Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Oct 1.
Published in final edited form as: Nat Rev Nephrol. 2019 Apr;15(4):199–219. doi: 10.1038/s41581-019-0116-9

Cellular signalling by primary cilia in development, organ function and disease

Zeinab Anvarian 1, Kirk Mykytyn 2, Saikat Mukhopadhyay 3, Lotte Bang Pedersen 1, Søren Tvorup Christensen 1
PMCID: PMC6426138  NIHMSID: NIHMS1011175  PMID: 30733609

Abstract

Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this review we highlight central mechanisms by which primary cilia coordinate HH, G-protein-coupled receptor, WNT, receptor tyrosine kinase and TGFβ/BMP signalling, and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.

Opening section

The primary cilium is a microtubule-based, non-motile organelle that extends as a solitary unit from the basal body (derived from the centrosomal mother centriole of most cell types in the human body 1. The cilium is enclosed by a membrane that is continuous with the plasma membrane but has a unique lipid and receptor composition that enables the cilium to detect changes in the extracellular environment and convey signalling information to the cell to regulate diverse cellular, developmental and physiological processes. Consequently, mutations that lead to dysfunction of primary cilia give rise to a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which can affect many different organs during embryonic development as well as in postnatal life 2. Primary cilia are dynamic organelles that are assembled and disassembled in coordination with cell cycle and developmental cues. Emerging evidence indicates that the constellation of signalling components within the cilium is also dynamic and closely coupled to the differentiation state and microenvironment of the cell 3. This versatility of the cilium might explain how specific cell types are able to receive and convert signalling inputs at different time points during development and under physiological conditions. Here we present an overview of the main signalling pathways, including those regulated by Hedgehog (HH), G-protein-coupled receptors (GPCR), WNT, receptor-tyrosine kinases (RTKs) and TGFβ/BMP receptors, that are coordinated by primary cilia to control developmental processes, tissue plasticity and organ function. We discuss the potential mechanisms by which primary cilia regulate signalling pathway interactions and organize spatial-temporal signalling networks during development as well as in the maintenance of tissue homeostasis, and describe how dysfunctional cilliary signalling can lead to a multitude of human diseases.

Intraflagellar transport and ciliopathies

Both motile and non-motile cilia, including primary cilia, comprise a microtubule-based axoneme that extends from a basal body and is covered by a bilayer lipid membrane enriched in specific signalling receptors and ion channels. The axoneme of a primary cilium contains a ring of nine outer microtubule doublets (known as a 9+0 axoneme), whereas the axoneme of a motile cilium has nine outer microtubule doublets around two central microtubule singlets (called a 9+2 axoneme). The basal body is a modified centriole that contains specialized structures at its distal end that regulate critical aspects of ciliary biogenesis and function. For example, transition fibres mediate docking of the basal body to the plasma membrane or vesicles during early stages of ciliogenesis 4, 5 whereas basal feet interact with the actin cytoskeleton of the cell to regulate basal body alignment in cells that contain multiple motile cilia, such as epithelial cells that line the mammalian respiratory tract, brain ventricles or oviduct 6. For cells that form a single primary cilium (Figure 1), the basal body is derived from the mother centriole of the centrosome, and depending on the cell type, axoneme extension can be initiated before or after docking of the basal body at the plasma membrane 4, 5. The length of cilia is controlled by the actions of various kinases and other proteins 7, 8; before mitosis the cilium is usually dismantled and centrioles are duplicated for participation in mitotic spindle pole formation 914. Quiescent cells can lose their cilium as a consequence of developmental programming 1519 or in response to environmental insults such as mechanical stress 20.

Figure 1. Overview of primary cilia, cellular signalling and ciliopathies.

Figure 1.

a| The primary cilium is a non-motile organelle that extends as a solitary unit from the centrosomal mother centriole (basal body). a | The cilium comprises a microtubule (MT)-based axoneme containing a ring of nine outer microtubule doublets. Between the basal body and cilium is the ciliary transition zone (TZ), which contains specialized gating structures such as Y-links that along with the basal body transition fibres control the entrance and exit of ciliary proteins, thereby contributing to compartmentalization of the organelle. The intraflagellar transport (IFT) system zips up (anterograde) and down (retrograde) axonemal microtubules to mediate the transport of specific ciliary cargo, such as receptors, into or out of the organelle, whereupon they are degraded or recycled. Cilia can also release ectosomes by shedding off membrane-enclosed material from the surface of the organelle. The function of these extracellular vesicles has been linked to maintenance of ciliary integrity, balancing of intraciliary signalling events and/or in transmission of signals across cells 28, 293. b| Image of a primary cilium in a mouse embryonic fibroblast analysed by scanning electron microscopy. c| Selected ciliopathies are caused by dysfunctional primary cilia. d| Overview of diverse sensory capabilities of primary cilia and their associated signalling pathways. BBSome: protein complex of eight Bardet–Biedl syndrome proteins; SDA, sub-distal appendages, TF, transition fibres.

Between the basal body and cilium proper is a region known as the ciliary transition zone (TZ), which contains specialized gating structures such as Y-links that along with the basal body transition fibres control the entrance and exit of ciliary proteins, and thereby contribute to compartmentalization of the organelle (Figure 1). Importantly, a number of genes mutated in ciliopathies such as Joubert syndrome, Meckel-Gruber syndrome (MKS), and nephronophthisis (NPHP), encode protein module components of the TZ or basal body transition fibres, highlighting the physiological importance of these structures 21. In addition to gating by the TZ and basal body transition fibres, ciliary composition and function are also regulated by active transport mechanisms (Figure 1). These include vesicular transport pathways that target specific receptors or signalling molecules from the Golgi or recycling endosome to the ciliary base where vesicles are exocytosed 22, 23, and the intraflagellar transport (IFT) system that zips up and down axonemal microtubules to mediate the transport of specific ciliary cargo proteins into or out of the organelle 24, 25 (Figure 1). Moreover, ciliary membrane content can be modulated by the ectocytosis of vesicles at the ciliary tip 2628 (Figure 1). Cilia are unable to synthesize proteins; therefore, cilia-associated transport pathways also function during cilium biogenesis and maintenance by delivering the building blocks required for ciliary axoneme and membrane extension 2225. Not surprisingly, mutations in genes that encode proteins involved in cilium-associated transport processes, such as IFT, typically result in absent or defective cilia and are associated with a wide range of human ciliopathies (Figure 1), including polycystic kidney disease (PKD), Bardet Biedl syndrome (BBS) and Short-rib thoracic dysplasia29. Indeed, studies of IFT have been instrumental for furthering our understanding of the importance of primary cilia in human health and disease. We therefore provide a brief overview of the process of IFT below. Comprehensive reviews on IFT are available elsewhere 25, 30, 31.

The IFT system

Discovery and characterization of the IFT system

The IFT system was discovered in 1993 following the observation of a continuous movement of particles, sandwiched between the flagellar membrane and axonemal outer doublet microtubules, from the flagellar base to tip (anterograde IFT) and back (retrograde IFT) in Chlamydomonas reinhardtii flagella32. A few years later, studies using temperature sensitive C. reinhardtii mutants with defects in flagellar assembly revealed that anterograde and retrograde IFT movements are powered by heterotrimeric kinesin-2 3335 and cytoplasmic dynein 2 motors 3638, respectively. A large number of studies in different organisms subsequently confirmed a universal requirement for these motors in IFT, but also demonstrated that some organisms additionally use ‘accessory’ kinesins to regulate certain aspects of ciliary assembly or function 23, 39. For example, metazoans such as Caenorhabditis elegans and zebrafish express a homodimeric kinesin-2 motor that functions somewhat redundantly with heterotrimeric kinesin-2 to mediate anterograde IFT and distal singlet axonemal microtubule assembly in certain types of cilia 4043.

Purification of the polypeptides within the particles moved by IFT 44, demonstrated that they sediment as two larger complexes, termed IFT-A and IFT-B 44, 45. We now know these polypeptides can be subdivided into IFT-A, IFT-B1 and IFT-B2 sub-complexes that contain 6, 10 and 6 different IFT particle proteins, respectively 25, 30, 31, 46, 47. The three dimensional structures of many IFT-B polypeptides have been solved, and progress has been made towards understanding the arrangement and interactions of individual IFT polypeptides within the IFT-A and IFT-B sub-complexes 25. Likewise, progress has been made towards understanding the interactions between specific IFT particle polypeptides and specific ciliary cargo proteins or motor subunits during anterograde or retrograde IFT, as well as how IFT motor activity is regulated 24, 4852.

Sequence analysis of IFT polypeptides isolated from C. reinhardtii flagella revealed that they are homologous to proteins required for ciliary assembly in sensory neurons of C. elegans 44, 53, providing the first evidence that IFT is an evolutionarily conserved process required for ciliary assembly in eukaryotes. Subsequent work in a range of additional organisms, including protists such as Tetrahymena thermophila and Trypanosoma brucei and metazoans like C. elegans, sea urchin, zebrafish, mouse, and human, have substantiated this view, and provided remarkable insight into the molecular workings and physiological roles of IFT 25, 30, 31, 46. For example, although early work using C. reinhardtii and C. elegans mutants suggested that IFT-A and IFT-B complexes function in retrograde and anterograde IFT, respectively 30, 46, we now know that these polypeptides participate in ciliary transport in both directions 5460. For instance, the IFT-B complex proteins IFT-81 and IFT-74 promote anterograde IFT of tubulin towards the ciliary tip during axoneme assembly 61, whereas the IFT-B complex proteins IFT25 and IFT27 are involved in ciliary export of hedgehog (HH) signalling components via retrograde IFT 59, 60, 62, 63. Furthermore, the IFT-A complex interacts with the tubby domain proteins TULP3 and TUB to mediate ciliary targeting of certain transmembrane proteins, including G protein-coupled receptors (GPCRs) 56, 58, 6469. In addition, a complex of BBS proteins called the BBSome 70, was shown to function as an IFT adapter 71, 72 in transporting ciliary proteins. The BBSome proteins were initially thought to facilitate the delivery of GPCRs to cilia 7375; however, it has been proposed that BBSome proteins primarily regulate ciliary export of signalling proteins 72, 76, 77, 78. For example, flagella in a Chlamydomonas bbs4 mutant show abnormal accumulation of several signalling proteins, resulting in disrupted phototaxis 72.

Linking IFT to human disease

The above-described studies of IFT in Chlamydomonas set the stage for a landmark study which revealed that a hypomorphic mutation in the mouse Tg737 gene, which encodes an orthologue of the Chlamydomonas IFT-B polypeptide IFT88, causes ciliary loss and autosomal recessive (AR) PKD 79, 80. This study provided the first evidence that defective primary cilia can lead to disease in mammals, and was supported by prior work in C. elegans demonstrating that homologs of the human autosomal dominant (AD) PKD1 and PKD2 gene products, the transmembrane proteins polycystin (PC) 1 and PC2, localize to neuronal sensory cilia and regulate male mating behaviour 81. In agreement with these observations, PC1 and PC2 were found to localize to primary cilia in mammalian cells such as those of the renal collecting duct 8284. The physiological importance of the polycystins is underscored by the fact that mutations in the corresponding genes lead to ADPKD — a disease characterized by the adult-onset development of kidney and liver cysts 85, 86. Furthermore, mutations in PKD2 have been linked to left-right laterality defects in vertebrates, including humans (add refs: PMID 12062060 and PMID 21719175). It is generally accepted that proper functioning of PC1 and PC2 relies on their appropriate targeting to the ciliary compartment, but the precise mechanism by which the polycystins function at cilia to prevent cyst formation and control left-right patterning during development is a matter of intense debate 85. For many years, the prevailing hypothesis was that PC1 and PC2 comprise a mechanosensitive receptor-Ca2+ channel complex, which is activated by fluid flow 87, 88. This hypothesis was challenged 89, 90, and it was subsequently suggested that PC2, independently of PC1, forms a homotetrameric K+ and Na+-conducting ciliary ion channel that is potentiated by intraciliary Ca2+ 9193. A report of the near-atomic-resolution structure of the human PC1-PC2 complex by single-particle cryo-electron microscopy, however, strongly favours a model in which PC1 and PC2 function together in regulating cation transport 94. Nevertheless, despite important advances, the molecular mechanisms by which cilia and polycystins cooperate to regulate development and tissue homeostasis remain incompletely understood (discussed elsewhere85). Further work in many different organisms, including mouse and human, has substantiated the link between primary cilia and kidney disease, and revealed a requirement for IFT and cilia in a range of other disease-relevant pathways (Figure 1).

