Lujan Wickman13R

10
New insights into the therapeutic potential of Girk channels Rafael Luja ´n 1 , Ezequiel Marron Fernandez de Velasco 2 , Carolina Aguado 1 , and Kevin Wickman 2 1 Instituto de Investigacio ´ n en Discapacidades Neurolo ´ gicas (IDINE), Departamento de Ciencias Me ´ dicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain 2 Department of Pharmacology, University of Minnesota, 321 Church Street South East, Minneapolis, MN 55455, USA G protein-dependent signaling pathways control the activity of excitable cells of the nervous system and heart, and are the targets of neurotransmitters, clinically relevant drugs, and drugs of abuse. G protein-gated inwardly rectifying potassium (K + ) (Girk/Kir3) channels are a key effector in inhibitory signaling pathways. Girk- dependent signaling contributes to nociception and an- algesia, reward-related behavior, mood, cognition, and heart-rate regulation, and has been linked to epilepsy, Down syndrome, addiction, and arrhythmias. We dis- cuss recent advances in our understanding of Girk chan- nel structure, organization in signaling complexes, and plasticity, as well as progress on the development of subunit-selective Girk modulators. These findings offer new hope for the selective manipulation of Girk channels to treat a variety of debilitating afflictions. Introduction to Girk signaling Signal transduction involving inhibitory (G i/o ) G proteins titrates the excitability of neurons, cardiac myocytes, and endocrine cells, actions crucial for regulating mood and cognition, nociception and antinociception, reward, energy homeostasis, motor activity and coordination, hormone secretion, and cardiac output. Not surprisingly, dysregula- tion of G i/o -dependent signaling has been linked to several neurological and cardiac disorders. Given this, and because the efficacy of many clinically relevant and abused drugs relates to their actions on G i/o -dependent signaling, it is important that we understand the cellular, subcellular, and molecular details of how such signaling is organized, how it is regulated, and how and when it goes awry. Girk/Kir3 channels are a common effector for G i/o -de- pendent signaling pathways in the heart and nervous system [1,2]. Studies of mutant mice and a more limited set of linkage analyses have suggested that dysregulation of Girk signaling may contribute to particular human diseases and disorders (Table 1). Although this work sug- gests that therapeutic approaches targeting Girk channels may prove beneficial in some settings, there is legitimate concern that manipulation of Girk signaling would trigger profound and widespread off-target effects. The goal of this review is to highlight recent developments related to our understanding of Girk channel diversity, compartmentali- zation, and plasticity. These studies suggest new opportu- nities for selective manipulation of Girk signaling, efforts that could eventually lead to novel treatments for debili- tating human afflictions. Girk channel structure Girk channels are tetramers formed by differential multi- merization among the products of four genes: Girk1/Kir3.1/ Kcnj3, Girk2/Kir3.2/Kcnj6, Girk3/Kir3.3/Kcnj9, and Girk4/ Kir3.4/Kcnj5 [1,2] (Figure 1A). Each Girk subunit pos- sesses intracellular N- and C-terminal domains, and two transmembrane segments that flank a hydrophobic pore domain. Random assembly theoretically allows the forma- tion of many distinct Girk channel subtypes, and alterna- tive splicing of Girk1 and Girk2 gene mRNAs potentially adds an additional spice of diversity (e.g., [3,4]). However, two observations suggest that a more limited number of Girk channel subtypes exist in vivo: (i) Girk1 does not form functional homomultimeric channels [5,6], an observation attributable at least in part to the absence of an endoplas- mic reticulum (ER) export signal that is found in Girk2 and Girk4 [7]. Surprisingly, however, introduction of a single point mutation into the pore domain of Girk1 (F137S) is sufficient to yield functional homomeric channels [8]. Col- lectively, these findings suggest that, although ER export signals are important, other factors such as protein folding and stability may also influence the trafficking of Girk channels. (ii) The four Girk subunits exhibit overlapping but distinct expression patterns, and this is particularly evident in the nervous system [9,10]. Indeed, work in the past two decades has clarified the cell type-specific expres- sion patterns of Girk subunits (Box 1). Moreover, recent crystallography studies have begun to provide insight into the 3D structure of Girk channels, with resolution of the membrane-spanning and large portions of the intracellular domains of homomeric channels [11–16] (Figure 1B). Functional implications of Girk channel diversity In expression systems, Girk channels of various composi- tion including heteromers (Girk1/2, Girk1/3, Girk1/4, Review 0166-2236/$ see front matter ß 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tins.2013.10.006 Corresponding authors: Luja ´n, R. ([email protected]); Wickman, K. ([email protected]). TINS-1022; No. of Pages 10 Trends in Neurosciences xx (2013) 1–10 1

Transcript of Lujan Wickman13R

Page 1: Lujan Wickman13R

TINS-1022; No. of Pages 10

New insights into the therapeuticpotential of Girk channelsRafael Lujan1, Ezequiel Marron Fernandez de Velasco2, Carolina Aguado1, andKevin Wickman2

1 Instituto de Investigacio n en Discapacidades Neurolo gicas (IDINE), Departamento de Ciencias Me dicas, Facultad de Medicina,

Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain2 Department of Pharmacology, University of Minnesota, 321 Church Street South East, Minneapolis, MN 55455, USA

Review

G protein-dependent signaling pathways control theactivity of excitable cells of the nervous system andheart, and are the targets of neurotransmitters, clinicallyrelevant drugs, and drugs of abuse. G protein-gatedinwardly rectifying potassium (K+) (Girk/Kir3) channelsare a key effector in inhibitory signaling pathways. Girk-dependent signaling contributes to nociception and an-algesia, reward-related behavior, mood, cognition, andheart-rate regulation, and has been linked to epilepsy,Down syndrome, addiction, and arrhythmias. We dis-cuss recent advances in our understanding of Girk chan-nel structure, organization in signaling complexes, andplasticity, as well as progress on the development ofsubunit-selective Girk modulators. These findings offernew hope for the selective manipulation of Girk channelsto treat a variety of debilitating afflictions.

Introduction to Girk signalingSignal transduction involving inhibitory (Gi/o) G proteinstitrates the excitability of neurons, cardiac myocytes, andendocrine cells, actions crucial for regulating mood andcognition, nociception and antinociception, reward, energyhomeostasis, motor activity and coordination, hormonesecretion, and cardiac output. Not surprisingly, dysregula-tion of Gi/o-dependent signaling has been linked to severalneurological and cardiac disorders. Given this, and becausethe efficacy of many clinically relevant and abused drugsrelates to their actions on Gi/o-dependent signaling, it isimportant that we understand the cellular, subcellular,and molecular details of how such signaling is organized,how it is regulated, and how and when it goes awry.

Girk/Kir3 channels are a common effector for Gi/o-de-pendent signaling pathways in the heart and nervoussystem [1,2]. Studies of mutant mice and a more limitedset of linkage analyses have suggested that dysregulationof Girk signaling may contribute to particular humandiseases and disorders (Table 1). Although this work sug-gests that therapeutic approaches targeting Girk channels

0166-2236/$ – see front matter

� 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tins.2013.10.006

Corresponding authors: Lujan, R. ([email protected]);Wickman, K. ([email protected]).

may prove beneficial in some settings, there is legitimateconcern that manipulation of Girk signaling would triggerprofound and widespread off-target effects. The goal of thisreview is to highlight recent developments related to ourunderstanding of Girk channel diversity, compartmentali-zation, and plasticity. These studies suggest new opportu-nities for selective manipulation of Girk signaling, effortsthat could eventually lead to novel treatments for debili-tating human afflictions.

Girk channel structureGirk channels are tetramers formed by differential multi-merization among the products of four genes: Girk1/Kir3.1/Kcnj3, Girk2/Kir3.2/Kcnj6, Girk3/Kir3.3/Kcnj9, and Girk4/Kir3.4/Kcnj5 [1,2] (Figure 1A). Each Girk subunit pos-sesses intracellular N- and C-terminal domains, and twotransmembrane segments that flank a hydrophobic poredomain. Random assembly theoretically allows the forma-tion of many distinct Girk channel subtypes, and alterna-tive splicing of Girk1 and Girk2 gene mRNAs potentiallyadds an additional spice of diversity (e.g., [3,4]). However,two observations suggest that a more limited number ofGirk channel subtypes exist in vivo: (i) Girk1 does not formfunctional homomultimeric channels [5,6], an observationattributable at least in part to the absence of an endoplas-mic reticulum (ER) export signal that is found in Girk2 andGirk4 [7]. Surprisingly, however, introduction of a singlepoint mutation into the pore domain of Girk1 (F137S) issufficient to yield functional homomeric channels [8]. Col-lectively, these findings suggest that, although ER exportsignals are important, other factors such as protein foldingand stability may also influence the trafficking of Girkchannels. (ii) The four Girk subunits exhibit overlappingbut distinct expression patterns, and this is particularlyevident in the nervous system [9,10]. Indeed, work in thepast two decades has clarified the cell type-specific expres-sion patterns of Girk subunits (Box 1). Moreover, recentcrystallography studies have begun to provide insight intothe 3D structure of Girk channels, with resolution of themembrane-spanning and large portions of the intracellulardomains of homomeric channels [11–16] (Figure 1B).

