Abstract
The primary cilium has been found to be associated with a number of cellular signaling pathways, such as vertebrate hedgehog signaling, and implicated in the pathogenesis of diseases affecting multiple organs, including the neural tube, kidney, and brain. The primary cilium is the site where a subset of the cell's membrane proteins is enriched. However, pathways that target and concentrate membrane proteins in cilia are not well understood. Processes determining the level of proteins in the ciliary membrane include entry into the compartment, removal, and retention by diffusion barriers such as the transition zone. Proteins that are concentrated in the ciliary membrane are also localized to other cellular sites. Thus it is critical to determine the particular role for ciliary compartmentalization in sensory reception and signaling pathways. Here we provide a brief overview of our current understanding of compartmentalization of proteins in the ciliary membrane and the dynamics of trafficking into and out of the cilium. We also discuss major unanswered questions regarding the role that defects in ciliary compartmentalization might play in disease pathogenesis. Understanding the trafficking mechanisms that underlie the role of ciliary compartmentalization in signaling might provide unique approaches for intervention in progressive ciliopathies.
INTRODUCTION: THE PRIMARY CILIUM AS A COMPARTMENTALIZED ORGANELLE
The primary cilium is a tiny, antenna-like projection from the apical membrane of most vertebrate cells (Rosenbaum and Witman, 2002). Most cilia are a few micrometers in length and are ∼200 nm in diameter. Long believed to be vestigial, the primary cilium has now been implicated in multiple cellular pathways, including vertebrate hedgehog signaling (Goetz and Anderson, 2010). Defects in primary cilia result in diseases (ciliopathies) affecting multiple tissues, including the neural tube, brain, and kidney (Hildebrandt et al., 2011).
The membrane of the primary cilium envelops the microtubular axoneme that templates from the basal body and is continuous with the rest of the plasma membrane. However, the ciliary membrane is believed to be partitioned from the rest of the plasma membrane by the transition zone (Reiter et al., 2012; Figure 1). At least 25 rhodopsin-family G protein–coupled receptors (GPCRs) have been reported to localize to cilia, particularly in neurons in the brain and in other cell types (Hilgendorf et al., 2016). Proteins linked to polycystic kidney disease, such as the TRP-channel family proteins polycystin-1 and 2 (PC1/2; Pazour et al., 2002; Yoder et al., 2002), and the single-pass transmembrane protein fibrocystin (Ward et al., 2003), also localize to cilia. In addition, sonic hedgehog (Shh) pathway components such as the Shh receptor Patched (Ptch1), the pathway activator Smoothened (Smo), and the orphan GPCR, Gpr161, a negative regulator of the pathway localize to ciliary membrane in a dynamic manner (Corbit et al., 2005; Rohatgi et al., 2007; Mukhopadhyay et al., 2013). Other cilia and ciliary pocket-coordinated signaling pathways involve transforming growth factor β, receptor tyrosine kinase, Wnt, and Notch signaling (Ezratty et al., 2011; Wallingford and Mitchell, 2011; Pedersen et al., 2016). Signaling mediated by cilia is an ancient phenomenon; for example, interactions between receptors (agglutinins) on plus and minus gamete cilia during fertilization in the green alga Chlamydomonas stimulate a signaling pathway involved in gamete activation that ultimately leads to cell–cell fusion (Wang et al., 2006). Thus the ciliary membrane serves as a compartment for subcellular localization of factors associated with sensory perception and multiple signaling pathways.
The lipid composition of the ciliary membrane is different from the rest of the plasma membrane (Lechtreck et al., 2013). In particular, phosphoinositide 5-phosphatases Inpp5e and Ocrl localize to cilia (Bielas et al., 2009; Jacoby et al., 2009; Luo et al., 2012), and the ciliary compartment lacks phosphoinositide 4,5-bisphosphate (PI(4,5)P2), similar to endosomes (Chavez et al., 2015; Garcia-Gonzalo et al., 2015). The ciliary pocket flanking the primary cilium is rich in coated vesicles and actin microfilaments (Rohatgi and Snell, 2010; Benmerah, 2013; Pedersen et al., 2016). In addition, there are distinct lipid barriers between cilia and rest of the plasma membrane (Vieira et al., 2006), and proteins such as septins that localize to the cilia and transition zone restrict ciliary membrane components from diffusing into the rest of the plasma membrane (Hu et al., 2010; Chih et al., 2012; Ghossoub et al., 2013). Certain membrane proteins are prevented from trafficking to cilia by being immobilized by the apical actin network outside cilia (Francis et al., 2011). Thus factors that affect ciliary pools include trafficking into cilia, removal from cilia, retention inside cilia, restriction outside cilia, and recycling of membrane components in the endosomal compartment (Figure 1; Bloodgood, 2012). Finally, loss of proteins in extracellular vesicles might also regulate ciliary content (Wang et al., 2014; Cao et al., 2015; Wood and Rosenbaum, 2015).
Localization of endogenous proteins in the ciliary membrane of vertebrate cells has been mostly determined by immunolabeling techniques. However, it is important to realize that these proteins are not exclusive to cilia; rather, they are enriched in cilia. The ciliary membrane is ∼1/1000–1/5000 of the total cellular surface, and the ciliary volume (∼0.5 fl) is about ∼1/30,000 of the total cellular volume (Delling et al., 2013). The small size of the cilium enables enrichment of proteins with respect to the rest of the plasma membrane and establishing an effective signaling compartment by local concentration of second messengers and effectors. However, the absolute amounts of cilia-localized proteins are likely to be minute in comparison to total cellular levels. Thus, to understand the role of compartmentalization in ciliary signaling, it is imperative to determine mechanisms underlying ciliary trafficking, and identify functional consequences upon disruption of ciliary localization. Unfortunately, we are lacking in understanding of signaling inside cilia, mostly due to the difficulty of working with such a tiny compartment. We are also extremely limited in the availability of tools that allow us to address the role of ciliary compartmentalization while maintaining the architecture of cilia and/or retaining the functionality of the studied proteins.
ITINERARY FOR MEMBRANE PROTEIN TRAFFICKING TO CILIA
Membrane biogenesis has to be closely coordinated with axonemal growth during ciliogenesis. Key players in this process have been identified and include a Rab cascade consisting of Rab11 and Rab8 (Moritz et al., 2001; Nachury et al., 2007; Westlake et al., 2011; Lu et al., 2015). Because disruption of these factors affects ciliogenesis per se, it is important to distinguish between factors that affect biogenesis of the ciliary membrane and those that affect trafficking. An increasing number of pathways linked to the secretory pathway have been implicated in trafficking of membrane proteins to cilia. These include the small G protein ARF4 for rhodopsin and fibrocystin trafficking (Mazelova et al., 2009; Follit et al., 2014) and the GGA1 adapters for PC1/2 trafficking (Kim et al., 2014a). The BBSome proteins regulate membrane composition (Lechtreck et al., 2009, 2013) in addition to regulating ciliary GPCR pools and removal of GPCRs, polycystins, and membrane-associated proteins from cilia (Berbari et al., 2008b; Lechtreck et al., 2009, 2013; Jin et al., 2010; Domire et al., 2011; Loktev and Jackson, 2013; Eguether et al., 2014; Liew et al., 2014; Xu et al., 2015). Thus the BBSome proteins have multiple effects on ciliary trafficking and in maintaining membrane composition.