Ciliary organization of HH signalling

HH signalling pathways regulate a number of cell fate and self-renewal processes in development and tissue homeostasis 95. The final transcriptional output of canonical HH signalling is determined by post-translational modifications of GLI transcription factors, which cause intracellular activation or basal repression of pathway targets in the presence or absence of HH morphogens, respectively. Defects in IFT were first shown to disrupt sonic hedgehog (SHH) signalling during mouse embryonic development in a forward genetic screen in 2003 96; the primary cilium is now known to be fundamentally important for canonical SHH signalling in vertebrates 97. Activation of the SHH pathway by formation of the GLI transcriptional activator (GLIA) and basal repression of the SHH pathway by GLI transcriptional repressor (GLIR) are both dependent on the primary cilium 97. Binding of SHH to its 12-transmembrane receptor Patched-1 (PTCH1) triggers endocytic clearance of PTCH1 from the cilium in a process that relies on ubiquitination of the receptor by the HECT domain E3 ubiquitin ligases SMURF1 and SMURF2, which co-localize with PTCH1 in caveolin-1 (CAV1)-positive lipid rafts and vesicles 98. CAV1 localizes to the base of cilia in cultured mammalian cells in a manner dependent on the kinesin-3 motor protein KIF13B; both CAV1 and KIF13B have been implicated in the regulation of SHH signalling 99. CAV1 has also been reported to have ciliary functions in C. elegans 100. Removal of ciliary PTCH1 in response to SHH binding is associated with ciliary enrichment of Smoothened (SMO) 101, 102 –a 7-transmembrane receptor belonging to the class F (frizzled) family of GPCRs – which ultimately leads to the formation of GLIA, predominantly by phosphorylation of GLI2 103 (Figure 2). In contrast, basal repression in the absence of SHH involves protein kinase A (PKA)-mediated phosphorylation, predominantly of GLI3, and formation of the truncated N-terminal form of GLI, GLIR, in a cilia-dependent manner 104. Additional ligands of the HH family includes desert (DHH) and Indian (IHH) hedgehog, which also operate via primary cilia but in a tissue-specific manner, such as in cells of the testis and in growth plate chondrocytes 105, respectively.

Figure 2. Overview of canonical hedgehog signalling in primary cilia.

Figure 2.

a| In the absence of sonic hedgehog (SHH; that is, under conditions of basal suppression) the receptor patched-1 (PTCH1) is enriched in the ciliary membrane, preventing ciliary accumulation of smoothened (SMO). A transmembrane sterol sensing domain (SSD) in PTCH1 can accommodate cholesterol or cholesterol derivatives. The class A GPCR, GPR161, is targeted to the cilium by tubby-like protein 3 (TULP3) and intraflagellar transport complex A (IFT-A) to activate adenylyl cyclases via G-proteins (Gαs), leading to increased ciliary levels of cAMP. Increased cAMP levels release protein kinase A (PKA) from regulatory subunits (RI), which in conjunction with glycogen synthase kinase 3 beta (GSK3-β) and casein kinases (CK) promote the limited proteolytic cleavage of full-length versions of GLI transcription factors (GLI-FL) into their repressor form (GLI-R) in a cilia-dependent manner. Suppressor of FUSED (SUFU) restrains GLI3 in the cytoplasm and promotes GLI3 processing. b| Binding of SHH to PTCH1 extracellular domains (ECDs) is regulated by cholesterol derivatives. In addition, SMURF1/2-mediated ubiquitination regulates PTCH1 exit from cilia and internalization at the ciliary base. Removal of PTCH1 causes concomitant enrichment and activation of SMO in cilia by cholesterol or derivatives. A longitudinal tunnel in the transmembrane region of SMO (dotted arrow) could move cholesterol from the ciliary membrane to its binding domain in the cysteine rich region (CRD). SHH stimulation results in dissociation of SUFU from GLI transcription factors, formation of full-length activator forms of GLIs (GLI-A), and accumulation of these proteins along with the microtubule-associated atypical kinesin KIF7 at ciliary tips. Downstream effectors for SMO include the EVC–EVC2 complex, which localizes in cilia distal to the transition zone. SHH stimulation further triggers the ciliary exit of GPR161 and ciliary entry of GPR175, which inhibits the production of cAMP. Both GLI-R and GLI-A translocate from the cilium into the nucleus to repress and induce transcriptional activation of HH target genes, respectively. Abbreviations: PI(4,5)P2: phosphatidylinositol 4,5-bisphosphate, Ub: ubiquitination.

In addition to canonical pathways, HH signalling can occur though so-called non-canonical pathways, which involve either GLI-independent mechanisms or SMO-independent regulation of GLI activity 106, 107. For example, the IFT-B complex protein, IFT80, can repress SHH-mediated noncanonical activation of the GTPase RHOA in differentiating mouse osteoblasts 108, and HH-stimulated chemotaxis is mediated by SMO localized outside of cilia 109. However, little is still known about the role of primary cilia in regulating non-canonical HH pathways and whether such pathways act in parallel to canonical HH signaling to control cellular processes during development and in tissue homeostasis 106, 108, 110. Below we summarize current knowledge regarding the organization of canonical HH signalling in primary cilia.

Activation of canonical HH signalling

The mechanism underlying either the repression of SMO by PTCH1 or the activation of SMO upon removal of PTCH1 from cilia is not well understood. However, the solved structures of PTCH1 bound or unbound to SHH 111, 112 and SMO bound or unbound to cholesterol derivatives 113 reveal important insights. First, PTCH1 shows similarity to members of the resistance-nodulation-cell division (RND) family of bacterial efflux transporters and to sterol sensing domains in the cholesterol trafficking protein, Niemann-Pick C1 (NPC1). The transmembrane segments 2–6 in PTCH1 constitute the sterol-sensing domain, and are sufficient to accommodate cholesterol derivatives 111, 112. The two extracellular domains of PTCH1 interact with SHH, although the two structural studies that investigated SHH binding identified different interacting interfaces with native lipidated SHH and an SHH N-fragment 111, 112. Interestingly, the extracellular domains also have an interface that accommodates cholesterol derivatives, and binding of PTCH1 to the N-fragment of SHH potentially limits access to or exit of these derivatives from this site. Binding of PTCH1 to the N-fragment of SHH is enhanced by cholesterol derivatives and it is predicted that binding of cholesterol to the extracellular domain interface might arise from cholesterol transport by PTCH1 111.

Inactive SMO is stabilized by a π-cation lock in the inner transmembrane interface. Binding of cholesterol to the extracellular cysteine rich domain of SMO dramatically changes the conformation of this domain with respect to the transmembrane domains, leading to release of the π-cation lock. Interestingly, a longitudinal tunnel capable of accommodating cholesterol was identified in the transmembrane region of active SMO, suggesting that SMO itself might move cholesterol from the plasma membrane to the extracellular domain 113. Oxysterols can also bind to the cysteine rich domain and activate SMO downstream and independent of PTCH1 removal from cilia 102, 114; however, the physiological role of oxysterols in HH pathway activation is not clear.

These findings suggest that SHH-mediated activation and removal of PTCH1 from cilia might increase the availability of endogenous SMO ligands — most likely cholesterol or cholesterol derivatives — in the ciliary membrane, thereby leading to activation of SMO in cilia. Downstream factors that mediate SMO-dependent activation of GLI2 are varied, and include G proteins and other proteins such as the EVC2 complex 104, 115. Furthermore, the 7-transmembrane receptor GPR175 (also known as TPRA1 or TPRA40) is enriched in cilia in the presence of SHH, and might regulate maximal activation of pathways downstream of SMO by coupling to the inhibitory Gα protein, Gαi, leading to decreased cAMP levels and inhibiting PKA-mediated formation of GLI3R 116. The intermediate steps between SMO activation, GLIA formation and translocation to the nucleus are not well understood, although GLI2 and GLI3 proteins accumulate in cilia tips upon HH pathway activation, suggesting that GLIA formation is regulated in the vicinity of cilia 117.

Basal Repression of canonical HH signalling

In the absence of SHH, PKA-mediated phosphorylation of GLI3 primes sequential phosphorylation events by casein kinase 1 (CK1) and glycogen synthase kinase-3 beta (GSK3β), which results in binding of GLI3 to the SCFβTrCP ubiquitin ligase, and its subsequent proteolysis into GLI3R 118121. The cilia-localized class A orphan GPCR, GPR161, was identified as a negative regulator of SHH signalling during early neural tube development in mice 122. GPR161 regulates formation of GLI3R possibly via constitutive activation of cAMP–PKA signalling. Another negative regulator of SHH signalling, Suppressor of FUSED (SUFU) restrains GLI3 in the cytoplasm and promotes GLI3R processing 123 in a cilia-independent step 124. Importantly, the absence of GPR161, PKA, and SUFU induces high levels of SHH signalling during mouse neural tube development 122, 125, 126, similar to the effect of Ptch1 deletion, which induces activation of SMO signalling 127. Interestingly, mutations in the genes encoding TULP3 or IFT-A subunits phenocopy Gpr161 mutants by increasing HH signalling in the caudal neural tube 58, 128131. The pre-ciliary function of the IFT-A core complex, together with phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), in binding and ciliary trafficking of TULP3 — an adapter protein involved in the gating of ciliary GPCRs including GPR161 — explains high SHH signalling observed in IFT-A mutants, despite the presence of abnormal cilia in these models 56, 64, 122. Thus, IFT-A-regulated trafficking of TULP3 and GPR161 regulates basal suppression of SHH signalling. However, the neural tube ventralization phenotypes of Tulp3 and IFT-A mutants, and Gpr161 mutants are weaker than those resulting from mutations in downstream negative regulators of HH signalling such as Sufu and PKA 132, suggesting the existence of additional inputs into PKA activation other than GPR161 133.

Studies from the past few years suggest that active suppression of HH pathway is as important as activation of the pathway during development and in maintenance of tissue homeostasis. Although SHH secretion from distinct regions, such as the purkinje neurons in the postnatal cerebellum and the zone of polarizing activity in limb buds, patterns these tissues, each of these tissues expresses low or no SHH at distinct stages of development. The importance of HH pathway repression is illustrated by the finding that premature SHH signalling resulting from disruption of GPR161 causes defects in limb and skeletal morphogenesis 134, cerebellar granule cell hyper-proliferation and formation of SHH-subtype medulloblastoma 135. Thus, active repression of HH pathway by GPR161 regulates tissue architecture, whereas pathway de-repression contributes to disease pathogenesis.

[H2] Ciliary coordination of HH signalling

Most components of canonical HH signalling are dynamically associated with cilia. In addition to PTCH1 removal, SMO enrichment and association of GLI2, GLI3 and SUFU with ciliary tips, the microtubule-associated atypical kinesin, KIF7, also becomes enriched in ciliary tips during activation of HH signalling 136, 137, where it. functions as both a positive and negative regulator of SHH signalling by modifying ciliary architecture 136, 137. GPR161 is also removed from primary cilia in a SMO-dependent and β-arrestin-dependent manner following activation of HH signalling 28, 138. β-arrestins are adaptor proteins that are recruited to the proximal C-terminus of GPR161 in a manner dependent on G protein-coupled receptor kinase 2 (GRK2)138. GRK2 and GRK3 also transduce high level SHH signals through mechanisms independent of GPR161133. Depletion of Inpp5e (which encodes a phosphoinositide 5-phosphatase responsible for removing the 5-phosphate from PI(4,5)P2), leads to accumulation of PI(4,5)P2 in cilia and increased steady-state levels of TULP3, IFT-A and TULP3-dependent cargo such as GPR161 and PC2 139, 140. In addition, removal of GPR161 from cilia upon activation of the SHH pathway is impaired in Inpp5e-knockout cells 139, 140, irrespective of levels of the pathway activator SMO suggesting that a number of factors are responsible for the dynamic regulation of HH signalling components in cilia.

Activation of PKA in different subcellular regions is mediated by local cAMP production and anchoring of PKA to A-kinase anchoring proteins (AKAPs) 141. The distal C-tail of GPR161 has a conserved amphipathic helix that directly binds to type I PKA regulatory subunits 142 (Figure 2). Since the type I PKA regulatory subunit alpha localizes to cilia 143, direct coupling of this subunit to PKA in this compartment might enhance the ability of GPR161 to activate PKA via constitutive cAMP signalling. nine adenylyl cyclases that regulate downstream cAMP signalling, at least three — ADCY3, ADCY5, and ADCY6 — are localized to cilia 143145. In particular, overexpression of ADCYC5 and ADCYC6 partially represses the HH pathway in the developing chicken neural tube 146. However, factors that regulate trafficking of adenylyl cyclases to cilia are currently unknown, and the role of such factors in SHH signalling has not been established.

