (2008). Higher magni- sirtuin 5 longevity gene polymor- Hodes, G. E., Brookshire, B. R., 10.1093/cercor/7.6.476 tude cash payments improve research phism. Neurobiol. Dis. 41, 279–290. Hill-Smith, T. E., Teegarden, S. Ke, M. T., Fujimoto, S., and follow-up rates without increasing doi: 10.1016/j.nbd.2010.09.016 L., Berton, O., and Lucki, I. Imai, T. (2013). SeeDB: a simple drug use or perceived coercion. Drug Glorioso, C., Sabatini, M., Unger, T., (2012). Strain differences in the and morphology-preserving optical Alcohol Depend. 96, 128–135. doi: Hashimoto, T., Monteggia, L. M., effects of chronic corticosterone clearing agent for neuronal circuit 10.1016/j.drugalcdep.2008.02.007 Lewis, D. A., et al. (2006). Specificity exposure in the hippocampus. reconstruction. Nat. Neurosci. 16, Figueredo-Cardenas, G., Morello, M., and timing of neocortical transcrip- Neuroscience 222, 269–280. doi: 1154–1161. doi: 10.1038/nn.3447 Sancesario, G., Bernardi, G., and tome changes in response to BDNF 10.1016/j.neuroscience.2012.06.017 Kling, M. A., Rubinow, D. R., Reiner, A. (1996). Colocaliza- gene ablation during embryogenesis Hollingworth, P., Harold, D., Sims, Doran, A. R., Roy, A., Davis, C. tion of somatostatin, neuropeptide or adulthood. Mol. Psychiatry 11, R., Gerrish, A., Lambert, J. L., Calabrese, J. R., et al. (1993). Y, neuronal nitric oxide synthase 633–648. doi: 10.1038/sj.mp.4001835 C., Carrasquillo, M. M., et al. Cerebrospinal fluid immunoreac- and NADPH-diaphorase in striatal Gonchar, Y., and Burkhalter, A. (1997). (2011). Common variants at ABCA7, tive somatostatin concentrations in interneurons in rats. Brain Res. Three distinct families of GABAergic MS4A6A/MS4A4E, EPHA1, CD33 patients with Cushing’s disease and 735, 317–324. doi: 10.1016/0006- neurons in rat visual cortex. Cereb. and CD2AP are associated with major depression: relationship to 8993(96)00801-3 Cortex 7, 347–358. doi: 10.1093/cer- Alzheimer’s disease. Nat. Genet. 43, indices of corticotropin-releasing Fishell, G., and Rudy, B. (2011). Mecha- cor/7.4.347 429–435. doi: 10.1038/ng.803 hormone and cortisol secretion. Neu- nisms of inhibition within the telen- Gonchar, Y., Wang, Q., and Burkhalter, Holsboer, F. (2000). The corti- roendocrinology 57, 79–88. doi: cephalon: “where the wild things A. (2007). Multiple distinct subtypes costeroid receptor hypothesis of 10.1159/000126345 are”. Annu.Rev.Neurosci.34, 535– of GABAergic neurons in mouse depression. Neuropsychopharmacol- Kluxen, F. W., Bruns, C., and Lubbert, 567. doi: 10.1146/annurev-neuro- visual cortex identified by triple ogy 23, 477–501. doi: 10.1016/S0893- H. (1992). Expression cloning of a rat 061010-113717 immunostaining. Front. Neuroanat. 133X(00)00159-7 brain somatostatin receptor cDNA. Fitz-Patrick, D., and Patel, Y. C. 1:3. doi: 10.3389/neuro.05.003.2007 Howren, M. B., Lamkin, D. M., and Suls, Proc. Natl. Acad. Sci. U.S.A. 89, 4618– (1981). Evidence for somatostatin Goshen, I., Kreisel, T., Ben-Menachem- J. (2009). Associations of depres- 4622. doi: 10.1073/pnas.89.10.4618 precursors in human stomach, pla- Zidon, O., Licht, T., Weiden- sion with C-reactive protein, IL-1, Koch, B. D., Blalock, J. B., and Schon- centa, and amniotic fluid. J. Clin. feld, J., Ben-Hur, T., et al. (2008). and IL-6: a meta-analysis. Psycho- brunn, A. (1988). Characterization of Endocrinol. Metab. 53, 372–376. doi: Brain interleukin-1 mediates chronic som. Med. 71, 171–186. doi: 10.1097/ the cyclic AMP-independent actions 10.1210/jcem-53-2-372 stress-induced depression in mice via PSY.0b013e3181907c1b of somatostatin in GH cells. I. Gardner, A., Johansson, A., Wibom, adrenocortical activation and hip- Hoyer, D., Nunn, C., Hannon, An increase in potassium conduc- R., Nennesmo, I., von Dobeln, U., pocampal neurogenesis suppression. J., Schoeffter, P., Feuerbach, D., tance is responsible for both the