Functional implications of Girk channel diversity

In expression systems, Girk channels of various composi-tion – including heteromers (Girk1/2, Girk1/3, Girk1/4,

Trends in Neurosciences xx (2013) 1–10 1

Page 2: Lujan Wickman13R

Table 1. Physiological and pathophysiological relevance of Girk signalinga

Girk gene(s) Mutation Phenotype Refs

Cardiovascular physiology

Heart rate Girk1, Girk4 Null Resting tachycardia [86,87]

Parasympathetic regulation Girk1, Girk4 Null Decreased chronotropic response [86,87]

Heart rate variability Girk4 Null Decreased variability [86]

Arrhythmia GIRK1, GIRK4 Multiple Long QT, atrial fibrillation [88–90]

Hypertension GIRK4 Multiple Associated with aldosteronism [91,92]

Nociception

Thermal Girk1, Girk2 Null Hyperalgesia [93,94]

Mechanical Girk2 Null Hyperalgesia [93]

Chemical Girk2 Null Hyperalgesia [93]

Antinociception

Opioid Girk1, Girk2 Null Decreased analgesia [93–97]

Girk3 Null Decreased sensitivity [98]

GIRK2 Polymorphism Increased dosing requirement [99,100]

a2 Adrenergic Girk2,Girk3 Null Decreased analgesia [93,95,98]

GABAB Girk2 Null Decreased analgesia [95]

Cholinergic Girk2 Null Decreased analgesia [95]

Cannabinoid Girk2, Girk3 Null Decreased sensitivity [95,98]

Reward

Motor activity Girk1, Girk2 Null Enhanced basal and cocaine-induced [101–103]

Natural rewards Girk2, Girk4 Null Elevated responding for food [104,105]

Girk2 Triploid Enhanced sucrose intake [58]

Self-administration Girk2, Girk3 Null Decreased (cocaine) [102]

Girk2 Null Enhanced consumption (ethanol) [106]

Dependence/withdrawal Girk2 & Girk3 Null Decreased opioid withdrawal [97]

Girk3 Null Decreased sedative-hypnotic withdrawal [107]

GIRK2 Polymorphism Association with ethanol intake and stress [108]

Learning/memory

Spatial learning/memory Girk4 Null Decreased recall [109]

Fear conditioning Girk2 Triploid Decreased contextual recall [58]

Anxiety Girk2 Null Anxiolysis [104,110]

Schizophrenia GIRK1 Polymorphism Genetic association [111]

Seizure/epilepsy Girk2 Null Increased spontaneous and PTZ-induced [74]

Energy homeostasis Girk4 Null Late-onset obesity [105]

Thermoregulation Girk2 Null Decreased drug-induced hypothermia [112]

Neurodevelopment Girk2 weaver Loss of granule and dopamine neurons [113]

aThe table lists outcomes from behavioral studies involving mice harboring mutant Girk subunits (null/knockout, triploid, or weaver) or from human linkage studies (GIRK

genes in uppercase) that identified polymorphisms or mutations in GIRK genes.

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

Girk2/3, Girk2/4) and homomers (Girk2 and Girk4) – dis-play K+ selectivity, inward rectification, and G protein-dependent gating [1,2]. Although it cannot form a func-tional homomer, Girk1 is an integral subunit of mostneuronal Girk channels and the cardiac Girk channel IKACh

[5,17]. Girk1 confers robust receptor-dependent activity toGirk heteromers, attributable in part to unique residuesfound in the pore and second transmembrane helix thatenhance single-channel conductance and opening proba-bility [8,18–20]. The intracellular C-terminal domain alsocontributes to the potentiating influence of Girk1 onGPCR-dependent heteromeric channel activity, likelydue to the presence of unique Gbg, Ga, and phosphatidy-linositol 4,5-bisphosphate (PIP2) interaction domains andphosphorylation sites [20–26].

The functional relevance of Girk channel subunit com-position is nicely illustrated in the ventral tegmental area(VTA). VTA dopamine neurons express Girk2 and Girk3,whereas VTA GABA neurons express Girk1, Girk2, andGirk3 [27,28]. VTA dopamine neurons are significantlyless sensitive than VTA GABA neurons to direct GABAB

2

receptor-dependent inhibition [27,28], consistent with therelatively low sensitivity of recombinant Girk2/3 hetero-mers to Gbg-dependent activation [29]. Interestingly, ec-topic expression of Girk1 or genetic ablation of Girk3enhanced the sensitivity of the Girk channel in VTA dopa-mine neurons to GABAB receptor-dependent inhibition[28]. The negative influence of Girk3 on the sensitivityof Girk2/3 heteromers to Gbg- and GABAB receptor-depen-dent activation may be linked to intrinsic structural ele-ments that weaken its interaction with Gbg or the couplingbetween Gbg binding and channel gating, an explanationsupported by the behavior of recombinant Girk2/3 hetero-mers [29]. Alternatively, Girk3-specific interactions withnegative regulatory proteins expressed in VTA dopamineneurons (Rgs2 and/or sorting nexin 27 [28,30], discussedbelow), interactions that are presumably precluded ormitigated by the presence of Girk1, may explain the dif-ferential sensitivity of Girk channels in VTA dopamine andGABA neurons to GABAB receptor activation. Regardlessof the mechanism, the molecular and cellular diversity ofGirk channels shapes the sensitivity of VTA dopamine

Page 3: Lujan Wickman13R

Box 1. Cellular and subcellular diversity of Girk channels

Although Girk channel expression has been reported in neuroendo-

crine cells and more recently in some cancer cells (e.g., [114–117]), the

expression and relevance of Girk channels is far better understood in

the heart and nervous system. Girk1/4 heteromers comprise the

muscarinic-gated atrial K+ channel IKACh, a crucial mediator of the

parasympathetic regulation of heart rate [5,86]. Although the proto-

typical neuronal Girk channel is thought to be the Girk1/2 heteromer,

multiple Girk channel subtypes are present in the rodent nervous

system. Indeed, Girk3 is expressed throughout the central nervous

system [9] and, although Girk4 expression is not prominent in the

brain, it is found in a few regions including the hypothalamus and

cerebellum [105,118]. Girk1, Girk2, and Girk3 are coexpressed in many

neuron populations, including hippocampal pyramidal neurons

[9,10]. By contrast, dopamine neurons of the VTA and substantia

nigra pars compacta (SNc) display Girk2/3 and Girk2a/c heteromers,

respectively [27,119]. The cerebellum exemplifies the molecular

diversity that can be achieved via differential subunit expression:

seven distinct Girk expression patterns were discerned within the

various neuronal subtypes in this brain region [118].

Girk channels are distributed mainly in the somato-dendritic

compartment of neurons (e.g., [10,96,118,120]). This distribution is

consistent with most studies showing that Girk channels selectively

mediate the postsynaptic inhibitory effects of neurotransmitters and

related drugs, while making little or no contribution to their

presynaptic inhibitory effects [121]. Some ultrastructural and

functional data, however, support the contention that Girk channels

contribute to presynaptic inhibition in some neurons [122–124]. Girk

channels of distinct subunit composition can also exist within

specific subcellular compartments of the same neuron. For exam-

ple, Girk1 and Girk2 show extensive colocalization across the

different dendritic layers of the hippocampus [120]. Their expression

levels vary among dendritic regions innervated by distinct synaptic

inputs in CA1 pyramidal cells, showing a significant increase from

proximal to distal dendrites. By contrast, Girk3 is more uniformly

distributed along the cell surface of pyramidal cells, including the

presynaptic terminal [10]. In addition, Girk2 and Girk3 are evenly

distributed along the postsynaptic density (PSD) of pyramidal cells

and Purkinje cells, whereas Girk1 is absent from excitatory synapses

and only observed at perisynaptic sites [10,96,120,123]. Finally, in

the subpopulation of parvalbumin-expressing interneurons consist-

ing mainly of basket and chandelier cells, Girk1, Girk2, and Girk3

share the same localization at very similar densities [125]. Collec-

tively, these data argue that distinct Girk channel subtypes exist in a

tissue/cell type- and subcellular compartment-dependent manner, a

scenario that enhances the prospects for selective and efficacious

therapeutic manipulation of Girk signaling.