Ciliary modulation of GPCR signalling

The cilium is recognized as an important nexus for GPCR signalling. GPCRs comprise the largest signalling receptor superfamily in the human genome, with more than 800 functional GPCRs 147, which can be subdivided into six different classes: class A (the Rhodopsin family), class B1 (the Secretin family), class B2 (the Adhesion family), class C (the Glutamate family), class F (the frizzled–smoothened family) and the Taste 2 family, based on sequence and phylogenetic analysis 147, 148. The vast majority of GPCRs fall into the class A rhodopsin family, which comprises approximately 700 receptors, including 460 olfactory receptors 147. GPCRs mediate numerous physiological functions in the human body by responding to a wide range of signals, such as photons, peptides, proteins, hormones, chemicals, lipids and sugars. GPCRs are also the largest group of therapeutic drug targets; about one-third of all US Food and Drug Administration (FDA) approved drugs act on GPCRs 149. Thus, understanding the molecular mechanism of GPCR signalling has great therapeutic importance.

Canonical signal transduction through GPCRs is mediated by activation of heterotrimeric G proteins composed of three associated subunits, Gα, Gβ, and Gγ. G proteins are classified on the basis of their Gα subunit and at least 20 Gα isotypes exist that are functionally categorized into four major families (Gαs, Gαi, Gαq, and Gα12) 150. Signalling through these proteins involves a very well characterized sequence of events. When inactive, a heterotrimeric G protein consists of a GDP-bound Gα subunit associated with a Gβγ dimer. Activated GPCRs engage the GDP-bound heterotrimer and facilitate GDP dissociation from Gα, which is rapidly followed by GTP binding to the G protein. Consequently, the Gα and Gβγ subunits dissociate and modulate the activity of downstream effectors (such as adenylyl cyclases by Gα and potassium channels by Gβγ). G protein activity is terminated when the Gα subunit hydrolyses GTP to GDP and re-associates with Gβγ.

Upon activation, GPCRs are phosphorylated at specific sites within their intracellular domains primarily by GRKs. Phosphorylated receptors are targets for the recruitment and binding of β-arrestins, which are scaffolding proteins that inhibit additional G protein activation and promote internalization of receptors by facilitating clathrin-mediated endocytosis. However, in some cases endocytosis leads to sustained or enhanced G protein signalling from endosomes 151. Moreover, β-arrestins bind to various signalling proteins, such as c-SRC and ERK1/2, and can promote G protein-independent signalling both at the plasma membrane and within endosomes 151.

Ciliary GPCR signalling in diverse cell types

Numerous GPCRs and their downstream effector molecules localize to cilia on a variety of mammalian cell types 152, 153 (Table 1, Figure 3). Intrahepatic bile ducts are lined with ciliated epithelial cells called cholangiocytes, which mediate bile acid transport and bicarbonate secretion 154. Primary cilia on these cholangiocytes have been proposed to provide mechanosensory, chemosensory and osmosensory functions to regulate cholangiocyte proliferation 155. Indeed, polycystic liver disease is a ciliopathy, characterized by the formation of fluid-filled hepatic cysts that originate from cholangiocytes 156. Reports suggest cholangiocyte cilia are enriched for two different GPCRs — purinergic receptor P2Y12 (P2RY12) 155 and G-protein coupled bile acid receptor 1 (GPBAR1, also known as TGR5) 157, 158, as well as the cAMP signalling proteins adenylyl cyclase, PKA, exchange protein directly activated by cAMP isoform 2 (EPAC2), and A-kinase anchoring protein (AKAP150) 155, suggesting that cholangiocyte cilia mediate cAMP signalling in response to biliary factors. In support of this proposal, activation of Gαi-coupled P2RY12 by its endogenous ligand, ADP, decreased forskolin-induced cAMP levels in ciliated cholangiocytes but not in non-ciliated cholangiocytes 155. The presence or absence of cilia might also affect GPBAR1 signalling 158. Activation of GPBAR1 on non-ciliated cholangiocytes induced co-localization of GPBAR1 with Gαs, increased cAMP signalling, inhibited ERK signalling, and increased cellular proliferation 155, whereas activation of GPBAR1 on ciliated cells induced co-localization of GPBAR1 with Gαi, decreased cAMP signalling, activated ERK signalling, and decreased cellular proliferation. These findings therefore suggest that GPCR-mediated cAMP signalling in cholangiocyte cilia is distinct from cAMP signalling on the plasma membrane of cholangiocytes.

Table 1:

Non-olfactory and non-visual class A and B G-protein-coupled receptors that localize to primary cilia

GPCR Cell type Reference(s)
β2-adrenergic receptor (β2AR) Neurons 274
Bile acid receptor 1 (GPBAR1 or TGR5) Cholangiocytes 158, 295
Dopamine receptor 1 (D1) Neurons 168
Dopamine receptor 5 (D5) Vascular endothelial cells, renal epithelial cells 160, 296
Galanin receptor 3 (GALR3) Neurons 74
GPR19 Neurons, Glial cells 64
GPR83 Neurons 74
GPR161 Neurons, mouse embryonic fibroblasts 122
GPR175 Mouse fibroblasts 297
Kisspeptin receptor 1 (KISS1R) Neurons 298
Melanin-concentrating hormone receptor 1 (MCHR1)
Melanocortin 4 receptor (MC4R)
Neurons
Neurons
299
180, 299
Muscarinic acetylcholine receptor 3 (M3R) Olfactory sensory neurons 300
Neuropeptide Y receptor 2 (NPY2R) Neurons 74
Neuropeptide Y receptor 5 (NPY5R) Neurons 74
Parathyroid hormone receptor 1 (PTH1R) Nucleus pulposus cells 301
Prokineticin receptor 1 (PROKR1) Human trophoblast cells, human placental tissue 164
Prolactin-releasing hormone receptor (PRLHR) Glial cells 302
Prostaglandin E receptor 4 (EP4) Human retinal pigment epithelial cells 303
Purinergic receptor P2Y12 (P2RY12) Cholangiocytes 155
Pyroglutamylated RFamide peptide receptor (QRFPR) Neurons 74
Serotonin receptor 6 (HTR6) Neurons 304
Somatostatin receptor 3 (SSTR3) Neurons 305
Trace amine-associated receptor 1 (TAAR1) Thyroid epithelial cells 306
Vasopressin receptor 2 (V2R) Renal epithelial cells 162

Figure 3. Overview of ciliary GPCR signalling.

Figure 3.

Binding of agonists to various G-protein-coupled receptors (GPCRs) expressed on cilia modulates a variety of signalling pathways on different cell types. For example, binding of endocrine gland-derived vascular endothelial growth factor to prokineticin receptor 1 (PROKR1) on trophoblast cell cilia activates mitogen-activated protein kinase kinase 1/2 (ERK1/2) at the ciliary base, presumably through a Gαq-mediated increase in calcium levels. Dopamine binding to dopamine receptor 5 (D5) on renal epithelial cell cilia facilitates Gβγ subunit dissociation and activation of an L-type calcium channel, resulting in an increase in ciliary calcium levels. ADP binding to the purinergic receptor P2Y12 (P2RY12) on cholangiocyte cilia stimulates Gαi, which inhibits adenylyl cyclase activity and subsequently reduces cAMP levels. Vasopressin binding to the type 2 vasopressin receptor (V2R) on renal epithelial cell cilia results in activation of adenylyl cyclase, increased cAMP levels, activation of PKA, and stimulation of a cation selective channel on the ciliary membrane. Increased cAMP levels and activated PKA can also activate exchange protein directly activated by cAMP (EPAC) and cAMP response element binding protein (CREB) in the cilium, respectively. Somatostatin binding to somatostatin receptor subtype 3 (SSTR3) on neuronal cilia stimulates β-arrestin (ARRB2) recruitment into the cilium where it mediates SSTR3 ciliary export in cooperation with the BBSome and intraflagellar transport (IFT) complex. Neuropeptide Y (NPY) binding to neuropeptide Y receptor subtype 2 (NPY2R) on neuronal cilia causes accumulation of the receptor at the ciliary tip and release through ectosomes.

Ciliary GPCR signalling in nephrons

A clear link also exists between cilia dysfunction and cystic kidney disease 159. Studies have further implicated ciliary GPCR signalling in renal cilia function and disease. Dopamine receptor type 5 (D5) localizes to cilia on renal epithelial cells, where it may functionally couple to the CaV1.2 L-type calcium channel 160. Activation of D5 caused an increase in calcium levels in the cilium along with an actin-mediated increase in cilia length, which might increase sensitivity to fluid flow 161 and inhibit cystogenesis. Type 2 vasopressin receptor (V2R) which regulates Na+ and water reabsorption in the mammalian nephron has also been reported to localize to cilia on renal epithelial cells 162. Upon activation, ciliary V2R seems to activate adenylyl cyclase, which increases local cAMP concentrations and activates a cation-selective channel to regulate intraciliary Ca+2 signals 162. Although the precise consequences of V2R ciliary signalling on renal epithelial cell function are unknown, the V2R receptor antagonist tolvaptan slows the decline in glomerular filtration rate in patients with ADPKD163. Tolvaptan is now the first FDA-approved treatment for PKD.

Ciliary GPCR signalling in trophoblasts

GPCR signalling in cilia also seems to have an important role in early human pregnancy. During embryo implantation, trophoblasts, which form the outer layer of a blastocyst, invade the uterus in order to secure an adequate supply of oxygen and nutrients for the foetus. Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is a critical regulator of embryo implantation and placental development. Interestingly, the EG-VEGF receptor, prokineticin receptor 1 (PROKR1) localizes to cilia on human trophoblast cell lines and human first-trimester placental tissue 164. Treatment of trophoblast cells with EG-VEGF activates ERK1/2 signalling to induce upregulation of matrix metalloproteinases (MMPs) and facilitate cell invasion 164. Disruption of cilia on trophoblast cells ameliorates EG-VEGF-induced activation of ERK1/2, MMP upregulatoin, and cell invasion 164. Thus, ciliary-mediated PROKR1 signalling might have an important role in embryo implantation.

Ciliary GPCR signalling on central neurons

Most neurons in the mammalian brain possess primary cilia. The importance of neuronal cilia is highlighted by the fact that ciliopathies are associated with numerous neuropathologies, including anatomical abnormalities and neuropsychiatric disorders 165. Neuronal cilia are enriched for certain GPCRs and downstream effector proteins 152, 166, 167, suggesting they act as specialized signalling hubs. The importance of GPCR ciliary localization is underscored by the fact that mouse models of the ciliopathy BBS show dysregulation of GPCR ciliary localization. Specifically, deletion of BBS proteins leads to failure of somatostatin receptor subtype 3 (SSTR3), melanin-concentrating hormone receptor 1 and neuropeptide Y (NPY) receptor subtype 2 (NPY2R) to localize to neuronal cilia, whereas, dopamine receptor 1 accumulates in neuronal cilia 73, 74, 168. In addition, tubby mice, which carry a mutation in the gene encoding the TUB protein and display ciliopathy phenotypes, exhibit defective localization of a number of GPCRs in neuronal cilia 74, 169. Thus, defects in localization of GPCRs to neuronal cilia likely disrupt ciliary signalling and contribute to ciliopathy phenotypes.

Numerous studies have implicated ciliary GPCR signalling in both neural development and function. Inhibitory interneurons originate in the telencephalon from where they migrate to numerous brain regions and integrate into local neural circuits to regulate the balance of excitatory and inhibitory inputs. Disruption of interneuronal circuits has been linked to neurodevelopmental disorders such as schizophrenia, autism and intellectual disabilities 170. Interestingly, conditional disruption of ARL13B, which is a GTPase required for proper ciliary signalling, in post-migratory interneurons perturbs circuit development in the mouse striatum 171. Specifically, loss of ARL13B in interneurons results in reduced dendritic and axonal complexity, disrupted synaptic connectivity, and functional deficits in synaptic activity 171. Interneurons lacking ARL13B also show disrupted ciliary calcium dynamics and a dramatic reduction in ciliary localization of SSTR3 171. Intriguingly, expression of SSTR3 in ARL13B-deficient interneuronal cilia rescues the morphological and synaptic connectivity defects, whereas expression of a non-ciliary form of ARL13B with normal GTPase activity does not rescue the developmental defects in ARL13B-deficient interneurons 171, suggesting that SSTR3 signalling within interneuronal cilia is critical for proper inhibitory network construction and function.