www.frontiersin.org September 2013 | Volume 4 | Article 110 | 9

“fphar-04-00110” — 2013/9/5 — 15:44 — page9—#9 Lin and Sibille Somatostatin deficits in mood disorders

hyperpolarization and the decrease in Le Magueresse, C., and Monyer, aging. Front. Endocrinol. (Lau- in somatostatin mRNA expression intracellular free calcium produced H. (2013). GABAergic interneurons sanne) 3:154. doi: 10.3389/fendo. in the dorsolateral prefrontal cor- by somatostatin. J. Biol. Chem. 263, shape the functional maturation of 2012.00154 tex of subjects with schizophrenia or 216–225. the cortex. Neuron 77, 388–405. doi: Martinowich, K., Schloesser, R. schizoaffective disorder. Cereb. Cor- Koch, B. D., and Schonbrunn, A. 10.1016/j.neuron.2013.01.011 J., Jimenez, D. V., Weinberger, tex 18, 1575–1587. doi: 10.1093/cer- (1984). The somatostatin receptor is Lin, M. T., and Beal, M. F. D. R., and Lu, B. (2011). cor/bhm186 directly coupled to adenylate cyclase (2006). Mitochondrial dysfunction Activity-dependent brain-derived Murayama, M., Perez-Garci, E., Nevian, in GH4C1 pituitary cell membranes. and oxidative stress in neurodegener- neurotrophic factor expression reg- T., Bock, T., Senn, W., and Endocrinology 114, 1784–1790. doi: ative diseases. Nature 443, 787–795. ulates cortistatin-interneurons and Larkum, M. E. (2009). Dendritic 10.1210/endo-114-5-1784 doi: 10.1038/nature05292 sleep behavior. Mol. Brain 4, 11. doi: encoding of sensory stimuli con- Konradi, C., Yang, C. K., Zim- Livet, J., Weissman, T. A., Kang, H., 10.1186/1756-6606-4-11 trolled by deep cortical interneu- merman, E. I., Lohmann, K. M., Draft, R. W., Lu, J., Bennis, R. McGuffin, P., Rijsdijk, F., Andrew, M., rons. Nature 457, 1137–1141. doi: Gresch, P., Pantazopoulos, H., et al. A., et al. (2007). Transgenic strate- Sham, P., Katz, R., and Cardno, 10.1038/nature07663 (2011a). Hippocampal interneu- gies for combinatorial expression of A. (2003). The heritability of bipo- Nanda, S. A., Qi, C., Roseboom, P. H., rons are abnormal in schizophrenia. fluorescent proteins in the nervous lar affective disorder and the genetic and Kalin, N. H. (2008). Predator Schizophr. Res. 131, 165–173. doi: system. Nature 450, 56–62. doi: relationship to unipolar depression. stress induces behavioral inhibition 10.1016/j.schres.2011.06.007 10.1038/nature06293 Arch. Gen. Psychiatry 60, 497–502. and amygdala somatostatin receptor Konradi, C., Zimmerman, E. I., Lotrich, F. E., Rabinovitz, M., doi: 10.1001/archpsyc.60.5.497 2 gene expression. Genes Brain Behav. Yang, C. K., Lohmann, K. M., Gironda, P., and Pollock, B. Miller, A. H., Maletic, V., and Raison, 7, 639–648. doi: 10.1111/j.1601- Gresch, P., Pantazopoulos, H., et al. G. (2007). Depression following C. L. (2009). Inflammation and its 183X.2008.00401.x (2011b). Hippocampal interneu- pegylated interferon-alpha: charac- discontents: the role of cytokines in Neggers, S. J., and van der Lely, rons in bipolar disorder. Arch. teristics and vulnerability. J. Psy- the pathophysiology of major depres- A. J. (2009). Somatostatin ana- Gen. Psychiatry 68, 340–350. chosom. Res. 63, 131–135. doi: sion. Biol. Psychiatry 65, 732–741. log and pegvisomant combina- doi: 10.1001/archgenpsychiatry. 10.1016/j.jpsychores.2007.05.013 doi: 10.1016/j.biopsych.2008.11.029 tion therapy for acromegaly. Nat. 2010.175 Maciag, D., Hughes, J., O’Dwyer, G., Molchan, S. E., Hill, J. L., Martinez, Rev. Endocrinol. 