Cilia are specialized surface regions of eukaryotic cells that serve a variety of functions, ranging from motility to sensation and to regulation of cell growth and differentiation. The discovery that a number of human diseases, collectively known as ciliopathies, result from defective cilium function has expanded interest in these structures. Among the many properties of cilia, motility and intraflagellar transport have been most extensively studied. The latter is the process by which multiprotein complexes associate with microtubule motors to transport structural subunits along the axoneme to and from the ciliary tip. By contrast, the mechanisms by which membrane proteins and lipids are specifically targeted to the cilium are still largely unknown. In this Commentary, we review the current knowledge of protein and lipid targeting to ciliary membranes and outline important issues for future study. We also integrate this information into a proposed model of how the cell specifically targets proteins and lipids to the specialized membrane of this unique organelle.

Cilia and flagella are ancient organelles found in organisms spanning the eukaryotic lineage. The basic structure and biogenesis of cilia and flagella are highly conserved. With few exceptions, these organelles consist of a microtubule-based axonemal complex that is assembled on a basal body and projects out from the cell surface, ensheathed in membrane. Because there are no fundamental differences between cilia and flagella, we use the term ‘cilium’ to refer to both structures.

Cilia can be motile or immotile. Motile cilia have a readily observable function, whether it be the propulsion of the cell through fluid (e.g. the protists) or the sweeping of material along the cell surface (e.g. respiratory epithelial cells). By contrast, since their discovery over 100 years ago (Zimmermann, 1898), immotile cilia have been mostly thought to be vestigial organelles. More recently, however, the finding that dysfunction of immotile cilia is linked to a variety of human diseases (collectively known as ciliopathies) has led to a resurgence of interest in investigating the physiological functions and biogenesis of cilia (Tobin and Beales, 2009). For example, the proteins in which mutations cause autosomal dominant polycystic kidney disease (European Polycystic Kidney Disease Consortium, 1994; Mochizuki et al., 1996) have been shown to function in ciliary signaling (Pazour et al., 2000; Torres and Harris, 2006). Renal tubular epithelial cells sense lumenal urine flow through deflection of their primary cilia. This in turn causes an intraciliary calcium flux, regulated by the transmembrane proteins polycystin-1 and polycystin-2, that modulates downstream STAT6 and Wnt cellular proliferation pathways. Dysregulation of this pathway as a result of mutations in the genes encoding the polycystins leads to the hallmark pathologic features of autosomal dominant polycystic kidney disease.

At the same time that investigations of the primary cilium illuminated its important sensory functions, studies of motile cilia revealed a surprising complexity in these ancient organelles. Putative signaling proteins, including protein kinases and phosphodiesterases, localize to motile cilia in a variety of cell types, indicating the potential for signal transduction. The best-understood signaling pathways that have been characterized in cilia include the guanylyl-cyclase-mediated chemotactic response of sea urchin spermatozoa (Kaupp, 2003) and an adenylyl-cyclase- and phosphorylation-dependent mating process activated by flagellar adhesion of Chlamydomonas (Pan and Snell, 2000). In humans, airway epithelial cilia have recently been shown to regulate their beat frequency in response to extracellular noxious stimuli through a calcium-regulated pathway (Shah, 2009). Phylogenetic analysis of the structural components of the eukaryotic cilium has led to a model whereby a common eukaryotic ancestor contained a single 9 + 2 cilium, probably derived from the microtubule intracellular transport system, with combined sensory and motor functions (Mitchell, 2007). In certain cell types, including vertebrate photoreceptors and olfactory neurons, cilia then further evolved to perform tissue-specific functions.

The recent increase in our understanding of the importance of ciliary function in physiology has led to renewed interest in the molecular mechanisms that underlie ciliary biogenesis and maintenance. Although the current understanding of protein and lipid targeting is not as advanced for the cilium as for organelles such as the endoplasmic reticulum or lysosome, several recent reports have begun to elucidate the mechanisms involved. In this Commentary, we draw on recent findings to present a general model for the transport of ciliary membrane proteins and lipids from the Golgi complex to the ciliary membrane. We focus on molecules known to play specific roles in ciliary targeting, rather than on those molecules that are generally involved in establishment of cell polarity or in mediating vesicular transport. We draw on examples from diverse organisms and assume that key themes, if not exact molecules, are probably common to ciliary biogenesis and targeting in all eukaryotes.

Nearly all ciliary axonemes contain microtubules in either the 9 + 2 arrangement, in which 9 outer doublets surround a central pair, or the 9 + 0 arrangement, in which the central pair of microtubules is absent. Motility is driven by dynein-mediated sliding between these microtubules. In either arrangement, axoneme assembly is driven by a process known as intraflagellar transport (IFT) (Kozminski et al., 1993). The ciliary axoneme and its associated cytoskeletal elements are not elongated by the addition of subunits to the base, but by extension from the growing tip (Johnson and Rosenbaum, 1992; Song and Dentler, 2001; Stephens, 2000). IFT is responsible for the delivery of structural subunits to the ciliary tip, as well as for their recycling back to the cell body (Qin et al., 2004) (Fig. 1). These processes are mediated by two distinct multiprotein complexes, known as the A and B complexes, whose movement is powered by microtubule motors of the kinesin 2 and dynein 1b families for anterograde and retrograde movement, respectively (Kozminski et al., 1995; Pazour et al., 1999; Porter et al., 1999; Signor et al., 1999). Immunogold electron microscopy experiments localize the IFT component IFT52 to the periphery of transition fibers, which suggests that IFT complexes assemble and dock at the base of the cilium before entry into the organelle. These fibers connect the basal body to the membrane at the base of the cilium and create an environment that restricts free entry of diffusible molecules (Deane et al., 2001). This implies that active mechanisms, currently uncharacterized, transport ciliary proteins through or around this barrier. An interesting candidate that might participate in this process is importin-β, a protein with a known role in the transit of nuclear proteins through nuclear pore complexes. Importin-β directly binds to the ciliary protein Crumbs3-CLPI (CRB3-CLPI), and the expression of a dominant-negative importin-β disrupts ciliogenesis, possibly by blocking the entry of proteins into the cilium (Fan et al., 2007).

Fig. 1.

