REVIEWS

Lipid raft microdomains and signalling

John A. Allen*, Robyn A. Halverson-Tamboli* and Mark M. Rasenick*‡ Abstract | Lipid rafts are specialized structures on the plasma membrane that have an altered lipid composition as well as links to the cytoskeleton. It has been proposed that these structures are membrane domains in which neurotransmitter signalling might occur through a clustering of receptors and components of -activated signalling cascades. The localization of these proteins in lipid rafts, which is affected by the cytoskeleton, also influences the potency and efficacy of neurotransmitter receptors and transporters. The effect of lipid rafts on neurotransmitter signalling has also been implicated in neurological and psychiatric diseases.

The classic Singer–Nicolson fluid mosaic model of the nature, enrichment in glycosylphosphatidylinositol cell membrane describes the lipid bilayer as a neutral (GPI)-anchored proteins, cytoskeletal association two-dimensional solvent in which proteins diffuse and resistance to detergent extraction. Caveolae are freely1. This membrane concept has been modified small, flask-shaped invaginations of the membrane substantially since it was first proposed, as membrane that contain caveolin proteins. Caveolins are a major compartmentalization has been shown to occur through component and marker of caveolae, and are widely lipid–lipid, lipid–protein and membrane–cytoskeletal expressed in the nervous system in brain micro- interactions2. Similarly, the notion that a neurotransmit- vessels, endothelial cells, astrocytes, oligodendrocytes, ter receptor, once activated, initiates a signalling cascade Schwann cells, dorsal root ganglia and hippocampal by randomly colliding with other membrane proteins neurons5. Caveolins and caveolae are absent from has given way to the view that signalling molecules are most and neuroblastoma cells; however, arranged in stable, possibly preformed, complexes at neurons possess planar lipid rafts and flotillin, a protein the membrane3. These emerging concepts of membrane that is analogous, but not homologous, to caveolin6. organization and mechanisms of signalling indicate that Planar lipid rafts possess many of the features of caveo- membrane heterogeneity might be important; however, lae (cholesterol- and sphingomyelin-rich cytoskeletal the role of such heterogeneity in regulating neurotrans- association) but are not invaginated. While caveolae are mitter response and responsiveness is just beginning to the most readily-observed structures associated with be explored. lipid rafts, the presence of caveolae is not required to Lipid rafts are a well-studied type of membrane assign ‘raftness’, and we will not attempt to subdivide microdomain. Numerous roles have been ascribed to these structures in this review. lipid rafts, but this review focuses on the hypothesis that Although lipid rafts have earned wide acceptance they organize and compartmentalize neurotransmitter among cell biologists as signalling platforms, the field signalling components to either increase or dampen sig- remains controversial. This controversy has stemmed *Department of Physiology nalling (FIG. 1). The involvement of lipid rafts as vehicles mainly from the technical challenges of studying these and Biophysics, ‡Department for and trafficking during signalling will submicroscopic, dynamic structures in the membranes of Psychiatry and the also be discussed. of living cells (BOX 1). The field is both developing and Program in Neuroscience, University of Illinois at Two common types of lipid raft have been proposed employing new techniques to investigate these dynamic Chicago, College of Medicine, and studied with respect to neurotransmitter signal membrane domains (as discussed below). Regardless of Chicago, Illinois, USA. transduction: planar lipid rafts (which are also referred this debate, data accumulated over the past 10–15 years Correspondence to M.M.R. to as non-caveolar, or glycolipid, rafts) and caveolae have suggested that lipid rafts provide both a spatial e-mail: [email protected] Published online (little caves), both of which are estimated to be 25–100 and a temporal meeting point for signalling molecules, and 2,4 29 December 2006 nm in diameter . These raft microdomains are often this review attempts to compile some of these data with doi:10.1038/nrn2059 defined by their cholesterol- and sphingomyelin-rich a focus on the nervous system.

128 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group

REVIEWS

a Disruption of lipid rafts can be used to determine the NT Plasma NT physiological relevance of the localization of proteins to membrane these domains. Agents that sequester, chelate or prevent the synthesis of cholesterol are commonly used to disrupt EElipid rafts (BOX 3), and the subsequent effects on neuro- transmitter signalling can be observed. In addition, the manipulation of other lipid components — for example, E adding exogenous fatty acids or gangliosides — has been Planar lipid raft effective for disrupting lipid rafts4. As with any pharma- cological treatment, cholesterol manipulation can result Caveola in pleiotropic effects. This can be controlled for by adding cholesterol back to the cells and determining bc whether this results in a reversal of the observed effects. NT NT Recently, genetic approaches to disrupt caveolae have been developed. For example, knockdown of caveolins E E by small interfering RNA (siRNA) in glial cells disrupts neurotransmitter signalling7, and caveolin 1-knockout mice possess abnormal behavioural and neurological 5 Receptors and effectors organized Receptors and effectors separated by phenotypes . in rafts to promote signalling raft organization to prevent signalling Although these commonly utilized techniques are informative, they have a number of limitations and the field is developing new and more sophisticated Ionotropic G protein-coupled G protein receptor receptor Caveolin methods to study these domains in living cells. These new techniques rely mainly on biophysical and imag- Lipid raft Effector E Flotillin Neurotransmitter NT ing methodologies including fluorescence correlation microdomains spectroscopy8, single molecule tracking microscopy9, Laurdan partitioning10 and fluorescence resonance Figure 1 | Lipid raft microdomains and membrane organization of 4,11 neurotransmitter signalling molecules. Lipid rafts are cholesterol- and sphingolipid- energy transfer . These new techniques are powerful enriched, highly dynamic, submicroscopic (25–100 nm diameter) assemblies, which float approaches for studying lipid rafts and their influence in the liquid-disordered lipid bilayer in cell membranes2,4,40. a | There are two common raft on signalling in living cells, and they are advantageous domains in mammalian cells: planar lipid rafts and caveolae. Both possess a similar lipid as they do not rely on cell disruption. However, they composition. Planar rafts are essentially continuous with the plane of the plasma also have limitations, such as often requiring protein membrane and lack distinguishing morphological features. By contrast, caveolae are labelling, which has the potential to disrupt signal- small, flask-shaped membrane invaginations of the plasma membrane that contain ling cascades. Regardless of the individual techniques caveolins. Caveolin molecules can oligomerize and are thought to be essential in forming used, the strongest approach for studying lipid rafts 108 these invaginated membrane structures . Caveolins and flotillin can recruit signalling and signal transduction is one that employs several molecules into lipid rafts. Many neurotransmitter receptors (both ionotropic and complementary methods. G-protein-coupled), G proteins, and signalling effectors such as second-messenger- generating enzymes are found in lipid rafts. might activate receptors These methods have been employed, in various com- that are located both within and outside lipid rafts. b | The lipid raft signalling hypothesis binations, in the studies reviewed here. The results of proposes that these microdomains spatially organize signalling molecules at the these studies indicate that lipid rafts influence several membrane, perhaps in complexes, to promote kinetically favourable interactions that are components of different neurotransmitter signalling necessary for signal transduction. c | Alternatively, lipid raft microdomains might inhibit cascades including neurotransmitter receptors and their interactions by separating signalling molecules, thereby dampening signalling responses. effectors, as well as transporters. We begin by considering lipid rafts and receptors.

Methods for studying lipid rafts Ionotropic neurotransmitter receptors There are several complementary approaches for study- Ionotropic receptors (ligand-gated ion channels) control ing lipid rafts and their putative involvement in signal neurotransmission by facilitating ion conductance and transduction. The most common definition of a raft- the subsequent generation or inhibition of membrane associated protein is one that cannot be extracted by electrical potentials. Lipid raft microdomains might Triton X-100 non-ionic detergents (BOX 2). Preparation of membranes modulate neuronal excitability by contributing to A commonly used non-ionic that are resistant to Triton X-100 by sucrose density gradient ionotropic receptor sensitivity and function. surfactant detergent that centrifugation is a simple and easy method to assess the solubilizes proteins and cell potential raft association of a protein. However, this Reported localization in lipid rafts. The lipid raft membranes. technique is not without pitfalls, as some proteins show localization status for nearly all ionotropic receptors Sucrose density gradient a facultative and variable raft association during deter- that are expressed in the nervous system has been centrifugation gent extraction4. In addition, the localization of signal- investigated; however, there is some confusion about the Separation of cell organelles ling proteins in fractionated raft membranes does not raft localization of some of their subunits and subtypes and subcellular components (TABLE 1) from crude cellular extracts provide sufficient evidence of raft involvement in a given . Given the lack of consistency in the methods based on their buoyant signalling process, and agreement among biophysical, and in the cell and tissue types used for studying rafts, density. biochemical and imaging studies has been elusive. the discrepancies probably result from procedural

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | FEBRUARY 2007 | 129 © 2007 Nature Publishing Group

REVIEWS

Box 1 | The debate regarding lipid rafts duality might function to compartmentalize differ- ent signal transduction pathways that are associated The concept of lipid rafts has received significant attention in recent years, but with the same receptor (see below). attendant with that attention has been controversy. Although the lateral heterogeneity of proteins in the plasma membrane is undisputed, the very existence of planar lipid Neurotransmitter binding and receptor localization rafts and their biological importance has been questioned103. These concerns are rooted in the current technology used to study lipid rafts and their biochemical and biophysical in rafts. Little is known about how agonist binding limitations (BOXES 2,3). It is noteworthy that whereas the unequivocal existence of affects the localization of ionotropic receptors to caveolae can be defined by the presence of caveolins and confirmed by electron membrane microdomains and how this localization,