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

output to GABAB receptor activation, and helps to explainthe intriguing pharmacodynamic differences between theGABAB receptor agonists baclofen (an anti-craving com-pound) and g-hydroxybutyric acid (GHB, a drug of abuse)[28].

Macromolecular organization of Girk signalingGirk channels are thought to exist in multiprotein com-plexes that include GPCRs, G proteins, and regulatoryproteins [1,2,31], a consensus that has emerged despitethe fact that relatively few protein–protein interactionsinvolving Girk channels have been demonstrated usingclassical biochemical approaches or native systems. In-deed, most reported interactions have involved overexpres-sion of Girk subunits and putative binding partners in celltypes that do not normally express Girk channels. Never-theless, data obtained using these approaches have beenvaluable in supporting a conceptual model that aligns withkey functional properties of GPCR–Girk signaling (e.g., Gi/o

coupling specificity, signaling kinetics).

A core signaling complex: GPCR–Gabg–Girk

The interaction with Gbg represents, from a structural andfunctional perspective, the best understood of the protein–protein interactions involving Girk channels. Gbg binds toGirk channels, strengthening the interaction between thechannel and PIP2, a required cofactor for channel gating[32,33]. Although biochemical and structure–functionapproaches have suggested multiple interaction domainsfor Gbg in all four Girk subunits [1,34], clear resolution ofthis crucial interaction was obtained recently with thecrystallization of a complex formed by Gbg and the Girk2homomer [16] (Figure 1B). Girk2 homomers possess fourbinding sites for Gbg found at the well-conserved cyto-plasmic interfaces between adjacent subunits that contrib-ute to formation of the extended cytoplasmic pore; Gbg

promotes a ‘pre-open’ state of the channel that is interme-diate between the closed state of the channel and the openconformation of a constitutively-active Girk2 homomer[15,16].

Girk channels also interact with Gai/o. The inactiveheterotrimeric G protein (Gai/o-GDP/Gbg), as well asGai/o-GDP and Gai/o-GTP, can bind to intracellulardomains of Girk channels [25,26,35–38]. Interactions be-tween Girk channels and Gai/o-GDP (either alone or in thecontext of the inactive heterotrimeric G protein) suppressbasal activity of Girk channels, while enhancing their Gprotein-dependent activation [25,39,40]. Moreover, the se-lective association between Girk channels and Gai/o likelyexplains in part the strict coupling specificity between Girkchannels and Gi/o-dependent signaling pathways in vivo[36,41]. This specificity may be reinforced further by selec-tive pre-coupling between certain GPCRs and Gai/o-GDP/Gbg [42], and/or by direct GPCR–Girk interactions [31].Indeed, several GPCRs that couple to Gi/o G proteins,including D2 and D4 dopamine receptors, and GABAB

receptors, have been shown to interact directly with Girkchannels [31,43,44].

Collectively, these studies support the vision of a com-pact core Girk signaling complex, wherein minor confor-mational changes triggered by agonist binding to GPCRunveils key protein–protein interaction interfaces [45].Organization of signaling elements within a multiproteincomplex affords several advantages over random, collision-based designs. The close spatial proximity of the relevantmolecules allows fast and efficient signaling, and the noiseor ‘crosstalk’ that might otherwise occur with nonspecificcollision events is minimized, creating a tailored intracel-lular response to an external stimulus. Moreover, thestrength or sensitivity of the signaling pathway can theo-retically be titrated, by altering macrocomplex composi-tion, to suit the needs of the cell under specificcircumstances. With respect to this latter point, twoGirk-interacting proteins warrant further discussion.

Sorting nexin 27 (SNX27)

Girk2c and Girk3 possess identical C-terminal class 1interaction motifs for PDZ domain-containing proteins (–ESKV) (Figure 1A). Using the distal C-terminal domain ofGirk3 as bait in an unbiased proteomic screen, sortingnexin 27 (SNX27) was identified as a Girk-interactingprotein [30]. SNX27 regulates the trafficking between cell

3

Page 4: Lujan Wickman13R

M1

M1

M1

M1

M1

M2

M2

M2

M2

M2

P

P

P

P

P

1

1

1

1

1

2

2

2

2

2

Girk1

N-terminus(A) (B)

0 100 200 300 400 500

Girk2A

Girk2C

Girk3

Girk4

ELETEEEE

EAEKAEAEDHEEEE

ESKV

Ser-9 VL

Amino acid sequenceC-terminus

PIP2

Turret

TM

CTD

Out

GγN-term Gβ

N-term

gg

Na+In

TRENDS in Neurosciences

Figure 1. Structure of G protein-gated inwardly rectifying potassium (K+) Girk channels. (A) Linear depiction of Girk channel subunits, including two prevalent Girk2 splice

variants expressed in the mouse nervous system. The four Girk subunits exhibit a high degree of homology in the membrane-spanning (M1, M2), extracellular (1, 2), and

pore (P) domains, with most inter-subunit differences observed in the distal N- and C-terminal domains. The Girk2 splice variants and Girk4 contain endoplasmic reticulum

(ER) export signals (ELETEEEE and EAEKAEAEDHEEEE, respectively) not found in the other subunits [7]. Girk2 also exhibits an N-terminal internalization motif (VL), whose

influence on channel trafficking is precluded by phosphorylation of Girk2(Ser9) [71]. Girk2C and Girk3 possess identical C-terminal PDZ interaction motifs (–ESKV). (B)

Structure of the Girk–Gbg complex from a side-view, with colors highlighting Girk2 (blue), Gb (red), Gg [green, with associated geranyl-geranyl (gg) lipid modification], PIP2

(yellow/orange sticks), Na+ ions (purple spheres), K+ ions (green spheres within the pore). Note that there are four Gbg binding sites per channel, and the Gbg facing the

viewer has been removed for clarity [16]. Other abbreviations: CTD, C-terminal domain; N-term, N-terminal domain; PIP2, phosphatidylinositol 4,5-bisphosphate; TM,

transmembrane domains.

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

surface and endosome of an array of neuronal signalingproteins [46], and has been implicated in Down syndromeand addiction [47,48]. SNX27 is the only member of thesorting nexin family that has a PDZ domain and it recog-nizes class I PDZ-binding motifs [49].

Amino acids immediately upstream from the ESKVmotif in Girk3 are crucial for promoting the Girk3–SNX27 interaction, and preclude its interaction withpost-synaptic density protein PSD95 [30,50]. SNX27 alsocontains a Ras association (RA) and a lipid-binding phox-homology (PX) domain. All three functional domains arecrucial for the proper function of SNX27 – which in thecontext of Girk signaling involves targeting Girk3-contain-ing channels to early endosomes, effectively reducing thesurface expression of Girk channels, and enhancing theexcitability of host neurons [30,50,51] (Figure 2). Interest-ingly, although SNX27 can bind to Girk2c, the surfacedistribution of homomeric Girk2c channels is unaffectedby SNX27 expression, suggesting that another factor(s)may influence the trafficking fates of Girk channels [52].

The upregulation of b isoform of SNX27 (SNX27b) inresponse to in vivo exposure to the psychostimulants co-caine and amphetamine is particularly intriguing in lightof recent observations that GABAB–Girk signaling is weak-ened by acute psychostimulant exposure in VTA dopamineand GABA neurons, and following chronic cocaine treat-ment in layer 5/6 pyramidal neurons of the medial prefron-tal cortex (mPFC) [47,53–55] (discussed below). Similarly,the association between loss of SNX27 and Down syndrome[48] is interesting given that Girk signaling is enhanced inthe hippocampus and cortex of a mouse model of Downsyndrome [56,57], and because Girk2 triploid mice recapit-ulated many of the neurological phenotypes associatedwith this syndrome [58]. Although altered SNX27 expres-sion and/or function will certainly impact upon a widearray of cell signaling pathways, these studies arguethat SNX27-dependent alterations in Girk signaling may

4

contribute to some of the cellular and behavioral deficitslinked to psychostimulant addiction and Down syndrome.