Protein targeting to the ciliary membrane. The eukaryotic cilium is a distinct organelle that is separated from the cytoplasm by transition fibers that connect the basal body to the membrane and separate the ciliary membrane (green) from the periciliary (pale orange) (Reiter and Mostov, 2006) and cell-body membranes (dark orange) (Sloboda and Rosenbaum, 2007). Although cilia in certain cell types differ in the fine details of their structures, functions, mechanisms of assembly and regulation, some general principles have emerged in recent years; this figure attempts to integrate these general concepts. The ciliary membrane has a lipid composition that is distinct from that of the periciliary and cell-body membranes because it is highly enriched in sterols, glycolipids and sphingolipids (Tyler et al., 2009). This specialized composition is probably formed in the Golgi (Ejsing et al., 2009; Schuck and Simons, 2004). Various cell membranes have unique complements of membrane proteins, and one mechanism that might contribute to this specialized composition is the association of certain types of proteins with vesicles of specific lipid composition. The model shown is based on the current literature and recent findings from our own laboratory and outlines six stages: (1).Vesicles of different lipid composition and containing specific cargos form in the Golgi (Klemm et al., 2009). For example, those vesicles rich in sterols and sphinoglipids might load certain lipid raft-associated proteins such as the calflagins. One or more cilium-specific palmitoyl acyltransferases might reside in these vesicles to confer lipid-raft association to their substrates in order to sort them into these vesicles. These ciliary vesicles also load with other proteins that contain ciliary targeting sequences, whereas vesicles of other compositions might load other types of cargo (e.g. proteins bound for the cell-body membrane, shown here as one example). IFT20 also loads with the ciliary-bound vesicles and might serve as an adaptor to recruit other ciliary cargo. (2).Vesicles destined for the cilium interact with GTP-Rab8, which is produced by GDP-GTP exchange catalyzed by Rabin8 and the BBSome. This facilitates movement of the vesicles to the base of the cilium near the transition fiber (Hao and Scholey, 2009; Jin and Nachury, 2009), although the molecular mechanism for transport is not known. (3).Vesicles fuse with the periciliary membrane, and ciliary lipids and proteins enter the cilium. Some vesicles accumulate at the base of the cilium, giving rise to the ciliary necklace structure. The periciliary membrane and transition fiber can take different forms in different cell types. For example, the trypanosome flagellar pocket is a unique dynamic structure through which all endocytosis and exocytosis occurs, and which is much larger than the periciliary membrane of most cells. (4).IFT complexes, which consist of protein subcomplexes IFT-A and IFT-B and are possibly associated with the BBSome (Ou et al., 2005), move cargo along the length of the cilium. The BBSome might also interact directly with the ciliary membrane (Hao and Scholey, 2009). IFT is driven in the anterograde direction by kinesin 2 and in the retrograde direction by dynein 1b. A key component of IFT-B is DYF-11, which might promote a separate membrane association of the IFT complex through Rabaptin5 and GTP-Rab8 (Omori et al., 2008), although these interactions need further investigation because they were not confirmed in other studies (Follit et al., 2009). Ciliary membrane proteins might also be associated with the IFT complexes. (5).IFT complexes disassociate from their cargo at the ciliary tip, where anterograde motors become inactivated and retrograde motors become active. (6).Turnover products are recycled back to the base of the cilium. Note that not all components of IFT are included in this model, and specific elements are not drawn to scale.

Fig. 1.

Protein targeting to the ciliary membrane. The eukaryotic cilium is a distinct organelle that is separated from the cytoplasm by transition fibers that connect the basal body to the membrane and separate the ciliary membrane (green) from the periciliary (pale orange) (Reiter and Mostov, 2006) and cell-body membranes (dark orange) (Sloboda and Rosenbaum, 2007). Although cilia in certain cell types differ in the fine details of their structures, functions, mechanisms of assembly and regulation, some general principles have emerged in recent years; this figure attempts to integrate these general concepts. The ciliary membrane has a lipid composition that is distinct from that of the periciliary and cell-body membranes because it is highly enriched in sterols, glycolipids and sphingolipids (Tyler et al., 2009). This specialized composition is probably formed in the Golgi (Ejsing et al., 2009; Schuck and Simons, 2004). Various cell membranes have unique complements of membrane proteins, and one mechanism that might contribute to this specialized composition is the association of certain types of proteins with vesicles of specific lipid composition. The model shown is based on the current literature and recent findings from our own laboratory and outlines six stages: (1).Vesicles of different lipid composition and containing specific cargos form in the Golgi (Klemm et al., 2009). For example, those vesicles rich in sterols and sphinoglipids might load certain lipid raft-associated proteins such as the calflagins. One or more cilium-specific palmitoyl acyltransferases might reside in these vesicles to confer lipid-raft association to their substrates in order to sort them into these vesicles. These ciliary vesicles also load with other proteins that contain ciliary targeting sequences, whereas vesicles of other compositions might load other types of cargo (e.g. proteins bound for the cell-body membrane, shown here as one example). IFT20 also loads with the ciliary-bound vesicles and might serve as an adaptor to recruit other ciliary cargo. (2).Vesicles destined for the cilium interact with GTP-Rab8, which is produced by GDP-GTP exchange catalyzed by Rabin8 and the BBSome. This facilitates movement of the vesicles to the base of the cilium near the transition fiber (Hao and Scholey, 2009; Jin and Nachury, 2009), although the molecular mechanism for transport is not known. (3).Vesicles fuse with the periciliary membrane, and ciliary lipids and proteins enter the cilium. Some vesicles accumulate at the base of the cilium, giving rise to the ciliary necklace structure. The periciliary membrane and transition fiber can take different forms in different cell types. For example, the trypanosome flagellar pocket is a unique dynamic structure through which all endocytosis and exocytosis occurs, and which is much larger than the periciliary membrane of most cells. (4).IFT complexes, which consist of protein subcomplexes IFT-A and IFT-B and are possibly associated with the BBSome (Ou et al., 2005), move cargo along the length of the cilium. The BBSome might also interact directly with the ciliary membrane (Hao and Scholey, 2009). IFT is driven in the anterograde direction by kinesin 2 and in the retrograde direction by dynein 1b. A key component of IFT-B is DYF-11, which might promote a separate membrane association of the IFT complex through Rabaptin5 and GTP-Rab8 (Omori et al., 2008), although these interactions need further investigation because they were not confirmed in other studies (Follit et al., 2009). Ciliary membrane proteins might also be associated with the IFT complexes. (5).IFT complexes disassociate from their cargo at the ciliary tip, where anterograde motors become inactivated and retrograde motors become active. (6).Turnover products are recycled back to the base of the cilium. Note that not all components of IFT are included in this model, and specific elements are not drawn to scale.

To form a fully functional organelle, IFT-mediated axonemal assembly must be coordinated with the delivery of membrane lipids and proteins. The ciliary membrane is not simply a homogeneous extension of the plasma membrane; extensive data support the notion that the ciliary membrane has a lipid and protein composition that differs from that of the cell-body membrane [see Tyler et al. (Tyler et al., 2009) and references therein]. In theory, the distinct composition of proteins and lipids of the plasma membrane versus the ciliary membrane could be achieved through multiple mechanisms. In a targeted delivery model, the molecules destined for either location would be sorted at an earlier point and then delivered separately to the specific membrane. Alternatively, in a diffusion-retention model, transported molecules would be delivered by a common pathway and freely exchange among different domains, but differ with respect to their selective retention at each target membrane. Experimental evidence has long favored the former model. Early ultrastructural studies of the ciliary membrane showed a ‘ciliary necklace’ in close proximity to axonemal transition fibers that appeared to form a diffusion barrier for entry into the cilium (Gilula and Satir, 1972). Instead, ciliary membrane proteins and lipids are probably delivered to the periciliary membrane directly adjacent the base of the cilium. The earliest ultrastructural studies also support this view; vesicles containing ciliary membrane proteins were visualized fusing with the periciliary membrane in algae (Bouck, 1971). Subsequent experiments in photoreceptors revealed that vesicles accumulate at the base of the connecting cilium before entry, suggesting that vesicles are specifically targeted to this membrane (Papermaster et al., 1985). Therefore, we propose a pathway for ciliary membrane trafficking based on the targeted delivery model, in which vesicle transport is crucial. The steps in this pathway include cargo selection and vesicle budding from the Golgi, vesicle targeting and fusion at the periciliary membrane, and finally active transport of membrane components into the ciliary membrane (Fig. 1).

A principal mechanism by which eukaryotic cells establish distinct compositions for different membrane-bound organelles is through selective incorporation of export cargo at the trans-Golgi into vesicles that are then targeted to specific compartments. This process is regulated by the assembly of several distinct multiprotein complexes on vesicle membranes (Guo, 2000). In particular, small guanosine triphosphate hydrolases (GTPases) of the Arf, Arl and Rab families play central roles in cargo sorting at the Golgi. These proteins cycle between active and inactive states that are determined by whether they are bound to GTP or GDP, as modulated by the activity of GTPase-activating proteins (GAPs) and guanine-nucleotide-exchange factors (GEFs). When GTP-bound, these GTPases recruit effector proteins that regulate membrane-membrane and membrane-cytoskeleton interactions.