microscopy, planar lipid rafts have been more difficult to both define and study. in turn, might affect ligand binding. The P2X3 purinergic The nanometre diameter of these microdomains is beyond the resolution capability of receptor retains its residence in the lipid rafts of light microscopy, making the study of these structures in intact, living cells complex. neuronal cells subsequent to agonist stimulation13, but In addition, the classic approaches for studying and isolating lipid rafts often rely on the consequences for raft localization following agonist detergent extraction, a method that is thought by some to introduce undue stimulation have not been reported for other ionotropic 12,103,104 artefacts . Some studies have also questioned the biological importance of raft receptor types. Raft localization does seem to affect the domains. One recent study removed lipid binding capabilities from signalling proteins binding of neurotransmitter to ionotropic receptors. and concluded that protein clusters are dependent on protein–protein interactions and 105 Caveolin 1 has been shown to indirectly reduce the that lipid rafts and actin are irrelevant to these clusters . However, despite the α observed clustering of signalling molecules in this study, there was no indication that binding of AMPA ( -amino-3-hydroxy-5-methyl-4- the signalling capabilities of these molecules were maintained in the absence of lipid isoxazole propionic acid) to its receptor in hippocam- raft association. pal synaptosomes14. Both depletion and enrichment of Recent investigations have introduced new, more sophisticated techniques for cholesterol, which is a defining component of lipid studying lipid rafts and have addressed some of these controversial issues. Biophysical rafts, decrease the potency of GABA (γ-aminobutyric studies of model membranes have confirmed the existence of nanoscale liquid-ordered acid) at the GABAA receptor, which suggests an and liquid-disordered microdomains predicted by the raft hypothesis11. Moreover, a optimal level of cholesterol for optimal agonist recent hypothesis synthesizing current results and differing views about membrane potency, whereas depletion of membrane cholesterol organization suggests a revised model in which rafts are dynamic associating and 11 had no effect on the potency of GABAA receptor dissociating nanodomains influenced by protein–protein interactions . Ultimately, 15 better methodology is needed to further elucidate the nature of these nanoscale, antagonists . Similarly, cholesterol is required for the dynamic membrane structures. agonist-induced ion-gating function of the nicotinic acetylcholine (nACh) receptor16. Exactly how lipid rafts alter neurotransmitter affinity for ionotropic receptors variations. This is confounded by the fact that relatively is unclear. Lipid rafts contain a number of proteins that few reports describing biochemical raft isolations give associate with receptors, modifying their affinity for sufficient information to precisely replicate the results. and capacity to bind ligands, as well as the properties For example, reports of detergent to protein ratios are of the channels themselves. Coincident localization of largely absent from methods sections, and this key these proteins with receptors in lipid rafts is transitory. ratio can greatly influence the outcome of results12. In addition, the specific lipid–protein interactions In most instances the biochemical isolation of lipid that occur in lipid rafts could influence ionotropic rafts also reveals a population of ionotropic receptors/ receptor conformation and therefore agonist potency subunits that is not associated with lipid rafts. This or efficacy. Finally, oligomerization of NMDA

Box 2 | Common methods to isolate lipid rafts Biochemical isolation of lipid raft membranes and their subsequent analysis is a useful and simple method to determine if signalling components are located (or change localization) in raft microdomains. Two common techniques described here demonstrate protein localization to rafts prior to, during and after neurotransmitter signalling. Preparation of detergent-resistant membranes (DRMs) Classically, lipid raft membranes are defined as being insoluble in cold non-ionic detergents such as Triton X-100. Detergent-resistant membrane fractions float into the buoyant fractions of sucrose gradients during ultracentrifugation and are distinguished as two or three opaque bands in the lighter density sucrose106. The DRM method of isolation has 12,104 received criticism by some investigators because it is prone to variable, and sometimes inconsistent, results . One A plasma membrane channel problem is that Triton X-100 can solubilize proteins that are only weakly associated with lipid raft membranes. that is activated by the binding Furthermore, although lipid rafts are often cytoskeletal-associated, the methods used to prepare DRMs disrupt of ATP and is permeant to cytoskeletal–membrane interactions and compromise this association. mono- and divalent cations. Non-detergent-based isolation of raft membranes There is also a non-detergent-based procedure for the isolation of lipid rafts based on pH and carbonate resistance107. Synaptosome A preparation of elements of Sodium carbonate, at high pH, separates proteins that are firmly attached to membranes from those that are more the presynaptic and peripherally associated. Rafts are isolated by centrifugation and fractions are harvested as for the detergent preparation. postsynaptic terminal, isolated As is the case with detergent-resistant preparations, the high pH method requires cell disruption — in this case by after subcellular fractionation. sonication. Such treatment of membranes alters the often delicate relationship between membrane, protein and Synaptosomes retain some cytoskeletal components that normally exists in cells. anatomical integrity and can Monitoring of the level of cholesterol present in sucrose fractions obtained from either method confirms that take up, store and release cholesterol-enriched membrane domains are present or absent within given fractions. neurotransmitters.

130 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group

REVIEWS

Box 3 | Common approaches for disrupting lipid rafts linked to lipid raft stability and is also thought to mediate the clustering of nAChRs (through rapsyn)25. Signalling processes that are promoted or inhibited by lipid rafts are often studied by However, despite the association between actin, disruption of the microdomains through chemical or genetic manipulation. Loss of nAChRs and lipid rafts, disruption of actin filaments regulation subsequent to raft disruption is evidence for a requirement of raft compart- did not prevent clustering of the receptors in rafts22. mentalization of signalling components. Owing to their enrichment in cholesterol, lipid rafts are sensitive to the removal of or reduction in membrane cholesterol, and This suggests that either actin monomers are active cholesterol manipulations can redistribute proteins away from lipid raft membrane in this process or that other factors also contribute to domains. Commonly used techniques to achieve these effects include: receptor clustering; this will be discussed below. • Sequestration of cholesterol using filipin, nystatin, amphotericin • Depletion and removal of cholesterol using methyl-β-cyclodextrin Raft-regulated signalling. Studies exploring the role of • Inhibition of cholesterol synthesis using HMG-CoA reductase inhibitors (statins) lipid rafts in ionotropic receptor signalling events have • Genetic approaches including caveolin 1 (Cav1)-knockout mice5 or caveolin RNA begun to emerge in recent years. The calcium-mediated interference7 inhibition of type VI adenylyl cyclase by nAChRα7 is These methods of manipulating cholesterol are particularly informative when dependent on raft integrity26, which supports the idea combined and confirmed with fractionation studies (BOX 2). With respect to the use of that lipid rafts can orchestrate specific signalling events cholesterol-disrupting drugs, which can result in pleiotropic effects, the restoration of cholesterol provides an important experimental control. Also of note is that when downstream of ionotropic receptors by influencing the genetic ablation of specific caveolin gene products is used, compensatory effects can inclusion or exclusion of receptors and their associated be observed. Studies designed to reveal the roles of lipid rafts in neurotransmitter effector molecules. Furthermore, the segregation of an signalling should ideally combine complementary approaches, including the evaluation ionotropic receptor into raft and non-raft fractions can of raft localization of components through fractionation, disruption of lipid rafts by be associated with specific signal transduction cascades biochemical and/or genetic means, assessment of signalling properties and the and cellular responses subsequent to receptor activa- examination of signalling protein localization by live-cell imaging. tion. For example, a population of P2X7 receptors in lipid rafts is responsible for activating phospholipase

A2 and generating ceramide, but P2X7 receptors resid- (N-methyl-d-aspartate) and nACh receptor subunits ing outside rafts form a non-selective cation channel modulates agonist affinity17,18, and raft localization that elevates levels of intracellular calcium27. Similarly, might have a role in the orientation and availability of NMDA receptors (NMDARs) located in lipid rafts ionotropic receptors for multimer formation. are reported to mediate neurotoxicity28,29, whereas NMDARs outside of lipid rafts are responsible for gluta- Lipid rafts and trafficking. Lipid rafts might also con- mate-mediated growth cone guidance30. The former tribute to the trafficking of ionotropic receptors to and observation is somewhat confounded by two studies from the cell membrane. Raft-depleted cells have fewer that report conflicting results on the requirement of nACh receptors on their cell surface, probably as a lipid rafts for calcium conductance by NMDARs28,29. result of impaired transport from the Golgi complex to These studies used different neuronal cultures the membrane and/or a reduced membrane stability19. (hippocampal versus cortical), raising the possibility Similarly, disrupting lipid rafts reduces the stability of that there might be regional variation in raft–receptor surface AMPA receptors, but also increases internali- interactions. zation of unstimulated receptors20. Internalized nACh receptors have been shown to colocalize with caveolin21, G-protein-coupled neurotransmitter receptors suggesting a caveolae-mediated route of internaliza- G-protein-coupled neurotransmitter receptors (GPCRs) tion. However, so far, a lipid raft/caveolae trafficking activate heterotrimeric binding pro- mechanism common to all ionotropic receptors has yet teins (G proteins), which act as signalling intermediates to be identified. between activated receptors and their intracellular effec- tors. Signalling through GPCRs is complex, requiring a Lipid rafts and clustering. Several ionotropic receptors minimum of three protein components: a receptor, a G show a clustered distribution at the membrane, and protein and a signalling effector; these components must such a distribution is considered integral to the for- physically interact to transmit information. Lipid rafts mation and maintenance of synapses. Likewise, intact can influence any one of these components and both lipid rafts seem to be necessary for the maintenance localization and trafficking/internalization of receptor of normal synaptic morphology and dendritic spine components by rafts can modify GPCR signalling. density20. Lipid rafts have also been shown to contrib- ute to both the size and the number of postsynaptic Stable signalling complexes in microdomains. In the Heparan sulphate 20 NMDA and GABAA receptor clusters , which further classic model for GPCR signalling, receptors dif- proteoglycans indicates an important role for lipid rafts in synaptic fuse randomly in the lipid bilayer of cells and activate A group of proteoglycans in which heparan sulphate chains neurotransmission. The integrity of lipid rafts is also their cognate G proteins and effectors through ‘colli- are attached near the cell necessary for maintaining the stability of nACh recep- sion coupling’ that is driven by high-affinity protein– surface or on extracellular tor (nAChR) clusters10,22,23, and is required for the clus- protein interactions. However, studies of these matrix proteins. Agrin is one tering of nAChRs that is induced by the heparan sulphate molecules in living cells indicate that stable complexes such proteoglycan, named for proteoglycan 24 its involvement in aggregation , agrin . Neural agrin also translocates are formed between many GPCRs and G proteins, and 3 of acetylcholine receptors nAChRs into lipid rafts where they are subsequently between G proteins and effectors . These results are most during synaptogenesis. anchored by rapsyn23. The cytoskeleton is intimately easily explained by the existence of GPCR signalling

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | FEBRUARY 2007 | 131 © 2007 Nature Publishing Group

REVIEWS

Table 1 | Localization of ionotropic receptors and neurotransmitter transporters Receptor or transporter Raft Non-raft References nACh receptor α, α α, α β, β 22–24,26,109 7 2, 5

GABA receptor GABAA 110 NMDA receptor NR1, NR2A, NR2B NR1, NR2B 20,29,30,111 AMPA receptor GluR1, GluR2/3, GluR4 20,111