R7 RGS proteins

GPCR–Girk signaling is negatively modulated by RGS(regulator of G protein signaling) proteins [1,2]. RGS pro-teins enhance the GTPase activity of Ga subunits, acceler-ating the deactivation of G protein signaling followingagonist removal [59]. Accordingly, RGS proteins acceleratereceptor-induced Girk current deactivation kinetics,among other influences [60]. Discrete protein modulesconfer unique functionality to specific RGS proteins [59],and these – together with their unique cell/tissue expres-sion patterns – appear to promote interactions with Girkchannels. For example, the selective expression of Rgs2 inVTA dopamine neurons, and its preferential associationwith Girk3, weakens the coupling between GABAB recep-tors and the Girk2/3 heteromer [28]. Interestingly, changesin the expression of Rgs2 in response to chronic GHB (andmorphine) treatment correlated with altered GABAB–Girkcoupling in VTA dopamine neurons, and this neuroadapta-tion may contribute to the development of tolerance toGHB and other drugs of abuse.

Recent data show that Girk channels are modulated byRGS proteins in the R7 subfamily, which includes Rgs6,Rgs7, Rgs9, and Rgs11. The R7 RGS proteins possess adomain resembling the G protein Gg subunit (Gg-likedomain, or GGL) that promotes association with the atypi-cal 5th member of the G protein Gb family, Gb5 [61]. Thecrystal structure of the Rgs9–Gb5 complex reveals that theinteraction between GGL and Gb5 resembles that ob-served in conventional Gbg dimers [62]. Not surprisingly,therefore, complexes formed between R7 RGS proteins andGb5 (Rgs/Gb5) bind to Girk channels and modulate thekinetics of M2 muscarinic receptor/Girk signaling in atrialcardiomyocytes (Rgs6/Gb5) [63,64] and GABAB receptor/Girk signaling in hippocampal CA1 pyramidal neurons

Page 5: Lujan Wickman13R

EE

Girk2Key:Girk2

Girk2 Girk2Girk3

Girk1Girk3 SNX27Girk1

EE

RE RE

LE-LyLE-Ly

TRENDS in Neurosciences

Figure 2. Subunit-dependent regulation of Girk channel trafficking by sorting nexin 27 (SNX27). Neuronal Girk channels are thought to internalize into early endosomes

(EE), at which point they can be recycled back to the cell surface via recycling endosomes (RE) or diverted to late endosomes/lysosomes (LE-Ly) for degradation. Via a

selective physical and functional association with Girk3, SNX27 enhances the trafficking of Girk3-containing channels to early endosomes, leading ultimately to increased

lysosomal degradation and consequently, reduced numbers of Girk channels on the cell surface (right side of schematic). By contrast, SNX27 does not influence the

trafficking of Girk2 homomers or Girk1/2 heteromers (left side of schematic).

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

(Rgs7/Gb5) [44,65]. Moreover, Gb5 ablation enhanced thesensitivity of Girk channels to GABAB receptor stimulationin hippocampal neurons, indicating that Rgs/Gb5 com-plexes can also negatively influence GPCR–Girk couplingefficiency [65]. Interestingly, Rgs6/Gb5 complexes appearto modulate GABAB–Girk signaling in cerebellar granulecells [66], showing that complexes containing either Rgs6or Rgs7 are relevant to neuronal Girk signaling. In addi-tion, the Rgs/Gb5–Girk channel interaction may be con-trolled by the R7 RGS-binding protein R7BP [67],providing another layer of fine regulation of GPCR–Girksignaling and another potential therapeutic target.

Loss of the Rgs6/Gb5-dependent modulation of Girksignaling in the heart correlates with bradycardia andenhanced parasympathetic influence [63,64]. Because dys-regulation of the parasympathetic control of cardiac outputhas been linked to sick sinus syndrome, heart failure, andarrhythmia [68], selective targeting of the Rgs6/Gb5–Girkaxis may prove beneficial in clinical settings involvingcardiac disorders. Although the relevance of the Rgs7/Gb5-dependent modulation of GABAB–Girk signaling inthe hippocampus is not understood, mice lacking Gb5 weremore sensitive to the sedative effect of the GABAB agonistbaclofen [65]. Moreover, mice lacking R7BP exhibited en-hanced thermal nociceptive thresholds and augmentedanalgesic effects of opioids and baclofen, consistent withits proposed role as a facilitator of the Rgs/Gb5-dependentregulation of GPCR–Girk signaling [67]. Because en-hanced Girk signaling has been linked to depotentiation[69], a particular form of excitatory synaptic plasticity, andwith cognitive deficits associated with Down syndrome[58], it will be particularly important to explore the rela-tionship between the Rgs7/Gb5 regulation of Girk signal-ing and of hippocampal-dependent learning and memory.Moreover, it will be interesting to probe the involvement ofRgs/Gb5 complexes in other neuronal GPCR–Girk signal-ing pathways.

Plasticity of Girk signalingRecent work has shown that the strength and sensitivity ofneuronal Girk signaling is titratable and subject to regu-lation by multiple stimuli. The first clear example of Girksignaling plasticity came with the demonstration thatstimulation protocols that evoked NMDA receptor-depen-dent long-term potentiation (LTP) of glutamatergic neuro-transmission in hippocampal CA1 neurons alsostrengthened synaptic GABAB–Girk signaling [70]. Subse-quent work in cultured hippocampal pyramidal neuronsrevealed that neuronal activity triggered by NMDA recep-tor activation leads to a rapid increase in the levels of Girkchannels (Girk1/2) on the somatodendritic membrane, andenhanced Girk signaling via the A1 adenosine receptor[71]. Pharmacologic or genetic ablation of Girk signalingsuggested that this neuroadaptation is crucial for thedepotentiation of excitatory LTP [69].

Drugs of abuse

As documented in Table 1, Girk signaling shapes many ofthe behavioral effects of drugs of abuse, including opioids,psychostimulants (cocaine), and ethanol. Exposure todrugs of abuse can alter neuronal Girk signaling in durablefashion in the reward circuitry, the core of which consists ofinterconnected neurons in the VTA, mPFC, and nucleusaccumbens (NAc) [72]. For example, a single non-contin-gent exposure to cocaine suppressed GABAB–Girk signal-ing by 50% in VTA dopamine (but not SNc dopamine)neurons for several days [53], paralleling the better-under-stood enhancement of glutamatergic neurotransmissionoccurring in the same neurons [72]. Acute psychostimulantexposure also persistently suppressed GABAB–Girk sig-naling in VTA GABA neurons [54]. Finally, repeated co-caine exposure suppressed GABAB–Girk signaling in layer5/6 glutamatergic pyramidal neurons of the mPFC [55], themain source of glutamatergic input to the VTA and NAc.This neuroadaptation was specific for pyramidal neurons

5

Page 6: Lujan Wickman13R

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

in the prelimbic cortex (as compared to pyramidal neuronsin the infralimbic and motor cortices), and persisted formore than 1 month after the final cocaine injection. More-over, persistent suppression of Girk signaling in layer 5/6of the mPFC presensitized mice to the motor-stimulatoryeffects of acute cocaine. Although more work is necessary tounderstand the behavioral relevance of drug-inducedadaptations in Girk signaling in the reward circuitry, theseearly insights, together with the anatomic and cellularspecificity of the neuroadaptations, and their durability,suggest that they drive and/or contribute to the persistentexpression of addictive behaviors including drug-seeking,craving, and relapse.

Mechanism(s) of plasticity

Plasticity of Girk signaling triggered by neuronal activityand drugs of abuse involves the redistribution of Girk2-containing channels to and from the surface membrane(Figure 3). Enhanced Girk signaling in hippocampal

Girk

(A) (B)

(D) (E)

Axonterminal

Key:

Saline

Saline

Baclofen

Baclofen

Cocaine

Cocaine

GABABR

Spine

D

Excitatoryimput

Excitatoryimput

VTA dopamine neuron

Layer 5/6 pyramidal neuron

Figure 3. Plasticity of neuronal Girk signaling. Cocaine-induced suppression of Girk sign

(D–F). (A) A single cocaine injection reduced baclofen-induced (GABAB receptor-depende

days, required activation of D2 dopamine receptors, and correlated with a redistribution

intracellular sites (B,C) [53]. (D) Repeated cocaine administration suppressed baclofen-in

more than 1 month, required activation of D1 dopamine receptors, and correlated with a

receptors from the cell surface to intracellular sites (D,F) [55]. The blue line in the current

antagonist.