Ciliopathies are also associated with cognitive deficits. SSTR3 localizes to neuronal cilia in the hippocampus, a region of the brain that is important for learning and memory. Treatment of mouse hippocampal neurons with the SSTR3 ligand, somatostatin, stimulates recruitment of endogenous β-arrestin into SSTR3-positive cilia, leading to a rapid β-arrestin-2-dependent decrease in the ciliary SSTR3 167, suggesting that ciliary export of activated SSTR3 is mediated by β-arrestin-2. In support of this finding, ciliary export of activated SSTR3 in cultured renal epithelial cells also requires β-arrestin-2 28, 172. Interestingly, mice lacking SSTR3, ADCY3, β-arrestin-2 or cilia in the hippocampus all show similar deficits in learning and memory 173176, implicating SSTR3 ciliary signalling in proper learning and memory.

GPCR signalling in neuronal cilia also contributes to the regulation of metabolic homeostasis. The first hint that cilia affect food intake and energy metabolism arose from the observation that some ciliopathies are associated with obesity. This connection was further supported by studies showing that conditional disruption of cilia in adult mice causes hyperphagia and obesity 177, 178. More recent studies have provided important mechanistic insights into the metabolic neural circuits that are affected by GPCR ciliary signalling. The Gαs-coupled GPCR melanocortin 4 receptor (MC4R) is an important component of the neurocircuitry that regulates food intake and energy expenditure. In fact, mutations in MC4R are the most common cause of monogenic obesity in humans, underlying up to 6% of early-onset or severe cases of adult obesity 179. A 2018 study showed that MC4R localizes to cilia on neurons in the mouse paraventricular nucleus of the hypothalamus — a region of the brain that is important for the regulation of energy homeostasis and metabolism 180. Interestingly, some MC4R mutations associated with obesity in humans impair ciliary localization of MC4R. Moreover, inhibition of ciliary cAMP signalling specifically in cilia on MC4R-expressing neurons of the paraventricular nucleus led to increased food intake and weight gain in mice 180. Thus, impaired ciliary signalling of MC4R could be a common cause of syndromic and non-syndromic obesity in humans.

NPY also has an important role in the central regulation of food intake and energy expenditure 181. NPY2R localizes to cilia on hypothalamic neurons in mice 74. Importantly, BBS mutant mice that lack NPY2R-positive cilia are obese and do not respond to NPY2R ligand 74, suggesting that receptor ciliary localization is required for ligand-dependent signalling in vivo. Moreover, quantification of NPY2R-mediated cAMP signalling in a ciliated cell line revealed that ligand treatment produced a stronger signal in cells with a cilium than in non-ciliated cells 74. Thus, NPY2R signalling is seemingly enhanced within the cilium. Similar to SSTR3, ligand treatment results in a decrease in NPY2R ciliary localization. NPY2R binds poorly to β-arrestin 182; however, using live-cell imaging in a ciliated renal epithelial cell line, Nager et al 28 revealed that activated NPY2R accumulates at the ciliary tip and is released in extracellular vesicles called ectosomes 28 (Figure 3), raising the fascinating possibility that ciliary GPCR signals might be transmitted between cells. Interestingly, Nager et al also showed that agonist-dependent ciliary export of heterologously-expressed SSTR3 in inner medullary collecting duct cells lacking either BBSome function or β-arrestin-2, involved active recruitment of SSTR3 to the ciliary tip from where a considerable fraction was released in ectosomes 28. These findings further support a role for the BBSome and β-arrestin-2 in GPCR ciliary export and led to the suggestion that loss of GPCR localization on neuronal cilia in BBS mutant mice 73 might result from constitutive ectocytosis, rather than a defect in GPCR trafficking to cilia. However, this proposal would suggest that hippocampal neurons that lack β-arrestin-2 would also lack ciliary localization of SSTR3, yet ciliary localization of SSTR3 is not affected in β-arrestin-2-deficient hippocampal neurons167. Why SSTR3 would be constitutively ectocytosed from cilia on BBS mutant neurons, whereas D1 accumulates in cilia on BBS mutant neurons even in the presence of agonist, is unclear 168. Additional studies are therefore required to define the precise roles of the BBSome in GPCR trafficking to and from neuronal cilia.

Ciliary modulation of WNT signalling

WNT signalling comprises an evolutionarily-conserved network of pathways that coordinate a multitude of cellular events over a lifespan 183. WNT ligands comprise a family of secreted lipoproteins that often activate frizzled receptors of the class F GPCRs in conjunction with a series of co-receptors 184, 185. Two prominent branches of the WNT signalling network are the so-called canonical WNT–β-catenin and non-canonical WNT–planar cell polarity (PCP) pathways. In the canonical pathway, WNT ligands bind to frizzled to trigger complex formation with the co-receptor LRP5/6 and Dishevelled (DVL), promoting stabilization of cytoplasmic β‐catenin, which enters the nucleus to regulate target gene expression with effects on cell proliferation, differentiation and survival 186, 187. By contrast, WNT–PCP signalling regulates cell morphology, migration and oriented cell division and relies on a multitude of receptor combinations and downstream signalling events 188, 189. For example, WNT ligands can bind frizzled and members of the RTK family, specifically ROR1/2, RYK or PTK7, which recruit DVL and activate the branches of RAC–JNK and DAAM-1–RHOA–ROCK signalling pathways that control polarity processes while inhibiting canonical WNT–β-catenin signalling 188, 190192.

Controversies in ciliary WNT signalling

Several core WNT pathway components localize to primary cilia (Figure 4) 193197 and although multiple lines of evidence from gene knockout or knockdown studies have suggested roles for cilia in regulating WNT signalling, the literature in this field is controversial. The first links between WNT signalling and primary cilia came from functional studies of nephrocystin-2 (NPHP2, also known as inversin, which when overexpressed inhibits DVL1-mediated activation of WNT reporter constructs in mammalian cells and rescues secondary axis formation induced by overexpression of Dsh or CK1μ in Xenopus laevis 198. In addition, morpholino-induced depletion of NPHP2 in X. laevis and zebrafish disrupted convergent extension — a process that was rescued by overexpression of mouse NPHP2 or Diversin, respectively 198. These findings led to the hypothesis that the cilium acts as a switch for diverse WNT pathways by modulating DVL. A subsequent study in X. laevis showed that NPHP2 promotes WNT–PCP signalling by facilitating fzd8-mediated recruitment of Dvl to the membrane, and that proximal pronephros extension is disrupted by morpholino-mediated depletion of NPHP2 199, although normal WNT–β-catenin signalling was unaffected in this study. In vitro, NPHP2 controlled fibroblast polarity and directional cell motility through modulation of ciliary WNT signalling pathways that control the activity and localization of polarity proteins to the leading edge of migrating cells 196. In support of a role for primary cilia in canonical WNT signalling, mouse embryos deficient in the ciliary microtubule-based kinesin-like protein KIF3A, which is a component of the kinesin-2 motor that facilitates anterograde IFT and ciliogenesis, displayed increased levels of canonical WNT reporter activity compared to levels in heterozygous embryos, and loss of the primary cilium in mouse embryonic fibroblasts lacking key ciliogenesis genes (Kif3A, Ift88- or Ofd1)was associated with hypersensitivity to a WNT3A ligand 193. These findings support the proposal that the primary cilium restrains canonical WNT signalling; however, another study showed that mouse embryos and mouse embryonic fibroblasts with mutations in Kif3a, Ift88, Ift72 and a gene that encodes a component of dynein-2 (Dync2h1) exhibited normal Axin2 mRNA expression and canonical WNT reporter activity 200. A 2016 study contradicted both these reports showing that in lung cancer cell lines, KIF3A restricts WNT canonical signalling, but independently of the primary cilium by restricting β-arrestin interaction with DVL2 and AXIN as well by modulating WNT ligand secretion 201. Conversely, the Drosophila homologue of KIF3A, Klp64D, promotes canonical WNT signalling in the non-ciliated Drosophila wing discs, forms a complex with Arm (the Drosophila homologue of β-catenin) and Dvl, and probably mediates correct trafficking of Arm during WNT signalling 202. Finally, a further study showed that a zebrafish maternal-zygotic mutant of ift88 has normal expression of canonical WNT target genes and normal convergent extension and body axis development, demonstrating that primary cilia are redundant for both branches of WNT signalling 203. Thus, despite intense investigation, the function of the primary cilium in fine-tuning WNT signalling remains unclear 204.

Figure 4.

Figure 4.

Overview of ciliary transition zone and basal body modulation of WNT signalling. The ciliary transition zone (TZ) is composed of several interacting protein modules or complexes, including the nephronophtisis (NPHP) and Meckel-Gruber Syndrome (MKS) modules that establish ciliary gating and contribute to the regulating of WNT signalling. The NPHP module protein retinitis pigmentosa GTPase regulator-interacting protein 1-like (RPGRIP1L) inhibits canonical WNT signalling through its interacting with a component of the 19S proteasome subunit, PSMD2, thereby promoting ubiquitin (Ub)-mediated proteasomal degradation of β-catenin and DVL. Further, β-catenin is ubiquitinated by the E3 ubiquitin ligase JADE1, which is present in the TZ as well as at the basal body. In the MKS module, transmemembrane protein 67 (TMEM67) recruits receptor tyrosine kinase-like orphan receptor 2 (ROR2) to the TZ to form a receptor complex that binds WNT5A, which is a ligand for non-canonical WNT signalling. In the presence of a cilium, the MKS module component Jouberin (JBN) recruits cytoplasmic β-catenin, which accumulates in response to WNT activation. In the absence of a cilium, JBN facilitates β-catenin nuclear entry. Several key components of the β-catenin destruction complex localize to the basal body, including the scaffold protein adenomatous polyposis coli (APC), GSK3-β glycogen synthase kinase 3 beta (GSK3-β) and casein kinases CK1-α, CK1-μ and CK1-δ 193195, 294. The main product of the destruction complex, phosphorylated β-catenin, is also concentrated at the basal body, where it undergoes degradation by the proteasome. Whether β-catenin is actively phosphorylated and ubiquitinated within the cilium, or whether the modified protein is recruited to the basal body, is unknown. Frizzled receptors can accumulate in the primary cilia but the functional relevance of this localization is unknown. RHOA: Ras homolog gene family member A.

WNT signalling: the TZ and the basal body

Dvl, β-catenin and several members of the β-catenin destruction complex have been shown to localize to the ciliary base and available evidence suggests that they can be modulated by components of the MKS and NPHP modules, which are protein complexes of the TZ, which forms a prominent part of the ciliary gate that regulate proteins entering and exiting the cilium (PMID: 27770015). For example, in the absence of a primary cilium, the MKS module component Jouberin (JBN, also known as AHI1) promoted WNT and β-catenin signalling by facilitating nuclear localization of β-catenin 205. The primary cilium induces depletion of nuclear and cytoplasmic JBN and thereby represses the effect of JBN on β-catenin. Ciliary JBN facilitates the ciliary localization of β-catenin following WNT3A stimulation and thereby negatively affects WNT signalling 206. JBN therefore has a dual function in regulating WNT–β-catenin signalling, depending on the presence or absence of the primary cilium. Another study showed that the cystic kidney and abnormal brain development phenotypes of mice deficient in JBN are largely caused by dysregulated WNT–β-catenin signalling 207. The transmembrane protein TMEM67 is another key component of the MKS module that recruits the tyrosine-protein kinase transmembrane receptor ROR2 to the TZ. Together TMEM67 and ROR2 respond to WNT5A to activate RHOA, modulate the actin cytoskeleton and promote epithelial branching morphogenesis; these proteins also partially facilitate WNT5A-mediated repression of the canonical pathway 208. In addition to MKS module components and NPHP2 (discussed above), evidence suggests that other NPHP module components including NPHP3 209, NPHP4 210 and RPGRIP1L (PMID 22927466, 212) modulate the WNT pathway through DVL. Similarly to NPHP2, NPHP3 and NPHP4 restrain DVL-mediated WNT–β-catenin activation in cell culture, and when depleted, induce WNT–PCP defects in vivo (PMID 18371931 and PMID 21498478). In addition, NPHP4 was reported to stabilize the E3 PHD finger domain E3 ubiquitin ligase JADE1 and promote its nuclear translocation 213. JADE1 is a negative regulator of WNT–β-catenin signalling that ubiquitinates cytoplasmic and nuclear β-catenin irrespective of its phosphorylation status, thereby promoting its proteasomal degradation 214. In ciliated cells, JADE1 is also localized at the TZ and the basal body, probably through its interaction with NPHP4. Thus, NPHP4, in addition to its negative effect on DVL, also negatively regulates β-catenin levels through JADE1. Similarly, RPGRIP1L regulates WNT–PCP pathways by modulating DVL levels 212, 215. RPGRIP1L was reported to regulate the proteasome function in a cilia and basal body-dependent manner 211. Mouse embryonic fibroblasts deficient in RPGRIP1L exhibit impaired proteasomal activity at the basal body — a phenotype that can also be induced by depletion of the 19S proteasome subunits PSMD2, PSMD3 and PSMD4, all of which localize at the TZ and basal body. Indeed a key manner by which cilia might regulate WNT pathway components is through direct proteasomal degradation 212, 216, 217 (PMID: 24691443). Cilia proteomics have identified several proteasomal subunits that bind to BBSome subunits 218, and depletion of BBS4 in cultured cells (which results in both canonical and WNT–PCP defects in zebrafish) reduces proteasomal activity and leads to an accumulation of cytoplasmic and nuclear β-catenin 216.