Irrespective of the role of factors in the secretory pathway and in ciliary membrane biogenesis, the final critical step in ciliary trafficking is the targeting of GPCRs into cilia from the plasma membrane or juxtaciliary vesicles. A ciliary targeting sequence needs to be carefully considered because lack of ciliary localization in mutants might result from defective transit or recycling through the secretory pathway, both of which are steps distinct from direct trafficking into the compartment. Multiple sequences that target proteins to ciliary membrane have been determined (Deretic et al., 1998; Jenkins et al., 2006; Berbari et al., 2008a, b; Follit et al., 2010; Loktev and Jackson, 2013; Mukhopadhyay et al., 2013). The lack of a consensus sequence that could exclusively predict ciliary localization (Loktev and Jackson, 2013) and the multiplicity of pathways implicated in trafficking argue for multiple ways for finally targeting proteins to cilia (Pazour and Bloodgood, 2008). Alternatively, binding of these motifs with a few adapters that is dictated by structural elements in these varied sequences could determine trafficking into cilia.
The tubby-family proteins Tulp3 and tubby (Tub) have been implicated as adapters in trafficking of multiple GPCRs into the ciliary membrane (Mukhopadhyay et al., 2010, 2013; Sun et al., 2012; Loktev and Jackson, 2013). These tubby-family proteins have an N-terminal intraflagellar complex A (IFT-A) core-binding conserved helix and a C-terminal tubby domain that binds to PI(4,5)P2 (Santagata et al., 2001; Mukhopadhyay et al., 2010). Disrupting either of these domains prevents trafficking of these GPCRs to cilia, suggesting that Tulp3 “bridges” the GPCRs with IFT-A core in targeting them into cilia (Mukhopadhyay et al., 2010). The generality of this model in targeting all cilia-localized rhodopsin-family GPCRs, the parallels between Tulp3 and Tub in ciliary trafficking, and the role of Tulp3/Tub as adapters in ciliary trafficking of other integral membrane proteins are important future directions to pursue.
In contrast to transmembrane protein trafficking to cilia, lipidated membrane-associated protein trafficking to cilia is mediated by a set of proteins that serve as carriers for the lipid modifications (Unc-119 and Pde6δ for myristolylated and prenylated proteins, respectively; Wright et al., 2011; Humbert et al., 2012). The lipid-binding carriers release the lipidated cargo into cilia in an Arl3-GTP–dependent cycle in which Arl13b functions as a guanine nucleotide exchange factor for Arl3 (Gotthardt et al., 2015). Disruption of trafficking of lipidated cargo to cilia causes profound defects in ciliary function, including disrupted Shh signaling, photodegeneration, and ciliopathies (Caspary et al., 2007; Cantagrel et al., 2008; Hanke-Gogokhia et al., 2016). An important future direction here is to determine factors that regulate trafficking of the Arl proteins, such as Arl13b, to cilia.
THE VERTEBRATE SHH PATHWAY, PRIMARY CILIUM, AND GPCR SIGNALING
The vertebrate Shh pathway is one of the best examples in which the primary cilium has been implicated in cellular signaling (Goetz and Anderson, 2010). The final output of the Shh pathway is the formation of Gli transcriptional repressors or activators, both of which occur in a cilia-dependent manner (Goetz and Anderson, 2010). Although the Gli3 repressor is critical in basal suppression of the pathway, the Gli2 activator is the major activator for signaling (Goetz and Anderson, 2010). The Gli2 transcriptional activator is formed by a Smo-dependent process, which is initiated upon binding of Shh to Ptch1, removal of Ptch1 from cilia (Rohatgi et al., 2007), and ciliary retention of Smo (Corbit et al., 2005). The Gli3 repressor is formed in a protein kinase A (PKA)–dependent manner by limited proteolysis, with the N-terminus acting as a transcriptional repressor (Chen et al., 2009; Jia et al., 2009; Humke et al., 2010; Wang et al., 2010; Wen et al., 2010). Phenotypes resulting from loss of cilia depend on the predominance of the role of repressor or activator in development and patterning of the particular tissue. For example, disruption of cilia in the neural tube results in decreased Shh signaling, predominantly in a Gli2-activator dependent manner (Goetz and Anderson, 2010).
Loss of Tulp3 and IFT-A complex results in increased Shh signaling in the neural tube as opposed to decreased signaling with loss of cilia (Norman et al., 2009; Ocbina et al., 2011; Qin et al., 2011). This suggests that there are Tulp3/IFT-A–regulated negative regulators of Shh signaling. The orphan GPCR Gpr161 localizes to cilia in a Tulp3/IFT-A–regulated manner and negatively regulates Shh signaling via cAMP signaling (Mukhopadhyay et al., 2013). A null murine allele of Gpr161 phenocopies Tulp3/IFT-A mutants in causing increased Shh signaling with concomitant lack of Gli3 repressor. Constitutive cAMP signaling by Gpr161 suggests that it regulates PKA-mediated Gli repressor formation, possibly by increasing cAMP levels in cilia (Mukhopadhyay et al., 2013). Because cAMP production by GPCRs is mediated by Gαs coupling and activation of adenylyl cyclases, it is interesting to note that Gnas (Gαs)-knockout mice exhibit increased Shh signaling (Regard et al., 2013). Finally, cAMP binds to PKA regulatory subunits, which are spatially restricted by A-kinase anchoring proteins (AKAPs), promoting release of PKA catalytic subunits in close vicinity. PKA catalytic subunit mutants demonstrate increased Shh signaling (Tuson et al., 2011). Of interest, PKA regulatory subunits that localize to cilia (Mick et al., 2015) directly bind to an amphipathic helix at the Gpr161-distal C-terminal tail (Bachmann et al., 2016), suggesting that Gpr161-PKA coupling occurs in cilia, with Gpr161 functioning as an AKAP. Key future directions here are to determine whether lack of Gpr161 localization to cilia results in a phenotype similar to lack of Gpr161. In addition, phenotypic characterization of Gpr161 conditional mutants should provide important clues regarding the role of this GPCR in the basal suppression of the hedgehog pathway in normal development and in pathogenesis of Shh-dependent tumors (Wong et al., 2009; Han and Alvarez-Buylla, 2010).