6

pyramidal neurons triggered by NMDA receptor activationwas linked to enhanced trafficking of Girk2-containingchannels from recycling endosomes to the cell surface[71]. By contrast, increased intracellular labeling at theexpense of surface labeling was seen for Girk2 in VTAdopamine and GABA neurons following acute cocaine andmethamphetamine treatment [53,54], respectively, and inlayer 5/6 pyramidal neurons following repeated cocaine[55]. Interestingly, a similar redistribution of the GABAB

receptor from the surface to inside the cell was observed inVTA GABA and layer 5/6 mPFC pyramidal neurons, butnot in VTA dopamine neurons, suggesting that differentmechanisms mediate Girk signaling plasticity in differentcell types. Consistent with this premise, the methamphet-amine-induced adaptation in VTA GABA neurons andcocaine-induced adaptation in layer 5/6 mPFC pyramidalneurons were both blocked by pretreatment with a D1

dopamine receptor antagonist, whereas the cocaine-induced adaptation in VTA dopamine neurons was D2

(C)

(F)

NMDA AMPAIntracellular

compartment

Axonterminal

SpineDendrite Dendrite

A dendrite DA dendrite

Excitatoryimput

Excitatoryimput

Control

Control

Cocaine

Cocaine

TRENDS in Neurosciences

aling in the ventral tegmental area (VTA) (A–C) and medial prefrontal cortex (mPFC)

nt) Girk currents in VTA dopamine neurons, an adaptation that persisted for up to 5

of Girk2-containing channels (but not of GABAB receptors) from the cell surface to

duced Girk currents in layer 5/6 pyramidal neurons, an adaptation that persisted for

phosphorylation-dependent redistribution of Girk2-containing channels and GABAB

traces shows that the baclofen-induced current was reversed by a GABAB receptor

Page 7: Lujan Wickman13R

Box 2. Outstanding questions

Pharmacology. The recent identification of the subunit-selective,

efficacious, and potent direct modulators of Girk signaling repre-

sents an important step forward in this field, and should provide a

foundation upon which efforts in synthetic chemistry, molecular

modeling/simulations, and crystallography can synergize to yield

new compounds with optimized pharmacodynamic and pharmaco-

kinetic properties. When available, these compounds will greatly

facilitate efforts directed at understanding the physiological rele-

vance of Girk channels, permitting us to move beyond studies in

mutant mice.

Macrocomplex formation. Although a large body of evidence

supports the existence of discrete signaling complexes containing

Girk channels in vivo, gaps in our understanding of Girk channel

trafficking and regulation argue that additional proteins that

influence Girk function – either via direct or indirect physical

interactions – remain to be discovered. Moreover, understanding

the molecular determinants of macrocomplex formation will be

helpful because it can potentially provide the means to manipulate

Girk signaling selectively in a subtle manner.

Physiological relevance. Further investigation into the physiolo-

gical relevance of Girk channels is necessary to understand better

the opportunities for beneficial therapeutic manipulation. The

development of novel pharmacological tools that can discriminate

among the various Girk channel subtypes existing in vivo will

facilitate this process, and will complement next-generation genetic

approaches that can give region- and cell type-dependent insights

into the function of Girk channels formed by distinct combinations

of subunits (including specific splice isoforms). In addition, efforts

that build on the growing evidence linking mutations or polymorph-

isms in GIRK genes to human disease will be invaluable.

Plasticity. We are only now beginning to understand the triggers

and mechanisms underlying the plasticity in Girk signaling. Going

forward, it will be important to understand more about the

mechanisms involved in the dynamic modulation of Girk channel

trafficking, including differences that may exist between distinct

drugs of abuse, neuron populations, Girk channel subtypes, and

GPCR–Girk combinations. It will be particularly interesting to

explore the potential relationship between SNX27, the phosphor-

egulation of Girk trafficking, and the drug-induced adaptations in

Girk signaling seen in the reward circuitry. Of course, understanding

the physiological (and perhaps pathophysiological) relevance of the

adaptations is the ultimate goal.

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

dopamine receptor-dependent. Moreover, GABAB–Girksignaling (but not somatodendritic inhibitory signalingvia D2 dopamine or a2 adrenergic receptors, or somatoden-dritic GABAB-dependent signaling that did not involveGirk channels) was suppressed by repeated cocaine inlayer 5/6 mPFC pyramidal neurons, whereas Girk signal-ing via both the GABAB and D2 dopamine receptor wassuppressed by acute cocaine in VTA dopamine neurons.These observations are reminiscent of the selective en-hancement by neuronal activity of Girk signaling via theA1 adenosine (but not GABAB) receptor [69], and suggestthat some forms of Girk signaling plasticity are compart-mentalized within neurons, and are presumably driven bythe GPCR or other proteins within the signaling macro-complex.

The trafficking of Girk channels and GPCR–Girk com-plexes to and from the cell surface that underlies the formsof plasticity described above appears to be regulated byphosphorylation. For example, the potentiation of synapticGABAB–Girk signaling triggered in parallel with LTP ofglutamatergic neurotransmission was dependent on Ca2+/calmodulin-dependent protein kinase (CaMKII) activity[70]. In addition, in cultured hippocampal neurons, CaM-KII activation (via prolonged morphine treatment, activa-tion of metabotropic glutamate receptors, or aconstitutively active CaMKII mutant) shifted the distribu-tion of Girk2 from dendritic shafts to spines, leading toenhanced Girk signaling via 5-HT receptors and decreasedGABAB–Girk signaling [73]. Although the direct moleculartarget of CaMKII was not determined in these studies,Girk2(Ser9) is a reasonable candidate. Phosphorylation ofGirk2(Ser9), which sits upstream of a unique internaliza-tion motif (VL) [7], suppresses surface trafficking of Girk2-containing channels [71]. Conversely, dephosphorylationof Girk2(Ser9) via protein phosphatase 1 (PP1) promotessurface trafficking of Girk2-containing channels from recy-cling endosomes, explaining the activity-dependent poten-tiation of Girk signaling linked to the depotentiation ofLTP [69,71]. Although dephosphorylation of Girk2(Ser9)promotes enhanced surface distribution of Girk channels,psychostimulant-induced suppression of GABAB–Girk sig-naling in VTA GABA and layer 5/6 mPFC neurons is morelikely related to the phosphorylation status of the GABAB

receptor [54,55]. Surprisingly, despite the durable natureof the drug-induced suppression of GABAB–Girk signalingin these pyramidal neurons, acute intracellular treatmentwith okadaic acid, an inhibitor of protein phosphatasesPP1/PP2a, restored normal Girk signaling.

Pharmacologic manipulation of Girk channelsGiven the broad distributions and roles of Girk channels inthe nervous system, and their contributions to cardiac andendocrine physiology, global and direct pharmacologicmanipulation of Girk signaling should evoke an array ofconsequences, many undesirable (Table 1). Indeed, globalconstitutive ablation of Girk2 triggers an array of pheno-types, most notably a shortened lifespan due to spontane-ous lethal seizures [74]. Although constitutive ablation ofother Girk subunits correlates with less-severe pheno-types, the full therapeutic potential associated with inhi-biting or enhancing Girk signaling will likely not be

achieved without regional and/or Girk subunit-selectivemanipulation. For example, an agonist with selectivity forGirk2/3 heteromers, the Girk channel subtype thatappears to be uniquely expressed in VTA dopamine neu-rons [28], could be useful as an anti-craving compound. Inaddition, drugs that selectively activate or inhibit Girk1/4heteromers, and which cannot pass the blood–brain barri-er, could represent efficacious therapies for particulartypes of arrhythmias. Indeed, two drugs that can inhibitGirk1/4 heteromers (NTC-801 and NIP-151) showed prom-ise in preclinical studies in the treatment of atrial fibrilla-tion [75,76].

Although Girk channels are activated in a G protein-independent manner by ethanol [77,78], volatile anes-thetics [79,80], and the flavonoid naringin [81], and areblocked by an array of psychoactive compounds (many ofwhich are clinically-relevant) (e.g., [82]), most of the com-pounds have other primary molecular targets and/or thereis little evidence for Girk subtype-specificity or pharmaco-kinetic advantages. Recently, however, a new class ofsubunit-selective, efficacious, potent, and direct-actingGirk channel agonists and antagonists was identified[83–85]. The prototype (ML297) is strikingly selective for

7

Page 8: Lujan Wickman13R

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

Girk1-containing heteromers, and was efficacious in micein delaying seizure onset in a maximal electroshock modelof epilepsy, and was able to prevent convulsions andlethality in a chemically induced epilepsy model [84].Accordingly, this family of compounds and derivativesshould afford an excellent opportunity to investigate thepotential therapeutic benefits associated with direct acti-vation or inhibition of Girk1-containing channels, and mayserve as a platform for the identification of compounds thatcan discriminate between a wide range of channel sub-types.