P2X receptor P2X1, P2X3, P2X7 P2X1, P2X2, P2X3, P2X4, P2X7 13,27,112

5-HT receptor 5-HT3A 94 Serotonin transporter Yes Yes 71,78 Norepinephrine transporter Yes Yes 76 Glutamate transporter EAAT1–4 EAAT1–4 73,113 Lipid raft localization was determined by various biochemical methods or by colocalization with lipid raft markers. 5-HT, 5-hydroxytryptamine; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid; EAAT, excitatory amino acid transporter; GABA, γ-aminobutyric acid; GluR, ; nACh, (nicotinic subtype); NMDA, N-methyl-d- aspartate; NR, NMDA receptor subunit; P2X, 2 class X.

complexes that are constrained in their lateral mobility receptors (mGluR1a)38. These caveolin–receptor inter- in the plasma membrane, and confined within mem- actions could function to localize receptors in lipid brane microdomains31. rafts, and caveolins are known to profoundly influence signalling from certain neurotransmitter receptors. Ligand binding and localization in rafts. The involve- For example, stable knockdown of caveolin 1 by RNA ment of lipid rafts in regulating and organizing neuro- interference (RNAi) in C6 glioma cells alters the signal- α transmitter signalling through GPCRs has been reported ling of only select G q-coupled GPCRs, as caveolin

for many of the main receptor classes and subtypes knockdown abolishes signalling through 5-HT2A and (TABLE 2); however, these raft-regulated processes are just P2Y receptors; however, thrombin signalling through beginning to be elucidated in the nervous system. As protease-activated receptor 1 (PAR1) is unaffected7. is the case with certain ionotropic receptors, lipid rafts By contrast, the constitutive signalling of mGluR1a is influence the binding of neurotransmitters to several inhibited when receptors and caveolin 2 (but not caveo- 39 HEK293 cells GPCRs. Disruption of rafts by cholesterol depletion lin 1) are heterologously expressed in HEK293 cells . A hypotriploid cell line significantly reduces agonist binding and G-protein So, although signalling from certain GPCRs (5-HT2A, derived from embryonic kidney coupling to 5-hydroxytryptamine 1A (5-HT1A) sero- P2Y) seems to be dependent on caveolin 1, some (for epithelial cells; commonly used tonin receptors in bovine hippocampal membranes32. example, PAR1) are unaffected, whereas others (such as as an expression system to Similarly, NK1 neurokinin receptors expressed in human mGluR1a) are inhibited by caveolin 2. One explanation study signalling and recombinant proteins. embryonic kidney cells (HEK293 cells) bind significantly for these varied effects is that caveolins can interact with less agonist after raft disruption33. Physical association several G proteins and effectors (as discussed below). CHO cells (coupling) between receptor and G protein represents A chinese hamster ovary cell the high-affinity state of agonist binding. These studies Rafts and GPCR trafficking. In addition to acting line often used as an expression system for studying suggest that for some receptor–G protein pairs, the as organizing centres for signalling molecules, both cell signalling and recombinant localization of receptor and G protein in lipid rafts planar lipid rafts and caveolae can facilitate - proteins. might facilitate their coupling, which subsequently influ- independent endocytosis40–42. During neurotransmitter ences agonist binding to receptors. By contrast, human signalling, many GPCRs undergo agonist-induced B max κ-opioid receptors stably expressed in Chinese hamster endocytosis, which can lead to receptor recycling, A measure of the total number of receptors, determined by ovary cells (CHO cells) are concentrated in lipid rafts, and receptor sequestration and/or receptor downregu- the binding of agonist or cholesterol depletion of these cells significantly increases lation. The agonist-induced trafficking of GPCRs antagonist ligands. This 34 maximal agonist binding (Bmax) and G-protein coupling . through arrestin-dependent and dynamin-dependent, number will reflect either This indicates that, in this case, receptor and G protein clathrin-mediated endocytosis is well established43. surface receptors or surface plus internalized receptors occupy different membrane compartments. Therefore, However, clathrin-independent lipid raft mecha- depending on the chemical raft domains differentially influence neurotransmitter nisms might also contribute to receptor trafficking. characteristics of the ligand. binding and the coupling of G proteins to GPCRs. At Trafficking by rafts during signalling could involve present, it is not clear whether these differences are due three distinct mechanisms: endocytosis mediated by RNA interference to receptor type or to the cell/tissue type in which the caveolae; endocytosis mediated by planar lipid rafts; (RNAi). A molecular method in which small interfering RNA receptors are expressed. and/or lateral trafficking into or out of these micro- sequences are introduced into domains (FIG. 2). Internalization of GPCRs through cells or tissues, and Caveolin–GPCR interactions. Caveolins have been shown caveolae or rafts has been suggested for several subsequently silence the to interact with and/or regulate signalling from several GPCRs (TABLE 2). For example, activated β -adrenergic expression of target genes. 1 35 β GPCRs including A1 receptors , muscarinic receptors ( 1-ARs), which commonly undergo 36 37 C6 glioma cells acetylcholine receptors , D1 dopamine receptors , clathrin-mediated endocytosis, switch their route of A diploid cell line cloned 7 5-HT2A receptors and metabotropic glutamate 1a endocytosis to caveolae/rafts when phosphorylated from a glial tumour.

132 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group

REVIEWS

Table 2 | Lipid raft localization, trafficking and regulation of G protein-coupled neurotransmitter signalling molecules Lipid raft localization Regulation of Neurotransmitter Endocytosis References function by raft effects on mediated by Anatomical Biochemical association localization in rafts rafts methods methods GPCR class Acetylcholine Yes (M2) Yes (M1, M2) ↑ (M3) ↑ (M2) Yes (M2) 36,47,64,114 Adrenergic β β β β ↑ β ↓ β β β 44,115 Yes ( 1, 2) Yes ( 1, 2) ( 1) ( 2) Yes ( 1, 2) ↓ β 47,48,60,115 ( 2) Cannabinoid nd nd ↓ nd Yes (CB ) 45,116 (CB1) 1 Dopamine Yes (D1, D2) Yes (D1) ↑ (D1) nd Yes (D2) 37,117 Glutamate Yes (mGluR1a) Variable results ↓ (mGluR1a) nd nd 38,39,118 GABA nd Yes ↓ nd nd 119

Histamine Yes (H1) nd none nd Yes (H1)46 ↑ Neurokinin Yes (NK1) Yes (NK1) nd nd 33 Opioid nd Yes (κ, μ) ↓ (κ) none (μ) nd 34,120 Purinergic Yes (A , A ) Yes (A ) ↑ ↑ Yes (A ) 35,117 1 2A 1 (P2Y) (A1) 1 Yes (P2Y) 7,64 Serotonin Yes (5-HT ) Yes (5-HT ) ↑ nd nd 32 2A 2A (5-HT1A) ↑ 7,121 (5-HT2A) GnRH nd Yes ↑ nd nd 122 Oxytocin nd Yes ↑ nd nd 123 G proteins α Yes Yes ↓↑ Yes 47,48,60 G s α nd Yes nd nd nd 53,59 G i α Yes Yes ↑↓ nd 7,58,59 G q α nd Yes nd nd nd 124 G 12 α nd Yes nd nd nd 125 G o α nd Yes ↓↑ nd 51 G t Gβγ Yes Yes nd nd nd 59 RGS proteins nd Yes nd ↑ nd 51,61 G-protein effectors Adenylyl cyclases nd Yes ↓ nd nd 47,62,63 Phospholipase C nd Yes ↑↑ nd 64 G-protein-gated nd Yes (Kir3) ↓ (Kir3) nd nd 65 ion channels ↓ (N-type-Ca2+) 66 cAMP-gated ion nd Yes ↑ nd nd 67 channels cGMP nd Yes nd ↑ nd 51,126 phosphodiesterase ↑, increase in; ↓, decrease in; 5-HT, 5-hydroxytryptamine; A , adenosine receptor 1; cAMP, cyclic AMP; cGMP, cyclic GMP; CB , cannabinoid receptor 1; D1, γ 1 1 dopamine receptor 1; GABA, -aminobutyric acid; GnRH, gonadotropin releasing hormone receptor; GPCR, G-protein-coupled receptor; H1, histamine receptor 1; Kir3, G-protein-gated inwardly rectifying potassium channel; M1, acetylcholine receptor muscarinic subtype 1; mGluR1a, metabotropic glutamate receptor 1a; nd,

not determined; NK1, neurokinin receptor 1; P2Y, purinergic receptor 2 class Y; RGS, regulators of G-protein signalling.

44 by protein kinase A . Similarly, cannabinoid CB1 recep- raft-mediated endocytic mechanism that is distinct tors have been suggested to undergo both clathrin- from caveolae-mediated internalization46. and raft/caveolae-induced endocytosis that leads Some receptors also translocate into or out of lipid to receptor downregulation45. In addition, hista- rafts during signalling. For example, agonist stimulation

mine H1 receptors undergo clathrin-independent of adenosine A1 receptors increases the pool of receptors internalization during signalling but do not colocalize in lipid rafts, which presumably facilitates the observed with caveolins after internalization, indicating a lipid clathrin-independent, raft-mediated internalization of

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | FEBRUARY 2007 | 133 © 2007 Nature Publishing Group