5, 546–552. doi: Koo, J. W., and Duman, R. S. (2008). Pride, Y., Stockmeier, C. A., Sana- R. A., Lawlor, B. A., Mellow, A. M., 10.1038/nrendo.2009.175 IL-1beta is an essential mediator cora, G., et al. (2010). Reduced Rubinow, D. R., et al. (1993). CSF Nilsson, A., Stroth, N., Zhang, X., of the antineurogenic and anhedo- density of calbindin immunoreactive somatostatin in Alzheimer’s disease Qi, H., Falth, M., Skold, K., nic effects of stress. Proc. Natl. GABAergic neurons in the occip- and major depression: relationship to et al. (2012). Neuropeptidomics Acad. Sci. U.S.A. 105, 751–756. doi: ital cortex in major depression: hypothalamic-pituitary-adrenal axis of mouse hypothalamus after 10.1073/pnas.0708092105 relevance to neuroimaging studies. and clinical measures. Psychoneu- imipramine treatment reveal Kornstein, S. G., Schatzberg, A. F., Biol. Psychiatry 67, 465–470. doi: roendocrinology 18, 509–519. doi: somatostatin as a potential mediator Thase, M. E., Yonkers, K. A., McCul- 10.1016/j.biopsych.2009.10.027 10.1016/0306-4530(93)90044-L of antidepressant effects. Neu- lough, J. P., Keitner, G. I., et al. (2000). Maes, M. (2011). Depression is Molchan, S. E., Lawlor, B. A., Hill, ropharmacology 62, 347–357. doi: Gender differences in chronic major an inflammatory disease, but cell- J. L., Martinez, R. A., Davis, C. 10.1016/j.neuropharm.2011.08.004 and double depression. J. Affect. Dis- mediated immune activation is L., Mellow, A. M., et al. (1991). Nyegaard, M., Borglum, A. D., Bruun, ord. 60, 1–11. doi: 10.1016/S0165- the key component of depres- CSF monoamine metabolites and T. G., Collier, D. A., Russ, C., Mors, 0327(99)00158-5 sion. Prog. Neuropsychopharmacol. somatostatin in Alzheimer’s dis- O., et al. (2002). Novel polymor- Kurnatowska, I., and Pawlikowski, Biol. Psychiatry 35, 664–675. doi: ease and major depression. Biol. phisms in the somatostatin receptor 5 M. (2000). Effect of somatostatin 10.1016/j.pnpbp.2010.06.014 Psychiatry 29, 1110–1118. doi: (SSTR5) gene associated with bipolar analog-octreotide on the adjuvant Maes, M., Mihaylova, I., Kubera, 10.1016/0006-3223(91)90253-I affective disorder. Mol. Psychiatry 7, arthritis in rat. Neuro Endocrinol. M., and Ringel, K. (2012). Acti- Molitch, M. E. (2008). Lanreotide 745–754. doi: 10.1038/sj.mp.4001049 Lett. 21, 121–126. vation of cell-mediated immunity Autogel in the management of Olias, G., Viollet, C., Kusserow, Lauritsen, M. B., Nyegaard, M., Betan- in depression: association with acromegaly. Drugs 68, 724. doi: H., Epelbaum, J., and Meyerhof, cur, C., Colineaux, C., Josiassen, T. L., inflammation, melancholia, clini- 10.2165/00003495-200868050-00014 W. (2004). Regulation and func- Kruse, T. A., et al. (2003). Analysis cal staging and the fatigue and Mollenholt, P., Rawal, N., Gordh, T. Jr., tion of somatostatin receptors. J. of transmission of novel polymor- somatic symptom cluster of depres- and Olsson,Y.(1994). Intrathecal and Neurochem. 89, 1057–1091. doi: phisms in the somatostatin receptor 5 sion. Prog. Neuropsychopharmacol. epidural somatostatin for patients 10.1111/j.1471-4159.2004.02402.x (SSTR5) gene in patients with autism. Biol. Psychiatry 36, 169–175. doi: with cancer. Analgesic effects and Padmos, R. C., Hillegers, M. H., Kni- Am. J. Med. Genet. B Neuropsy- 10.1016/j.pnpbp.2011.09.006 postmortem neuropathologic inves- jff, E. M., Vonk, R., Bouvy, A., chiatr. Genet. 