Whereas the characterization of Tulp3/IFT-A–regulated Gpr161 and downstream factors such as Gαs and PKA provide important clues to the role of maintaining Gli repressors in basal suppression of Shh pathway activity, the role of cAMP-generating adenylyl cyclases in cilia is not clear. At least three of the adenylyl cyclases (ACIII, ACV, ACVI) localize to cilia in cultured cells and in brain (Berbari et al., 2007; Choi et al., 2011; Vuolo et al., 2015); however, their role in cilia is difficult to ascertain because of redundancy (Vuolo et al., 2015). Levels of cAMP in cilia have been measured using cilia-localized sensors; however, the results are controversial. Whereas high levels of cAMP in cilia with respect to rest of the cytoplasm were detected using an intensiometric sensor (Moore et al., 2016), no differences were found using a fluorescence resonance energy transfer–based sensor (Marley et al., 2013). Phosphatidylinositol 3,4,5-triphosphate (PI(3,4,5)P3) levels in cilia, instead of Gαs, have been recently implicated in tonic regulation of ciliary cAMP levels by adenylyl cyclases (Moore et al., 2016). However, the presence of 5’ inositol phosphatases Inpp5e and Ocrl in cilia would be counterproductive to PI(3,4,5)P3 generation inside this compartment (Bielas et al., 2009; Jacoby et al., 2009; Luo et al., 2012; Chavez et al., 2015; Garcia-Gonzalo et al., 2015). In addition, factors that target adenylyl cyclases to cilia are unknown, and ciliary localization signals are not well defined. If cAMP signaling in cilia is critical, loss of trafficking of adenylyl cyclases to cilia should result in increased Shh signaling, phenocopying other factors in the basal suppression machinery. Identification of factors important in trafficking of adenylyl cyclases to cilia and their role in development and disease are important directions to pursue in the future.
MEMBRANE PROTEIN FLUX IN CILIA
One of the best examples of transmembrane protein flux in the ciliary membrane in vertebrates is provided by Smo (Corbit et al., 2005). Smo is a seven-transmembrane receptor that has an external cysteine-rich domain similar to Frizzled (Byrne et al., 2016; Huang et al., 2016). A potential route for Smo trafficking into the ciliary membrane is by lateral diffusion from the plasma membrane (Milenkovic et al., 2009). Unlike certain class A GPCRs, trafficking of Smo to cilia is Tulp3 independent (Mukhopadhyay et al., 2010; Qin et al., 2011). The Smo ciliary targeting sequence has been mapped to the C-terminal tail of the protein (Kim et al., 2015). However, upon activation of Shh signaling, Smo is retained in cilia. Recent exciting results suggest that cholesterol binding to Smo at its extracellular cysteine-rich domain could be the agonist-induced event that results in activation of Smo in the Shh pathway (Byrne et al., 2016; Huang et al., 2016). Upon Shh pathway activation, accumulation of endogenous Smo occurs gradually in cilia over hours (Wen et al., 2010). However, the determination of ciliary pools is dictated by detection limits of available antibodies. Of interest, similar rates of Smo entry and loss (t1/2 ≈ 100 min) maintain steady-state Smo levels in the compartment upon pathway activation (Kim et al., 2014b). Although these rates are based on overexpressed and tagged Smo fusions, they nicely reflect the process of simultaneous trafficking of proteins in and out of the compartment while maintaining steady-state ciliary levels.
Ciliary pools of the orphan GPCR Gpr161 are also dynamically regulated during Shh signaling. Although Gpr161 is normally localized to cilia, its levels are reduced sharply upon Shh signaling (t1/2 ≈ 30 min; Mukhopadhyay et al., 2013). The rate of entry of Gpr161 to cilia in the endogenous context has been determined by depleting the ciliary pools by Shh signaling and tracking reversible entry. These experiments suggest that similar to Smo, entry of Gpr161 occurs gradually over hours (Pal et al., 2016). Once inside cilia, the rate of GPCR loss is minimal under basal conditions (Hu et al., 2010; Chih et al., 2012). However, addition of agonists (in the case of somatostatin receptor 3 [Sstr3]; Green et al., 2016) and activation of the Shh pathway (in the case of Gpr161; Pal et al., 2016) result in their rapid removal from cilia. Removal of Sstr3 from cilia occurs through β-arrestin recruitment upon addition of agonists, similar to agonist-induced endocytosis of GPCRs from plasma membrane (Green et al., 2016). However, Gpr161 removal involves Smo trafficking to cilia (Pal et al., 2016). Whereas basal β-arrestin recruitment by Gpr161 depends on both its constitutive cAMP signaling and the Grk2 kinase, trafficking of Smo into cilia results in increased recruitment of β-arrestins to the Gpr161-proximal C-terminal tail. The β-arrestin–bound Gpr161 is finally removed by clathrin-mediated endocytosis upon exit from cilia. The mechanism underlying Smo-mediated enhancement of β-arrestin recruitment by Gpr161 is unknown but does not involve either Gαi recruitment by Smo or Grk2-mediated phosphorylation at the Smo C-tail. Instead, direct interactions between Gpr161 and Smo might promote simultaneous exit of the Gpr161-Smo bipartite receptor complex (Kim et al., 2015; Pal et al., 2016). Lack of removal of Gpr161 is associated with decreased Shh signaling (Pal et al., 2016). This is apparent from decreased Shh signaling in β-arrestin double-knockout mouse embryonic fibroblasts (MEFs), which prevents Gpr161 removal without affecting trafficking of endogenous Smo, or upon stable overexpression of the β-arrestin–binding Gpr161 mutant that is retained in cilia (Pal et al., 2016).
Ptch1 is also removed from cilia upon Shh addition (Rohatgi et al., 2007). Shh binding to Ptch1 results in its removal in a caveolin-dependent process (Yue et al., 2014). Ptch1 undergoes Smurf1/2 (HECT-domain ubiquitin E3 ligase)–dependent proteolysis, which regulates recycling in the endosomal compartment, and double knockouts of Smurf1/2 increase Ptch1 levels in cilia. The cerebellar external granule neurons proliferate in a Shh- and cilia-dependent manner postnatally in mice (Wechsler-Reya and Scott, 1999; Spassky et al., 2008). Double knockouts of Smurf1/2 in cerebellar slices inhibit Shh-dependent proliferation of granule progenitors (Yue et al., 2014), suggesting that lack of Ptch1 removal from cilia blocks high Shh signaling. However, a Ptch1 mutant that is retained in cilia restores signaling in Ptch1-knockout cells, suggesting no adverse consequences upon ciliary retention in cultured MEFs (Kim et al., 2015). Thus the consequences of lack of removal of Ptch1 are context dependent.
Dynamic redistribution of proteins also occurs in the flagella of Chlamydomonas plus gametes in a signaling-dependent manner during fertilization. Signaling in plus gametes induces rapid redistribution of the plus agglutinin (SAG1) from the plasma membrane to the periciliary region and the ciliary membrane (Belzile et al., 2013). Of interest, the entire complement of cellular SAG1 is shed during signaling in the form of ciliary ectosomes (Cao et al., 2015).