Targeting of proteins to different cellular locations can be mediated by recognition of targeting sequences within the cargo by specific receptors. These receptors might be the GTPases themselves or the coat protein complexes whose assembly they promote. The benefits of identifying organelle-specific targeting sequences are twofold. First, it enables prediction of localization for other proteins that have the same the targeting sequence. Second, it provides a means of identifying the molecular interactions that cause this targeting (e.g. binding of ER-resident proteins to the KDEL receptor). A variety of ciliary targeting sequences have been identified that contain diverse polypeptide and acylation motifs (Pazour and Bloodgood, 2008). However, the manner in which these sequences confer ciliary targeting remains largely obscure. An exception is the direct binding of the GTPase Arf4 by the rhodopsin VxPx motif, a ciliary targeting sequence that is common to the ciliary proteins polycystin-2 in renal tubule epithelial cells (Geng, 2006) and cyclic-nucleotide-gated channel β1 (CNG-β1) in olfactory neurons (Jenkins et al., 2006). In the Golgi, Arf4 binding to rhodopsin via this motif promotes the assembly of a complex that includes Rab11, ASAP1 (ArfGAP with SH3 domain, ankyrin repeat and PH domain 1) and FIP3 (Rab11-family-interacting protein 3) and in turn triggers vesicle budding from the trans-Golgi network (Inoue et al., 2008; Mazelova et al., 2009a). ASAP1 seems to be a key player in these events because it possesses a Bin-Amphiphysin-Rvs (BAR) domain that binds to curved membranes and promotes further membrane deformation (Nie et al., 2006). ASAP1 can actually promote tubulation of vesicles in vitro (Krauss et al., 2008), suggesting that it can facilitate vesicle budding from the trans-Golgi network. Therefore, this multiprotein complex might be involved in a mechanism for the selective sorting of ciliary proteins into vesicles and vesicular budding. Further investigation of the mechanisms by which ciliary targeting sequences confer localization will undoubtedly elucidate additional molecules involved in ciliary targeting. It should be noted that the divergence of ciliary targeting sequences probably reflects coevolution with binding partners; the nature of the sequence itself might not be as important as the physiological interaction that facilitates proper trafficking.

Another binding partner for a ciliary targeting motif has also recently been described. CNG-β1 was shown to both colocalize and coimmunoprecipitate with the membrane adaptor ankyrin-G through its C-terminal domain. This interaction is both necessary and sufficient for the targeting of human CNG-β1 to the ciliary membrane when expressed in Xenopus photoreceptors (Kizhatil et al., 2009). However, the stage of transport at which this interaction confers targeting to the ciliary membrane remains unclear.

In addition to transport mediated by the direct binding of vesicle membrane proteins, ciliary cargo might be recruited into ciliary-bound vesicles indirectly through interactions with other ciliary proteins that serve as adaptors. A promising candidate for such an adaptor is the IFT protein IFT20. In contrast to all other known IFT components, which localize to the cilium and/or basal body, IFT20 also localizes to the Golgi (Follit et al., 2006). The pools of Golgi-localized IFT20 and IFT20 localized to the basal body and/or cilium are in dynamic exchange. Total inhibition of IFT20 expression prevents ciliogenesis, whereas partial inhibition that depletes the amount of IFT20 in the Golgi pool but not the ciliary pool results in defective transport of polycystin-2 to the ciliary membrane. This suggests that IFT20 serves as an adaptor or chaperone for the targeting of polycystin-2 to the ciliary membrane.

IFT20 is anchored to the Golgi through its interaction with GMAP210 (also known as thyroid hormone receptor interacting protein 11, TRIP11) (Follit et al., 2008), a member of the golgin family of proteins. GMAP210 contains both an ArfGAP1 lipid-packing sensor (ALPS) domain that preferentially binds to curved membranes (Drin et al., 2008) and GRIP-related Arf-binding (GRAB) domains that bind to small GTPases (Short et al., 2005). Notably, although cells from GMAP210-null mice exhibit normal Golgi architecture, they have reduced levels of polycystin-2 in their ciliary membranes. Both IFT20 and GMAP210 therefore have clear roles in the trafficking of polycystin-2 to the ciliary membrane, and are ideally positioned to mediate cargo sorting within the Golgi. Given these findings, it will be interesting to know whether these proteins perform a similar function in the transport of other ciliary cargo, and whether they do so through their interaction with specific GTPases.

Following cargo sorting and vesicle budding from the Golgi, vesicles are delivered to their target membranes. This is a multistep process involving transport, tethering and fusion. The molecular mediators that govern these steps for vesicles bound for ciliary locations are only recently coming to light. It appears that a key player in targeting vesicles to their sites of fusion is the exocyst, a multiprotein complex whose components were originally discovered in yeast for their roles in secretion (Novick, 1980; TerBush, 1996). In polarized epithelial cells, the exocyst localizes to basolateral membranes, where it mediates the delivery of vesicles. An analogous function in ciliary protein targeting was suggested by the observation that exocyst components were localized to the membrane of the primary cilium in MDCK cells (Rogers et al., 2004). The exocyst is an effector of the GTPase Rab11 (Guo, 1999), whose recruitment to rhodopsin-containing vesicles at the Golgi was discussed above. During cytokinesis in yeast, the exocyst guides the tethering of Rab11- and FIP3-containing vesicles to the cleavage furrow (Fielding, 2005). An analogous role in ciliated cells might therefore promote the targeting of vesicles to the ciliary membrane. Consistent with this idea, knocking down the expression of the exocyst protein Sec10 in MDCK cells abolishes ciliogenesis (Zuo et al., 2009).

In addition to Rab11, several other GTPases play a role in ciliary protein targeting. Rab8 associates with ciliary-bound vesicles soon after they bud from the Golgi. Expression of dominant-negative Rab8 impairs rhodopsin delivery in Xenopus photoreceptors (Moritz et al., 2001), as well as the delivery of membrane to the primary cilia of immortalized human retinal pigment epithelial cells (Nachury et al., 2007). The work by Nachury and colleagues also showed that the activity of Rab8 requires the expression of a GEF (Rabin8) that interacts with a multiprotein complex called the BBSome. The exact function of the BBSome is unclear, although it is known that mutations in the proteins that it comprises cause Bardet-Biedl syndrome (BBS), a pleiotropic disease associated with multiple ciliary signaling defects (Blacque and Leroux, 2006). One of the BBSome proteins is Arl6, a GTPase that was previously found to be involved in ciliary trafficking (Fan et al., 2004). Other GTPases implicated in ciliary targeting include IFT27 in Chlamydomonas (Qin et al., 2007) and RabL5 in Trypanosoma brucei (Adhiambo et al., 2009). Finally, systematic screening of 46 human Rabs demonstrated a requirement for Rab23 and Rab17, in addition to Rab8, in ciliogenesis (Yoshimura et al., 2007). Therefore, multiple GTPases have been identified that probably contribute to ciliary membrane trafficking through the assembly of multiprotein complexes on vesicle coats.

Vesicle delivery to target membranes is guided by the cytoskeleton. Very little is known, however, about the cytoskeletal interactions that guide vesicle delivery to the ciliary membrane. In photoreceptors, Rab8 colocalizes with actin filaments near the vesicle fusion site, and this colocalization seems to be mediated by ezrin and/or moesin and Rac1 (Deretic et al., 2004). In addition, it has been shown that inhibition of the actin-based motor myosin-VIIa disrupts rhodopsin delivery (Liu, 1999). The actin network thus might be involved in transporting vesicles to the ciliary membrane. More research is needed to clarify the extent of actin involvement in guiding vesicle transport to the cilium, and how this process might intersect with microtubule-based transport.