121B,100–104. doi: Mancuso, C., Scapagini, G., Curro, D., tigations of spinal cord and nerve Staal, F. J., et al. (2008). A dis- 10.1002/ajmg.b.20050 Giuffrida Stella, A. M., De Marco, roots. Anesthesiology 81, 534–542. criminating messenger RNA signa- Leach, L. S., Christensen, H., Mack- C., Butterfield, D. A., et al. (2007). doi: 10.1097/00000542-199409000- ture for bipolar disorder formed by innon, A. J., Windsor, T. D., and Mitochondrial dysfunction, free rad- 00004 an aberrant expression of inflam- Butterworth, P. (2008). Gender dif- ical generation and cellular stress Moller, L. N., Stidsen, C. E., Hartmann, matory genes in monocytes. Arch. ferences in depression and anxiety response in neurodegenerative dis- B., and Holst, J. J. (2003). Somato- Gen. Psychiatry 65, 395–407. doi: across the adult lifespan: the role of orders. Front. Biosci. 12:1107–1123. statin receptors. Biochim. Biophys. 10.1001/archpsyc.65.4.395 psychosocial mediators. Soc. Psychia- doi: 10.2741/2130 Acta 1616, 1–84. doi: 10.1016/S0005- Pallis, E., Vasilaki, A., Fehlmann, D., try Psychiatr. Epidemiol. 43, 983–998. Markram, H., Toledo-Rodriguez, M., 2736(03)00235-9 Kastellakis, A., Hoyer, D., Spyraki, C., doi: 10.1007/s00127-008-0388-z Wang, Y., Gupta, A., Silberberg, G., Montminy, M. R., Sevarino, K. A., et al. (2009). Antidepressants influ- Lee, S., Hjerling-Leffler, J., Zagha, and Wu, C. (2004). Interneurons Wagner, J. A., Mandel, G., and Good- ence somatostatin levels and receptor E., Fishell, G., and Rudy, B. of the neocortical inhibitory system. man, R. H. (1986). Identification pharmacology in brain. Neuropsy- (2010). The largest group of Nat. Rev. Neurosci. 5, 793–807. doi: of a cyclic-AMP-responsive element chopharmacology 34, 952–963. doi: superficial neocortical GABAergic 10.1038/nrn1519 within the rat somatostatin gene. 10.1038/npp.2008.133 interneurons expresses ionotropic Martel, G., Dutar, P., Epelbaum, J., Proc. Natl. Acad. Sci. U.S.A. 83, 6682– Pallis, E. G., Spyraki, C., and Thermos, serotonin receptors. J. Neu- and Viollet, C. (2012). Somatostatin- 6686. doi: 10.1073/pnas.83.18.6682 K. (2006). Chronic antidepressant rosci. 30, 16796–16808. doi: ergic systems: an update on brain Morris, H. M., Hashimoto, T., and treatment modulates the release of 10.1523/JNEUROSCI.1869-10.2010 functions in normal and pathological Lewis, D. A. (2008). Alterations somatostatin in the rat nucleus

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 110 | 10

“fphar-04-00110” — 2013/9/5 — 15:44 — page 10 — #10 Lin and Sibille Somatostatin deficits in mood disorders

accumbens. Neurosci. Lett. 395, et al. (2003). Novel sst(4)-selective somatostatin receptor in rat brain. J. Res. Brain Res. Rev. 33, 228–241. doi: 76–81. doi: 10.1016/j.neulet.2005. somatostatin (SRIF) agonists. 1. Lead Neurosci. 20, 3785–3797. 10.1016/S0165-0173(00)00031-X 10.055 identification using a betide scan. J. Schulz, S., Handel, M., Schreff, M., Stafford, R. S., Ausiello, J. C., Misra, Patel, Y. C. (1999). Somatostatin Med. Chem. 46, 5579–5586. doi: Schmidt, H., and Hollt, V. (2000). B., and Saglam, D. (2000). National and its receptor family. Front. 10.1021/jm030243c Localization of five somatostatin patterns of depression treatment in Neuroendocrinol. 20:157–198. doi: Robas, N., Mead, E., and Fidock, M. receptors in the rat central nervous primary care. Prim. Care Compan- 10.1006/frne.1999.0183 (2003). MrgX2 is a high potency system using subtype-specific anti- ion J. Clin. Psychiatry 2, 211–216. doi: Paus, T., Keshavan, M., and Giedd, J. N. cortistatin receptor expressed in bodies. J. Physiol. Paris 94, 259– 10.4088/PCC.v02n0603 (2008). Why do many psychiatric dis- dorsal root ganglion. J. Biol. 264. doi: 10.1016/S0928-4257(00) Stefanescu, C., and Ciobica, A. (2012). orders emerge during adolescence? Chem. 278, 44400–44404. doi: 00212-6 The relevance of oxidative stress Nat. Rev. Neurosci. 