CONSEQUENCES OF LACK OF TRAFFICKING TO CILIA
To understand the role of cilia in cellular pathways, it is critical to determine whether localization of proteins in the ciliary membrane is important for signaling. GPCRs that localize to cilia also localize to the plasma membrane and in the recycling endosomal compartment (Marley and von Zastrow, 2010; Leaf and Von Zastrow, 2015). Similarly, the polycystin PC2 is present mostly in the endoplasmic reticulum (ER), and PC1 promotes its exit from ER and trafficking to cilia (Cai et al., 2014; Kim et al., 2014a; Gainullin et al., 2015). Apart from localizing to cilia, the polycystins PC1/2 and fibrocystin are also present in urinary exosomes (Pazour et al., 2002; Yoder et al., 2002; Ward et al., 2003; Hu et al., 2007; Hogan et al., 2009; Chapin and Caplan, 2010). Localization of GPCRs or polycystins in cilia does not necessarily imply that they function in this compartment in the context of a particular pathway.
To understand the role of trafficking of a protein to the ciliary membrane in relation to a cellular pathway, it is important to maintain intact cilia and prevent ciliary localization without affecting other functions of the trafficked protein. Affecting the IFT machinery or transition zone complex proteins results in gross ciliary defects. In addition, in many ciliary membrane proteins, such as Ptch1, the function of the protein is still speculative (Bazan and de Sauvage, 2009). Thus it is difficult to ascertain whether a Ptch1 mutant that does not traffic to cilia retains its native function. In such cases, ciliary-trafficking mutants can be fused with heterologous ciliary-targeting sequences and tested for rescue in knockout cells. If the mutant chimeric construct rescues the knockout phenotype, the mutant nonciliary form possibly retains native function and can be assessed for its role in the pathway. Similar experiments suggest that a Ptch1 mutant lacking in trafficking to cilia is ineffective in Shh signaling (Kim et al., 2015). Certain mutants in the polycystin PC2 suggest that trafficking to cilia is important. A mutation in the highly conserved extracellular polycystin domain in PC2 (PC2W414G) in polycystic kidney disease patients prevents trafficking to cilia but has wild-type channel properties (Cai et al., 2014). A mutant allele in mouse (PC2lrm4, E442G within the conserved ion channel region of PC2) that is not trafficked to cilia retains channel activity but causes left–right asymmetry similar to a germline PC2 knockout (Ermakov et al., 2009; Yoshiba et al., 2012). In the case of GPCRs, a similar approach would be to test whether mutants that are defective in ciliary trafficking but are otherwise functional result in phenotypes similar to the germline knockouts.
Phenotypes resulting from a lack of cilia-localized proteins are further modified in the background of cilia mutants. Whereas lack of PC1/2 causes severe polycystic kidney disease, the cysts are suppressed in the absence of cilia (Ma et al., 2013). Thus phenotypes arising from lack of PC1/2 require cilia, and a cilia-dependent cystogenesis pathway is suppressed by PC1/2 ciliary localization. Unlike PC1/2, the related polycystins PKD1L1/PKD2L1 function as calcium-selective ion channels in cilia, as detected by patch clamping of cilia (DeCaen et al., 2013; Delling et al., 2013). However, PC1/2 might be functioning in cilia as “regulated” cation channels, with yet-unidentified ligands. Determining factors that target polycystins to cilia and identifying the cilia-regulated cystogenic pathway are important future directions in studying polycystic kidney disease pathogenesis.
CONCLUSIONS
Although the cilium is a tiny cellular compartment, it has profound implications in signaling pathways. Because ciliary membrane–enriched proteins are not exclusive to this compartment, it is critical to identify factors important in trafficking them to the ciliary membrane. It is also important to determine the role of ciliary localization in the respective pathways both in GPCR-regulated signaling and in polycystic kidney disease. Furthermore, measurement of ciliary levels of second messengers and lipids are exciting newer directions in this rapidly evolving field. Finally, identifying factors in trafficking of ciliary membrane proteins and determining their role in the pathophysiology of ciliopathies might provide unique approaches to targeting these debilitating diseases.
Acknowledgments
We are indebted to anonymous reviewers for comments and suggestions on earlier versions of the manuscript. Work in our laboratory is funded by a recruitment grant from the Cancer Prevention Research Institute of Texas (R1220; S.M.), a R01 grant from the National Institutes of Health (1R01GM113023-01; S.M.), and a Welch Foundation grant (I-1906; S.M.).
Abbreviations used:
- AKAP
A-kinase anchoring protein
- BBS
Bardet-Biedl syndrome
- GPCR
G protein–coupled receptor
- IFT
intraflagellar transport
- MEF
mouse embryonic fibroblast
- PC
polycystin
- PKA
protein kinase A
- Ptch1
Patched1
- Smo
smoothened
- TRP-channel
transient receptor potential channel
Footnotes
REFERENCES
- Bachmann VA, Mayrhofer JE, Ilouz R, Tschaikner P, Raffeiner P, Rock R, Courcelles M, Apelt F, Lu TW, Baillie GS, et al. Gpr161 anchoring of PKA consolidates GPCR and cAMP signaling. Proc Natl Acad Sci USA. 2016;113:7786–7791. doi: 10.1073/pnas.1608061113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bazan JF, de Sauvage FJ. Structural ties between cholesterol transport and morphogen signaling. Cell. 2009;138:1055–1056. doi: 10.1016/j.cell.2009.09.006. [DOI] [PubMed] [Google Scholar]
- Belzile O, Hernandez-Lara CI, Wang Q, Snell WJ. Regulated membrane protein entry into flagella is facilitated by cytoplasmic microtubules and does not require IFT. Curr Biol. 2013;23:1460–1465. doi: 10.1016/j.cub.2013.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benmerah A. The ciliary pocket. Curr Opin Cell Biol. 2013;25:78–84. doi: 10.1016/j.ceb.2012.10.011. [DOI] [PubMed] [Google Scholar]
- Berbari NF, Bishop GA, Askwith CC, Lewis JS, Mykytyn K. Hippocampal neurons possess primary cilia in culture. J Neurosci Res. 2007;85:1095–1100. doi: 10.1002/jnr.21209. [DOI] [PubMed] [Google Scholar]