Once vesicles are tethered to target membranous compartments, soluble NSF attachment protein receptor (SNARE) proteins on vesicle (v-SNARE) and target (t-SNARE) membranes drive membrane fusion. Although the pairing of specific v-SNAREs with cognate t-SNAREs does not confer specificity to vesicle targeting, particular SNARE pairings are associated with specific fusion events (Bethani et al., 2007; Brandhorst et al., 2006; Guo, 2000; ter Beest et al., 2005). In vertebrate photoreceptors, the membrane at the base of the cilium is enriched in the t-SNARE syntaxin 3 in a manner that depends on microtubules (Mazelova et al., 2009b). Modulation of the cell lipid composition that promotes the association of syntaxin 3 with another t-SNARE, SNAP-25, correlates with increased delivery of rhodopsin to the ciliary membrane. Therefore, syntaxin 3 and SNAP-25 are promising candidates for mediating the fusion of vesicles at the base of the ciliary membrane and, together with the GTPases, multiprotein complexes and cytoskeletal elements discussed above, are probably key players in this crucial step of ciliary targeting.

Following vesicle fusion at the periciliary membrane, lipids and membrane proteins enter the cilium. As discussed above, it appears that a diffusion barrier restricts passive diffusion into the cilium, indicating that an active means of transport into the cilium probably occurs. An obvious candidate for this function is the IFT complex. However, although electron micrographs of IFT complexes show a tightly apposed overlying membrane, none of the IFT proteins discovered to date possesses transmembrane domains, lipid modifications or lipid-binding domains. Moreover, investigation of an IFT-membrane interaction has been hindered by the detergent-based treatments that are involved in most biochemical purifications of IFT-complex proteins (Shogomori and Brown, 2003).

Despite the difficulties in detecting an interaction between IFT complexes and membrane proteins, several lines of evidence suggest that IFT plays a role in the transport of ciliary membrane proteins. Cyclic-nucleotide-gated channels in mammalian cells (Jenkins et al., 2006) and a membrane-bound aurora kinase in Chlamydomonas reinhardtii have both been shown to require IFT for their transport into the cilium (Pan and Snell, 2003), and polycystin-2 accumulates in cilia when retrograde IFT is inhibited in a variety of cell types (Huang et al., 2007; Pazour et al., 2002). Perhaps most convincingly, ciliary transient receptor potential vanilloid (TRPV) channels have been visualized translocating within Caenorhabditis elegans cilia at speeds that are characteristic of IFT (Qin et al., 2005).

Evidence of a biochemical association between IFT complexes and ciliary membrane proteins has also emerged. For example, IFT complexes co-purify with DnaJ chaperones and the membrane-associated proteins rhodopsin and guanylyl cyclase 1 in photoreceptors (Bhowmick et al., 2009). In addition, an indirect association between Rab8 and IFT20 through Elipsa and rabaptin 5 has recently been reported, and the inhibition of Elipsa expression causes defective ciliogenesis in zebrafish (Omori et al., 2008). In C. elegans, the Elipsa homolog DYF-11 translocates within sensory cilia via IFT (Kunitomo and Iino, 2008), supporting the link between IFT and this putative binding partner of Rab8. Therefore, both functional and biochemical evidence implicating IFT in the transport of membrane proteins within the cilium is accumulating.

If ciliary membrane proteins indeed traffic into cilia with IFT complexes, an equally important consideration becomes how they disassociate from these complexes, lest they recycle back out of the organelle. The logical place to look for molecular regulators of IFT disassociation is at the tip of the cilium because this is where axonemal subunits are incorporated. The plus-end microtubule-binding protein EB1 localizes to the tip of the cilium (Pedersen, 2003), where it interacts with the retrograde complex protein IFT172 (Pedersen et al., 2005). The ciliary tip is probably also the site where anterograde and retrograde IFT transport are coordinated. This coordination might involve the BBSome because inhibiting the expression of BBS proteins, which transit with IFT complexes in C. elegans, leads to uncoupling of the anterograde and retrograde movement (Ou et al., 2005).

Early studies of ciliary membrane lipid composition indicated that the lipids contain a high content of sterols (Chailley and Boisvieux-Ulrich, 1985; Kaneshiro, 1990; Souto-Padron and de Souza, 1983; Tetley, 1986), glycolipids (Bloodgood et al., 1995) and sphingolipids (Kaneshiro et al., 1984; Kaya et al., 1984), which now are known to be components of canonical lipid-raft microdomains. This suggested to us (Tyler et al., 2009) that cilia might be enriched in lipid rafts and that this special lipid composition might serve to recruit or retain ciliary membrane proteins, such as the dually acylated flagellar calcium-binding protein (FCaBP) in Trypanosoma cruzi (Godsel and Engman, 1999) and its homolog in Trypanosoma brucei, calflagin (Emmer et al., 2009). These proteins require both myristoylation and palmitoylation for lipid-raft association and ciliary-membrane targeting. Microscopic and biochemical analysis of the trypanosome flagella showed that they are highly enriched in sterols, sphingolipids and some dually acylated proteins, and that they possess high liquid order as determined by laurdan two-photon microscopy (Tyler et al., 2009) [which involves the intercalation of laurdan into membranes and its fluorescence according to the molecular freedom of the water molecules surrounding it (Gaus et al., 2003)]. Interestingly, mutation of the palmitoylation site of calflagin, or inhibition of the acyltransferase that palmitoylates calflagin (both of which lead to the production of a protein that has only a myristoyl group), leads to protein mislocalization to the cell-body membrane (Emmer et al., 2009). Thus, although myristoylation is sufficient for cell-body-membrane targeting, palmitoylation serves as a secondary signal for calflagin lipid-raft association and flagellar targeting. Disruption of lipid rafts also leads to protein mislocalization (Tyler et al., 2009).

An association between lipid rafts and many other cilia-targeted proteins has also been described in other systems, including vertebrate photoreceptors (Senin et al., 2004), Chlamydomonas reinhardtii (Iomini et al., 2006), mammalian spermatozoa (Travis et al., 2001) and Leishmania major (Tull et al., 2004). Similarly, certain proteins that are thought to be involved in targeting proteins to ciliary membranes have been shown to require protein-lipid interactions for their localization in non-ciliated cells. For example, the SNARE protein syntaxin 3, whose involvement in rhodopsin trafficking was discussed above (Mazelova et al., 2009b), requires cholesterol for its clustering in lipid rafts at the plasma membrane of epithelial cells (Low, 2006). In a similar way, the localization of the exocyst in MDCK cells is mediated by a direct interaction with phosphatidylinositol (4,5)-bisphosphate (Liu et al., 2007). Whether these same protein-lipid associations are involved in localizing syntaxin 3 and the exocyst to ciliary membranes remains to be determined.

Collectively, these findings illustrate the distinct lipid composition of the eukaryotic ciliary membrane relative to the plasma membrane and provide a potential mechanism for downstream protein recruitment. However, the mechanisms by which the cell establishes and maintains the discrete lipid composition of the ciliary membrane are unknown. It should also be noted that, although it is tempting to speculate that lipid-raft enrichment occurs upstream of protein sorting, it is equally plausible that ciliary proteins drive lipid-raft coalescence and their enrichment in ciliary membranes. These two models are not mutually exclusive, however, and perhaps the most likely scenario is that protein and lipid sorting have reciprocal effects on each other during the trafficking of these molecules to and within cilia. A major advance in this area was the finding that a transmembrane lipid-raft protein is sorted in the Golgi complex specifically into vesicles that are enriched in sphingolipids and sterols and that exhibit high liquid order by laurdan microscopy (Klemm et al., 2009). This work indicates that lipid sorting and formation of vesicles with distinct lipid composition can occur in the trans-Golgi network. It is therefore reasonable to hypothesize that lipid-raft vesicles containing specific cargo can form here as well, a subset of which might be destined for the ciliary membrane. This notion is consistent with a targeted delivery model for the delivery of ciliary components.