9, 947–957. doi: 10.1074/jbc.M302456200 Sharif, N., Gendron, L., Wowchuk, status in first episode and recur- 10.1038/nrn2513 Rocheville, M., Lange, D. C., Kumar, U., J., Sarret, P., Mazella, J., Beaudet, rent depression. J. Affect. Disord. Petrozzi, L., Ricci, G., Giglioli, N. Patel, S. C., Patel, R. C., and Patel,Y.C. A., et al. (2007). Coexpression 143, 34–38. doi: 10.1016/j.jad.2012. J., Siciliano, G., and Mancuso, M. (2000). Receptors for dopamine and of somatostatin receptor subtype 5 05.022 (2007). Mitochondria and neurode- somatostatin: formation of hetero- affects internalization and trafficking Stroh, T., Kreienkamp, H. J., and generation. Biosci. Rep. 27, 87–104. oligomers with enhanced functional of somatostatin receptor subtype 2. Beaudet, A. (1999). Immuno- doi: 10.1007/s10540-007-9038-z activity. Science 288, 154–157. doi: Endocrinology 148, 2095–2105. doi: histochemical distribution of the Pivonello, R., Ferone, D., Lam- 10.1126/science.288.5463.154 10.1210/en.2006-1266 somatostatin receptor subtype berts, S. W., and Colao, A. Rossor, M. N., Emson, P. C., Moun- Sharma, R. P., Bissette, G., Janicak, P. 5 in the adult rat brain: pre- (2005). Cabergoline plus lanreotide tjoy, C. Q., Roth, M., and Iversen, G., Davis, J. M., and Nemeroff, C. dominant expression in the basal for ectopic Cushing’s syndrome. N. L. L. (1980). Reduced amounts of B. (1995). Elevation of CSF somato- forebrain. J. Comp. Neurol. 412, Engl. J. Med. 352, 2457–2458. doi: immunoreactive somatostatin in the statin concentrations in mania. Am. 69–82. doi: 10.1002/(SICI)1096- 10.1056/NEJM200506093522322 temporal cortex in senile dementia J. Psychiatry 152, 1807–1809. 9861(19990913)412:1<69::AID- Ponomarev, I., Rau, V., Eger, E. I., Har- of Alzheimer type. Neurosci. Lett. Sheppard, M., Shapiro, B., Pimstone, CNE5>3.0.CO;2-V ris, R. A., and Fanselow, M. S. (2010). 20, 373–377. doi: 10.1016/0304- B., Kronheim, S., Berelowitz, M., and Strowski, M. Z., Kohler, M., Chen, Amygdala transcriptome and cellu- 3940(80)90177-9 Gregory, M. (1979). Metabolic clear- H. Y., Trumbauer, M. E., Li, Z., lar mechanisms underlying stress- Rubinow, D. R., Gold, P. W., Post, ance and plasma half-disappearance Szalkowski, D., et al. (2003). Somato- enhanced fear learning in a rat model R. M., and Ballenger, J. C. (1985). time of exogenous somatostatin in statin receptor subtype 5 regulates of posttraumatic stress disorder. CSF somatostatin in affective ill- man. J. Clin. Endocrinol. Metab. 48, secretion and glucose home- Neuropsychopharmacology 35, 1402– ness and normal volunteers. Prog. 50–53. doi: 10.1210/jcem-48-1-50 ostasis. Mol. Endocrinol. 17, 93–106. 1411. doi: 10.1038/npp.2010.10 Neuropsychopharmacol. Biol. Psychi- Sibille, E., Morris, H. M., Kota, doi: 10.1210/me.2001-0035 Post, R. M., Rubinow, D. R., Kling, atry 9, 393–400. doi: 10.1016/0278- R. S., and Lewis, D. A. (2011). Szolcsanyi, J., Helyes, Z., Oroszi, M. A., Berrettini, W., and Gold, P. 5846(85)90192-7 GABA-related transcripts in the G., Nemeth, J., and Pinter, E. W. (1988). Neuroactive substances Rubinow, D. R., Gold, P.W., Post, R. M., dorsolateral prefrontal cortex in (1998). Release of somatostatin and in cerebrospinal fluid. Normal and Ballenger, J. C., Cowdry, R., Bollinger, mood disorders. Int. J. Neuropsy- its role in the mediation of the pathological regulatory mechanisms. J., et al. (1983). CSF somatostatin in chopharmacol. 