REVIEWS

Plasma Caveola Planar lipid raft localization in lipid rafts in the nervous system is lacking, membrane G proteins have been found in lipid raft membranes NT NT isolated from whole brain lysates50, glia48, photorecep- tors51 and cardiac muscle52, as well as many other tissues. Various G protein subtypes, including Gα , Gα , Gα , c Lateral movement s i q NT α α α βγ G 12, G o, G t and G are all enriched, to a varying b Raft- degree, in lipid rafts (TABLE 2). mediated a Caveolae- endocytosis mediated Raft targeting of G proteins. G proteins could be targeted endocytosis to raft domains by several mechanisms. The most plau- sible mechanism is that Gα subunits are subject to fatty acylations such as palmitoylation and/or myristoylation, which might localize the proteins to rafts. For example, in NT NT NT NT reconstituted membranes or liposomes, palmitoylation or α myristoylation of G i is required for it to be targeted to lipid rafts, and lipid rafts are sites of increased palmitoyl- CaveosomeRecycling Lipid raft-derived transferase activity for Gα 53,54. Interactions of G proteins Sequestration vesicle i Degradation with caveolins or other scaffolding proteins might also α α α contribute to raft targeting, as G s, G o and G i interact directly with the amino (N) terminus of caveolin 1, G-protein- Cholera 55 coupled Caveolin Dynamin Neurotransmitter NT which could target the proteins to rafts . Furthermore, toxin B receptor tubulin, a scaffold protein that is also a component of lipid rafts, shows high-affinity interactions with Figure 2 | Lipid rafts as sites for endocytosis and trafficking of neurotransmitter G proteins (G , G and G )56,57. receptors. During neurotransmitter signalling, many G-protein-coupled receptors s i q (GPCRs) undergo agonist-induced endocytosis, leading to receptor recycling, receptor G protein trafficking. As is the case for many receptors, sequestration and receptor downregulation. Clathrin-independent lipid raft mechanisms some G proteins also undergo signal-dependent traf- might contribute to this process. Both caveolae and planar lipid rafts can facilitate clathrin-independent endocytosis from the plasma membrane40,42, although the precise ficking into or out of lipid rafts. For example, chronic components and intracellular trafficking pathways that are involved have yet to be stimulation of thyrotropin-releasing hormone receptors 40,41 α 58 definitively determined . Caveolae or raft endocytosis can be assessed using induces G q/11 to exit detergent-resistant membranes . fluorescence microscopy, and cellular uptake of cholera toxin B is often used to By contrast, during stimulation of β-ARs in glioma cells, α distinguish clathrin-independent, raft-mediated endocytosis; however, this toxin can G s localization in lipid rafts increases and it undergoes also be taken up by other pathways41. a | Several GPCRs have been reported to be agonist-induced endocytosis that is dependent on raft internalized through the caveolae pathway (TABLE 2). A signal-dependent event leads to domains48. So, rafts can function as sites for G-protein dynamin-dependent fission of the invaginated caveola and subsequent endocytosis and trafficking and internalization, as well as platforms for 40 vesicle trafficking to caveolin-containing caveosomes . The molecular mechanisms signalling. responsible for GPCR internalization through caveolae are largely undefined and warrant further investigation. b | Planar lipid rafts can also facilitate endocytosis; however, less evidence is available for this route of GPCR internalization. Raft regulation of signalling. Different G proteins prob- c | Neurotransmitter signalling can also result in the movement of receptors into or out of ably associate selectively with caveolae or planar lipid lipid rafts. This translocation and lateral movement in the membrane could either rafts and this might help explain the complex regulation activate or diminish neurotransmitter signalling by altering the coupling of receptors of G-protein signalling by raft domains. For example, with G proteins and/or other signalling effectors. isolation of caveolae from other lipid raft membranes α in endothelial cells indicates that G q interacts directly α with caveolins, targeting G q to caveolae; by contrast, 35 β α α β the receptors . By contrast, -ARs are removed from G s, G i and G are targeted to lipid rafts that do not 59 Palmitoylation lipid rafts during signalling in cardiac myocytes and contain caveolins . Stable knockdown of caveolin 1 in 47,48 α The covalent attachment of glioma cells . Also of note, some GPCRs, such as kinin glioma cells prevents G q-coupled signalling from the a palmitate (16-carbon B1 receptors, translocate into lipid raft domains but do 5-HT2A receptor, indicating that lipid rafts and/or caveo- saturated fatty acid) to a not undergo endocytosis49, indicating that some activated lins are required7. By contrast, disrupting raft domains cysteine residue through α β a thioester bond. GPCRs move into raft domains and remain there, perhaps by cholesterol chelation elevates G s-coupled, -AR to promote or prevent interactions with downstream signalling47,60. Therefore, lipid raft domains seem to Myristoylation signalling molecules. These examples indicate that GPCRs spatially organize specific G proteins at the membrane, The covalent (and, in the case undergo agonist-induced endocytosis through lipid rafts which could facilitate or prevent interactions with their of a G protein, cotranslational) and also traffic into or out of these domains — events that receptors and/or effectors, serving to either diminish or attachment of a hydrophobic myristoyl group to the amino- could ultimately regulate receptor signalling. promote neurotransmitter signalling. terminal glycine residue of a nascent polypeptide. G proteins Rafts and RGS proteins. Regulators of G-protein Many G proteins, in addition to their cognate receptors, signalling (RGS) proteins associate with Gα subunits Liposome A lipid vesicle artificially have been reported to localize to lipid rafts, and these associa- and inactivate them by increasing the hydrolysis of GTP. formed by sonicating lipids in tions differentially alter neurotransmitter signalling. Palmitoylation of RGS16 targets it to lipid rafts, and this an aqueous solution. Although a definitive consensus about G-protein raft localization might be necessary for the involvement of

134 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group

REVIEWS

α α 61 RGS16 in turning off G i or G q signalling . Exposure of their resident accessory proteins might regulate the α β retinal photoreceptors to light causes a G t–RGS9–G 5 functional availability of ion channels at the membrane complex to translocate to lipid raft membranes, resulting and also possibly at synapses. Overexpression of caveo- α in the decreased coupling of G t to, and the subsequent lins in NG108-15 neuroblastoma/glioma cells prevented reduced activation of, its receptor rhodopsin51. These the inhibition of N-type currents by δ reports raise the possibility that the G-protein inactiva- both the -opioid receptor and G proteins, indicating tion cycle could be regulated, in part, by partitioning RGS that rafts and caveolins prevent G-protein-coupling to proteins in lipid raft microdomains. N-type calcium channels66. CNG ion channels, effectors of light and olfactory receptors, are located in lipid rafts Downstream effector systems isolated from olfactory epithelia67. When HEK293 cells Common neurotransmitter effector systems include phos- expressing CNG ion channels are depleted of cholesterol, phodiesterases, ion channels and enzymes that generate the channels’ affinity for cAMP is significantly reduced second messengers. Activated G proteins modulate signal- and channel activity is abolished. These studies reveal ling effectors such as adenylyl cyclases, phospholipase C that lipid raft domains influence ion channel effectors in (PLC), ion channels and retinal phosphodiesterase. Several a diverse manner — regulating the targeting of channels of these effectors are located in lipid rafts (TABLE 2), where to the cell surface, altering the coupling of G proteins to these microdomains might facilitate or inhibit interac- the channels and changing ligand binding affinity. tions among the effectors and their cognate GPCRs and G proteins to propagate or dampen signalling. Neurotransmitter transporters Neurotransmitter transporters control neuronal signal- Adenylyl cyclases. There is considerable evidence that ling by regulating the levels of neurotransmitter in the several adenylyl cyclases, which are regulated by G pro- synapse. A number of antidepressant and psychoactive teins and calcium to produce cyclic AMP (cAMP) during drugs modulate transporter activity — and thereby also neurotransmission, are associated with lipid rafts. There neurotransmitter signalling — underscoring the impor- are nine membrane-associated adenylyl cyclase isoforms, tance of understanding how transporters are regulated68. each of which is uniquely regulated. For example, adeny- Several transporters have been found to be localized to lyl cyclase type VI is enriched in lipid rafts in glial cells, lipid rafts (TABLE 1), and these membrane microdomains and its inhibition by calcium influx is dependent on raft seem to significantly influence transporter function and integrity62. Adenylyl cyclase type V can be co-immuno- availability at the membrane. precipitated with caveolin 3 in cardiac myocytes52, and N-terminal caveolin peptides inhibit the enzyme activ- Cholesterol and transporter function. Disruption of lipid ity of purified type V adenylyl cyclase as well as a pool rafts by cholesterol-interfering agents produces, on aver- 63 of adenylyl cyclases isolated from brain membranes . age, a 50% decrease in the transport rate (Vmax) of the Furthermore, forskolin-stimulated adenylyl cyclase activ- GABA transporter69, 5-HT transporter (5-HTT)70,71 and ity is significantly increased after disrupting lipid rafts glutamate transporters72,73. Reconstitution of GABA by depleting cholesterol47,60. These studies collectively and glutamate transporter activity in liposomes requires indicate that some adenylyl cyclases are localized in lipid cholesterol74, and glutamate uptake activity is almost raft domains where their activity is attenuated. completely confined to lipid raft fractions prepared from mouse forebrain extracts73. Cholesterol levels in synaptic Phospholipases and calcium signalling. Although there plasma membrane vesicles closely parallel the amount is substantial evidence to indicate that lipid rafts spatially of GABA that is bound to its transporter69 and the rate of and temporally regulate calcium signalling in cardiac GABA and glutamate uptake69,73. So, most transporters myocytes and other tissues, this has yet to be well estab- seem to exert maximal neurotransmitter uptake while lished in the nervous system. However, one recent study residing in lipid rafts. The ratio of raft-associated to non- in astrocytes shows that several proteins that form the raft-associated transporter might represent a mechanism

Scaffolding proteins -1,4,5-trisphosphate (Ins(1,4,5)P3)-dependent for regulating the rate and extent of neurotransmitter

Proteins that organize groups calcium cascade — metabotropic P2Y1, Gq, Ins(1,4,5)P3 uptake. Interestingly, glial-conditioned medium increases of interacting intracellular receptor 2, PLCβ and protein kinase Cα (PKCα) — are glutamate transport in primary neuronal cultures, and signalling proteins into signalling all enriched in lipid rafts64. In this study, stimulation of cholesterol is implicated as an active factor75. These complexes. cells with a purinergic agonist recruited PLCβ and PKCα data predict that an external mechanism for altering Second messengers to raft fractions, whereas lipid raft disruption slowed cholesterol levels in rafts could regulate the amount of a Small intracellular signalling propagation of agonist-evoked calcium waves. neurotransmitter available to interact with its receptor. molecules generated in large The reduction in the transport rate for 5-HTT numbers in response to the G-protein- and cyclic nucleotide-gated ion channels. The after cholesterol depletion occurs with a concomitant primary message of a hormone or neurotransmitter. lipid raft compartmentalization of G-protein-gated or reduction in the affinity of the transporter for 5-HT, cyclic nucleotide-gated (CNG) ion channels also has func- indicating that lipid rafts might promote a high-affinity Calcium wave tional consequences. G-protein-gated inwardly rectifying state of the transporter70,71. Several studies support the A phenomena by which K+ (Kir3) channels are localized in lipid rafts and the lipid idea of specific cholesterol–transporter interactions over calcium released from intracellular stores diffuses raft-associated neural cell adhesion molecule (NCAM) the effects of cholesterol on general membrane properties. within a cell in a pattern of impairs the delivery of functional 5-HT1A-coupled Optimal 5-HTT and GABA transporter activity is not waves. Kir3 channels to the cell surface65. So, lipid rafts and observed when cholesterol is replaced with sterols that

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | FEBRUARY 2007 | 135 © 2007 Nature Publishing Group

REVIEWS

a internalization of the noradrenaline transporter is a Plasma Caveola Planar lipid raft dynamin- and clathrin-independent process that is membrane blocked by the cholesterol-disrupting agents filipin and E nystatin, which is an indication of a lipid raft-mediated process76. By contrast, similar experiments reveal that PKC-induced internalization of the dopamine trans- E porter (DAT) is dynamin- and clathrin-dependent, and is independent of lipid rafts77. Internalization of 5-HTT in rat midbrain synaptosomes also redistributes Cytoskeletal 5-HTT from lipid rafts78; however, the ramifications of disruption this movement for internalization is unclear, as a defining b study for the requirement of lipid rafts in 5-HTT traffick- ing is lacking. Lipid rafts might also function to insert E E transporters into the plasma membrane, as demonstrated for the EAAT2 and EAAT3 glutamate transporters73, and it would be interesting to determine if lipid rafts target transporters to specific regions of the neuronal plasma membrane.