In this Commentary, we have attempted to trace the synthesis, assembly, packaging and trafficking of ciliary proteins and lipids from their site of initial assembly to their final destination in ciliary membranes. The key steps in this process are illustrated in Fig. 1. One interesting aspect, illustrated in this figure but not discussed above, is the possibility that the cilium is a secretory organelle in addition to being a motility and sensory organelle. This is a notion that we have had for some time — since the discovery nearly two decades ago that T. cruzi FCaBP can be found in vesicles shed from the cell (Ouaissi et al., 1992). This possibility is eloquently described in a recent review by Baldari and Rosenbaum (Baldari and Rosenbaum, 2009), which integrates information from various fields and posits that vesicle secretion could be a natural and important extension of normal ciliary processes. Clearly, it is important to determine the mechanisms regulating this exocytosis and the functions of shed vesicles in intercellular communication and host-pathogen interactions. It will also be interesting to know how the unique lipid composition of the ciliary membrane is established and maintained, and how vesicles of different lipid composition and containing specific cargo are formed in the Golgi. Where are the enzymes involved in the synthesis and assembly of these unique lipid vesicles located, and how are they regulated? In addition, the molecular mechanisms underlying the differential trafficking of specific vesicle populations from the Golgi to their intracellular destinations are still largely unknown. As the known functions of cilia in basic cell physiology and regulation, as well as in health and disease, increase each month, it will be fascinating to follow this field as it evolves in the coming years.

We thank the reviewers of this manuscript and Mark Field (University of Cambridge) and Kent Hill (UCLA) for their very helpful suggestions. This work was supported in part by NIH R01 AI46781 (D.M.E.) and Predoctoral Fellowships from the NIH (B.T.E.) and American Heart Association (B.T.E. and D.M.). Deposited in PMC for release after 12 months.

This article is part of a Minifocus on cilia and flagella. For further reading, please see related articles: ‘The primary cilium at a glance’ by Peter Satir et al. (J. Cell Sci.123, 499-503), ‘Sensory reception is an attribute of both primary cilia and motile cilia’ by Robert A. Bloodgood (J. Cell Sci.123, 505-509), ‘The perennial organelle: assembly and disassembly of the primary cilium’ by E. Scott Seeley and Maxence V. Nachury (J. Cell Sci.123, 511-518) and ‘Flagellar and ciliary beating: the proven and the possible’ by Charles B. Lindemann and Kathleen A. Lesich (J. Cell Sci.123, 519-528).