14, 721–734. doi: anti-inflammatory effect induced by Ann. N. Y. Acad. Sci. 531, 15– affective illness. Arch. Gen. Psychia- 10.1017/S1461145710001616 antidromic stimulation of sensory 28. doi: 10.1111/j.1749-6632.1988. try 40, 409–412. doi: 10.1001/arch- Sicuteri, F., Geppetti, P., Marabini, S., fibres of rat sciatic nerve. Br. tb31808.x psyc.1983.01790040063009 and Lembeck, F. (1984). Pain relief J. Pharmacol. 123, 936–942. doi: Prosperini, E., Rizzi, M., Fuma- Rudy, B., Fishell, G., Lee, S., and by somatostatin in attacks of clus- 10.1038/sj.bjp.0701685 galli, F., Tarizzo, G., Samanin, R., Hjerling-Leffler, J. (2011). Three ter headache. Pain 18, 359–365. doi: Tallent, M. K., and Qiu, C. and Bendotti, C. (1997). Acute groups of interneurons account for 10.1016/0304-3959(84)90048-4 (2008). Somatostatin: an endoge- and chronic treatments with citalo- nearly 100% of neocortical GABAer- Siehler, S., Nunn, C., Hannon, J., Feuer- nous antiepileptic. Mol. Cell. pram lower somatostatin levels in gic neurons. Dev. Neurobiol. 71, bach, D., and Hoyer, D. (2008). Endocrinol. 286, 96–103. doi: rat brain striatum through dif- 45–61. doi: 10.1002/dneu.20853 Pharmacological profile of somato- 10.1016/j.mce.2007.12.004 ferent mechanisms. J. Neurochem. Saveanu, A., Lavaque, E., Gunz, G., Bar- statin and cortistatin receptors. Mol. Tamminga, C. A., Foster, N. L., Fedio, 69, 206–213. doi: 10.1046/j.1471- lier, A., Kim, S., Taylor, J. E., et al. Cell. Endocrinol. 286, 26–34. doi: P., Bird, E. D., and Chase, T. N. 4159.1997.69010206.x (2002). Demonstration of enhanced 10.1016/j.mce.2007.12.007 (1987). Alzheimer’s disease: low cere- Rajkowska, G., O’Dwyer, G., Teleki, potency of a chimeric somatostatin- Sohal, R. S., and Weindruch, R. (1996). bral somatostatin levels correlate with Z., Stockmeier, C. A., and Miguel- dopamine molecule, BIM-23A387, Oxidative stress, caloric restriction, impaired cognitive function and cor- Hidalgo, J. J. (2007). GABAergic in suppressing growth hormone and and aging. Science 273, 59–63. doi: tical metabolism. Neurology 37, 161– neurons immunoreactive for prolactin secretion from human pitu- 10.1126/science.273.5271.59 165. doi: 10.1212/WNL.37.1.161 calcium binding proteins are itary somatotroph adenoma cells. Song, C., and Wang, H. (2011). Tan, Z., Hu, H., Huang, Z. J., reduced in the prefrontal cortex J. Clin. Endocrinol. Metab. 87, Cytokines mediated inflamma- and Agmon, A. (2008). Robust in major depression. Neuropsy- 5545–5552. doi: 10.1210/jc.2002- tion and decreased neurogenesis but delayed thalamocortical activa- chopharmacology 32, 471–482. doi: 020934 in animal models of depres- tion of dendritic-targeting inhibitory 10.1038/sj.npp.1301234 Schmid, H. A., and Schoeffter, P.(2004). sion. Prog. Neuropsychopharmacol. interneurons. Proc. Natl. Acad. Reinikainen, K. J., Koponen, Functional activity of the multi- Biol. Psychiatry 35, 760–768. doi: Sci. U.S.A. 105, 2187–2192. doi: H., Jolkkonen, J., and Riekki- ligand analog SOM230 at human 10.1016/j.pnpbp.2010.06.020 10.1073/pnas.0710628105 nen, P. J. (1990). Decreased recombinant somatostatin receptor Sorce, S., and Krause, K. H. (2009). Taura, P., Planella, V., Balust, J., Bel- somatostatin-like immunoreac- subtypes supports its usefulness in NOX enzymes in the central nervous tran, J., Anglada, T., Carrero, E., tivity in the cerebrospinal fluid neuroendocrine tumors. Neuroen- system: from signaling to disease. et al. (1994). Epidural somatostatin of chronic schizophrenic patients docrinology 80(Suppl. 1), 47–50. doi: Antioxid. Redox. Signal. 11, 2481– as an analgesic in upper abdomi- with cognitive impairment. Psy- 10.1159/000080741 2504. doi: 10.1089/ars.2009.2578 nal surgery: a double-blind study. chiatry Res. 33, 307–312. doi: Schreff, M., Schulz, S., Handel, M., Spier, A. D., and de Lecea, L. (2000). Pain 59, 135–140. doi: 10.1016/0304- 10.1016/0165-1781(90)90047-9 Keilhoff, G., Braun, H., Pereira, G., Cortistatin: a member of the somato- 3959(94)90057-4 Rivier, J., Erchegyi, J., Hoeger, C., et al. (2000). Distribution, target- statin neuropeptide family with dis- Tostivint, H., Lihrmann, I., and Miller, C., Low, W., Wenger, S., ing, and internalization of the sst4 tinct physiological functions. Brain Vaudry, H. (2008). New insight