Concluding remarksAlthough several outstanding questions remain (Box 2),efforts by many research groups over the last two decadeshave revealed key functional, structural, and regulatoryfeatures of Girk channels. This body of evidence, combinedwith our evolving understanding of Girk channel contribu-tions to physiology and disease, and a continually improv-ing capacity for pharmacologic manipulation, set the stagefor an exciting future of investigations into the therapeuticpotential of this interesting and important channel class.Such efforts hold the promise of yielding novel therapeuticapproaches to the treatment of many forms of neurological,cardiovascular, and neuroendocrine disorders.

AcknowledgmentsThis work was supported by National Institutes of Health (NIH) grants toK.W. (MH061933, HL105550, and DA034696) and by the SpanishMinistry of Science and Innovation BFU2012-38348 and CONSOLI-DER-Ingenio CSD2008-0000 (to R.L.). The authors thank members of theWickman and Lujan laboratories for their suggestions for the manuscript.

References1 Luscher, C. and Slesinger, P.A. (2010) Emerging roles for G protein-

gated inwardly rectifying potassium (GIRK) channels in health anddisease. Nat. Rev. Neurosci. 11, 301–315

2 Hibino, H. et al. (2010) Inwardly rectifying potassium channels: theirstructure, function, and physiological roles. Physiol. Rev. 90, 291–366

3 Wei, J. et al. (1998) Characterization of murine Girk2 transcriptisoforms: structure and differential expression. Genomics 51, 379–390

4 Zhu, L. et al. (2001) Cloning and characterization of G protein-gatedinward rectifier K+ channel (GIRK1) isoforms from heart and brain. J.Mol. Neurosci. 16, 21–32

5 Krapivinsky, G. et al. (1995) The G-protein-gated atrial K+ channelIKACh is a heteromultimer of two inwardly rectifying K+-channelproteins. Nature 374, 135–141

6 Kennedy, M.E. et al. (1999) GIRK4 confers appropriate processing andcell surface localization to G-protein-gated potassium channels. J.Biol. Chem. 274, 2571–2582

7 Ma, D. et al. (2002) Diverse trafficking patterns due to multiple trafficmotifs in G protein-activated inwardly rectifying potassium channelsfrom brain and heart. Neuron 33, 715–729

8 Chan, K.W. et al. (1996) Control of channel activity through a uniqueamino acid residue of a G protein-gated inwardly rectifying K+

channel subunit. Proc. Natl. Acad. Sci. U.S.A. 93, 14193–141989 Karschin, C. et al. (1996) IRK(1–3) and GIRK(1–4) inwardly rectifying

K+ channel mRNAs are differentially expressed in the adult rat brain.J. Neurosci. 16, 3559–3570

10 Fernandez-Alacid, L. et al. (2011) Developmental regulation of Gprotein-gated inwardly-rectifying K+ (GIRK/Kir3) channel subunitsin the brain. Eur. J. Neurosci. 34, 1724–1736

11 Nishida, M. and MacKinnon, R. (2002) Structural basis of inwardrectification: cytoplasmic pore of the G protein-gated inward rectifierGIRK1 at 1.8 A resolution. Cell 111, 957–965

12 Inanobe, A. et al. (2007) Structural diversity in the cytoplasmic regionof G protein-gated inward rectifier K+ channels. Channels 1, 39–45

8

13 Nishida, M. et al. (2007) Crystal structure of a Kir3.1-prokaryotic Kirchannel chimera. EMBO J. 26, 4005–4015

14 Inanobe, A. et al. (2010) A structural determinant for the control ofPIP2 sensitivity in G protein-gated inward rectifier K+ channels. J.Biol. Chem. 285, 38517–38523

15 Whorton, M.R. and MacKinnon, R. (2011) Crystal structure of themammalian GIRK2 K+ channel and gating regulation by G proteins,PIP2, and sodium. Cell 147, 199–208

16 Whorton, M.R. and MacKinnon, R. (2013) X-ray structure of themammalian GIRK2–betagamma G-protein complex. Nature 498,190–197

17 Liao, Y.J. et al. (1996) Heteromultimerization of G-protein-gatedinwardly rectifying K+ channel proteins GIRK1 and GIRK2 andtheir altered expression in weaver brain. J. Neurosci. 16, 7137–7150

18 Slesinger, P.A. et al. (1995) Identification of structural elementsinvolved in G protein gating of the GIRK1 potassium channel.Neuron 15, 1145–1156

19 Chan, K.W. et al. (1997) Specific regions of heteromeric subunitsinvolved in enhancement of G protein-gated K+ channel activity. J.Biol. Chem. 272, 6548–6555

20 Wydeven, N. et al. (2012) Structural elements in the Girk1 subunitthat potentiate G protein-gated potassium channel activity. Proc.Natl. Acad. Sci. U.S.A. 109, 21492–21497

21 Medina, I. et al. (2000) A switch mechanism for G beta gammaactivation of I(KACh). J. Biol. Chem. 275, 29709–29716

22 Ivanina, T. et al. (2003) Mapping the Gbetagamma-binding sites inGIRK1 and GIRK2 subunits of the G protein-activated K+ channel. J.Biol. Chem. 278, 29174–29183

23 Thomas, A.M. et al. (2006) Differential phosphoinositide binding tocomponents of the G protein-gated K+ channel. J. Membr. Biol. 211,43–53

24 Rusinova, R. et al. (2009) Mass spectrometric analysis reveals afunctionally important PKA phosphorylation site in a Kir3 channelsubunit. Pflugers Arch. 458, 303–314

25 Rubinstein, M. et al. (2009) Divergent regulation of GIRK1 andGIRK2 subunits of the neuronal G protein gated K+ channel byGalphaiGDP and Gbetagamma. J. Physiol. 587, 3473–3491

26 Berlin, S. et al. (2010) G alpha(i) and G betagamma jointly regulatethe conformations of a G betagamma effector, the neuronal G protein-activated K+ channel (GIRK). J. Biol. Chem. 285, 6179–6185

27 Cruz, H.G. et al. (2004) Bi-directional effects of GABA(B) receptoragonists on the mesolimbic dopamine system. Nat. Neurosci. 7, 153–159

28 Labouebe, G. et al. (2007) RGS2 modulates coupling between GABABreceptors and GIRK channels in dopamine neurons of the ventraltegmental area. Nat. Neurosci. 10, 1559–1568

29 Jelacic, T.M. et al. (2000) Functional and biochemical evidence for G-protein-gated inwardly rectifying K+ (GIRK) channels composed ofGIRK2 and GIRK3. J. Biol. Chem. 275, 36211–36216

30 Lunn, M.L. et al. (2007) A unique sorting nexin regulates trafficking ofpotassium channels via a PDZ domain interaction. Nat. Neurosci. 10,1249–1259

31 Doupnik, C.A. (2008) GPCR-Kir channel signaling complexes:defining rules of engagement. J. Recept. Signal Transduct. Res. 28,83–91

32 Huang, C.L. et al. (1998) Direct activation of inward rectifierpotassium channels by PIP2 and its stabilization by Gbetagamma.Nature 391, 803–806

33 Sui, J.L. et al. (1998) Activation of the atrial KACh channel by thebetagamma subunits of G proteins or intracellular Na+ ions dependson the presence of phosphatidylinositol phosphates. Proc. Natl. Acad.Sci. U.S.A. 95, 1307–1312

34 Yokogawa, M. et al. (2011) NMR analyses of the Gbetagamma bindingand conformational rearrangements of the cytoplasmic pore of Gprotein-activated inwardly rectifying potassium channel 1 (GIRK1).J. Biol. Chem. 286, 2215–2223

35 Huang, C.L. et al. (1995) Evidence that direct binding of G betagamma to the GIRK1 G protein-gated inwardly rectifying K+

channel is important for channel activation. Neuron 15, 1133–114336 Clancy, S.M. et al. (2005) Pertussis-toxin-sensitive Galpha subunits

selectively bind to C-terminal domain of neuronal GIRK channels:evidence for a heterotrimeric G-protein-channel complex. Mol. Cell.Neurosci. 28, 375–389

User
Highlight
Page 9: Lujan Wickman13R

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

37 Rebois, R.V. et al. (2006) Heterotrimeric G proteins form stablecomplexes with adenylyl cyclase and Kir3.1 channels in living cells.J. Cell Sci. 119, 2807–2818

38 Mase, Y. et al. (2012) Structural basis for modulation of gatingproperty of G protein-gated inwardly rectifying potassium ionchannel (GIRK) by i/o-family G protein alpha subunit (Galphai/o).J. Biol. Chem. 287, 19537–19549

39 Peleg, S. et al. (2002) G(alpha)(i) controls the gating of the G protein-activated K+ channel, GIRK. Neuron 33, 87–99

40 Rubinstein, M. et al. (2007) Galphai3 primes the G protein-activatedK+ channels for activation by coexpressed Gbetagamma in intactXenopus oocytes. J. Physiol. 581, 17–32