Adhiambo
C.
,
Blisnick
T.
,
Toutirais
G.
,
Delannoy
E.
,
Bastin
P.
(
2009
).
A novel function for the atypical small G protein Rab-like 5 in the assembly of the trypanosome flagellum
.
J. Cell Sci.
122
,
834
-
841
.
Baldari
C. T.
,
Rosenbaum
J.
(
2009
).
Intraflagellar transport: it's not just for cilia anymore
.
Curr. Opin. Cell Biol.
[Epub ahead of print] doi:10.1016/j.ceb.2009.10.010
.
Bethani
I.
,
Lang
T.
,
Geumann
U.
,
Sieber
J. J.
,
Jahn
R.
,
Rizzoli
S. O.
(
2007
).
The specificity of SNARE pairing in biological membranes is mediated by both proof-reading and spatial segregation
.
EMBO J.
26
,
3981
-
3992
.
Bhowmick
R.
,
Li
M.
,
Sun
J.
,
Baker
S. A.
,
Insinna
C.
,
Besharse
J. C.
(
2009
).
Photoreceptor IFT complexes containing chaperones, guanylyl cyclase 1 and rhodopsin
.
Traffic
10
,
648
-
663
.
Blacque
O. E.
,
Leroux
M. R.
(
2006
).
Bardet-Biedl syndrome: an emerging pathomechanism of intracellular transport
.
Cell. Mol. Life Sci.
63
,
2145
-
2161
.
Bloodgood
R. A.
,
Woodward
M. P.
,
Young
W. W.
(
1995
).
Unusual distribution of a glycolipid antigen in the flagella of Chamydomonas
.
Protoplasma
185
,
123
-
130
.
Bouck
G. B.
(
1971
).
The structure, origin, isolation, and composition of the tubular mastigonemes of the Ochromas flagellum
.
J. Cell Biol.
50
,
362
-
384
.
Brandhorst
D.
,
Zwilling
D.
,
Rizzoli
S. O.
,
Lippert
U.
,
Lang
T.
,
Jahn
R.
(
2006
).
Homotypic fusion of early endosomes: SNAREs do not determine fusion specificity
.
Proc. Natl. Acad. Sci. USA
103
,
2701
-
2706
.
Chailley
B.
,
Boisvieux-Ulrich
E.
(
1985
).
Detection of plasma membrane cholesterol by filipin during microvillogenesis and ciliogenesis in quail oviduct
.
J. Histochem. Cytochem.
33
,
1
-
10
.
Deane
J. A.
,
Cole
D. G.
,
Seeley
E. S.
,
Diener
D. R.
,
Rosenbaum
J. L.
(
2001
).
Localization of intraflagellar transport protein IFT52 identifies basal body transitional fibers as the docking site for IFT particles
.
Curr. Biol.
11
,
1586
-
1590
.
Deretic
D.
,
Traverso
V.
,
Parkins
N.
,
Jackson
F.
,
Rodriguez de Turco
E. B.
,
Ransom
N.
(
2004
).
Phosphoinositides, ezrin/moesin, and rac1 regulate fusion of rhodopsin transport carriers in retinal photoreceptors
.
Mol. Biol. Cell
15
,
359
-
370
.
Drin
G.
,
Morello
V.
,
Casella
J. F.
,
Gounon
P.
,
Antonny
B.
(
2008
).
Asymmetric tethering of flat and curved lipid membranes by a golgin
.
Science
320
,
670
-
673
.
Ejsing
C. S.
,
Sampaio
J. L.
,
Surendranath
V.
,
Duchoslav
E.
,
Ekroos
K.
,
Klemm
R. W.
,
Simons
K.
,
Shevchenko
A.
(
2009
).
Global analysis of the yeast lipidome by quantitative shotgun mass spectrometry
.
Proc. Natl. Acad. Sci. USA
106
,
2136
-
2141
.
Emmer
B. T.
,
Souther
C.
,
Toriello
K. M.
,
Olson
C. L.
,
Epting
C. L.
,
Engman
D. M.
(
2009
).
Identification of a palmitoyl acyltransferase required for protein sorting to the flagellar membrane
.
J. Cell Sci.
122
,
867
-
874
.
European Polycystic Kidney Disease Consortium
(
1994
).
The polycystic kidney disease 1 gene encodes a 14 kb transcript and lies within a duplicated region on chromosome 16
.
Cell
77
,
881
-
894
.
Fan
S.
,
Fogg
V.
,
Wang
Q.
,
Chen
X. W.
,
Liu
C. J.
,
Margolis
B.
(
2007
).
A novel Crumbs3 isoform regulates cell division and ciliogenesis via importin beta interactions
.
J. Cell Biol.
178
,
387
-
398
.
Fan
Y.
,
Esmail
M. A.
,
Ansley
S. J.
,
Blacque
O. E.
,
Boroevich
K.
,
Ross
A. J.
,
Moore
S. J.
,
Badano
J. L.
,
May-Simera
H.
,
Compton
D. S.
, et al. 
. (
2004
).
Mutations in a member of the Ras superfamily of small GTP-binding proteins causes Bardet-Biedl syndrome
.
Nat. Genet.
36
,
989
-
993
.
Fielding
A. B.
,
Schonteich
E.
,
Matheson
J.
,
Wilson
G.
,
Yu
X.
,
Hickson
G. R.
,
Srivaslava
S.
,
Baldwin
S. A.
,
Prekeris
R.
,
Gould
G. W.
(
2005
).
Rab11-FIP3 and FIP4 interact with Arf6 and the exocyst to control membrane traffic in cytokinesis
.
EMBO J.
24
,
3389
-
3399
.
Follit
J. A.
,
Tuft
R. A.
,
Fogarty
K. E.
,
Pazour
G. J.
(
2006
).
The intraflagellar transport protein IFT20 is associated with the Golgi complex and is required for cilia assembly
.
Mol. Biol. Cell
17
,
3781
-
3792
.
Follit
J. A.
,
San Agustin
J. T.
,
Xu
F.
,
Jonassen
J. A.
,
Samtani
R.
,
Lo
C. W.
,
Pazour
G. J.
(
2008
).
The golgin GMAP210/TRIP11 anchors IFT20 to the Golgi complex
.
PLoS Genet.
4
,
e1000315
.
Follit
J. A.
,
Xu
F.
,
Keady
B. T.
,
Pazour
G. J.
(
2009
).
Characterization of mouse IFT complex B
.
Cell Motil. Cytoskeleton
66
,
457
-
468
.
Gaus
K.
,
Gratton
E.
,
Kable
E. P.
,
Jones
A. S.
,
Gelissen
I.
,
Kritharides
L.
,
Jessup
W.
(
2003
).
Visualizing lipid structure and raft domains in living cells with two-photon microscopy
.
Proc. Natl. Acad. Sci. USA
100
,
15554
-
15559
.
Geng
L.
,
Okuhara
D.
,
Yu
Z.
,
Tian
X.
,
Cai
Y.
,
Shibazaki
S.
,
Somlo
S.
(
2006
).
Polycystin-2 traffics to cillia independently of polycystin-1 by using an N-terminal RVxP motif
.
J. Cell Sci.
119
,
1383
-
1395
.
Gilula
N. B.
,
Satir
P.
(
1972
).
The ciliary necklace. A ciliary membrane specialization
..
J. Cell Biol.
53
,
494
-
509
.
Godsel
L. M.
,
Engman
D. M.
(
1999
).
Flagellar protein localization mediated by a calcium-myristoyl/palmitoyl switch mechanism
.
EMBO J.
18
,
2057
-
2065
.
Guo
W.
,
Roth
D.
,
Walch-Solimena
C.
,
Novick
P.
(
1999
).
The exocyst is an effector for Sec4p, targeting secretory vesicles to sites of exocytosis
.
EMBO J.
18
,
1071
-
1080
.
Guo
W.
,
Sacher
M.
,
Barrowman
J.
,
Ferro-Novick
S.
,
Novick
P.
(
2000
).
Protein complexes in transport vesicle targeting
..
Trends Cell Biol.
10
,
251
-
255
.
Hao
L.
,
Scholey
J. M.
(
2009
).
Intraflagellar transport at a glance
.
J. Cell Sci.
122
,
889
-
892
.
Huang
K.
,
Diener
D. R.
,
Mitchell
A.
,
Pazour
G. J.
,
Witman
G. B.
,
Rosenbaum
J. L.
(
2007
).
Function and dynamics of PKD2 in Chlamydomonas reinhardtii flagella
.
J. Cell Biol.
179
,
501
-
514
.
Inoue
H.
,
Ha
V. L.
,
Prekeris
R.
,
Randazzo
P. A.
(
2008
).
Arf GTPase-activating protein ASAP1 interacts with Rab11 effector FIP3 and regulates pericentrosomal localization of transferring receptor-positive recycling endosome
.
Mol. Biol. Cell
19
,
4224
-
4237
.
Iomini
C.
,
Li
L.
,
Mo
W.
,
Dutcher
S. K.
,
Piperno
G.
(
2006
).
Two flagellar genes, AGG2 and AGG3, mediate orientation to light in Chlamydomonas
.
Curr. Biol.
16
,
1147
-
1153
.
Jenkins
P. M.
,
Hurd
T. W.
,
Zhang
L.
,
McEwen
D. P.
,
Brown
R. L.
,
Margolis
B.
,
Verhey
K. J.
,
Martens
J. R.
(
2006
).
Ciliary targeting of olfactory CNG channels requires the CNGB1b subunit and the kinesin-2 motor protein, KIF17
.
Curr. Biol.
16
,
1211