- Berbari NF, Johnson AD, Lewis JS, Askwith CC, Mykytyn K. Identification of ciliary localization sequences within the third intracellular loop of G protein-coupled receptors. Mol Biol Cell. 2008a;19:1540–1547. doi: 10.1091/mbc.E07-09-0942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berbari NF, Lewis JS, Bishop GA, Askwith CC, Mykytyn K. Bardet-Biedl syndrome proteins are required for the localization of G protein-coupled receptors to primary cilia. Proc Natl Acad Sci USA. 2008b;105:4242–4246. doi: 10.1073/pnas.0711027105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bielas SL, Silhavy JL, Brancati F, Kisseleva MV, Al-Gazali L, Sztriha L, Bayoumi RA, Zaki MS, Abdel-Aleem A, Rosti RO, et al. Mutations in INPP5E, encoding inositol polyphosphate-5-phosphatase E, link phosphatidyl inositol signaling to the ciliopathies. Nat Genet. 2009;41:1032–1036. doi: 10.1038/ng.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bloodgood RA. The future of ciliary and flagellar membrane research. Mol Biol Cell. 2012;23:2407–2411. doi: 10.1091/mbc.E12-01-0073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Byrne EF, Sircar R, Miller PS, Hedger G, Luchetti G, Nachtergaele S, Tully MD, Mydock-McGrane L, Covey DF, Rambo RP, et al. Structural basis of Smoothened regulation by its extracellular domains. Nature. 2016;535:517–522. doi: 10.1038/nature18934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Y, Fedeles SV, Dong K, Anyatonwu G, Onoe T, Mitobe M, Gao JD, Okuhara D, Tian X, Gallagher AR, et al. Altered trafficking and stability of polycystins underlie polycystic kidney disease. J Clin Invest. 2014;124:5129–5144. doi: 10.1172/JCI67273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cantagrel V, Silhavy JL, Bielas SL, Swistun D, Marsh SE, Bertrand JY, Audollent S, Attie-Bitach T, Holden KR, Dobyns WB, et al. Mutations in the cilia gene ARL13B lead to the classical form of Joubert syndrome. Am J Hum Genet. 2008;83:170–179. doi: 10.1016/j.ajhg.2008.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao M, Ning J, Hernandez-Lara CI, Belzile O, Wang Q, Dutcher SK, Liu Y, Snell WJ. Uni-directional ciliary membrane protein trafficking by a cytoplasmic retrograde IFT motor and ciliary ectosome shedding. Elife. 2015;4 doi: 10.7554/eLife.05242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caspary T, Larkins CE, Anderson KV. The graded response to Sonic Hedgehog depends on cilia architecture. Dev Cell. 2007;12:767–778. doi: 10.1016/j.devcel.2007.03.004. [DOI] [PubMed] [Google Scholar]
- Chapin HC, Caplan MJ. The cell biology of polycystic kidney disease. J Cell Biol. 2010;191:701–710. doi: 10.1083/jcb.201006173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chavez M, Ena S, Van Sande J, de Kerchove d'Exaerde A, Schurmans S, Schiffmann SN. Modulation of ciliary phosphoinositide content regulates trafficking and Sonic Hedgehog signaling output. Dev Cell. 2015;34:338–350. doi: 10.1016/j.devcel.2015.06.016. [DOI] [PubMed] [Google Scholar]
- Chen MH, Wilson CW, Li YJ, Law KK, Lu CS, Gacayan R, Zhang X, Hui CC, Chuang PT. Cilium-independent regulation of Gli protein function by Sufu in Hedgehog signaling is evolutionarily conserved. Genes Dev. 2009;23:1910–1928. doi: 10.1101/gad.1794109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chih B, Liu P, Chinn Y, Chalouni C, Komuves LG, Hass PE, Sandoval W, Peterson AS. A ciliopathy complex at the transition zone protects the cilia as a privileged membrane domain. Nat Cell Biol. 2012;14:61–72. doi: 10.1038/ncb2410. [DOI] [PubMed] [Google Scholar]
- Choi YH, Suzuki A, Hajarnis S, Ma Z, Chapin HC, Caplan MJ, Pontoglio M, Somlo S, Igarashi P. Polycystin-2 and phosphodiesterase 4C are components of a ciliary A-kinase anchoring protein complex that is disrupted in cystic kidney diseases. Proc Natl Acad Sci USA. 2011;108:10679–10684. doi: 10.1073/pnas.1016214108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corbit KC, Aanstad P, Singla V, Norman AR, Stainier DY, Reiter JF. Vertebrate Smoothened functions at the primary cilium. Nature. 2005;437:1018–1021. doi: 10.1038/nature04117. [DOI] [PubMed] [Google Scholar]
- DeCaen PG, Delling M, Vien TN, Clapham DE. Direct recording and molecular identification of the calcium channel of primary cilia. Nature. 2013;504:315–318. doi: 10.1038/nature12832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delling M, DeCaen PG, Doerner JF, Febvay S, Clapham DE. Primary cilia are specialized calcium signalling organelles. Nature. 2013;504:311–314. doi: 10.1038/nature12833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deretic D, Schmerl S, Hargrave PA, Arendt A, McDowell JH. Regulation of sorting and postGolgi trafficking of rhodopsin by its C-terminal sequence QVS(A)PA. Proc Natl Acad Sci USA. 1998;95:10620–10625. doi: 10.1073/pnas.95.18.10620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Domire JS, Green JA, Lee KG, Johnson AD, Askwith CC, Mykytyn K. Dopamine receptor 1 localizes to neuronal cilia in a dynamic process that requires the Bardet-Biedl syndrome proteins. Cell Mol Life Sci. 2011;68:2951–2960. doi: 10.1007/s00018-010-0603-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eguether T, San Agustin JT, Keady BT, Jonassen JA, Liang Y, Francis R, Tobita K, Johnson CA, Abdelhamed ZA, Lo CW, Pazour GJ. IFT27 links the BBSome to IFT for maintenance of the ciliary signaling compartment. Dev Cell. 2014;31:279–290. doi: 10.1016/j.devcel.2014.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ermakov A, Stevens JL, Whitehill E, Robson JE, Pieles G, Brooker D, Goggolidou P, Powles-Glover N, Hacker T, Young SR, et al. Mouse mutagenesis identifies novel roles for left-right patterning genes in pulmonary, craniofacial, ocular, and limb development. Dev Dyn. 2009;238:581–594. doi: 10.1002/dvdy.21874. [DOI] [PubMed] [Google Scholar]