41 Rusinova, R. et al. (2007) Specificity of Gbetagamma signaling to Kir3channels depends on the helical domain of pertussis toxin-sensitiveGalpha subunits. J. Biol. Chem. 282, 34019–34030

42 Nobles, M. et al. (2005) Heterotrimeric G proteins precouple with Gprotein-coupled receptors in living cells. Proc. Natl. Acad. Sci. U.S.A.102, 18706–18711

43 Ciruela, F. et al. (2010) Evidence for oligomerization between GABABreceptors and GIRK channels containing the GIRK1 and GIRK3subunits. Eur. J. Neurosci. 32, 1265–1277

44 Fajardo-Serrano, A. et al. (2013) Association of Rgs7/Gbeta5complexes with Girk channels and GABA receptors in hippocampalCA1 pyramidal neurons. Hippocampus http://dx.doi.org/10.1002/hipo.22161

45 Raveh, A. et al. (2009) Elucidation of the gating of the GIRK channelusing a spectroscopic approach. J. Physiol. 587, 5331–5335

46 Pfeffer, S.R. (2013) A nexus for receptor recycling. Nat. Cell Biol. 15,446–448

47 Kajii, Y. et al. (2003) A developmentally regulated andpsychostimulant-inducible novel rat gene mrt1 encoding PDZ-PXproteins isolated in the neocortex. Mol. Psychiatry 8, 434–444

48 Wang, X. et al. (2013) Loss of sorting nexin 27 contributes to excitatorysynaptic dysfunction by modulating glutamate receptor recycling inDown’s syndrome. Nat. Med. 19, 473–480

49 Carlton, J.G. and Cullen, P.J. (2005) Sorting nexins. Curr. Biol. 15,R819–R820

50 Balana, B. et al. (2011) Mechanism underlying selective regulation ofG protein-gated inwardly rectifying potassium channels by thepsychostimulant-sensitive sorting nexin 27. Proc. Natl. Acad. Sci.U.S.A. 108, 5831–5836

51 Balana, B. et al. (2013) Ras-association domain of sorting Nexin 27 iscritical for regulating expression of GIRK potassium channels. PLoSONE 8, e59800

52 Nassirpour, R. and Slesinger, P.A. (2007) Subunit-specific regulationof Kir3 channels by sorting nexin 27. Channels 1, 331–333

53 Arora, D. et al. (2011) Acute cocaine exposure weakens GABA(B)receptor-dependent G-protein-gated inwardly rectifying K+

signaling in dopamine neurons of the ventral tegmental area. J.Neurosci. 31, 12251–12257

54 Padgett, C.L. et al. (2012) Methamphetamine-evoked depression ofGABA(B) receptor signaling in GABA neurons of the VTA. Neuron 73,978–989

55 Hearing, M. et al. (2013) Repeated cocaine weakens GABA–Girksignaling in layer 5/6 pyramidal neurons in the prelimbic cortex.Neuron 80, 159–170

56 Harashima, C. et al. (2006) Abnormal expression of the G-protein-activated inwardly rectifying potassium channel 2 (GIRK2) inhippocampus, frontal cortex, and substantia nigra of Ts65Dnmouse: a model of Down syndrome. J. Comp. Neurol. 494, 815–833

57 Best, T.K. et al. (2007) Ts65Dn, a mouse model of Down syndrome,exhibits increased GABAB-induced potassium current. J.Neurophysiol. 97, 892–900

58 Cooper, A. et al. (2012) Trisomy of the G protein-coupled K+ channelgene, Kcnj6, affects reward mechanisms, cognitive functions, andsynaptic plasticity in mice. Proc. Natl. Acad. Sci. U.S.A. 109, 2642–2647

59 Sjogren, B. (2011) Regulator of G protein signaling proteins as drugtargets: current state and future possibilities. Adv. Pharmacol. 62,315–347

60 Doupnik, C.A. et al. (1997) RGS proteins reconstitute the rapid gatingkinetics of gbetagamma-activated inwardly rectifying K+ channels.Proc. Natl. Acad. Sci. U.S.A. 94, 10461–10466

61 Anderson, G.R. et al. (2009) The R7 RGS protein family: multi-subunitregulators of neuronal G protein signaling. Cell Biochem. Biophys. 54,33–46

62 Cheever, M.L. et al. (2008) Crystal structure of the multifunctionalGbeta5-RGS9 complex. Nat. Struct. Mol. Biol. 15, 155–162

63 Posokhova, E. et al. (2010) RGS6/Gb5 complex accelerates IKACh

gating kinetics in atrial myocytes and modulates parasympatheticregulation of heart rate. Circ. Res. 107, 1350–1354

64 Yang, J. et al. (2010) RGS6, a modulator of parasympatheticactivation in heart. Circ. Res. 107, 1345–1349

65 Xie, K. et al. (2010) Gbeta5 recruits R7 RGS proteins to GIRKchannels to regulate the timing of neuronal inhibitory signaling.Nat. Neurosci. 13, 661–663

66 Maity, B. et al. (2012) Regulator of G protein signaling 6 (RGS6)protein ensures coordination of motor movement by modulatingGABAB receptor signaling. J. Biol. Chem. 287, 4972–4981

67 Zhou, H. et al. (2012) GIRK channel modulation by assembly withallosterically regulated RGS proteins. Proc. Natl. Acad. Sci. U.S.A.109, 19977–19982

68 Stewart, A. et al. (2012) RGS proteins in heart: brakes on the vagus.Front. Physiol. 3, 95

69 Chung, H.J. et al. (2009) G protein-activated inwardly rectifyingpotassium channels mediate depotentiation of long-termpotentiation. Proc. Natl. Acad. Sci. U.S.A. 106, 635–640

70 Huang, C.S. et al. (2005) Common molecular pathways mediate long-term potentiation of synaptic excitation and slow synaptic inhibition.Cell 123, 105–118

71 Chung, H.J. et al. (2009) Neuronal activity regulates phosphorylation-dependent surface delivery of G protein-activated inwardly rectifyingpotassium channels. Proc. Natl. Acad. Sci. U.S.A. 106, 629–634

72 Luscher, C. and Malenka, R.C. (2011) Drug-evoked synaptic plasticityin addiction: from molecular changes to circuit remodeling. Neuron69, 650–663

73 Nassirpour, R. et al. (2010) Morphine- and CaMKII-dependentenhancement of GIRK channel signaling in hippocampal neurons.J. Neurosci. 30, 13419–13430

74 Signorini, S. et al. (1997) Normal cerebellar development butsusceptibility to seizures in mice lacking G protein-coupled,inwardly rectifying K+ channel GIRK2. Proc. Natl. Acad. Sci.U.S.A. 94, 923–927

75 Hashimoto, N. et al. (2008) Characterization of in vivo and in vitroelectrophysiological and antiarrhythmic effects of a novel IKACh

blocker, NIP-151: a comparison with an IKr-blocker dofetilide. J.Cardiovasc. Pharmacol. 51, 162–169

76 Machida, T. et al. (2011) Effects of a highly selective acetylcholine-activated K+ channel blocker on experimental atrial fibrillation. Circ.Arrhythm. Electrophysiol. 4, 94–102

77 Kobayashi, T. et al. (1999) Ethanol opens G-protein-activatedinwardly rectifying K+ channels. Nat. Neurosci. 2, 1091–1097

78 Lewohl, J.M. et al. (1999) G-protein-coupled inwardly rectifyingpotassium channels are targets of alcohol action. Nat. Neurosci. 2,1084–1090

79 Weigl, L.G. and Schreibmayer, W. (2001) G protein-gated inwardlyrectifying potassium channels are targets for volatile anesthetics.Mol. Pharmacol. 60, 282–289

80 Yamakura, T. et al. (2001) Differential effects of general anestheticson G protein-coupled inwardly rectifying and other potassiumchannels. Anesthesiology 95, 144–153

81 Yow, T.T. et al. (2011) Naringin directly activates inwardly rectifyingpotassium channels at an overlapping binding site to tertiapin-Q. Br.J. Pharmacol. 163, 1017–1033

82 Kobayashi, T. et al. (2011) Inhibition of G protein-activated inwardlyrectifying K+ channels by different classes of antidepressants. PLoSONE 6, e28208

83 Wen, W. et al. (2013) Discovery of ‘molecular switches’ within a GIRKactivator scaffold that afford selective GIRK inhibitors. Bioorg. Med.Chem. Lett. 23, 4562–4566