-
1216
.
Jin
H.
,
Nachury
M. V.
(
2009
).
The BBSome
.
Curr. Biol.
19
,
R472
-
R473
.
Johnson
K. A.
,
Rosenbaum
J. L.
(
1992
).
Polarity of flagellar assembly in Chlamydomonas
.
J. Cell Biol.
119
,
1605
-
1611
.
Kaneshiro
E. S.
(
1990
).
Lipids of ciliary and flagellar membranes
. In
Ciliary and Flagellar Membranes
(ed.
Bloodgood
R. A.
), pp.
241
-
265
.
New York
:
Plenum Press
.
Kaneshiro
E. S.
,
Matesic
D. F.
,
Jayasimhulu
K.
(
1984
).
Characterizations of six ethanolamine sphingophospholipids from Paramecium cells and cilia
.
J. Lipid Res.
25
,
369
-
377
.
Kaupp
U. B.
,
Solzin
J.
,
Hildebrand
E.
,
Brown
J. E.
,
Helbig
A.
,
Hagen
V.
,
Beyermann
M.
,
Pampaloni
F.
,
Weyand
I.
(
2003
).
The signal flow and motor response controlling chemotaxis of sea urchin sperm
.
Nat. Cell Biol.
5
,
109
-
117
.
Kaya
K.
,
Ramesha
C. S.
,
Thompson
G. A.
Jr
(
1984
).
On the formation of alpha-hydroxy fatty acids. Evidence for a direct hydroxylation of nonhydroxy fatty acid-containing sphingolipids
.
J. Biol. Chem.
259
,
3548
-
3553
.
Kizhatil
K.
,
Baker
S. A.
,
Arshavsky
V. Y.
,
Bennett
V.
(
2009
).
Ankyrin-G promotes cyclic nucleotide-gated channel transport to rod photoreceptor sensory cilia
.
Science
323
,
1614
-
1617
.
Klemm
R. W.
,
Ejsing
C. S.
,
Surma
M. A.
,
Kaiser
H. J.
,
Gerl
M. J.
,
Sampaio
J. L.
,
de Robillard
Q.
,
Ferguson
C.
,
Proszynski
T. J.
,
Shevchenko
A.
, et al. 
. (
2009
).
Segregation of sphingolipids and sterols during formation of secretory vesicles at the trans-Golgi network
.
J. Cell Biol.
185
,
601
-
612
.
Kozminski
K. G.
,
Johnson
K. A.
,
Forscher
P.
,
Rosenbaum
J. L.
(
1993
).
A motility in the eukaryotic flagellum unrelated to flagellar beating
.
Proc. Natl. Acad. Sci. USA
90
,
5519
-
5523
.
Kozminski
K. G.
,
Beech
P. L.
,
Rosenbaum
J. L.
(
1995
).
The Chlamydomonas kinesin-like protein FLA10 is involved in motility associated with the flagellar membrane
.
J. Cell Biol.
131
,
1517
-
1527
.
Krauss
M.
,
Jia
J. Y.
,
Roux
A.
,
Beck
R.
,
Wieland
F. T.
,
De Camilli
P.
,
Haucke
V.
(
2008
).
Arf1-GTP-induced tubule formation suggests a function of Arf family proteins in curvature acquisition at sites of vesicle budding
.
J. Biol. Chem.
283
,
27717
-
27723
.
Kunitomo
H.
,
Iino
Y.
(
2008
).
Caenorhabditis elegans DYF-11, an orthologue of mammalian Traf3ip1/MIP-T3, is required for sensory cilia formation
.
Genes Cells
13
,
13
-
25
.
Liu
J.
,
Zuo
X.
,
Yue
P.
,
Guo
W.
(
2007
).
Phosphatidylinositol 4,5-bisphosphate mediates the targeting of the exocyst to the plasma membrane for exocytosis in mammalian cells
.
Mol. Biol. Cell
18
,
4483
-
4492
.
Liu
X.
,
Udovichenko
I. P.
,
Brown
S. D.
,
Steel
K. P.
,
Williams
D. S.
(
1999
).
Myosin VIIa participates in opsin transport through the photoreceptor cilium
.
J. Neurosci.
19
,
6267
-
6274
.
Low
S. H.
,
Vasanji
A.
,
Nanduri
J.
,
He
M.
,
Sharma
N.
,
Koo
M.
,
Drazba
J.
,
Weimbs
T.
(
2006
).
Syntaxins 3 and 4 are concentrated in separate clusters on the plasma membrane before the establishment of cell polarity
.
Mol. Biol. Cell
17
,
977
-
989
.
Mazelova
J.
,
Astuto-Gribble
L.
,
Inoue
H.
,
Tam
B. M.
,
Schonteich
E.
,
Prekeris
R.
,
Moritz
O. L.
,
Randazzo
P. A.
,
Deretic
D.
(
2009a
).
Ciliary targeting motif VxPx directs assembly of a trafficking module through Arf4
.
EMBO J.
28
,
183
-
192
.
Mazelova
J.
,
Ransom
N.
,
Astuto-Gribble
L.
,
Wilson
M. C.
,
Deretic
D.
(
2009b
).
Syntaxin 3 and SNAP-25 pairing, regulated by omega-3 docosahexaenoic acid, controls the delivery of rhodopsin for the biogenesis of cilia-derived sensory organelles, the rod outer segments
.
J. Cell Sci.
122
,
2003
-
2013
.
Mitchell
D. R.
(
2007
).
The evolution of eukaryotic cilia and flagella as motile and sensory organelles
.
Adv. Exp. Med. Biol.
607
,
130
-
140
.
Mochizuki
T.
,
Wu
G.
,
Hayashi
T.
,
Xenophontos
S. L.
,
Veldhuisen
B.
,
Saris
J. J.
,
Reynolds
D. M.
,
Cai
Y.
,
Gabow
P. A.
,
Pierides
A.
, et al. 
. (
1996
).
PKD2, a gene for polycystic kidney disease that encodes an integral membrane protein
.
Science
272
,
1339
-
1342
.
Moritz
O. L.
,
Tam
B. M.
,
Hurd
L. L.
,
Peranen
J.
,
Deretic
D.
,
Papermaster
D. S.
(
2001
).
Mutant rab8 impairs docking and fusion of rhodopsin-bearing post-Golgi membranes and causes cell death of transgenic Xenopus rods
.
Mol. Biol. Cell
12
,
2341
-
2351
.
Nachury
M. V.
,
Loktev
A. V.
,
Zhang
Q.
,
Westlake
C. J.
,
Peranen
J.
,
Merdes
A.
,
Slusarski
D. C.
,
Scheller
R. H.
,
Bazan
J. F.
,
Sheffield
V. C.
, et al. 
. (
2007
).
A core complex of BBS proteins cooperates with the GTPase Rab8 to promote ciliary membrane biogenesis
.
Cell
129
,
1201
-
1213
.
Nie
Z.
,
Hirsch
D. S.
,
Luo
R.
,
Jian
X.
,
Stauffer
S.
,
Cremesti
A.
,
Andrade
J.
,
Lebowitz
J.
,
Marino
M.
,
Ahvazi
B.
, et al. 
. (
2006
).
A BAR domain in the N terminus of the Arf GAP ASAP1 affects membrane structure and trafficking of epidermal growth factor receptor
.
Curr. Biol.
16
,
130
-
139
.
Novick
P.
,
Field
C.
,
Schekman
R.
(
1980
).
Identification of 23 complementation groups required for post-translational events in the yeast secretory pathway
.
Cell
21
,
205
-
215
.
Omori
Y.
,
Zhao
C.
,
Saras
A.
,
Mukhopadhyay
S.
,
Kim
W.
,
Furukawa
T.
,
Sengupta
P.
,
Veraksa
A.
,
Malicki
J.
(
2008
).
Elipsa is an early determinant of ciliogenesis that links the IFT particle to membrane-associated small GTPase Rab8
.
Nat. Cell Biol.
10
,
437
-
444
.
Ou
G.
,
Blacque
O. E.
,
Snow
J. J.
,
Leroux
M. R.
,
Scholey
J. M.
(
2005
).
Functional coordination of intraflagellar transport motors
.
Nature
436
,
583
-
587
.
Ouaissi
A.
,
Aguirre
T.
,
Plumas-Marty
B.
,
Piras
M.
,
Schoneck
R.
,
Gras-Masse
H.
,
Taibi
A.
,
Loyens
M.
,
Tartar
A.
,
Capron
A.
, et al. 
. (
1992
).
Cloning and sequencing of a 24-kDa Trypanosoma cruzi specific antigen released in association with membrane vesicles and defined by a monoclonal antibody
.
Biol. Cell
75
,
11
-
17
.
Pan
J.
,
Snell
W. J.
(
2000
).
Signal transduction during fertilization in the unicellular green alga, Chlamydomonas
.
Curr. Opin. Microbiol.
3
,
596
-
602
.
Pan
J.
,
Snell
W. J.
(
2003
).
Kinesin II and regulated intraflagellar transport of Chlamydomonas aurora protein kinase
.
J. Cell Sci.
116
,
2179
-
2186
.
Papermaster
D. S.
,
Schneider
B. G.
,
Besharse
J. C.
(
1985
).
Vesicular transport of newly synthesized opsin from the Golgi apparatus toward the rod outer segment. Ultrastructural immunocytochemical and autoradiographic evidence in Xenopus retinas
.
Invest. Ophthalmol. Vis. Sci.
26
,
1386
-
1404
.
Pazour
G. J.
,
Bloodgood
R. A.
(
2008