www.frontiersin.org September 2013 | Volume 4 | Article 110 | 11

“fphar-04-00110” — 2013/9/5 — 15:44 — page 11 — #11 Lin and Sibille Somatostatin deficits in mood disorders

into the molecular evolution of Vezzani,A., and Hoyer, D. (1999). Brain C. (2003). Opportunities in somato- and intra-septal microinfusions. the somatostatin family. Mol. somatostatin: a candidate inhibitory statin research: biological, chemi- Psychopharmacology (Berl.) 216, Cell. Endocrinol. 286, 5–17. doi: role in seizures and epileptogene- cal and therapeutic aspects. Nat. 557–567. doi: 10.1007/s00213- 10.1016/j.mce.2008.02.029 sis. Eur. J. Neurosci. 11, 3767– Rev. Drug Discov. 2, 999–1017. doi: 011-2248-x Tringali, G., Greco, M. C., Lisi, L., 3776. doi: 10.1046/j.1460-9568.1999. 10.1038/nrd1255 Zeyda, T., Diehl, N., Paylor, R., Pozzoli, G., and Navarra, P. (2012). 00838.x Wei, J., Xu, H., Davies, J. L., and Brennan, M. B., and Hochgeschwen- Cortistatin modulates the expres- Viollet, C., Lepousez, G., Loudes, C., Hemmings, G. P. (1992). Increase der, U. (2001). Impairment in sion and release of corticotrophin Videau, C., Simon, A., and Epel- of plasma IL-6 concentration with motor learning of somatostatin null releasing hormone in rat brain. baum, J. (2008). Somatostatiner- age in healthy subjects. Life Sci. mutant mice. Brain Res. 906, 107– Comparison with somatostatin gic systems in brain: networks and 51, 1953–1956. doi: 10.1016/0024- 114. doi: 10.1016/S0006-8993(01) and octreotide. Peptides 34, 353– functions. Mol. Cell. Endocrinol. 3205(92)90112-3 02563-X 359. doi: 10.1016/j.peptides.2012. 286, 75–87. doi: 10.1016/j.mce.2007. Weiss, S. R., Nguyen, T., Rubinow, Zeyda, T., and Hochgeschwender, U. 02.004 09.007 D. R., Helke, C. J., Narang, P. K., (2008). Null mutant mouse mod- Tripp, A., Kota, R. S., Lewis, D. Viollet, C., Vaillend, C., Videau, Post, R. M., et al. (1987). Lack els of somatostatin and cortis- A., and Sibille, E. (2011). Reduced C., Bluet-Pajot, M. T., Ungerer, of effect of chronic carbamazepine tatin, and their receptors. Mol. somatostatin in subgenual anterior A., L’Héritier, A., et al. (2000). on brain somatostatin in the rat. J. Cell. Endocrinol. 286, 18–25. doi: cingulate cortex in major depression. Involvement of sst2 somatostatin Neural. Transm. 68, 325–333. doi: 10.1016/j.mce.2007.11.029 Neurobiol. Dis. 42, 116–124. doi: receptor in locomotor, exploratory 10.1007/BF02098507 10.1016/j.nbd.2011.01.014 activity and emotional reactivity in Wilson, N. R., Runyan, C. A., Conflict of Interest Statement: The Tripp, A., Oh, H., Guilloux, J. P., mice. Eur. J. Neurosci. 