84 Kaufmann, K. et al. (2013) ML297 (VU0456810), the first potent andselective activator of the GIRK potassium channel, displaysantiepileptic properties in mice. ACS Chem. Neurosci. 4, 1278–1286

85 Ramos-Hunter, S.J. et al. (2013) Discovery and SAR of a novel series ofGIRK1/2 and GIRK1/4 activators. Bioorg. Med. Chem. Lett. 23,5195–5198

9

Page 10: Lujan Wickman13R

Review Trends in Neurosciences xxx xxxx, Vol. xxx, No. x

TINS-1022; No. of Pages 10

86 Wickman, K. et al. (1998) Abnormal heart rate regulation in GIRK4knockout mice. Neuron 20, 103–114

87 Bettahi, I. et al. (2002) Contribution of the Kir3.1 subunit to themuscarinic-gated atrial potassium channel IKACh. J. Biol. Chem. 277,48282–48288

88 Yang, Y. et al. (2010) Identification of a Kir3.4 mutation in congenitallong QT syndrome. Am. J. Hum. Genet. 86, 872–880

89 Jabbari, J. et al. (2011) Common polymorphisms in KCNJ5 areassociated with early-onset lone atrial fibrillation in Caucasians.Cardiology 118, 116–120

90 Wang, F. et al. (2013) The phenotype characteristics of type-13 longQT syndrome with mutation in KCNJ5 (Kir3.4-G387R). HeartRhythm 10, 1500–1506

91 Scholl, U.I. and Lifton, R.P. (2013) New insights into aldosterone-producing adenomas and hereditary aldosteronism: mutations in theK+ channel KCNJ5. Curr. Opin. Nephrol. Hypertens. 22, 141–147

92 Mulatero, P. et al. (2013) Role of KCNJ5 in familial and sporadicprimary aldosteronism. Nat. Rev. Endocrinol. 9, 104–112

93 Mitrovic, I. et al. (2003) Contribution of GIRK2-mediated postsynapticsignaling to opiate and alpha 2-adrenergic analgesia and analgesicsex differences. Proc. Natl. Acad. Sci. U.S.A. 100, 271–276

94 Marker, C.L. et al. (2004) Spinal G-protein-gated K+ channels formedby GIRK1 and GIRK2 subunits modulate thermal nociception andcontribute to morphine analgesia. J. Neurosci. 24, 2806–2812

95 Blednov, Y.A. et al. (2003) A pervasive mechanism for analgesia:activation of GIRK2 channels. Proc. Natl. Acad. Sci. U.S.A. 100, 277–282

96 Marker, C.L. et al. (2005) Spinal G-protein-gated potassium channelscontribute in a dose-dependent manner to the analgesic effect of mu-and delta- but not kappa-opioids. J. Neurosci. 25, 3551–3559

97 Cruz, H.G. et al. (2008) Absence and rescue of morphine withdrawal inGIRK/Kir3 knock-out mice. J. Neurosci. 28, 4069–4077

98 Smith, S.B. et al. (2008) Quantitative trait locus and computationalmapping identifies Kcnj9 (GIRK3) as a candidate gene affectinganalgesia from multiple drug classes. Pharmacogenet. Genomics 18,231–241

99 Nishizawa, D. et al. (2009) Association between KCNJ6 (GIRK2) genepolymorphisms and postoperative analgesic requirements after majorabdominal surgery. PLoS ONE 4, e7060

100 Lotsch, J. et al. (2010) A KCNJ6 (Kir3.2, GIRK2) gene polymorphismmodulates opioid effects on analgesia and addiction but not on pupilsize. Pharmacogenet. Genomics 20, 291–297

101 Blednov, Y.A. et al. (2002) Hyperactivity and dopamine D1 receptoractivation in mice lacking Girk2 channels. Psychopharmacology 159,370–378

102 Morgan, A.D. et al. (2003) Decreased cocaine self-administration in Kir3potassium channel subunit knockout mice. Neuropsychopharmacology28, 932–938

103 Arora, D. et al. (2010) Altered neurotransmission in the mesolimbicreward system of Girk mice. J. Neurochem. 114, 1487–1497

104 Pravetoni, M. and Wickman, K. (2008) Behavioral characterization ofmice lacking GIRK/Kir3 channel subunits. Genes Brain Behav. 7,523–531

105 Perry, C.A. et al. (2008) Predisposition to late-onset obesity in GIRK4knockout mice. Proc. Natl. Acad. Sci. U.S.A. 105, 8148–8153

106 Blednov, Y.A. et al. (2001) Potassium channels as targets for ethanol:studies of G-protein-coupled inwardly rectifying potassium channel 2(GIRK2) null mutant mice. J. Pharmacol. Exp. Ther. 298, 521–530

107 Kozell, L.B. et al. (2009) Mapping a barbiturate withdrawal locus to a0.44 Mb interval and analysis of a novel null mutant identify a role for

10

Kcnj9 (GIRK3) in withdrawal from pentobarbital, zolpidem, andethanol. J. Neurosci. 29, 11662–11673

108 Clarke, T.K. et al. (2011) KCNJ6 is associated with adult alcoholdependence and involved in gene � early life stress interactions inadolescent alcohol drinking. Neuropsychopharmacology 36, 1142–1148

109 Wickman, K. et al. (2000) Brain localization and behavioral impact ofthe G-protein-gated K+ channel subunit GIRK4. J. Neurosci. 20,5608–5615

110 Blednov, Y.A. et al. (2001) GIRK2 deficient mice. Evidence forhyperactivity and reduced anxiety. Physiol. Behav. 74, 109–117

111 Yamada, K. et al. (2012) Association study of the KCNJ3 gene as asusceptibility candidate for schizophrenia in the Chinese population.Hum. Genet. 131, 443–451

112 Costa, A.C. et al. (2005) G-protein-gated potassium (GIRK) channelscontaining the GIRK2 subunit are control hubs for pharmacologicallyinduced hypothermic responses. J. Neurosci. 25, 7801–7804

113 Patil, N. et al. (1995) A potassium channel mutation in weaver miceimplicates membrane excitability in granule cell differentiation. Nat.Genet. 11, 126–129

114 Gregerson, K.A. et al. (2001) Identification of G protein-coupled,inward rectifier potassium channel gene products from the ratanterior pituitary gland. Endocrinology 142, 2820–2832

115 Plummer, H.K., 3rd et al. (2005) Expression of G-protein inwardlyrectifying potassium channels (GIRKs) in lung cancer cell lines. BMCCancer 5, 104

116 Iwanir, S. and Reuveny, E. (2008) Adrenaline-inducedhyperpolarization of mouse pancreatic islet cells is mediated by Gprotein-gated inwardly rectifying potassium (GIRK) channels.Pflugers Arch. 456, 1097–1108

117 Wagner, V. et al. (2010) Cloning and characterisation of GIRK1variants resulting from alternative RNA editing of the KCNJ3gene transcript in a human breast cancer cell line. J. Cell.Biochem. 110, 598–608

118 Aguado, C. et al. (2008) Cell type-specific subunit composition of Gprotein-gated potassium channels in the cerebellum. J. Neurochem.105, 497–511

119 Inanobe, A. et al. (1999) Characterization of G-protein-gated K+

channels composed of Kir3.2 subunits in dopaminergic neurons ofthe substantia nigra. J. Neurosci. 19, 1006–1017

120 Koyrakh, L. et al. (2005) Molecular and cellular diversity ofneuronal G-protein-gated potassium channels. J. Neurosci. 25,11468–11478

121 Luscher, C. et al. (1997) G protein-coupled inwardly rectifying K+

channels (GIRKs) mediate postsynaptic but not presynaptictransmitter actions in hippocampal neurons. Neuron 19, 687–695

122 Ladera, C. et al. (2008) Pre-synaptic GABA receptors inhibitglutamate release through GIRK channels in rat cerebral cortex. J.Neurochem. 107, 1506–1517

123 Fernandez-Alacid, L. et al. (2009) Subcellular compartment-specificmolecular diversity of pre- and post-synaptic GABA-activated GIRKchannels in Purkinje cells. J. Neurochem. 110, 1363–1376

124 Michaeli, A. and Yaka, R. (2010) Dopamine inhibits GABA(A)currents in ventral tegmental area dopamine neurons viaactivation of presynaptic G-protein coupled inwardly-rectifyingpotassium channels. Neuroscience 165, 1159–1169

125 Booker, S.A. et al. (2013) Differential GABAB-receptor-mediatedeffects in perisomatic- and dendrite-targeting parvalbumininterneurons. J. Neurosci. 33, 7961–7974