- Ezratty EJ, Stokes N, Chai S, Shah AS, Williams SE, Fuchs E. A role for the primary cilium in Notch signaling and epidermal differentiation during skin development. Cell. 2011;145:1129–1141. doi: 10.1016/j.cell.2011.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Follit JA, Li L, Vucica Y, Pazour GJ. The cytoplasmic tail of fibrocystin contains a ciliary targeting sequence. J Cell Biol. 2010;188:21–28. doi: 10.1083/jcb.200910096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Follit JA, San Agustin JT, Jonassen JA, Huang T, Rivera-Perez JA, Tremblay KD, Pazour GJ. Arf4 is required for Mammalian development but dispensable for ciliary assembly. PLoS Genet. 2014;10:e1004170. doi: 10.1371/journal.pgen.1004170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francis SS, Sfakianos J, Lo B, Mellman I. A hierarchy of signals regulates entry of membrane proteins into the ciliary membrane domain in epithelial cells. J Cell Biol. 2011;193:219–233. doi: 10.1083/jcb.201009001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gainullin VG, Hopp K, Ward CJ, Hommerding CJ, Harris PC. Polycystin-1 maturation requires polycystin-2 in a dose-dependent manner. J Clin Invest. 2015;125:607–620. doi: 10.1172/JCI76972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Gonzalo FR, Phua SC, Roberson EC, Garcia G, Abedin M, Schurmans S, Inoue T, Reiter JF. Phosphoinositides regulate ciliary protein trafficking to modulate Hedgehog signaling. Dev Cell. 2015;34:400–409. doi: 10.1016/j.devcel.2015.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghossoub R, Hu Q, Failler M, Rouyez MC, Spitzbarth B, Mostowy S, Wolfrum U, Saunier S, Cossart P, Jamesnelson W, Benmerah A. Septins 2, 7 and 9 and MAP4 colocalize along the axoneme in the primary cilium and control ciliary length. J Cell Sci. 2013;126:2583–2594. doi: 10.1242/jcs.111377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goetz SC, Anderson KV. The primary cilium: a signalling centre during vertebrate development. Nat Rev Genet. 2010;11:331–344. doi: 10.1038/nrg2774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gotthardt K, Lokaj M, Koerner C, Falk N, Giessl A, Wittinghofer A. A G-protein activation cascade from Arl13B to Arl3 and implications for ciliary targeting of lipidated proteins. Elife. 2015;4:e11859. doi: 10.7554/eLife.11859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green JA, Schmid CL, Bley E, Monsma PC, Brown A, Bohn LM, Mykytyn K. Recruitment of beta-arrestin into neuronal cilia modulates somatostatin receptor subtype 3 ciliary localization. Mol Cell Biol. 2016;36:223–235. doi: 10.1128/MCB.00765-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han YG, Alvarez-Buylla A. Role of primary cilia in brain development and cancer. Curr Opin Neurobiol. 2010;20:58–67. doi: 10.1016/j.conb.2009.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanke-Gogokhia C, Wu Z, Gerstner CD, Frederick JM, Zhang H, Baehr W. Arf-like Protein 3 (ARL3) regulates protein trafficking and ciliogenesis in mouse photoreceptors. J Biol Chem. 2016;291:7142–7155. doi: 10.1074/jbc.M115.710954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hildebrandt F, Benzing T, Katsanis N. Ciliopathies. N Engl J Med. 2011;364:1533–1543. doi: 10.1056/NEJMra1010172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hilgendorf KI, Johnson CT, Jackson PK. The primary cilium as a cellular receiver: organizing ciliary GPCR signaling. Curr Opin Cell Biol. 2016;39:84–92. doi: 10.1016/j.ceb.2016.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hogan MC, Manganelli L, Woollard JR, Masyuk AI, Masyuk TV, Tammachote R, Huang BQ, Leontovich AA, Beito TG, Madden BJ, et al. Characterization of PKD protein-positive exosome-like vesicles. J Am Soc Nephrol. 2009;20:278–288. doi: 10.1681/ASN.2008060564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu J, Wittekind SG, Barr MM. STAM and Hrs down-regulate ciliary TRP receptors. Mol Biol Cell. 2007;18:3277–3289. doi: 10.1091/mbc.E07-03-0239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Q, Milenkovic L, Jin H, Scott MP, Nachury MV, Spiliotis ET, Nelson WJ. A septin diffusion barrier at the base of the primary cilium maintains ciliary membrane protein distribution. Science. 2010;329:436–439. doi: 10.1126/science.1191054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang P, Nedelcu D, Watanabe M, Jao C, Kim Y, Liu J, Salic A. Cellular cholesterol directly activates smoothened in hedgehog signaling. Cell. 2016;166:1176–1187. doi: 10.1016/j.cell.2016.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Humbert MC, Weihbrecht K, Searby CC, Li Y, Pope RM, Sheffield VC, Seo S. ARL13B, PDE6D, and CEP164 form a functional network for INPP5E ciliary targeting. Proc Natl Acad Sci USA. 2012;109:19691–19696. doi: 10.1073/pnas.1210916109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Humke EW, Dorn KV, Milenkovic L, Scott MP, Rohatgi R. The output of Hedgehog signaling is controlled by the dynamic association between Suppressor of Fused and the Gli proteins. Genes Dev. 2010;24:670–682. doi: 10.1101/gad.1902910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacoby M, Cox JJ, Gayral S, Hampshire DJ, Ayub M, Blockmans M, Pernot E, Kisseleva MV, Compere P, Schiffmann SN, et al. INPP5E mutations cause primary cilium signaling defects, ciliary instability and ciliopathies in human and mouse. Nat Genet. 2009;41:1027–1031. doi: 10.1038/ng.427. [DOI] [PubMed] [Google Scholar]
- Jenkins PM, Hurd TW, Zhang L, McEwen DP, Brown RL, Margolis B, Verhey KJ, Martens JR. Ciliary targeting of olfactory CNG channels requires the CNGB1b subunit and the kinesin-2 motor protein, KIF17. Curr Biol. 2006;16:1211–1216. doi: 10.1016/j.cub.2006.04.034. [DOI] [PubMed] [Google Scholar]
- Jia J, Kolterud A, Zeng H, Hoover A, Teglund S, Toftgard R, Liu A. Suppressor of Fused inhibits mammalian Hedgehog signaling in the absence of cilia. Dev Biol. 2009;330:452–460. doi: 10.1016/j.ydbio.2009.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin H, White SR, Shida T, Schulz S, Aguiar M, Gygi SP, Bazan JF, Nachury MV. The conserved Bardet-Biedl syndrome proteins assemble a coat that traffics membrane proteins to cilia. Cell. 2010;141:1208–1219. doi: 10.1016/j.cell.2010.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim H, Xu H, Yao Q, Li W, Huang Q, Outeda P, Cebotaru V, Chiaravalli M, Boletta A, Piontek K, et al. Ciliary membrane proteins traffic through the Golgi via a Rabep1/GGA1/Arl3-dependent mechanism. Nat Commun. 2014a;5:5482. doi: 10.1038/ncomms6482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, Hsia EY, Brigui A, Plessis A, Beachy PA, Zheng X. The role of ciliary trafficking in Hedgehog receptor signaling. Sci Signal. 2015;8:ra55. doi: 10.1126/scisignal.aaa5622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J, Hsia EY, Kim J, Sever N, Beachy PA, Zheng X. Simultaneous measurement of smoothened entry into and exit from the primary cilium. PLoS One. 2014b;9:e104070. doi: 10.1371/journal.pone.0104070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leaf A, Von Zastrow M. Dopamine receptors reveal an essential role of IFT-B, KIF17, and Rab23 in delivering specific receptors to primary cilia. Elife. 2015;4 doi: 10.7554/eLife.06996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lechtreck KF, Brown JM, Sampaio JL, Craft JM, Shevchenko A, Evans JE, Witman GB. Cycling of the signaling protein phospholipase D through cilia requires the BBSome only for the export phase. J Cell Biol. 2013;201:249–261. doi: 10.1083/jcb.201207139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lechtreck KF, Johnson EC, Sakai T, Cochran D, Ballif BA, Rush J, Pazour GJ, Ikebe M, Witman GB. The Chlamydomonas reinhardtii BBSome is an IFT cargo required for export of specific signaling proteins from flagella. J Cell Biol. 2009;187:1117–1132. doi: 10.1083/jcb.200909183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liew GM, Ye F, Nager AR, Murphy JP, Lee JS, Aguiar M, Breslow DK, Gygi SP, Nachury MV. The intraflagellar transport protein IFT27 promotes BBSome exit from cilia through the GTPase ARL6/BBS3. Dev Cell. 2014;31:265–278. doi: 10.1016/j.devcel.2014.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loktev AV, Jackson PK. Neuropeptide Y family receptors traffic via the Bardet-Biedl syndrome pathway to signal in neuronal primary cilia. Cell Rep. 2013;5:1316–1329. doi: 10.1016/j.celrep.2013.11.011. [DOI] [PubMed] [Google Scholar]