Thus, available data suggest that various proteins in the TZ and basal body might act as a processing platform to which various WNT signalling components are recruited for post-translational modification and/or elimination through the basal body-associated proteasome pathway. However, it is important to remember that many TZ proteins may have extra-ciliary functions 219 and further work is needed to decipher and distinguish their ciliary and non-ciliary functions and the context in which they regulate the WNT pathways.

Ciliary RTK signalling

Many growth factors and hormones operate through RTKs, which belong the largest family of >50 enzyme-linked receptors. RTKs can be subdivided into different classes based on their structure, domain organization and requirement for co-receptors in signal transduction 220. In many cases, RTKs are activated by homodimerization or hetero-dimerization, which leads to activation of their intracellular tyrosine kinase domains followed by autophosphorylation of specific tyrosine residues, which in a sequence-specific context recruit a multitude of adaptor and effector proteins involved in signal transduction primarily through Src homology 2 (SH2) and pTyr-binding (PTB) domains. Prominent RTK downstream signalling components and pathways include ERK1/2, p38 and JNK in the family of mitogen-activated protein (MAP) kinases, PI3K-AKT-mTOR and PLCγ as well as a branch of STAT signalling 220. In addition, RTKs extensively cross-talk with other receptor systems and may signal through G proteins, GRKs, and β-arrestins to control various cellular responses 221. Designated subtypes of class II and class III RTKs, including platelet-derived growth factor alpha receptor (PDFGRα), the insulin receptor (IR) and insulin-like growth factor receptor (IGFIR) operate in primary cilia to control specific processes in cells and tissues 222, 223 (Figure 5). A series of other RTKs, including epidermal growth factor receptor (EGFR) 224, 225, fibroblast growth factor receptor 3 (FGFR3) 226, tropomyosin receptor kinase B (TRKB, also known as neurotrophic tyrosine kinase receptor 2 227 and angiopoetin-1 receptor (TIE2) 228 also localize to primary cilia. Stimulation of cultured retinal pigmented epithelial (RPE-1) cells with brain-derived neurotrophic factor (BDNF) — a secreted neurotrophin that is a ligand for TRKB and required for neuronal development and synaptic plasticity — induced recruitment and activation of TRKB in the primary cilium in a BBS4-dependent manner, thereby linking ciliary TRKB signalling to neuronal phenotypes associated with BBS 227. FGFR3 regulates ciliary length and IFT20 trafficking in the cilium, and ciliary expression of constitutively active FRFR3 in chondrocytes is linked to skeletal disorders 226, 229. Finally, RTKs can localize to specified sub-compartments of the primary cilium. For example, ROR, which as described above, might function as a co-receptor for WNT5A-regulated PCP signalling at the ciliary TZ. However, many of these receptors can also localize outside the primary cilium, indicating that cilia could constitute a specialized site for organizing cellular signalling events that might affect cellular processes differently from those organized at extra-ciliary sites.

Figure 5. Overview of ciliary PDGFRα, insulin and IGF-1 signalling.

Figure 5.

Targeting of platelet-derived growth factor receptor alpha (PDGFRα) to the cilium relies on intraflagellar transport protein 20 (IFT20) in complex with the E3 ubiquitin ligases c-CBL and CBL-b; activation of the receptor in the cilium activates the MEK1/2–ERK1/2–RSK and PI3K–AKT pathways, which in turn control activation and translocation of the Na+/H+ exchanger 1 (NHE1) to leading edge of the cell for directional migration. AKT might also become activated at the ciliary base in complex with NPHP2 (also known as inversin). Feedback inhibition of PDGFRα signalling might be controlled by CBL-mediated ubiquitination (Ub) of the receptor in the cilium as well as by inositol 1,4,5-trisphosphate 5-phosphatase (INPP5E), which inhibits AKT signalling. PDGFRβ localizes at the plasma membrane to induce aurora kinase A (AURKA)-mediated disassembly of the primary cilium, which promotes cell cycle re-entry. Similarly, expression of the oncogenic mutant PDGFRα Asp842Val, which localizes to the Golgi, promotes ciliary disassembly and cell proliferation via activation of AURKA. Insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF-1R) also localize to primary cilia to balance the regulation of various cellular processes, including adipogenesis, neuronal differentiation, mitogenic signalling and insulin production in islet β-cells of the pancreas. Ciliary IGF-1R activation also induces ciliary resorption and cell cycle entry possibly via IGF-1-mediated recruitment of phosphorylated TCTEX-1 to the ciliary TZ via non-canonical G-protein signalling, PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; IRS-1: insulin receptor substrate 1; PLCγ, phospholipase C gamma; PIP2, Phosphatidylinositol 4,5-bisphosphate; IP3, Inositol trisphosphate; HSP90α, heat shock protein 90 alpha.

Ciliary IR and IGF-1R signalling

The family of insulin and insulin-like receptors have critical roles in glucose storage and uptake, protein and lipid synthesis, cell differentiation and mitogenic responses. A series of studies mostly based on cell culture experiments and mutant mouse models indicated links between many of these functions and IR or IGF-1R signalling at the level of the primary cilium. An important contribution to this understanding came with the discovery that although IGF-IR is localized and activated at the plasma membrane, adipocyte differentiation, which is associated with transient ciliogenesis 197, 230, requires activation of IGF-IR in the primary cilium in cultures of 3T3-L1 preadipocytes 230. Indeed, extra-ciliary receptor populations seemed to be less sensitive to insulin stimulation than those in the cilium, where receptor activation led to activation of insulin receptor substrate 1 (IRS-1) and AKT at the ciliary base, instigating the adipocyte differentiation programme 230. Likewise, differentiation of human mesenchymal stem cells into adipocytes was demonstrated to involve elongation of primary cilia, associated with recruitment of IGF-1Rβ to the cilia 231. Ciliary IGF-1R activation also induced ciliary resorption and cell cycle entry through various downstream signalling pathways, including via IGF-1-mediated recruitment of phosphorylated TCTEX-1 to the ciliary TZ via non-canonical G-protein signalling, marking a mitogenic signalling cascade that accelerates ciliary resorption and G1/S progression in cultured mouse embryonic fibroblasts and RPE-1 cells 232. Perturbation of this signalling cascade in cortical neuron progenitors induces premature neuronal differentiation at the expense of proliferation 232 – a scenario that is linked to developmental brain abnormalities 233. Similarly, insulin-mediated resorption of cilia in mouse 3T3-L1 fibroblasts operates primarily through activation of ciliary IGF-1R to which IRS-1 is recruited and activated, followed by re-localization of IRS-1 to the ciliary neck region, where heat shock protein Hsp90α might function as a hub for activation of AKT 234. Evidence also suggests that primary cilia in islet β-cells of the pancreas are implicated in insulin production and that dysfunctional cilia in these cells contribute to susceptibility to metabolic diseases such type 2 diabetes mellitus (T2DM) 235, which is present in a subset of ciliopathies such as BBS and Alström syndrome (reviewed elsewhere 236, 237). Young Bbs4-knockout mice demonstrate impaired glucose handling before the onset of obesity, and ciliary dysfunction induced by knockdown of BBS4 or oral-facial-digital syndrome 1 protein (OFD-1) attenuates first-phase insulin secretion in pancreatic islet cells independent of glucose metabolism 235. These defects in glucose handling are linked to defects in insulin-mediated activation of the PI3K–AKT pathway in β-cells, which relies on ciliary recruitment of activated IR subtype A (IR-A), which controls the production of insulin in these cells 235, 238. However, the mechanisms by which primary cilia regulate metabolic processes are very complex and probably involve many different signalling systems, which in a spatial-temporal manner regulate cellular processes in multiple tissues, including the hypothalamus as well as adipose and muscle tissues 236,237.

Ciliary PDGFRα signalling

Two isoforms of PDGF receptors exist (PDGFRα and PDGFRβ), which function as either homodimers or heterodimers to control diverse cellular and developmental processes. Mutations in the genes expressing PDGFRα or its specific ligand, PDGF-AA, are associated with kidney and liver pathophysiologies, gastrointestinal stromal tumours, glioblastoma and a variety of other cancers 239. PDGFRα is principally localized to primary cilia 225, 240243, although non-ciliary localizations for this isoform have been reported for specific cell types 243245. PDGFRβ is primarily located and activated outside the cilium 240, 241, but can induce AURKA-mediated disassembly of the primary cilium in cultured fibroblasts and RPE-1 cells through activation of the PLCγ pathway upon stimulation with PDGF-DD 246. In fibroblasts, ciliary PDGFRα signalling activates the PI3K–AKT and MEK1/2–ERK1/2–RSK pathways to control directional cell migration, which is achieved by targeting the Na+/H+ exchanger 1, NHE1, to the leading edge of the migrating cells, which project their cilium towards the leading edge and parallel to the path of motility 240, 247249. Consequently, defects in the formation of primary cilia by depletion of IFT-B proteins such as IFT88 and IFT172 block PDGFRα activation 247, 250, rendering the cells unable to respond and move towards a gradient of PDGF-AA 247. In this context, AKT was shown to be activated at the ciliary base in complex with NPHP2 in a PDGF-AA-dependent manner 251; these interactions could provide a platform for cross-talk between PCP and PDGFRα signalling pathways in regulating directional cell migration, although further work will be required to understand the potential interaction between these two signalling systems at the level of the cilium. In addition, knockdown of the serine/threonine-protein kinase Ataxia telangiectasia and RAD3-related protein (ATR), which underlies Seckel syndrome 252, is associated with ciliary shortening and reduced responsiveness to PDGF-AA in cultures of fibroblasts 253, and depletion of the gene that encodes the ciliary TZ NPHP module component, RPGRIP1L, in mouse embryos leads to ventricular septal defects associated with loss of localization, which display cardiac ventricular septal defects 254 is associated with loss of localization of PDGFRα to primary cilia and reduced expression of the PDGFRα target gene, Hifα, in cardiac ventricular tissue 243. The latter observation supports the general concept that that dysfunctional cardiac cilia cause congenital heart defects 255.

Further studies have revealed diverse mechanisms that regulate ciliary targeting of PDGFRα and balance the output of PDGF-AA-mediated signalling. IFT20 appears to have a central role in this context by stabilizing the RING-finger domain family of CBL E3 ubiquitin ligases, c-CBL and CBL-b, which are tumour suppressor proteins that target active PDGFRs for internalization and degradation 256; dysfunctional CBL E3 ubiquitin ligases are associated with cancer development 257. Upon PDGF-AA stimulation CBL proteins are recruited to the primary cilium to mediate the ubiquitination and internalization of PDGFRα for feedback inhibition of signalling, whereas in IFT20-depleted cells, which lack the cilium, PDGFRα mislocalizes to the general cell surface, where PDGF-AA-mediated signalling is greatly overactivated due to autoubiquitination and proteasomal degradation of the CBL proteins 258. Similarly, co-depletion of c-CBL and CBL-b leads to reduced levels of IFT20 in combination with mislocalization and overactivation of PDGFRα 258. These results suggest the existence of a biochemical and functional relationship between IFT20 and CBL proteins in ciliary receptor sorting and modulation of PDGFRα signalling, which when defective underlies tumorigenic signalling in various tissues 239. Indeed, expression of the oncogenic mutant PDGFRα Asp842Val, which localizes to the Golgi 239 and is the most common PDGFRα mutation in gastrointestinal stromal tumours 259, promotes ciliary disassembly and cell proliferation by phenocoyping PDGF-DD-mediated signalling 246. Furthermore, rapamycin-mediated inhibition of mTOR signalling, which is upregulated in cells deficient in both c-CBL and CBL-b 260, rescues PDGF-AA–mediated signalling in IFT88 and IFT172 mutant fibroblasts that are devoid of primary cilia 250, although further studies are required to understand the relationship between aberrant mTOR signalling and mislocalization of PDGFRα in the context of ciliary signalling and tumorigenesis. Finally, INPP5E, was suggested to control ciliary PDGFRα signaling by inhibiting PDGF-mediated AKT activation261, 262, 263. These findings could indicate that INPP5E contributes to regulation of the balanced output of PDGF-AA-mediated AKT signalling at the cilium and that ciliopathies associated with mutations in INPP5E, such as Joubert syndrome, are partly linked to defects in this pathway.