G-protein-coupled Cytoskeletal components and lipid rafts CaveolinMicrotubule Effector receptor E Lipid rafts are defined as being both rich in choles- terol and associated with the cytoskeleton. But which Ionotropic G protein Tubulin Actin filament cytoskeletal elements are associated with neuronal receptor lipid rafts, and how do they contribute to raft-mediated signalling? And, consequently, how do neurotransmitter Figure 3 | Cytoskeletal and lipid raft organization of neurotransmitter signalling molecules. a | Actin and tubulin associate with lipid rafts and caveolae either as signals, emanating from rafts, modify the cytoskeleton? polymerized structures or their building block components (actin monomers or tubulin The relationship between cytoskeletal elements and lipid dimers). These cytoskeletal elements might help to organize lipid raft domains and the rafts is still emerging; however, it seems that microtubules neurotransmitter molecules that are present in these structures (G-protein-coupled and actin filaments are the primary interacting partners receptors, ionotropic receptors, effectors, G proteins). b | Disruption of actin filaments or of lipid rafts. Each is considered below and in FIG. 3. microtubules reorganizes certain receptors or G proteins present in lipid rafts, as described in the main text. It is suggested that tubulin-binding or actin-binding drugs Rafts and microtubule dynamics. Tubulin, the build- could compromise the association between dimeric tubulin and receptors or G proteins. ing block of microtubules, is resident in lipid rafts Depending on the system, cytoskeletal elements present in rafts would be expected to and can be co-immunoprecipitated with CAV1 in rat facilitate or inhibit neurotransmitter signalling by contributing to the raft organization of forebrain extracts79. One possible mechanism for the the signalling molecules. Adapted, with permission, from REF. 87 © (2006) American Society for Biochemistry and Molecular Biology. contribution of lipid rafts to neurotransmitter-induced alterations in microtubules is indicated by experiments in cells. In these cells, caveolins might stabilize microtubules by interfering with the interaction have similar effects on membrane fluidity70,74, and cho- between the microtubule-destabilizing protein stathmin lesterol depletion by various distinct mechanisms all and tubulin80. Unfortunately, this model contains no reduce the affinity of 5-HTT for its substrate70. These data provision for agonist-mediated modifications of the α indicate that specific cholesterol–transporter interactions microtubule cytoskeleton. G proteins, specifically G s, exist independant of the effects of cholesterol on general could provide such a link, as they reside in lipid rafts membrane properties. There is also the possibility that (TABLE 2) and, more importantly, internalize through different transporters differentially interact with lipid these rafts in response to agonist activation48. Certain G rafts. For example, the glutamate transporter can func- proteins have been shown to promote the GTPase activ- tion optimally at four- to fivefold lower cholesterol levels ity of tubulin and to increase the dynamic behaviour of than the GABA transporter can74, and the -specific microtubules81. Microtubules are particularly dynamic in 82 α excitatory amino acid transporter 2 (EAAT2) glutamate growth cones , and translocation of G s to the cytosol transporter is more tightly associated with lipid rafts through lipid rafts after activation of a GPCR might be and more sensitive to cholesterol depletion than other a mechanism for neurotransmitter-mediated changes in glutamate transporter subtypes73. Further investigation is neurons. Although dendritic spines lack microtubules, needed to determine the molecular mechanisms involved the microtubule apparatus reaches the base of the in the regulation of transporter function by lipid rafts and dendritic spine and might participate in neuronal Microtubules whether these mechanisms are transporter specific. extension and retraction of spines. Hollow tubes, 25 nm in Phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) diameter, formed by the Regulating transporter levels. One mechanism for con- serves as a tubulin anchor on the plasma membrane83. lateral association of 13 protofilaments which are trolling the amount of neurotransmitter in the synapse Acetylcholine-mediated hydrolysis of PtdIns(4,5)P2 themselves polymers of α- is to alter the amount of functional transporters at the releases tubulin from the plasma membrane and directs and β-tubulin subunits. plasma membrane in neurons or glia. PKC-mediated its internalization84 for subsequent re-use in microtubule

136 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group

REVIEWS

formation. This mechanism could promote micro- treat mood disorders (for example, unipolar depression,

tubule formation in regions subjacent to PtdIns(4,5)P2 - bipolar affective disorder) and schizophrenia accumulate 94 enriched lipid rafts. in lipid rafts and affect the signalling of resident pro- teins. Although chronic antidepressant treatment does Tubulin and neurotransmitter signalling. The role not alter membrane cholesterol content, treatment with of rafts in mediating microtubule dynamics remains chemically distinct antidepressants results in the move- α unclear, but the tubulin that is present in lipid rafts ment of G s (but not other G proteins) out of lipid rafts might well participate in neurotransmitter signalling. and into a closer association with adenylyl cyclase50,86,95. This could occur through the role of tubulin as a scaf- This might contribute to the increased cAMP tone and folding protein in rafts, through a dynamic interaction synaptic changes that are observed subsequent to chronic of tubulin with signalling proteins, or both. An associa- antidepressant treatment96. The microtubule-disrupting α 86 tion between tubulin and heterotrimeric G proteins has drug colchicine similarly removes G s from lipid rafts 56 α 88 been demonstrated , and this potentiates signalling and enhances G s-stimulated adenylyl cyclase activity . of both adrenergic and cholinergic neurotransmitters In concert, these data indicate that antidepressants by directly activating their respective G proteins57,85. might exert some of their therapeutic efficacy by alter- Curiously, treatment of glial cells or cardiac myocytes ing a component of the membrane or cytoskeleton that with microtubule-disrupting agents such as colchicine is associated with lipid rafts. results in the loss of many signalling molecules from There is also evidence in the literature for the involve- lipid raft membranes — in particular, those involved in ment of lipid rafts in the pathogenesis of several neu- AR signalling86,87. Colchicine treatment also significantly rodegenerative diseases. Lipid rafts might function as β α increases signalling through the -AR–G s–adenylyl platforms for the production of neurotoxic proteins, such cyclase system87,88. A putative reason for this might be as amyloid-β in Alzheimer’s disease97 and prion protein α 98 that colchicine disrupts the complexes between G s in transmissible spongiform encephalopathies , which α and tubulin in lipid rafts, releasing G s from raft micro- could then be poised to modulate raft-associated signal- α β domains. This ‘liberated’ G s might facilitate increased ling cascades. For example, amyloid- can modulate the α α 99 signalling from GPCRs that couple to G s and activate activity of the nACh 7 subunit of the nACh receptor , adenylyl cyclase. So, microtubules and tubulin might both of which are raft-associated proteins; however, a help to sequester and anchor signalling molecules in raft direct relationship has yet to be uncovered. Prion protein domains where their activity is diminished. can redistribute caveolins from lipid rafts100, and over- expression of α-synuclein — a protein which aberrantly Actin and lipid rafts. The actin cytoskeleton seems accumulates in Parkinson’s disease — can upregulate to have a bidirectional relationship with lipid rafts. caveolin 1 (REF. 101). Furthermore, Cav1-knockout mice

Phosphoinositide lipids such as PtdIns(4,5)P2 and show abnormal neurological phenotypes including clasp-

PtdIns(3,4)P2 have been shown to accumulate in lipid ing, muscle weakness and reduced activity — results that rafts, and these lipids are also known to bind actin and are consistent with altered motor control5. Given the role direct actin assembly into filaments89,90. Neurotransmitters of caveolins as scaffolding proteins and in the regula-

that induce the hydrolysis of PtdIns(4,5)P2 promote the tion of numerous signalling molecules, disease-related removal of actin from these anchoring sites at the mem- alterations in caveolin localization and expression could brane91. Dendritic spines show actin attachment on their globally affect neurotransmitter signalling. distal end, and rapid remodelling of these spines in living

neurons might result from the turnover of PtdIns(4,5)P2 Conclusions modulating either the dynamics of actin filaments or This review has synthesized existing information on lipid their binding sites on the membrane20. rafts and their role in neurotransmitter signalling. We However, actin also has a role in clustering signal- have identified unique instances where lipid rafts regulate ling molecules in lipid rafts. Small G proteins cluster in the processes of neurotransmitter receptor (ionotropic lipid rafts, and this seems to be dependent on the actin or GPCR) activation/inactivation or neurotransmitter cytoskeleton92. Small G proteins influence cell shape at the uptake and have illustrated how lipid rafts modulate the membrane and various developmental and survival pro- aggregation of signalling components. Curiously, there is grammes inside the cell by controlling kinase cascades. no clear definition for the role of rafts in neurotransmitter These GTPases change their raft localization in response signalling, as they seem equally likely to be domains in to the external signals that modify the actin cytoskele- which signalling is enhanced or inhibited. This allows ton93. Therefore, agents that modify the raft association for a rich complexity in signalling, suggesting that differ- of actin can utilize small G proteins and other signalling ent lipid raft compartments might have opposing effects molecules to further modify the cell shape or to evoke on different neurotransmitters. Furthermore, the unique sustained cell signalling pathways in the cell interior89. spatial segregation afforded by raft domains could allow a single neurotransmitter–receptor pair to channel Lipid rafts and disorders of the brain different signalling pathways along different time courses Colchicine The burgeoning interest in lipid rafts by neuroscien- or in limited areas of the same . Alkaloid used to inhibit the polymerization of tubulin and tists has also prompted studies to explore if and how This complexity is enhanced when further consid- cause the depolymerization of these microdomain components contribute to various eration is given to the relationship between neuronal- microtubules. neurological and psychiatric diseases. Drugs used to specific structures such as the postsynaptic density and

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | FEBRUARY 2007 | 137 © 2007 Nature Publishing Group

REVIEWS

Postsynaptic density lipid rafts. Even though many neurotransmitter receptors the process are rooted in experimental methodologies An electron-dense thickening are highly enriched and organized in the postsynaptic that have failed to deliver a holistic determination of underneath the postsynaptic density, it is unclear whether lipid rafts interact with, the physio logy of rafts as they relate to neurotransmitter membrane at excitatory or are part of, this structure. The raft marker flotillin signalling (BOXES 1,2). Future studies designed to synapses that contains is present in dendritic spines of the rat cerebral cortex define the relationship between these unique synaptic receptors, structural proteins linked to the actin cytoskeleton (which commonly contain postsynaptic densities), and structures and lipid raft microdomains with respect and signalling elements, such detergent-resistant raft-like fractions have been isolated to organizing both spatial and temporal aspects of as kinases and phosphatases. from preparations of synaptic membranes102. A func- neuronal signalling are essential, and these studies need tional relationship between rafts, the postsynaptic density to combine biochemical, biophysical, genetic and imag- and the cytoskeletal networks comprising them remains ing techniques — or develop new ones — to advance our to be established. Limitations on our understanding of understanding.