).
Targeting proteins to the ciliary membrane
.
Curr. Top. Dev. Biol.
85
,
111
-
145
.
Pazour
G. J.
,
Dickert
B. L.
,
Witman
G. B.
(
1999
).
The DHC1b (DHC2) isoform of cytoplasmic dynein is required for flagellar assembly
.
J. Cell Biol.
144
,
473
-
481
.
Pazour
G. J.
,
Dickert
B. L.
,
Vucica
Y.
,
Seeley
E. S.
,
Rosenbaum
J. L.
,
Witman
G. B.
,
Cole
D. G.
(
2000
).
Chlamydomonas IFT88 and its mouse homologue, polycystic kidney disease gene Tg737, are required for assembly of cilia and flagella
.
J. Cell Biol.
151
,
709
-
718
.
Pazour
G. J.
,
San Agustin
J. T.
,
Follit
J. A.
,
Rosenbaum
J. L.
,
Witman
G. B.
(
2002
).
Polycystin-2 localizes to kidney cilia and the ciliary level is elevated in orpk mice with polycystic kidney disease
.
Curr. Biol.
12
,
R378
-
R380
.
Pedersen
L. B.
,
Geimer
S.
,
Sloboda
R. D.
,
Rosenbaum
J. L.
(
2003
).
The microtubule plus end-tracking protein EB1 is localized to the flagellar tip and basal bodies in Chlamydomonas reinhardtii
.
Curr. Biol.
13
,
1969
-
1974
.
Pedersen
L. B.
,
Miller
M. S.
,
Geimer
S.
,
Leitch
J. M.
,
Rosenbaum
J. L.
,
Cole
D. G.
(
2005
).
Chlamydomonas IFT172 is encoded by FLA11, interacts with CrEB1, and regulates IFT at the flagellar tip
.
Curr. Biol.
15
,
262
-
266
.
Porter
M. E.
,
Bower
R.
,
Knott
J. A.
,
Byrd
P.
,
Dentler
W.
(
1999
).
Cytoplasmic dynein heavy chain 1b is required for flagellar assembly in Chlamydomonas
.
Mol. Biol. Cell
10
,
693
-
712
.
Qin
H.
,
Diener
D. R.
,
Geimer
S.
,
Cole
D. G.
,
Rosenbaum
J. L.
(
2004
).
Intraflagellar transport (IFT) cargo: IFT transports flagellar precursors to the tip and turnover products to the cell body
.
J. Cell Biol.
164
,
255
-
266
.
Qin
H.
,
Burnette
D. T.
,
Bae
Y. K.
,
Forscher
P.
,
Barr
M. M.
,
Rosenbaum
J. L.
(
2005
).
Intraflagellar transport is required for the vectorial movement of TRPV channels in the ciliary membrane
.
Curr. Biol.
15
,
1695
-
1699
.
Qin
H.
,
Wang
Z.
,
Diener
D.
,
Rosenbaum
J.
(
2007
).
Intraflagellar transport protein 27 is a small G protein involved in cell-cycle control
.
Curr. Biol.
17
,
193
-
202
.
Reiter
J. F.
,
Mostov
K.
(
2006
).
Vesicle transport, cilium formation, and membrane specialization: the origins of a sensory organelle
.
Proc. Natl. Acad. Sci. USA
103
,
18383
-
18384
.
Rogers
K. K.
,
Wilson
P. D.
,
Snyder
R. W.
,
Zhang
X.
,
Guo
W.
,
Burrow
C. R.
,
Lipschutz
J. H.
(
2004
).
The exocyst localizes to the primary cilium in MDCK cells
.
Biochem. Biophys. Res. Commun.
319
,
138
-
143
.
Schuck
S.
,
Simons
K.
(
2004
).
Polarized sorting in epithelial cells: raft clustering and the biogenesis of the apical membrane
.
J. Cell Sci.
117
,
5955
-
5964
.
Senin
I. I.
,
Hoppner-Heitmann
D.
,
Polkovnikova
O. O.
,
Churumova
V. A.
,
Tikhomirova
N. K.
,
Philippov
P. P.
,
Koch
K. W.
(
2004
).
Recoverin and rhodopsin kinase activity in detergent-resistant membrane rafts from rod outer segments
.
J. Biol. Chem.
279
,
48647
-
48653
.
Shah
A. S.
,
Ben-Shahar
Y.
,
Moninger
T. O.
,
Kline
J. N.
,
Weish
M. J.
(
2009
).
Motile cilia of human airway epithelia are chemosensory
.
Science
325
,
1131
-
1134
.
Shogomori
H.
,
Brown
D. A.
(
2003
).
Use of detergents to study membrane rafts: the good, the bad, and the ugly
.
Biol. Chem.
384
,
1259
-
1263
.
Short
B.
,
Haas
A.
,
Barr
F. A.
(
2005
).
Golgins and GTPases, giving identity and structure to the Golgi apparatus
.
Biochim. Biophys. Acta
1744
,
383
-
395
.
Signor
D.
,
Wedaman
K. P.
,
Orozco
J. T.
,
Dwyer
N. D.
,
Bargmann
C. I.
,
Rose
L. S.
,
Scholey
J. M.
(
1999
).
Role of a class DHC1b dynein in retrograde transport of IFT motors and IFT raft particles along cilia, but not dendrites, in chemosensory neurons of living Caenorhabditis elegans
.
J. Cell Biol.
147
,
519
-
530
.
Sloboda
R. D.
,
Rosenbaum
J. L.
(
2007
).
Making sense of cilia and flagella
.
J. Cell Biol.
179
,
575
-
582
.
Song
L.
,
Dentler
W. L.
(
2001
).
Flagellar protein dynamics in Chlamydomonas
.
J. Biol. Chem.
276
,
29754
-
29763
.
Souto-Padron
T.
,
de Souza
W.
(
1983
).
Freeze-fracture localization of filipin-cholesterol complexes in the plasma membrane of Trypanosoma cruzi
.
J. Parasitol.
69
,
129
-
137
.
Stephens
R. E.
(
2000
).
Preferential incorporation of tubulin into the junctional region of ciliary outer doublet microtubules: a model for treadmilling by lattice dislocation
.
Cell Motil. Cytoskeleton
47
,
130
-
140
.
ter Beest
M. B.
,
Chapin
S. J.
,
Avrahami
D.
,
Mostov
K. E.
(
2005
).
The role of syntaxins in the specificity of vesicle targeting in polarized epithelial cells
.
Mol. Biol. Cell
16
,
5784
-
5792
.
TerBush
D. R.
,
Maurice
T.
,
Roth
D.
,
Novick
P.
(
1996
).
The Exocyst is a multiprotein complex required for exocytosis in Saccharomyces cerevisiae
.
EMBO J.
15
,
6483
-
6494
.
Tetley
L.
(
1986
).
Freeze-fracture studies on the surface membranes of pleomorphic bloodstream and in vitro transformed procyclic Trypanosoma brucei
.
Acta Trop.
43
,
307
-
317
.
Tobin
J. L.
,
Beales
P. L.
(
2009
).
The nonmotile ciliopathies
.
Genet. Med.
11
,
386
-
402
.
Torres
V. E.
,
Harris
P. C.
(
2006
).
Mechanisms of disease: autosomal dominant and recessive polycystic kidney diseases
.
Nat. Clin. Pract. Nephrol.
2
,
40
-
55
;
quiz 55
.
Travis
A. J.
,
Merdiushev
T.
,
Vargas
L. A.
,
Jones
B. H.
,
Purdon
M. A.
,
Nipper
R. W.
,
Galatioto
J.
,
Moss
S. B.
,
Hunnicutt
G. R.
,
Kopf
G. S.
(
2001
).
Expression and localization of caveolin-1, and the presence of membrane rafts, in mouse and Guinea pig spermatozoa
.
Dev. Biol.
240
,
599
-
610
.
Tull
D.
,
Vince
J. E.
,
Callaghan
J. M.
,
Naderer
T.
,
Spurck
T.
,
McFadden
G. I.
,
Currie
G.
,
Ferguson
K.
,
Bacic
A.
,
McConville
M. J.
(
2004
).
SMP-1, a member of a new family of small myristoylated proteins in kinetoplastid parasites, is targeted to the flagellum membrane in Leishmania
.
Mol. Biol. Cell
15
,
4775
-
4786
.
Tyler
K. M.
,
Fridberg
A.
,
Toriello
K. M.
,
Olson
C. L.
,
Cieslak
J. A.
,
Hazlett
T. L.
,
Engman
D. M.
(
2009
).
Flagellar membrane localization via association with lipid rafts
.
J. Cell Sci.
122
,
859
-
866
.
Yoshimura
S.
,
Egerer
J.
,
Fuchs
E.
,
Haas
A. K.
,
Barr
F. A.
(
2007
).
Functional dissection of Rab GTPases involved in primary cilium formation
.
J. Cell Biol.
178
,
363
-
369
.
Zimmermann
K. W.
(
1898
).
Beiträge zur Kenntnis einiger Drüsen und Epithelien
.
Arch. Mikrosk. Entwickl. Mech.
52
,
552
-
706
.
Zuo
X.
,
Guo
W.
,
Lipschutz
J. H.
(
2009
).
The exocyst protein Sec10 is necessary for primary ciliogenesis and cystogenesis in vitro
.
Mol. Biol. Cell
20
,
2522
-
2529
.