TGFβ/BMP signalling in primary cilia

The transforming growth factor-β (TGFβ) superfamily comprises a highly pleiotropic group of ligands that signal via hetero-tetrameric receptor complexes of type I (RI) and type II (RII) serine/threonine kinases and a series of co-receptors to control a multitude of cellular processes during development and in the maintenance of tissue homeostasis in the adult 264, 265. In canonical signalling, transcription factors of the R-SMAD family, typically SMAD2/3 and SMAD1/5/8 in the TGFβ–activin–nodal or the bone morphogenetic protein (BMP) –müllerian inhibiting substance (MIS) branches of ligand signalling, respectively, are phosphorylated and activated by the receptors to form a complex with SMAD4, which translocates to the nucleus for targeted gene expression 265. By contrast, ligands of the growth and differentiation factor (GDF) subfamily can operate either through SMAD2/3 or SMAD1/5/8, whereas other ligand subtypes may antagonize R-SMAD signalling 265.

The TGFβ superfamily also operates through so-called non-canonical pathways, including NF-κB signalling, Rho-like GTPases and PI3K-AKT pathways as well as MAP kinases (for example, ERK1/2, p38 and JNK), which may cross-talk with R-SMAD signalling and/or become integrated into larger signalling networks that contribute to the diverse mechanisms by which ligand stimulation controls diverse complex cellular responses 265, 266. Finally, several studies have indicated a major role of receptor internalization in modulating the balanced output of TGFβ/BMP signalling, through clathrin-mediated and caveolin-mediated endocytosis. Clathrin-mediated endocytosis compartmentalizes the ligand–receptor complex in early endosomes, where SMAD anchor for receptor activation (SARA) binds to the PI(3)P-enriched membrane of the endosomes via its FYVE zinc finger domain to facilitate the association between RI and R-SMADs for robust SMAD2/3 signalling 265. However, the function of SARA in the context of R-SMAD activation, nuclear translocation and SMAD-dependent gene expression might not be equivalent in all cell types 267. Caveolin-mediated endocytosis has an inhibitory role in canonical TGFβ–BMP signalling through the proteasomal degradation of internalized receptors 265.

Consequences of TGFβ/BMP signalling

Increasing body of evidence indicates a prominent role for the primary cilium in co-organizing the balanced output of TGFβ/BMP signalling. The first report of active TGFβ signalling in the primary cilium was based on studies in cultured mouse and human fibroblasts in which TGFβ-RI and TGFβ-RII were shown to localize along and at the tip of the cilium 268. Upon ligand stimulation, the receptors accumulated at the ciliary base region, which is enriched in SMAD4, after which the receptors were internalized by clathrin-mediated endocytosis at the ciliary pocket to activate SMAD2/3 268 (Figure 6). Similarly, TGFβ signalling was associated with activation of ERK1/2 at the ciliary base, but in contrast to SMAD2/3, TGFβ1-induced phosphorylation of this MAP kinase seemed to occur independently of clathrin-mediated endocytosis 268. These findings indicate that the primary cilium uses diverse mechanisms to fine-tune the output of canonical and non-canonical TGFβ signalling to control varied cellular responses, which could be relevant for processes such as those related to heart development, where TGFβ-dependent differentiation of mouse cancer stem cells and human embryonic stem cells into cardiomyocytes is associated with the temporal accumulation of TGFβ receptors and activation of SMAD2/3 at the ciliary base 268. TGFβ and BMP receptors also localize to primary cilia in other cell types 241, 269273 to regulate mouse heart development in vivo 273, migration of human bone mesenchymal stem cells in bone remodelling assays 270 and osteogenic differentiation in vitro and bone formation in vivo 269, 271, 274. Primary cilia were also identified as major regulators of TGFβ-mediated activation of R-SMADs in the differentiation of human adipose progenitors into myofibroblasts 275. Further, endothelial primary cilia, which function as flow sensors in the vasculature and contribute to blood–brain barrier integrity 276279, counteract endothelial-to-mesenchymal transition in a process that is associated with reduced canonical TGFβ signalling in the endocardial cushion area 280, whereas endothelial primary cilia cooperate with BMP signalling to stabilize vessel connections in the developing mouse retina 281. More directly, shortening of primary cilia by TGFβ signalling 282284 is associated with epithelial-to-mesenchymal transition in mouse kidney epithelial cells 284 and impairment of mechanosensation and maturation in human osteoblasts 283.

Figure 6. Overview of ciliary TGFβ/BMP signalling.

Figure 6.

Receptors of the transforming growth factor beta (TGFβ) and bone morphogenic protein (BMP) family are recruited to the primary cilium to activate receptor (R)–SMAD transcription factors (SMAD2/3 and SMAD1/5/8) partly via internalization of active receptors by clathrin-mediated endocytosis (CME) at the ciliary pocket. Activated R-SMADs form a trimeric complex with the co-SMAD, SMAD4, which translocate to the nucleus for targeted gene expression. TGFβ may activate ERK1/2 in the cilium independently of CME. RAB11-mediated recycling of TGFβ receptors to the primary cilium may be regulated by the subdistal appendage protein, CEP128. Feedback inhibition of ciliary TGFβ/BMP signalling is under the control of the inhibitor SMAD, SMAD7, as well as the E3 ubiquitin ligase, SMURF1. Ciliary TGFβ receptors may further stimulate the hedgehog pathway component, smoothened (SMO), leading to activation of GLI transcription factors. Abbreviations: TGFβ-RI/II, TGFβ receptors I and II; BMP-RI/II, BMP receptors I and II; GLI-A: activator form of GLI transcription factors; ERK1/2: extracellular signal–regulated kinase 1/2; EE: early endosome.

The importance of the cilium in coordinating TGFβ/BMP signalling is underscored by the fact that additional sets of proteins, which regulate receptor trafficking and mechanisms in feedback inhibition, are enriched at the cilia-centrosome axis (Figure 6). RAB11 285, which is responsible for endosomal recycling of TGFβ receptors 286, is recruited to the cilium in a process that is mediated by the subdistal appendage protein, CEP128 287. Consequently, loss of CEP128 leads to impaired phosphorylation of R-SMADs, which in zebrafish is associated with defective organ development 287. Conversely, feedback inhibitors of TGFβ/BMP signalling, including SMAD7 and SMURF1 265, 288, localize to the ciliary base 268, 273 and restrain excessive signalling from the primary cilium. SMAD7 has been proposed to interact with TGFβ receptors to limit ARL6-mediated ciliary localization of the receptors and suppress tumour cell migration and invasion by restricting cross-talk between TGFβ receptors and SMO in HH signalling 272. In this context, hamartin (also known as tuberous sclerosis complex protein 1, TSC1, which originally was found to form a heterodimeric complex with TSC2 that negatively regulates mTOR signaling, is required for TGFβ-induced phosphorylation of SMAD2/3 and subsequent expression of GLI2 and WNT5A in an mTOR-independent manner 289, signifying an additional layer of cross-talk between multiple signaling pathways at the primary cilium. Finally, the E3 ubiquitin ligase SMURF1 negatively regulates phosphorylation of SMAD1/5/8 at primary cilia to control cell-type specification during in vivo heart development, which in Smurf1−/− mouse embryos is associated with delayed outflow tract septation 273.

Conclusions and future directions

A growing body of research over the past 15 years has uncovered important cellular signalling pathways that function via primary cilia. Cilia are present on most cell types, but not all cell types in a given tissue or organ will be ciliated. In addition, the ciliary composition of receptors and regulatory proteins seems to be cell and tissue specific. In the individual cell the constellation of receptors and regulatory proteins might change over time, enabling the cell to carry out specific functions during development and in the maintenance of tissue homeostasis in the adult. The importance of cilia in signalling is demonstrated by the fact that mutations in >200 genes that lead to dysfunction of primary cilia underlie a pleiotropic group of >30 ciliopathies, which affect many different tissues and organs during embryonic and postnatal development as well as in adulthood 29.

Current evidence suggest that primary cilia coordinate a variety of signalling pathways, including those regulated by HH, GPCR, WNT, RTK and TGFβ/BMP, to control developmental processes, tissue plasticity and organ function. In this regard it is important to recognize that while some pathways are considered to be bona fide ciliary pathways, such as canonical HH signalling, other pathways might be only partially associated with cilia and probably act in cell type-specific and developmentally dedicated contexts. A growing body of evidence points suggests that temporally regulated trafficking as well as activation or deactivation of receptors and/or regulatory signalling modules at the cilium–centrosome axis are critical for orchestrating the balanced output of these pathways. Some of these events are regulated within different sub-compartments of the cilium–centrosome axis, such as through E3 ubiquitin ligases, which in conjunction with endocytic pathways at the ciliary pocket provide feedback loops to control the balanced output of HH, PDGFRα and TGFβ/BMP signalling pathways. The role of IFT proteins, the BBSome, kinesin motor proteins and endocytic events in regulating ciliary signalling has been discussed elsewhere 23, 77, 78, 290.

The mechanisms that control the balanced output of ciliary signalling described in this Review might also apply to other pathways such as Hippo and Notch signalling pathways or those regulated by receptors of extracellular matrix molecules, which also have been linked to primary cilia in various cell types and tissues 23, 291, 292. Indeed, the growing list of signalling pathways that are connected to primary cilia is indicative of the importance of these organelles in orchestrating the integration and cross-talk between these pathways in a spatiotemporal manner. However, we still know very little as to how the temporal and spatial dynamics of major signalling networks are encoded by the primary cilium and how the integration of information in such networks is able to generate diversity in signalling outputs. In this context, it is important to recognize that primary cilia are highly dynamic organelles whose configuration is tightly coupled to the differentiation state and microenvironment of the cell, and that this flexibility of the cilium might explain how cells are able to receive and convert signalling inputs in different cellular, developmental and homeostatic settings. Despite enormous progress in this field, several key questions remain to be addressed including the mechanisms by which primary cilia orchestrate individual signalling pathways in a temporal manner, the mechanisms by which multiple ciliary pathways integrate into higher order signalling networks, the mechanisms by which cilia dictate cellular activity and fate in the context of stem cell microenvironments, cell differentiation states and organ development, and how ciliary protein composition differs at different stages of the cilia life cycle. Addressing these questions is crucial to improve our understanding of fundamental cilia biology and disease etiology as well as for developing new treatment approaches for ciliopathies.

Key points.

  • Primary cilia emanate in a single copy from the centrosomal mother centriole (basal body) at the surface of most vertebrate cell types.

  • Primary cilia possess a unique lipid and receptor composition and detect and convey extracellular cues to control cellular processes during development and in tissue homeostasis.

  • Current evidence suggest primary cilia coordinate a variety of signalling pathways, including those regulated by HH, GPCR, WNT, RTK and TGFβ/BMP, to control developmental processes, tissue plasticity and organ function.

  • The ability of primary cilia to balance the output of cellular signalling is dynamic and relies on the differentiation state and microenvironment of the cell.

  • Dysfunction of primary cilia underlies a pleiotropic group of diseases and syndromic disorders termed ciliopathies, affecting many different organs in the body.

  • Mechanistic insight into ciliary coordination of spatiotemporal signalling networks is critical for understanding the etiology of ciliopathies and for the discovery of novel ciliopathy disease genes and drug targets.

Acknowledgements

The authors’ work presented in this Review was supported by Independent Research Fund Denmark (6108–00457B and 8020–00162B to S.T.C and L.B.P), the Danish Cancer Society (R146-A9590–16-S2 to L.B.P. and Z.A.), Brødrene Hartmann’s Fond (A31662 to L.B.P.), research project grant R21 MH107021 from the NIH/NIMH (to K.M.), A-grant from the Alex’s Lemonade Foundation (to S.M.), a Welch Foundation Grant (I-1906 to S.M.), and a R01 grant from the National Institutes of Health (1R01GM113023 to S.M.). We are grateful to Stine K. Morthorst, University of Copenhagen, for help with formatting the references and the three reviewers for their insightful and constructive comments. We apologize to those authors whose work has not been cited because of space limitations.