1. Singer, S. J. & Nicolson, G. L. The fluid mosaic model 16. Burger, K., Gimpl, G. & Fahrenholz, F. Regulation of 33. Monastyrskaya, K., Hostettler, A., Buergi, S. & of the structure of cell membranes. Science 175, receptor function by cholesterol. Cell. Mol. Life Sci. Draeger, A. The NK1 receptor localizes to the plasma 720–731 (1972). 57, 1577–1592 (2000). membrane microdomains, and its activation is 2. Nicolau, D. V., Burrage, K., Parton, R. G. & 17. Laube, B., Kuhse, J. & Betz, H. Evidence for a dependent on lipid raft integrity. J. Biol. Chem. 280, Hancock, J. F. Identifying optimal lipid raft tetrameric structure of recombinant NMDA receptors. 7135–7146 (2005). characteristics required to promote nanoscale J. Neurosci. 18, 2954–2961 (1998). 34. Xu, W. et al. Localization of the κ opioid receptor in protein–protein interactions on the plasma 18. Wang, Z. Z., Hardy, S. F. & Hall, Z. W. Assembly of the lipid rafts. J. Pharmacol. Exp. Ther. 317, 1295–1306 membrane. Mol. Cell Biol. 26, 313–323 (2006). nicotinic acetylcholine receptor. The first (2006). 3. Rebois, R. V. & Hebert, T. E. Protein complexes transmembrane domains of truncated α and δ 35. Gines, S. et al. Involvement of caveolin in ligand-induced

involved in heptahelical receptor-mediated signal subunits are required for heterodimer formation recruitment and internalization of A1 adenosine transduction. Receptors Channels 9, 169–194 (2003). in vivo. J. Biol. Chem. 271, 27575–27584 (1996). receptor and adenosine deaminase in an epithelial cell 4. Simons, K. & Toomre, D. Lipid rafts and signal 19. Pediconi, M. F., Gallegos, C. E., De Los Santos, E. B. & line. Mol. Pharmacol. 59, 1314–1323 (2001). transduction. Nature Rev. Mol. Cell Biol. 1, 31–39 Barrantes, F. J. Metabolic cholesterol depletion 36. Lai, H. H. et al. Loss of caveolin-1 expression is (2000). hinders cell-surface trafficking of the nicotinic associated with disruption of muscarinic cholinergic This review article regarding lipid rafts and signal acetylcholine receptor. Neuroscience 128, 239–249 activities in the . Neurochem. Int. 45, transduction provides keen insights into lipid raft (2004). 1185–1193 (2004). microdomains and their involvement in cell signalling. 20. Hering, H., Lin, C. C. & Sheng, M. Lipid rafts in the 37. Yu, P. et al. D1 dopamine receptor signaling involves 5. Trushina, E., Du Charme, J., Parisi, J. & McMurray, C. T. maintenance of synapses, dendritic spines, and caveolin-2 in HEK-293 cells. Kidney Int. 66, Neurological abnormalities in caveolin-1 knock out surface AMPA receptor stability. J. Neurosci. 23, 2167–2180 (2004). mice. Behav. Brain Res. 172, 24–32 (2006). 3262–3271 (2003). 38. Burgueno, J. et al. Metabotropic glutamate type 1α Evaluates Cav1-knockout mice and their associated These authors demonstrate that, in rat receptor localizes in low-density caveolin-rich plasma neurological phenotypes. The results show that the hippocampal neurons, lipid rafts enriched in membrane fractions. J. Neurochem. 86, 785–791 knockout animals develop phenotypes which postsynaptic proteins are abundant in dendrites (2003). dictate impaired motor control. and are important for maintenance of synaptic 39. Burgueno, J. et al. Mutual regulation between 6. Lang, D. M. et al. Identification of reggie-1 and reggie- morphology and density. metabotropic glutamate type 1α receptor and 2 as plasmamembrane-associated proteins which 21. Trinidad, J. C. & Cohen, J. B. Neuregulin inhibits caveolin proteins: from traffick to constitutive activity. cocluster with activated GPI-anchored cell adhesion acetylcholine receptor aggregation in myotubes. Exp. Cell Res. 300, 23–34 (2004). molecules in non-caveolar micropatches in neurons. J. Biol. Chem. 279, 31622–31628 (2004). 40. Rajendran, L. & Simons, K. Lipid rafts and membrane J. Neurobiol. 37, 502–523 (1998). 22. Bruses, J. L., Chauvet, N. & Rutishauser, U. dynamics. J. Cell Sci. 118, 1099–1102 (2005). 7. Bhatnagar, A., Sheffler, D. J., Kroeze, W. K., Membrane lipid rafts are necessary for the 41. Nichols, B. Caveosomes and endocytosis of lipid rafts. Compton-Toth, B. & Roth, B. L. Caveolin-1 interacts maintenance of the α 7 nicotinic acetylcholine receptor J. Cell Sci. 116, 4707–4714 (2003). with 5-HT2A serotonin receptors and profoundly in somatic spines of ciliary neurons. J. Neurosci. 21, 42. Le Roy, C. & Wrana, J. L. Clathrin- and non-clathrin- α modulates the signaling of selected G q-coupled 504–512 (2001). mediated endocytic regulation of cell signalling. protein receptors. J. Biol. Chem. 279, 34614–34623 23. Zhu, D., Xiong, W. C. & Mei, L. Lipid rafts serve as a Nature Rev. Mol. Cell Biol. 6, 112–126 (2005). (2004). signaling platform for nicotinic acetylcholine receptor 43. Claing, A., Laporte, S. A., Caron, M. G. & Lefkowitz, R. J. Utilizes a novel molecular approach to stably clustering. J. Neurosci. 26, 4841–4851 (2006). Endocytosis of G protein-coupled receptors: roles of G knock-down caveolin 1/2 by RNAi in glial cells, Describes the raft-dependent signalling events protein-coupled receptor kinases and β-arrestin resulting in impaired serotonin signalling. involved in the recruitment and anchoring of the proteins. Prog. Neurobiol. 66, 61–79 (2002). 8. Lenne, P. F. et al. Dynamic molecular confinement in AChR into lipid rafts, thereby providing a 44. Rapacciuolo, A. et al. Protein kinase A and G protein- the plasma membrane by microdomains and the mechanism for AChR clustering. coupled receptor kinase phosphorylation mediates β-1 cytoskeleton meshwork. EMBO J. 25, 3245–3256 24. Campagna, J. A. & Fallon, J. Lipid rafts are involved in adrenergic receptor endocytosis through different (2006). C95 (4,8) agrin fragment-induced acetylcholine pathways. J. Biol. Chem. 278, 35403–35411 (2003). 9. Kusumi, A., Koyama-Honda, I. & Suzuki, K. Molecular receptor clustering. Neuroscience 138, 123–132 45. Keren, O. & Sarne, Y. Multiple mechanisms of CB1 dynamics and interactions for creation of stimulation- (2006). cannabinoid receptors regulation. Brain Res. 980, induced stabilized rafts from small unstable steady- 25. Fuhrer, C., Gautam, M., Sugiyama, J. E. & Hall, Z. W. 197–205 (2003). state rafts. Traffic 5, 213–230 (2004). Roles of rapsyn and agrin in interaction of 46. Self, T. J., Oakley, S. M. & Hill, S. J. Clathrin- 10. Stetzkowski-Marden, F., Gaus, K., Recouvreur, M., postsynaptic proteins with acetylcholine receptors. independent internalization of the human histamine Cartaud, A. & Cartaud, J. Agrin elicits membrane lipid J. Neurosci. 19, 6405–6416 (1999). H1-receptor in CHO-K1 cells. Br. J. Pharmacol. 146, condensation at sites of acetylcholine receptor clusters 26. Oshikawa, J. et al. Nicotinic acetylcholine receptor α 7 612–624 (2005). in C2C12 myotubes. J. Lipid Res. 47, 2121–2133 regulates cAMP signal within lipid rafts. Am. J. 47. Rybin, V. O., Xu, X., Lisanti, M. P. & Steinberg, S. F. (2006). Physiol. Cell Physiol. 285, C567–C574 (2003). Differential targeting of β-adrenergic receptor subtypes