12, 3761– Wang, F. L., and Sur, M. (2012). authors declare that the research was Martinowich, K., Lewis, D. A., and 3770. doi: 10.1046/j.1460-9568.2000. Division and subtraction by dis- conducted in the absence of any com- Sibille, E. (2012). Brain-derived 00249.x tinct cortical inhibitory networks in mercial or financial relationships that neurotrophic factor signaling Wang, A. Y., Lohmann, K. M., Yang, vivo. Nature 488, 343–348. doi: could be construed as a potential con- and subgenual anterior cingu- C. K., Zimmerman, E. I., Panta- 10.1038/nature11347 flict of interest. late cortex dysfunction in major zopoulos, H., Herring, N., et al. Xu, H., Jeong, H. Y., Tremblay, depressive disorder. Am. J. Psy- (2011). Bipolar disorder type 1 and R., and Rudy, B. (2013). Neo- Received: 25 May 2013; accepted: 13 chiatry 169, 1194–1202. doi: schizophrenia are accompanied by cortical somatostatin-expressing August 2013; published online: 09 10.1176/appi.ajp.2012.12020248 decreased density of parvalbumin- GABAergic interneurons disin- September 2013. Trivedi, M. H., Rush, A. J., Wisniewski, and somatostatin-positive interneu- hibit the thalamorecipient layer Citation: Lin L-C and Sibille E (2013) S. R., Nierenberg, A. A., Warden, D., rons in the parahippocampal 4. Neuron 77, 155–167. doi: Reduced brain somatostatin in mood Ritz, L., et al. (2006). Evaluation of region. Acta Neuropathol. 122, 10.1016/j.neuron.2012.11.004 disorders: a common pathophysiologi- outcomes with citalopram for depres- 615–626. doi: 10.1007/s00401-011- Xu, X., Roby, K. D., and Call- cal substrate and drug target? Front. sion using measurement-based care 0881-4 away, E. M. (2010). Immunochemical Pharmacol. 4:110. doi: 10.3389/ in STAR*D: implications for clinical Wang, X. M., Tresham, J. J., Coghlan, J. characterization of inhibitory mouse fphar.2013.00110 practice. Am. J. Psychiatry 163, 28–40. P., and Scoggins, B. A. (1987). Intrac- cortical neurons: three chemically This article was submitted to Neurophar- doi: 10.1176/appi.ajp.163.1.28 erebroventricular infusion of a cyclic distinct classes of inhibitory cells. J. macology, a section of the Frontiers in Vela, J., Gutierrez, A., Vitorica, J., and hexapeptide analogue of somato- Comp. Neurol. 518, 389–404. doi: Pharmacology. Ruano, D. (2003). Rat hippocam- statin inhibits hemorrhage-induced 10.1002/cne.22229 Copyright © 2013 Lin and Sibille. This is pal GABAergic molecular markers ACTH release. Neuroendocrinol- Xue, S., Jia, L., and Jia, J. an open-access article distributed under are differentially affected by ageing. ogy 45, 325–327. doi: 10.1159/ (2009). Association between somato- the terms of the Creative Commons J. Neurochem. 85, 368–377. doi: 000124747 statin gene polymorphisms and spo- Attribution License (CC BY). The use, 10.1046/j.1471-4159.2003.01681.x Warren, T. G., and Shields, D. (1984). radic Alzheimer’s disease in Chinese distribution or reproduction in other Vepsalainen, S., Helisalmi, S., Koivisto, Expression of preprosomatostatin population. Neurosci. Lett. 465, forums is permitted, provided the orig- A. M., Tapaninen, T., Hiltunen, M., in heterologous cells: biosynthe- 181–183. doi: 10.1016/j.neulet.2009. inal author(s) or licensor are credited and Soininen, H. (2007). Somato- sis, posttranslational processing, and 09.002 and that the original publication in statin genetic variants modify the secretion of mature somatostatin. Yeung, M., Engin, E., and Treit, D. this journal is cited, in accordance with risk for Alzheimer’s disease among Cell 39, 547–555. doi: 10.1016/0092- (2011). Anxiolytic-like effects of accepted academic practice. No use, Finnish patients. J. Neurol. 254, 8674(84)90461-6 somatostatin isoforms SST 14 and distribution or reproduction is permit- 1504–1508. doi: 10.1007/s00415- Weckbecker, G., Lewis, I., Albert, R., SST 28 in two animal models (Rattus ted which does not comply with these 007-0539-2 Schmid, H. A., Hoyer, D., and Bruns, norvegicus) after intra-amygdalar terms.

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 110 | 12

“fphar-04-00110” — 2013/9/5 — 15:44 — page 12 — #12