Footnotes

Competing interests

The authors declare no competing interests.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

REFERENCES

  • 1.Satir P & Christensen ST Overview of structure and function of mammalian cilia. Annu Rev Physiol 69, 377–400 (2007). [DOI] [PubMed] [Google Scholar]
  • 2.Ciliopathies: A reference for clinicians. (eds. Kenny TD & Beales PL) 1–282 (Oxford University Press, Oxford, Oxford; 2014). [Google Scholar]
  • 3.Heydeck W, Fievet L, Davis EE & Katsanis N The complexity of the cilium: spatiotemporal diversity of an ancient organelle. Current Opinion in Cell Biology 55, 139–149 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sorokin S Centrioles and the formation of rudimentary cilia by fibroblasts and smooth muscle cells. J Cell Biol 15, 363–377 (1962). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Sorokin SP Reconstructions of centriole formation and ciliogenesis in mammalian lungs. J Cell Sci 3, 207–230 (1968). [DOI] [PubMed] [Google Scholar]
  • 6.Meunier A & Azimzadeh J Multiciliated Cells in Animals. Cold Spring Harbor perspectives in biology 8 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Avasthi P & Marshall WF Stages of ciliogenesis and regulation of ciliary length. Differentiation; research in biological diversity 83, S30–42 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Broekhuis JR, Leong WY & Jansen G Regulation of cilium length and intraflagellar transport. International review of cell and molecular biology 303, 101–138 (2013). [DOI] [PubMed] [Google Scholar]
  • 9.Tucker RW, Pardee AB & Fujiwara K Centriole ciliation is related to quiescence and DNA synthesis in 3T3 cells. Cell 17, 527–535 (1979). [DOI] [PubMed] [Google Scholar]
  • 10.Rieder CL, Jensen CG & Jensen LC The resorption of primary cilia during mitosis in a vertebrate (PtK1) cell line. Journal of ultrastructure research 68, 173–185 (1979). [DOI] [PubMed] [Google Scholar]
  • 11.Tucker RW, Scher CD & Stiles CD Centriole deciliation associated with the early response of 3T3 cells to growth factors but not to SV40. Cell 18, 1065–1072 (1979). [DOI] [PubMed] [Google Scholar]
  • 12.Pugacheva EN, Jablonski SA, Hartman TR, Henske EP & Golemis EA HEF1-dependent Aurora A activation induces disassembly of the primary cilium. Cell 129, 1351–1363 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Spalluto C, Wilson DI & Hearn T Evidence for reciliation of RPE1 cells in late G1 phase, and ciliary localisation of cyclin B1. FEBS open bio 3, 334–340 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ford MJ et al. A Cell/Cilia Cycle Biosensor for Single-Cell Kinetics Reveals Persistence of Cilia after G1/S Transition Is a General Property in Cells and Mice. Developmental Cell 47, 509–523.e505 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Das RM & Storey KG Apical abscission alters cell polarity and dismantles the primary cilium during neurogenesis. Science 343, 200–204 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.McDermott KM, Liu BY, Tlsty TD & Pazour GJ Primary cilia regulate branching morphogenesis during mammary gland development. Current biology : CB 20, 731–737 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Blitzer AL et al. Primary cilia dynamics instruct tissue patterning and repair of corneal endothelium. Proceedings of the National Academy of Sciences of the United States of America 108, 2819–2824 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bangs FK, Schrode N, Hadjantonakis AK & Anderson KV Lineage specificity of primary cilia in the mouse embryo. Nature cell biology 17, 113–122 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.May-Simera HL et al. Primary Cilium-Mediated Retinal Pigment Epithelium Maturation Is Disrupted in Ciliopathy Patient Cells. Cell Rep 22, 189–205 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Iomini C, Tejada K, Mo W, Vaananen H & Piperno G Primary cilia of human endothelial cells disassemble under laminar shear stress. The Journal of cell biology 164, 811–817 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Garcia-Gonzalo FR & Reiter JF Open Sesame: How Transition Fibers and the Transition Zone Control Ciliary Composition. Cold Spring Harbor perspectives in biology (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sung CH & Leroux MR The roles of evolutionarily conserved functional modules in cilia-related trafficking. Nature cell biology 15, 1387–1397 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Morthorst SK, Christensen ST & Pedersen LB Regulation of ciliary membrane protein trafficking and signalling by kinesin motor proteins. The FEBS journal (2018). [DOI] [PubMed] [Google Scholar]
  • 24.Lechtreck KF IFT-Cargo Interactions and Protein Transport in Cilia. Trends in biochemical sciences 40, 765–778 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Taschner M & Lorentzen E The Intraflagellar Transport Machinery. Cold Spring Harb Perspect Biol 8 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wood CR, Huang K, Diener DR & Rosenbaum JL The cilium secretes bioactive ectosomes. Curr Biol 23, 906–911 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cao M et al. Uni-directional ciliary membrane protein trafficking by a cytoplasmic retrograde IFT motor and ciliary ectosome shedding. eLife 4, e05242 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nager AR et al. An Actin Network Dispatches Ciliary GPCRs into Extracellular Vesicles to Modulate Signaling. Cell 168, 252–263 e214 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Reiter JF & Leroux MR Genes and molecular pathways underpinning ciliopathies. Nature reviews. Molecular cell biology 18, 533–547 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pedersen LB & Rosenbaum JL Intraflagellar transport (IFT) role in ciliary assembly, resorption and signalling. Curr. Top. Dev. Biol. 85, 23–61 (2008). [DOI] [PubMed] [Google Scholar]
  • 31.Prevo B, Scholey JM & Peterman EJG Intraflagellar transport: mechanisms of motor action, cooperation, and cargo delivery. The FEBS journal 284, 2905–2931 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kozminski KG, Johnson KA, Forscher P & Rosenbaum JL A motility in the eukaryotic flagellum unrelated to flagellar beating. Proc Natl Acad Sci USA 90, 5519–5523 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Walther Z, Vashishtha M & Hall JL The Chlamydomonas FLA10 gene encodes a novel kinesin-homologous protein. The Journal of cell biology 126, 175–188 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kozminski KG, Beech PL & Rosenbaum JL The Chlamydomonas kinesin-like protein FLA10 is involved in motility associated with the flagellar membrane. The Journal of cell biology 131, 1517–1527 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vashishtha M, Walther Z & Hall JL The kinesin-homologous protein encoded by the Chlamydomonas FLA10 gene is associated with basal bodies and centrioles. Journal of cell science 109 ( Pt 3), 541–549 (1996). [DOI] [PubMed] [Google Scholar]
  • 36.Pazour GJ, Wilkerson CG & Witman GB A dynein light chain is essential for retrograde particle movement in intraflagellar transport (IFT). The Journal of cell biology 141, 979–992 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pazour GJ, Dickert BL & Witman GB The DHC1b (DHC2) isoform of cytoplasmic dynein is required for flagellar assembly. The Journal of cell biology 144, 473–481 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Porter ME, Bower R, Knott JA, Byrd P & Dentler W Cytoplasmic dynein heavy chain 1b is required for flagellar assembly in Chlamydomonas. Molecular biology of the cell 10, 693–712 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Verhey KJ, Dishinger J & Kee HL Kinesin motors and primary cilia. Biochemical Society transactions 39, 1120–1125 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ou G, Blacque OE, Snow JJ, Leroux MR & Scholey JM Functional coordination of intraflagellar transport motors. Nature 436, 583–587 (2005). [DOI] [PubMed] [Google Scholar]
  • 41.Snow JJ et al. Two anterograde intraflagellar transport motors cooperate to build sensory cilia on C. elegans neurons. Nature cell biology 6, 1109–1113 (2004). [DOI] [PubMed] [Google Scholar]
  • 42.Zhao C, Omori Y, Brodowska K, Kovach P & Malicki J Kinesin-2 family in vertebrate ciliogenesis. Proceedings of the National Academy of Sciences of the United States of America 109, 2388–2393 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Prevo B, Mangeol P, Oswald F, Scholey JM & Peterman EJ Functional differentiation of cooperating kinesin-2 motors orchestrates cargo import and transport in C. elegans cilia. Nature cell biology 17, 1536–1545 (2015). [DOI] [PubMed] [Google Scholar]
  • 44.Cole DG et al. Chlamydomonas kinesin-II-dependent intraflagellar transport (IFT): IFT particles contain proteins required for ciliary assembly in Caenorhabditis elegans sensory neurons. The Journal of cell biology 141, 993–1008 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Piperno G & Mead K Transport of a novel complex in the cytoplasmic matrix of Chlamydomonas flagella. Proc Natl Acad Sci USA 94, 4457–4462 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rosenbaum JL & Witman GB Intraflagellar transport. Nature reviews. Molecular cell biology 3, 813–825 (2002). [DOI] [PubMed] [Google Scholar]
  • 47.Taschner M, Kotsis F, Braeuer P, Kuehn EW & Lorentzen E Crystal structures of IFT70/52 and IFT52/46 provide insight into intraflagellar transport B core complex assembly. The Journal of cell biology 207, 269–282 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bhogaraju S, Engel BD & Lorentzen E Intraflagellar transport complex structure and cargo interactions. Cilia 2, 10 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Toropova K, Mladenov M & Roberts AJ Intraflagellar transport dynein is autoinhibited by trapping of its mechanical and track-binding elements. Nature structural & molecular biology 24, 461–468 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Funabashi T, Katoh Y, Okazaki M, Sugawa M & Nakayama K Interaction of heterotrimeric kinesin-II with IFT-B-connecting tetramer is crucial for ciliogenesis. The Journal of cell biology 217, 2867–2876 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mohamed MAA, Stepp WL & Okten Z Reconstitution reveals motor activation for intraflagellar transport. Nature 557, 387–391 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Liang Y, Zhu X, Wu Q & Pan J Ciliary Length Sensing Regulates IFT Entry via Changes in FLA8/KIF3B Phosphorylation to Control Ciliary Assembly. Current biology : CB 28, 2429–2435 e2423 (2018). [DOI] [PubMed] [Google Scholar]
  • 53.Perkins LA, Hedgecock EM, Thomson JN & Culotti JG Mutant sensory cilia in the nematode Caenorhabditis elegans. Developmental biology 117, 456–487 (1986). [DOI] [PubMed] [Google Scholar]
  • 54.Pedersen LB et al. Chlamydomonas IFT172 is encoded by FLA11, interacts with CrEB1, and regulates IFT at the flagellar tip. Current biology : CB 15, 262–266 (2005). [DOI] [PubMed] [Google Scholar]
  • 55.Qin H et al. Intraflagellar transport is required for the vectorial movement of TRPV channels in the ciliary membrane. Current biology : CB 15, 1695–1699 (2005). [DOI] [PubMed] [Google Scholar]
  • 56.Mukhopadhyay S et al. TULP3 bridges the IFT-A complex and membrane phosphoinositides to promote trafficking of G protein-coupled receptors into primary cilia. Genes Dev 24, 2180–2193 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Behal RH et al. Subunit interactions and organization of the Chlamydomonas reinhardtii intraflagellar transport complex A proteins. The Journal of biological chemistry 287, 11689–11703 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Liem KF Jr. et al. The IFT-A complex regulates Shh signaling through cilia structure and membrane protein trafficking. The Journal of cell biology 197, 789–800 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Keady BT et al. IFT25 links the signal-dependent movement of Hedgehog components to intraflagellar transport. Dev Cell 22, 940–951 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Eguether T et al. IFT27 links the BBSome to IFT for maintenance of the ciliary signaling compartment. Dev Cell 31, 279–290 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bhogaraju S et al. Molecular basis of tubulin transport within the cilium by IFT74 and IFT81. Science 341, 1009–1012 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Eguether T, Cordelieres FP & Pazour GJ Intraflagellar transport is deeply integrated in hedgehog signaling. Molecular biology of the cell 29, 1178–1189 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mourão A, Christensen ST & Lorentzen E The intraflagellar transport machinery in ciliary signaling. Current opinion in structural biology 41, 98–108 (2016). [DOI] [PubMed] [Google Scholar]
  • 64.Badgandi HB, Hwang SH, Shimada IS, Loriot E & Mukhopadhyay S Tubby family proteins are adapters for ciliary trafficking of integral membrane proteins. J Cell Biol 216, 743–760 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Takahara M et al. Ciliopathy-associated mutations of IFT122 impair ciliary protein trafficking but not ciliogenesis. Human Molecular Genetics 27, 516–528 (2018). [DOI] [PubMed] [Google Scholar]