11. Hancock, J. F. Lipid rafts: contentious only from 27. Garcia-Marcos, M. et al. Coupling of two pools of P2X7 and adenylyl cyclase to cardiomyocyte caveolae. A simplistic standpoints. Nature Rev. Mol. Cell Biol. 7, receptors to distinct intracellular signaling pathways in mechanism to functionally regulate the cAMP signaling 456–462 (2006). rat submandibular gland. J. Lipid Res. 47, 705–714 pathway. J. Biol. Chem. 275, 41447–41457 (2000). 12. Babiychuk, E. B. & Draeger, A. Biochemical (2006). 48. Allen, J. A., Yu, J. Z., Donati, R. J. & Rasenick, M. M. β α characterization of detergent-resistant membranes: a 28. Abulrob, A. et al. Protection by cholesterol-extracting -adrenergic receptor stimulation promotes G s systematic approach. Biochem. J. 397, 407–416 cyclodextrins: a role for N-methyl-D-aspartate receptor internalization through lipid rafts: a study in living (2006). redistribution. J. Neurochem. 92, 1477–1486 (2005). cells. Mol. Pharmacol. 67, 1493–1504 (2005). 13. Vacca, F., Amadio, S., Sancesario, G., Bernardi, G. & 29. Frank, C., Giammarioli, A. M., Pepponi, R., Fiorentini, C. Examines the endocytic trafficking of activated β α α Volonte, C. P2X3 receptor localizes into lipid rafts in & Rufini, S. Cholesterol perturbing agents inhibit -ARs and G s, and find that G s is endocytosed neuronal cells. J. Neurosci. Res. 76, 653–661 (2004). NMDA-dependent calcium influx in rat hippocampal by a lipid raft-mediated mechanism leading to a 14. Gaudreault, S. B., Chabot, C., Gratton, J. P. & Poirier, J. primary culture. FEBS Lett. 566, 25–29 (2004). putative reduction in cAMP signalling. The caveolin scaffolding domain modifies 2-amino-3- 30. Guirland, C., Suzuki, S., Kojima, M., Lu, B. & 49. Sabourin, T., Bastien, L., Bachvarov, D. R. & hydroxy-5-methyl-4-isoxazole propionate receptor Zheng, J. Q. Lipid rafts mediate chemotropic guidance Marceau, F. Agonist-induced translocation of the kinin

binding properties by inhibiting phospholipase A2 of nerve growth cones. Neuron 42, 51–62 (2004). B1 receptor to caveolae-related rafts. Mol. Pharmacol. activity. J. Biol. Chem. 279, 356–362 (2004). 31. Neubig, R. R. Membrane organization in G-protein 61, 546–553 (2002). 15. Sooksawate, T. & Simmonds, M. A. Effects of mechanisms. FASEB J. 8, 939–946 (1994). 50. Toki, S., Donati, R. J. & Rasenick, M. M. Treatment of

membrane cholesterol on the sensitivity of the GABAA 32. Pucadyil, T. J. & Chattopadhyay, A. Cholesterol C6 glioma cells and with antidepressant drugs α receptor to GABA in acutely dissociated rat modulates ligand binding and G-protein coupling to increases the detergent extraction of Gs from

hippocampal neurones. Neuropharmacology 40, serotonin1A receptors from bovine hippocampus. plasma membrane. J. Neurochem. 73, 1114–1120 178–184 (2001). Biochim. Biophys. Acta 1663, 188–200 (2004). (1999).

138 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group

REVIEWS

51. Nair, K. S., Balasubramanian, N. & Slepak, V. Z. 73. Butchbach, M. E., Tian, G., Guo, H. & Lin, C. L. 92. Plowman, S. J., Muncke, C., Parton, R. G. & Signal-dependent translocation of transducin, RGS9– Association of excitatory amino acid transporters, Hancock, J. F. H-ras, K-ras, and inner plasma 1-Gβ5L complex, and arrestin to detergent-resistant especially EAAT2, with cholesterol-rich lipid raft membrane raft proteins operate in nanoclusters with membrane rafts in photoreceptors. Curr. Biol. 12, microdomains: importance for excitatory amino acid differential dependence on the actin cytoskeleton. 421–425 (2002). transporter localization and function. J. Biol. Chem. Proc. Natl Acad. Sci. USA 102, 15500–15505 52. Head, B. P. et al. G-protein-coupled receptor signaling 279, 34388–34396 (2004). (2005). components localize in both sarcolemmal and 74. Shouffani, A. & Kanner, B. I. Cholesterol is required 93. Jaksits, S. et al. Lipid raft-associated GTPase signaling intracellular caveolin-3-associated microdomains in for the reconstruction of the sodium- and chloride- controls morphology and CD8+ T cell stimulatory adult cardiac myocytes. J. Biol. Chem. 280, coupled, γ-aminobutyric acid transporter from rat capacity of human dendritic cells. J. Immunol. 173, 31036–31044 (2005). brain. J. Biol. Chem. 265, 6002–6008 (1990). 1628–1639 (2004). 53. Dunphy, J. T., Greentree, W. K. & Linder, M. E. 75. Canolle, B. et al. Glial soluble factors regulate the 94. Eisensamer, B. et al. Antidepressants and Enrichment of G-protein palmitoyltransferase activity activity and expression of the neuronal glutamate antipsychotic drugs colocalize with 5-HT3 receptors in low density membranes: in vitro reconstitution of transporter EAAC1: implication of cholesterol. in raft-like domains. J. Neurosci. 25, 10198–10206 α G i to these domains requires palmitoyltransferase J. Neurochem. 88, 1521–1532 (2004). (2005). activity. J. Biol. Chem. 276, 43300–43304 (2001). 76. Jayanthi, L. D., Samuvel, D. J. & Ramamoorthy, S. 95. Menkes, D. B., Rasenick, M. M., Wheeler, M. A. & 54. Moffett, S., Brown, D. A. & Linder, M. E. Lipid- Regulated internalization and phosphorylation of the Bitensky, M. W. triphosphate activation of dependent targeting of G proteins into rafts. J. Biol. native norepinephrine transporter in response to brain adenylate cyclase: enhancement by long-term Chem. 275, 2191–2198 (2000). phorbol esters. Evidence for localization in lipid rafts antidepressant treatment. Science 219, 65–67 55. Li, S. et al. Evidence for a regulated interaction and lipid raft-mediated internalization. J. Biol. Chem. (1983). between heterotrimeric G proteins and caveolin. 279, 19315–19326 (2004). 96. Donati, R. J. & Rasenick, M. M. G protein signaling J. Biol. Chem. 270, 15693–15701 (1995). 77. Sorkina, T., Hoover, B. R., Zahniser, N. R. & Sorkin, A. and the molecular basis of antidepressant action. 56. Wang, N., Yan, K. & Rasenick, M. M. Tubulin binds Constitutive and protein kinase C-induced Life Sci. 73, 1–17 (2003). specifically to the signal-transducing proteins, Gs α internalization of the dopamine transporter is 97. Ehehalt, R., Keller, P., Haass, C., Thiele, C. & Simons, K. and Gi α 1. J. Biol. Chem. 265, 1239–1242 (1990). mediated by a clathrin-dependent mechanism. Traffic Amyloidogenic processing of the Alzheimer β-amyloid 57. Popova, J. S., Garrison, J. C., Rhee, S. G. & 6, 157–170 (2005). precursor protein depends on lipid rafts. Rasenick, M. M. Tubulin, Gq, and phosphatidylinositol 78. Samuvel, D. J., Jayanthi, L. D., Bhat, N. R. & J. Cell Biol. 160, 113–123 (2003). 4, 5-bisphosphate interact to regulate phospholipase Ramamoorthy, S. A role for p38 mitogen-activated 98. Taylor, D. R. & Hooper, N. M. The prion protein and Cβ1 signaling. J. Biol. Chem. 272, 6760–6765 (1997). protein kinase in the regulation of the serotonin lipid rafts. Mol. Membr. Biol. 23, 89–99 (2006). 58. Pesanova, Z., Novotny, J., Cerny, J., Milligan, G. & transporter: evidence for distinct cellular mechanisms 99. Dineley, K. T., Bell, K. A., Bui, D. & Sweatt, J. D. Svoboda, P. Thyrotropin-releasing hormone-induced involved in transporter surface expression. J. Neurosci. β-Amyloid peptide activates α 7 nicotinic acetylcholine α α depletion of Gq /G11 proteins from detergent- 25, 29–41 (2005). receptors expressed in Xenopus oocytes. J. Biol. insensitive membrane domains. FEBS Lett. 464, 79. Dremina, E. S., Sharov, V. S. & Schoneich, C. Protein Chem. 277, 25056–25061 (2002). 35–40 (1999). tyrosine nitration in rat brain is associated with raft 100. Russelakis-Carneiro, M., Hetz, C., Maundrell, K. & 59. Oh, P. & Schnitzer, J. E. Segregation of heterotrimeric proteins, flotillin-1 and α-tubulin: effect of biological Soto, C. Prion replication alters the distribution of

G proteins in cell surface microdomains. Gq binds aging. J. Neurochem. 93, 1262–1271 (2005). synaptophysin and caveolin 1 in neuronal lipid rafts.

caveolin to concentrate in caveolae, whereas Gi and Gs 80. Kawabe, J., Okumura, S., Nathanson, M. A., Hasebe, N. Am. J. Pathol. 165, 1839–1848 (2004). target lipid rafts by default. Mol. Biol. Cell 12, & Ishikawa, Y. Caveolin regulates microtubule 101. Hashimoto, M., Takenouchi, T., Rockenstein, E. & 685–698 (2001). polymerization in the vascular smooth muscle cells. Masliah, E. α-synuclein up-regulates expression of