- Lu Q, Insinna C, Ott C, Stauffer J, Pintado PA, Rahajeng J, Baxa U, Walia V, Cuenca A, Hwang YS, et al. Early steps in primary cilium assembly require EHD1/EHD3-dependent ciliary vesicle formation. Nat Cell Biol. 2015;17:228–240. doi: 10.1038/ncb3109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo N, West CC, Murga-Zamalloa CA, Sun L, Anderson RM, Wells CD, Weinreb RN, Travers JB, Khanna H, Sun Y. OCRL localizes to the primary cilium: a new role for cilia in Lowe syndrome. Hum Mol Genet. 2012;21:3333–3344. doi: 10.1093/hmg/dds163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma M, Tian X, Igarashi P, Pazour GJ, Somlo S. Loss of cilia suppresses cyst growth in genetic models of autosomal dominant polycystic kidney disease. Nat Genet. 2013;45:1004–1012. doi: 10.1038/ng.2715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marley A, Choy RW, von Zastrow M. GPR88 reveals a discrete function of primary cilia as selective insulators of GPCR cross-talk. PLoS One. 2013;8:e70857. doi: 10.1371/journal.pone.0070857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marley A, von Zastrow M. DISC1 regulates primary cilia that display specific dopamine receptors. PLoS One. 2010;5:e10902. doi: 10.1371/journal.pone.0010902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazelova J, Astuto-Gribble L, Inoue H, Tam BM, Schonteich E, Prekeris R, Moritz OL, Randazzo PA, Deretic D. Ciliary targeting motif VxPx directs assembly of a trafficking module through Arf4. EMBO J. 2009;28:183–192. doi: 10.1038/emboj.2008.267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mick DU, Rodrigues RB, Leib RD, Adams CM, Chien AS, Gygi SP, Nachury MV. Proteomics of primary cilia by proximity labeling. Dev Cell. 2015;35:497–512. doi: 10.1016/j.devcel.2015.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milenkovic L, Scott MP, Rohatgi R. Lateral transport of Smoothened from the plasma membrane to the membrane of the cilium. J Cell Biol. 2009;187:365–374. doi: 10.1083/jcb.200907126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore BS, Stepanchick AN, Tewson PH, Hartle CM, Zhang J, Quinn AM, Hughes TE, Mirshahi T. Cilia have high cAMP levels that are inhibited by Sonic Hedgehog-regulated calcium dynamics. Proc Natl Acad Sci USA. 2016;113:13069–13074. doi: 10.1073/pnas.1602393113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moritz OL, Tam BM, Hurd LL, Peranen J, Deretic D, Papermaster DS. Mutant rab8 Impairs docking and fusion of rhodopsin-bearing postGolgi membranes and causes cell death of transgenic Xenopus rods. Mol Biol Cell. 2001;12:2341–2351. doi: 10.1091/mbc.12.8.2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukhopadhyay S, Wen X, Chih B, Nelson CD, Lane WS, Scales SJ, Jackson PK. TULP3 bridges the IFT-A complex and membrane phosphoinositides to promote trafficking of G protein-coupled receptors into primary cilia. Genes Dev. 2010;24:2180–2193. doi: 10.1101/gad.1966210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukhopadhyay S, Wen X, Ratti N, Loktev A, Rangell L, Scales SJ, Jackson PK. The ciliary G-protein-coupled receptor Gpr161 negatively regulates the Sonic hedgehog pathway via cAMP signaling. Cell. 2013;152:210–223. doi: 10.1016/j.cell.2012.12.026. [DOI] [PubMed] [Google Scholar]
- Nachury MV, Loktev AV, Zhang Q, Westlake CJ, Peranen J, Merdes A, Slusarski DC, Scheller RH, Bazan JF, Sheffield VC, Jackson PK. A core complex of BBS proteins cooperates with the GTPase Rab8 to promote ciliary membrane biogenesis. Cell. 2007;129:1201–1213. doi: 10.1016/j.cell.2007.03.053. [DOI] [PubMed] [Google Scholar]
- Norman RX, Ko HW, Huang V, Eun CM, Abler LL, Zhang Z, Sun X, Eggenschwiler JT. Tubby-like protein 3 (TULP3) regulates patterning in the mouse embryo through inhibition of Hedgehog signaling. Hum Mol Genet. 2009;18:1740–1754. doi: 10.1093/hmg/ddp113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ocbina PJ, Eggenschwiler JT, Moskowitz I, Anderson KV. Complex interactions between genes controlling trafficking in primary cilia. Nat Genet. 2011;43:547–553. doi: 10.1038/ng.832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pal K, Hwang SH, Somatilaka B, Badgandi H, Jackson PK, DeFea K, Mukhopadhyay S. Smoothened determines beta-arrestin-mediated removal of the G protein-coupled receptor Gpr161 from the primary cilium. J Cell Biol. 2016;212:861–875. doi: 10.1083/jcb.201506132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pazour GJ, Bloodgood RA. Targeting proteins to the ciliary membrane. Curr Top Dev Biol. 2008;85:115–149. doi: 10.1016/S0070-2153(08)00805-3. [DOI] [PubMed] [Google Scholar]
- Pazour GJ, San Agustin JT, Follit JA, Rosenbaum JL, Witman GB. Polycystin-2 localizes to kidney cilia and the ciliary level is elevated in orpk mice with polycystic kidney disease. Curr Biol. 2002;12:R378–R380. doi: 10.1016/s0960-9822(02)00877-1. [DOI] [PubMed] [Google Scholar]
- Pedersen LB, Mogensen JB, Christensen ST. Endocytic control of cellular signaling at the primary cilium. Trends Biochem Sci. 2016;41:784–797. doi: 10.1016/j.tibs.2016.06.002. [DOI] [PubMed] [Google Scholar]