60. Miura, Y., Hanada, K. & Jones, T. L. Gs signaling is Biochem. Biophys. Res. Commun. 342, 164–169 caveolin-1 and down-regulates extracellular signal- intact after disruption of lipid rafts. Biochemistry 40, (2006). regulated kinase activity in B103 neuroblastoma 15418–15423 (2001). 81. Roychowdhury, S., Panda, D., Wilson, L. & cells: role in the pathogenesis of Parkinson’s disease. 61. Hiol, A. et al. Palmitoylation regulates regulators of Rasenick, M. M. G protein α subunits activate tubulin J. Neurochem. 85, 1468–1479 (2003). G-protein signaling (RGS) 16 function. I. Mutation of GTPase and modulate microtubule polymerization 102. Suzuki, T. Lipid rafts at postsynaptic sites: distribution, amino-terminal cysteine residues on RGS16 prevents dynamics. J. Biol. Chem. 274, 13485–13490 function and linkage to postsynaptic density. Neurosci. its targeting to lipid rafts and palmitoylation of an (1999). Res. 44, 1–9 (2002). internal cysteine residue. J. Biol. Chem. 278, 82. Suter, D. M., Schaefer, A. W. & Forscher, P. 103. Munro, S. Lipid rafts: elusive or illusive? Cell 115, 19301–19308 (2003). Microtubule dynamics are necessary for SRC family 377–388 (2003). 62. Fagan, K. A., Smith, K. E. & Cooper, D. M. Regulation kinase-dependent growth cone steering. Curr. Biol. 104. Lichtenberg, D., Goni, F. M. & Heerklotz, H. of the Ca2+-inhibitable adenylyl cyclase type VI by 14, 1194–1199 (2004). Detergent-resistant membranes should not be 2+ capacitative Ca entry requires localization in 83. Golub, T. & Caroni, P. PI(4,5)P2-dependent identified with membrane rafts. Trends Biochem. Sci. cholesterol-rich domains. J. Biol. Chem. 275, microdomain assemblies capture microtubules to 30, 430–436 (2005). 26530–26537 (2000). promote and control leading edge motility. J. Cell Biol. 105. Douglass, A. D. & Vale, R. D. Single-molecule 63. Toya, Y., Schwencke, C., Couet, J., Lisanti, M. P. & 169, 151–165 (2005). microscopy reveals plasma membrane microdomains Ishikawa, Y. Inhibition of adenylyl cyclase by caveolin 84. Popova, J. S. & Rasenick, M. M. Clathrin-mediated created by protein–protein networks that exclude or peptides. Endocrinology 139, 2025–2031 (1998). endocytosis of m3 muscarinic receptors. Roles for Gβγ trap signaling molecules in T cells. Cell 121, 937–950 64. Weerth, S. H., Holtzclaw, L. A. & Russell, J. T. Signaling and tubulin. J. Biol. Chem. 279, 30410–30418 (2005). proteins in raft-like microdomains are essential for (2004). 106. Brown, D. A. & Rose, J. K. Sorting of GPI-anchored Ca2+ wave propagation in glial cells. Cell Calcium 85. Yan, K., Popova, J. S., Moss, A., Shah, B. & proteins to glycolipid-enriched membrane subdomains 14 Aug 2006 (doi:10.1016/j.ceca.2006.06.006). Rasenick, M. M. Tubulin stimulates adenylyl cyclase during transport to the apical cell surface. Cell 68, 65. Delling, M. et al. The neural cell adhesion molecule activity in C6 glioma cells by bypassing the 533–544 (1992). regulates cell-surface delivery of G-protein-activated β-adrenergic receptor: a potential mechanism of G Determined that, on entering the Golgi complex, inwardly rectifying potassium channels via lipid rafts. protein activation. J. Neurochem. 76, 182–190 GPI-anchored proteins are localized to glycolipid- J. Neurosci. 22, 7154–7164 (2002). (2001). rich membranes isolated by differential solubility 66. Toselli, M. et al. Attenuation of G protein-mediated 86. Donati, R. J. & Rasenick, M. M. Chronic in non-ionic detergents. This work provided the inhibition of N-type calcium currents by expression of antidepressant treatment prevents accumulation of first reported method to isolate detergent- caveolins in mammalian NG108-15 cells. J. Physiol. gsα in cholesterol-rich, cytoskeletal-associated, plasma resistant membranes. 536, 361–373 (2001). membrane domains (lipid rafts). 107. Song, K. S. e. a. Co-purification and direct interaction 67. Brady, J. D. et al. Functional role of lipid raft Neuropsychopharmacology 30, 1238–1245 (2005). of Ras with caveolin, an integral membrane protein of microdomains in cyclic nucleotide-gated channel 87. Head, B. P. et al. Microtubules and actin caveolae microdomains. Detergent-free purification activation. Mol. Pharmacol. 65, 503–511 (2004). microfilaments regulate lipid raft/caveolae localization of caveolae microdomains. J. Biol. Chem. 271, 68. Richelson, E. Interactions of antidepressants with of adenylyl cyclase signaling components. J. Biol. 9690–9697 (1996). neurotransmitter transporters and receptors and Chem. 281, 26391–26399 (2006). Introduced the non-detergent method of isolating their clinical relevance. J. Clin. Psychiatry 64, 5–12 Demonstrates that disrupting either the actin or lipid raft/caveolae membranes using sodium (2003). the microtubule cytoskeleton in cardiac myocytes carbonate. 69. North, P. & Fleischer, S. Alteration of synaptic reduces the number of caveolae and also removes 108. Parton, R. G., Hanzal-Bayer, M. & Hancock, J. F. membrane cholesterol/phospholipid ratio using a lipid adenylyl cyclase molecules from rafts/caveolae, Biogenesis of caveolae: a structural model for caveolin- transfer protein. Effect on γ-aminobutyric acid uptake. which enhances cAMP signalling. induced domain formation. J. Cell Sci. 119, 787–796 J. Biol. Chem. 258, 1242–1253 (1983). 88. Rasenick, M. M., Stein, P. J. & Bitensky, M. W. The (2006). 70. Scanlon, S. M., Williams, D. C. & Schloss, P. regulatory subunit of adenylate cyclase interacts with 109. Marchand, S., Devillers-Thiery, A., Pons, S., Membrane cholesterol modulates serotonin cytoskeletal components. Nature 294, 560–562 Changeux, J. P. & Cartaud, J. Rapsyn escorts the transporter activity. Biochemistry 40, 10507–10513 (1981). nicotinic acetylcholine receptor along the exocytic (2001). 89. Caroni, P. New EMBO members’ review: actin pathway via association with lipid rafts. J. Neurosci. 71. Magnani, F., Tate, C. G., Wynne, S., Williams, C. & cytoskeleton regulation through modulation of 22, 8891–8901 (2002).

Haase, J. Partitioning of the serotonin transporter PI(4,5)P2 rafts. EMBO J. 20, 4332–4336 (2001). 110. Dalskov, S. M. et al. Lipid raft localization of GABAA into lipid microdomains modulates transport of 90. Pollard, T. D. & Borisy, G. G. Cellular motility driven by receptor and Na+, K+-ATPase in discrete microdomain serotonin. J. Biol. Chem. 279, 38770–38778 assembly and disassembly of actin filaments. Cell 112, clusters in rat cerebellar granule cells. Neurochem. Int. (2004). 453–465 (2003). 46, 489–499 (2005). 72. Butchbach, M. E., Guo, H. & Lin, C. L. Methyl- 91. Bittner, M. A. & Holz, R. W. Phosphatidylinositol-4, 111. Suzuki, T. et al. Biochemical evidence for localization β-cyclodextrin but not retinoic acid reduces EAAT3- 5-bisphosphate: actin dynamics and the regulation of of AMPA-type glutamate receptor subunits in the mediated glutamate uptake and increases GTRAP3-18 ATP-dependent and-independent secretion. Mol. dendritic raft. Brain Res. Mol. Brain Res. 89, 20–28 expression. J. Neurochem. 84, 891–894 (2003). Pharmacol. 67, 1089–1098 (2005). (2001).

NATURE REVIEWS | NEUROSCIENCE VOLUME 8 | FEBRUARY 2007 | 139 © 2007 Nature Publishing Group

REVIEWS

112. Vial, C. & Evans, R. J. Disruption of lipid rafts inhibits 119. Becher, A. et al. Ectopically expressed γ-aminobutyric 126. Seno, K. et al. Light- and guanosine 5’-3-O-

P2X1 receptor-mediated currents and arterial acid receptor B is functionally down-regulated in (thio)triphosphate-sensitive localization of a G protein . J. Biol. Chem. 280, 30705–30711 isolated lipid raft-enriched membranes. Biochem. and its effector on detergent-resistant membrane rafts (2005). Biophys. Res. Commun. 321, 981–987 (2004). in rod photoreceptor outer segments. J. Biol. Chem. 113. Zschocke, J., Bayatti, N. & Behl, C. Caveolin and GLT-1 120. Zhao, H., Loh, H. H. & Law, P. Y. Adenylyl cyclase 276, 20813–20816 (2001). gene expression is reciprocally regulated in primary superactivation induced by long-term treatment with astrocytes: association of GLT-1 with non-caveolar lipid opioid agonist is dependent on receptor localized Acknowledgements This work was supported by research grants from the National rafts. Glia 49, 275–287 (2005). within lipid rafts and is independent of receptor Institutes of Health to M.M.R and by a National Institutes of 114. Dessy, C., Kelly, R. A., Balligand, J. L. & Feron, O. internalization. Mol. Pharmacol. 69, 1421–1432 Health Training Grant to J.A.A. and R.H. We thank R. Donati Dynamin mediates caveolar sequestration of (2006). for valuable comments about this manuscript. muscarinic cholinergic receptors and alteration in NO 121. Dreja, K. et al. Cholesterol depletion disrupts caveolae signaling. EMBO J. 19, 4272–4280 (2000). and differentially impairs agonist-induced arterial Competing interests statement 115. Ostrom, R. S. et al. Receptor number and caveolar contraction. Arterioscler. Thromb. Vasc. Biol. 22, The authors declare no competing financial interests. co-localization determine receptor coupling efficiency to 1267–1272 (2002). adenylyl cyclase. J. Biol. Chem. 276, 42063–42069 122. Navratil, A. M. et al. Constitutive localization of the (2001). gonadotropin-releasing hormone (GnRH) receptor to DATABASES 116. Bari, M., Battista, N., Fezza, F., Finazzi-Agro, A. & low density membrane microdomains is necessary for The following terms in this article are linked online to: Maccarrone, M. Lipid rafts control signaling of type-1 GnRH signaling to ERK. J. Biol. Chem. 278, Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query. cannabinoid receptors in neuronal cells. Implications 31593–31602 (2003). fcgi?db=gene for anandamide-induced . J. Biol. Chem. 123. Rimoldi, V. et al. Oxytocin receptor elicits different α caveolin 1 | caveolin 2 | caveolin 3 | CB1 | EAAT2 | G s | NCAM | 280, 12212–12220 (2005). EGFR/MAPK activation patterns depending on its RGS16 | tubulin 117. Genedani, S. et al. Computer-assisted image analysis localization in caveolin-1 enriched domains. Oncogene OMIM: http://www.ncbi.nlm.nih.gov/entrez/query. of caveolin-1 involvement in the internalization 22, 6054–6060 (2003). fcgi?db=OMIM process of adenosine A -dopamine D receptor 124. Waheed, A. A. & Jones, T. L. Hsp90 interactions and 2A 2 Alzheimer’s disease | bipolar affective disorder | Parkinson’s α α heterodimers. J. Mol. Neurosci. 26, 177–184 acylation target the G protein G 12 but not G 13 to disease | schizophrenia | unipolar depression (2005). lipid rafts. J. Biol. Chem. 277, 32409–32412 (2002). 118. Becher, A., White, J. H. & McIlhinney, R. A. The 125. Arni, S., Keilbaugh, S. A., Ostermeyer, A. G. & FURTHER INFORMATION γ-aminobutyric acid receptor B, but not the Brown, D. A. Association of GAP-43 with detergent- Rasenick’s laboratory: http://www.uic.edu/depts/mcpb/ metabotropic glutamate receptor type-1, associates resistant membranes requires two palmitoylated pages/rasenick/rasenick.htm with lipid rafts in the rat cerebellum. J. Neurochem. cysteine residues. J. Biol. Chem. 273, 28478–28485 Access to this links box is available online. 79, 787–795 (2001). (1998).

140 | FEBRUARY 2007 | VOLUME 8 www.nature.com/reviews/neuro © 2007 Nature Publishing Group