- Qin J, Lin Y, Norman RX, Ko HW, Eggenschwiler JT. Intraflagellar transport protein 122 antagonizes Sonic Hedgehog signaling and controls ciliary localization of pathway components. Proc Natl Acad Sci USA. 2011;108:1456–1461. doi: 10.1073/pnas.1011410108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Regard JB, Malhotra D, Gvozdenovic-Jeremic J, Josey M, Chen M, Weinstein LS, Lu J, Shore EM, Kaplan FS, Yang Y. Activation of Hedgehog signaling by loss of GNAS causes heterotopic ossification. Nat Med. 2013;19:1505–1512. doi: 10.1038/nm.3314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reiter JF, Blacque OE, Leroux MR. The base of the cilium: roles for transition fibres and the transition zone in ciliary formation, maintenance and compartmentalization. EMBO Rep. 2012;13:608–618. doi: 10.1038/embor.2012.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohatgi R, Milenkovic L, Scott MP. Patched1 regulates hedgehog signaling at the primary cilium. Science. 2007;317:372–376. doi: 10.1126/science.1139740. [DOI] [PubMed] [Google Scholar]
- Rohatgi R, Snell WJ. The ciliary membrane. Curr Opin Cell Biol. 2010;22:541–546. doi: 10.1016/j.ceb.2010.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenbaum JL, Witman GB. Intraflagellar transport. Nat Rev Mol Cell Biol. 2002;3:813–825. doi: 10.1038/nrm952. [DOI] [PubMed] [Google Scholar]
- Santagata S, Boggon TJ, Baird CL, Gomez CA, Zhao J, Shan WS, Myszka DG, Shapiro L. G-protein signaling through tubby proteins. Science. 2001;292:2041–2050. doi: 10.1126/science.1061233. [DOI] [PubMed] [Google Scholar]
- Spassky N, Han YG, Aguilar A, Strehl L, Besse L, Laclef C, Ros MR, Garcia-Verdugo JM, Alvarez-Buylla A. Primary cilia are required for cerebellar development and Shh-dependent expansion of progenitor pool. Dev Biol. 2008;317:246–259. doi: 10.1016/j.ydbio.2008.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun X, Haley J, Bulgakov OV, Cai X, McGinnis J, Li T. Tubby is required for trafficking G protein-coupled receptors to neuronal cilia. Cilia. 2012;1:21. doi: 10.1186/2046-2530-1-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tuson M, He M, Anderson KV. Protein kinase A acts at the basal body of the primary cilium to prevent Gli2 activation and ventralization of the mouse neural tube. Development. 2011;138:4921–4930. doi: 10.1242/dev.070805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vieira OV, Gaus K, Verkade P, Fullekrug J, Vaz WL, Simons K. FAPP2, cilium formation, and compartmentalization of the apical membrane in polarized Madin-Darby canine kidney (MDCK) cells. Proc Natl Acad Sci USA. 2006;103:18556–18561. doi: 10.1073/pnas.0608291103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vuolo L, Herrera A, Torroba B, Menendez A, Pons S. Ciliary adenylyl cyclases control the Hedgehog pathway. J Cell Sci. 2015;128:2928–2937. doi: 10.1242/jcs.172635. [DOI] [PubMed] [Google Scholar]
- Wallingford JB, Mitchell B. Strange as it may seem: the many links between Wnt signaling, planar cell polarity, and cilia. Genes Dev. 2011;25:201–213. doi: 10.1101/gad.2008011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Pan Y, Wang B. Suppressor of fused and Spop regulate the stability, processing and function of Gli2 and Gli3 full-length activators but not their repressors. Development. 2010;137:2001–2009. doi: 10.1242/dev.052126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J, Silva M, Haas LA, Morsci NS, Nguyen KC, Hall DH, Barr MM. C. elegans ciliated sensory neurons release extracellular vesicles that function in animal communication. Curr Biol. 2014;24:519–525. doi: 10.1016/j.cub.2014.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Pan J, Snell WJ. Intraflagellar transport particles participate directly in cilium-generated signaling in Chlamydomonas. Cell. 2006;125:549–562. doi: 10.1016/j.cell.2006.02.044. [DOI] [PubMed] [Google Scholar]
- Ward CJ, Yuan D, Masyuk TV, Wang X, Punyashthiti R, Whelan S, Bacallao R, Torra R, LaRusso NF, Torres VE, et al. Cellular and subcellular localization of the ARPKD protein; fibrocystin is expressed on primary cilia. Hum Mol Genet. 2003;12:2703–2710. doi: 10.1093/hmg/ddg274. [DOI] [PubMed] [Google Scholar]
- Wechsler-Reya RJ, Scott MP. Control of neuronal precursor proliferation in the cerebellum by Sonic Hedgehog. Neuron. 1999;22:103–114. doi: 10.1016/s0896-6273(00)80682-0. [DOI] [PubMed] [Google Scholar]
- Wen X, Lai CK, Evangelista M, Hongo JA, de Sauvage FJ, Scales SJ. Kinetics of hedgehog-dependent full-length Gli3 accumulation in primary cilia and subsequent degradation. Mol Cell Biol. 2010;30:1910–1922. doi: 10.1128/MCB.01089-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Westlake CJ, Baye LM, Nachury MV, Wright KJ, Ervin KE, Phu L, Chalouni C, Beck JS, Kirkpatrick DS, Slusarski DC, et al. Primary cilia membrane assembly is initiated by Rab11 and transport protein particle II (TRAPPII) complex-dependent trafficking of Rabin8 to the centrosome. Proc Natl Acad Sci USA. 2011;108:2759–2764. doi: 10.1073/pnas.1018823108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong SY, Seol AD, So PL, Ermilov AN, Bichakjian CK, Epstein EH, Dlugosz AA, Reiter JF. Primary cilia can both mediate and suppress Hedgehog pathway-dependent tumorigenesis. Nat Med. 2009;15:1055–1061. doi: 10.1038/nm.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wood CR, Rosenbaum JL. Ciliary ectosomes: transmissions from the cell's antenna. Trends Cell Biol. 2015;25:276–285. doi: 10.1016/j.tcb.2014.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright KJ, Baye LM, Olivier-Mason A, Mukhopadhyay S, Sang L, Kwong M, Wang W, Pretorius PR, Sheffield VC, Sengupta P, et al. An ARL3-UNC119-RP2 GTPase cycle targets myristoylated NPHP3 to the primary cilium. Genes Dev. 2011;25:2347–2360. doi: 10.1101/gad.173443.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Q, Zhang Y, Wei Q, Huang Y, Li Y, Ling K, Hu J. BBS4 and BBS5 show functional redundancy in the BBSome to regulate the degradative sorting of ciliary sensory receptors. Sci Rep. 2015;5:11855. doi: 10.1038/srep11855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoder BK, Hou X, Guay-Woodford LM. The polycystic kidney disease proteins, polycystin-1, polycystin-2, polaris, and cystin, are colocalized in renal cilia. J Am Soc Nephrol. 2002;13:2508–2516. doi: 10.1097/01.asn.0000029587.47950.25. [DOI] [PubMed] [Google Scholar]
- Yoshiba S, Shiratori H, Kuo IY, Kawasumi A, Shinohara K, Nonaka S, Asai Y, Sasaki G, Belo JA, Sasaki H, et al. Cilia at the node of mouse embryos sense fluid flow for left-right determination via Pkd2. Science. 2012;338:226–231. doi: 10.1126/science.1222538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yue S, Tang LY, Tang Y, Tang Y, Shen QH, Ding J, Chen Y, Zhang Z, Yu TT, Zhang YE, Cheng SY. Requirement of Smurf-mediated endocytosis of Patched1 in sonic hedgehog signal reception. Elife. 2014;3 doi: 10.7554/eLife.02555. [DOI] [PMC free article] [PubMed] [Google Scholar]