  • 66.Hirano T, Katoh Y & Nakayama K Intraflagellar transport-A complex mediates ciliary entry and retrograde trafficking of ciliary G protein-coupled receptors. Molecular biology of the cell 28, 429–439 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fu W, Wang L, Kim S, Li J & Dynlacht BD Role for the IFT-A Complex in Selective Transport to the Primary Cilium. Cell reports 17, 1505–1517 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Caparrós-Martín JA et al. Specific variants in WDR35 cause a distinctive form of Ellis-van Creveld syndrome by disrupting the recruitment of the EvC complex and SMO into the cilium. Human molecular genetics 24, 4126–4137 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Boubakri M et al. Loss of ift122, a Retrograde Intraflagellar Transport (IFT) Complex Component, Leads to Slow, Progressive Photoreceptor Degeneration Due to Inefficient Opsin Transport. The Journal of biological chemistry 291, 24465–24474 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nachury MV et al. A core complex of BBS proteins cooperates with the GTPase Rab8 to promote ciliary membrane biogenesis. Cell 129, 1201–1213 (2007). [DOI] [PubMed] [Google Scholar]
  • 71.Ou G, Blacque OE, Snow JJ, Leroux MR & Scholey JM Functional coordination of intraflagellar transport motors. Nature 436, 583–587 (2005). [DOI] [PubMed] [Google Scholar]
  • 72.Lechtreck KF et al. The Chlamydomonas reinhardtii BBSome is an IFT cargo required for export of specific signaling proteins from flagella. J Cell Biol 187, 1117–1132 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.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 U S A 105, 4242–4246 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Loktev AV & Jackson PK Neuropeptide Y family receptors traffic via the Bardet-Biedl syndrome pathway to signal in neuronal primary cilia. Cell Rep 5, 1316–1329 (2013). [DOI] [PubMed] [Google Scholar]
  • 75.Jin H et al. The conserved Bardet-Biedl syndrome proteins assemble a coat that traffics membrane proteins to cilia. Cell 141, 1208–1219 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lechtreck KF et al. Cycling of the signaling protein phospholipase D through cilia requires the BBSome only for the export phase. J Cell Biol 201, 249–261 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Nachury MV The molecular machines that traffic signaling receptors into and out of cilia. Current Opinion in Cell Biology 51, 124–131 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wingfield Jenna L., Lechtreck K-F, Lorentzen E Trafficking of ciliary membrane proteins by the intraflagellar transport/BBSome machinery. Essays In Biochemistry (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pazour GJ et al. Chlamydomonas IFT88 and its mouse homologue, polycystic kidney disease gene tg737, are required for assembly of cilia and flagella. The Journal of cell biology 151, 709–718 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Moyer J et al. Candidate gene associated with a mutation causing recessive polycystic kidney disease in mice. Science 264, 1329–1333 (1994). [DOI] [PubMed] [Google Scholar]
  • 81.Barr MM & Sternberg PW A polycystic kidney-disease gene homologue required for male mating behaviour in C. elegans. Nature 401, 386–389 (1999). [DOI] [PubMed] [Google Scholar]
  • 82.Yoder BK, Hou X & Guay-Woodford LM The polycystic kidney disease proteins, polycystin-1, polycystin-2, polaris, and cystin, are co-localized in renal cilia. J Am Soc Nephrol 13, 2508–2516 (2002). [DOI] [PubMed] [Google Scholar]
  • 83.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 12, R378–380 (2002). [DOI] [PubMed] [Google Scholar]
  • 84.Nauli SM et al. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat Genet 33, 129–137 (2003). [DOI] [PubMed] [Google Scholar]
  • 85.Ma M, Gallagher AR & Somlo S Ciliary Mechanisms of Cyst Formation in Polycystic Kidney Disease. Cold Spring Harb Perspect Biol 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Norris DP Cilia, calcium and the basis of left-right asymmetry. BMC biology 10, 102 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Qian F et al. PKD1 interacts with PKD2 through a probable coiled-coil domain. Nat Genet 16, 179–183 (1997). [DOI] [PubMed] [Google Scholar]
  • 88.Nauli SM et al. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat. Genet. 33, 129–137 (2003). [DOI] [PubMed] [Google Scholar]
  • 89.Delling M et al. Primary cilia are not calcium-responsive mechanosensors. Nature 531, 656–660 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Norris DP & Jackson PK Cell biology: Calcium contradictions in cilia. Nature 531, 582–583 (2016). [DOI] [PubMed] [Google Scholar]
  • 91.Shen PS et al. The Structure of the Polycystic Kidney Disease Channel PKD2 in Lipid Nanodiscs. Cell 167, 763–773 e711 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Grieben M et al. Structure of the polycystic kidney disease TRP channel Polycystin-2 (PC2). Nature structural & molecular biology 24, 114–122 (2017). [DOI] [PubMed] [Google Scholar]
  • 93.Liu X et al. Polycystin-2 is an essential ion channel subunit in the primary cilium of the renal collecting duct epithelium. Elife 7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Su Q et al. Structure of the human PKD1-PKD2 complex. Science 361 (2018). [DOI] [PubMed] [Google Scholar]
  • 95.Briscoe J & Thérond PP The mechanisms of Hedgehog signalling and its roles in development and disease. Nature Reviews Molecular Cell Biology 14, 416 (2013). [DOI] [PubMed] [Google Scholar]
  • 96.Huangfu D et al. Hedgehog signalling in the mouse requires intraflagellar transport proteins. Nature 426, 83–87 (2003). [DOI] [PubMed] [Google Scholar]
  • 97.Goetz SC & Anderson KV The primary cilium: a signalling centre during vertebrate development. Nature reviews. Genetics 11, 331–344 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Yue S et al. Requirement of Smurf-mediated endocytosis of Patched1 in sonic hedgehog signal reception. Elife 3 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Schou KB et al. KIF13B establishes a CAV1-enriched microdomain at the ciliary transition zone to promote Sonic hedgehog signalling. Nat Commun 8, 14177 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Scheidel N, Kennedy J & Blacque OE Endosome maturation factors Rabenosyn-5/VPS45 and caveolin-1 regulate ciliary membrane and polycystin-2 homeostasis. Embo j (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Corbit KC et al. Vertebrate Smoothened functions at the primary cilium. Nature 437, 1018–1021 (2005). [DOI] [PubMed] [Google Scholar]
  • 102.Rohatgi R, Milenkovic L & Scott MP Patched1 regulates hedgehog signaling at the primary cilium. Science 317, 372–376 (2007). [DOI] [PubMed] [Google Scholar]
  • 103.Niewiadomski P et al. Gli protein activity is controlled by multisite phosphorylation in vertebrate Hedgehog signaling. Cell Rep 6, 168–181 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mukhopadhyay S & Rohatgi R G-protein-coupled receptors, Hedgehog signaling and primary cilia. Seminars in cell & developmental biology 33, 63–72 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Bitgood MJ & McMahon AP Hedgehog and Bmp genes are coexpressed at many diverse sites of cell-cell interaction in the mouse embryo. Dev Biol 172, 126–138 (1995). [DOI] [PubMed] [Google Scholar]
  • 106.Carballo GB, Honorato JR, de Lopes GPF & Spohr T.C.L.d.S.E. A highlight on Sonic hedgehog pathway. Cell communication and signaling : CCS 16, 11–11 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bijlsma MF & Roelink H Non-cell-autonomous signaling by Shh in tumors: challenges and opportunities for therapeutic targets. Expert Opinion on Therapeutic Targets 14, 693–702 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Yuan X et al. Ciliary IFT80 balances canonical versus non-canonical hedgehog signalling for osteoblast differentiation. Nature communications 7, 11024–11024 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bijlsma MF, Damhofer H & Roelink H Hedgehog-stimulated chemotaxis is mediated by smoothened located outside the primary cilium. Science signaling 5, ra60–ra60 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ho Wei L, Arastoo M, Georgiou I, Manning DR & Riobo-Del Galdo NA Activation of the Gi protein-RHOA axis by non-canonical Hedgehog signaling is independent of primary cilia. PloS one 13, e0203170–e0203170 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Gong X et al. Structural basis for the recognition of Sonic Hedgehog by human Patched1. Science 361 (2018). [DOI] [PubMed] [Google Scholar]
  • 112.Qi X, Schmiege P, Coutavas E, Wang J & Li X Structures of human Patched and its complex with native palmitoylated sonic hedgehog. Nature 560, 128–132 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Huang P et al. Structural Basis of Smoothened Activation in Hedgehog Signaling. Cell 174, 312–324 e316 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Nachtergaele S et al. Oxysterols are allosteric activators of the oncoprotein Smoothened. Nat Chem Biol 8, 211–220 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Dorn KV, Hughes CE & Rohatgi R A Smoothened-Evc2 complex transduces the Hedgehog signal at primary cilia. Dev Cell 23, 823–835 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Singh J, Wen X & Scales SJ The Orphan G Protein-Coupled Receptor Gpr175 (TPRA40) Enhances Hedgehog Signaling by Modulating cAMP Levels. J Biol Chem (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Haycraft CJ et al. Gli2 and Gli3 localize to cilia and require the intraflagellar transport protein polaris for processing and function. PLoS Genet 1, e53 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Jiang J & Struhl G Regulation of the Hedgehog and Wingless signalling pathways by the F-box/WD40-repeat protein Slimb. Nature 391, 493–496 (1998). [DOI] [PubMed] [Google Scholar]
  • 119.Tempe D, Casas M, Karaz S, Blanchet-Tournier MF & Concordet JP Multisite protein kinase A and glycogen synthase kinase 3beta phosphorylation leads to Gli3 ubiquitination by SCFbetaTrCP. Mol Cell Biol 26, 4316–4326 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Pan Y & Wang B A novel protein-processing domain in Gli2 and Gli3 differentially blocks complete protein degradation by the proteasome. J Biol Chem 282, 10846–10852 (2007). [DOI] [PubMed] [Google Scholar]
  • 121.Wang B, Fallon JF & Beachy PA Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb. Cell 100, 423–434 (2000). [DOI] [PubMed] [Google Scholar]
  • 122.Mukhopadhyay S et al. The ciliary G-protein-coupled receptor Gpr161 negatively regulates the Sonic hedgehog pathway via cAMP signaling. Cell 152, 210–223 (2013). [DOI] [PubMed] [Google Scholar]
  • 123.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 24, 670–682 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Jia J et al. Suppressor of Fused inhibits mammalian Hedgehog signaling in the absence of cilia. Dev Biol 330, 452–460 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.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 138, 4921–4930 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Svard J et al. Genetic elimination of Suppressor of fused reveals an essential repressor function in the mammalian Hedgehog signaling pathway. Dev Cell 10, 187–197 (2006). [DOI] [PubMed] [Google Scholar]
  • 127.Goodrich LV, Milenkovic L, Higgins KM & Scott MP Altered neural cell fates and medulloblastoma in mouse patched mutants. Science 277, 1109–1113 (1997). [DOI] [PubMed] [Google Scholar]
  • 128.Norman RX et al. Tubby-like protein 3 (TULP3) regulates patterning in the mouse embryo through inhibition of Hedgehog signaling. Hum Mol Genet 18, 1740–1754 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Patterson VL et al. Mouse hitchhiker mutants have spina bifida, dorso-ventral patterning defects and polydactyly: identification of Tulp3 as a novel negative regulator of the Sonic hedgehog pathway. Hum Mol Genet 18, 1719–1739 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.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. Proceedings of the National Academy of Sciences of the United States of America 108, 1456–1461 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ocbina PJR, Eggenschwiler JT, Moskowitz I & Anderson KV Complex interactions between genes controlling trafficking in primary cilia. Nature genetics 43, 547–553 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Hwang SH & Mukhopadhyay S G-protein-coupled receptors and localized signaling in the primary cilium during ventral neural tube patterning. Birth Defects Res A Clin Mol Teratol 103, 12–19 (2015). [DOI] [PubMed] [Google Scholar]
  • 133.Pusapati GV et al. G protein-coupled receptors control the sensitivity of cells to the morphogen Sonic Hedgehog. Sci Signal 11 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hwang SH et al. The G protein-coupled receptor Gpr161 regulates forelimb formation, limb patterning and skeletal morphogenesis in a primary cilium-dependent manner. Development 145 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Shimada IS et al. Basal Suppression of the Sonic Hedgehog Pathway by the G-Protein-Coupled Receptor Gpr161 Restricts Medulloblastoma Pathogenesis. Cell Rep 22, 1169–1184 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.He M et al. The kinesin-4 protein Kif7 regulates mammalian Hedgehog signalling by organizing the cilium tip compartment. Nat Cell Biol 16, 663–672 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Liem KF Jr., He M, Ocbina PJ & Anderson KV Mouse Kif7/Costal2 is a cilia-associated protein that regulates Sonic hedgehog signaling. Proc Natl Acad Sci U S A 106, 13377–13382 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Pal K et al. Smoothened determines beta-arrestin-mediated removal of the G protein-coupled receptor Gpr161 from the primary cilium. J Cell Biol 212, 861–875 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Garcia-Gonzalo FR et al. Phosphoinositides Regulate Ciliary Protein Trafficking to Modulate Hedgehog Signaling. Dev Cell 34, 400–409 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Chavez M et al. Modulation of Ciliary Phosphoinositide Content Regulates Trafficking and Sonic Hedgehog Signaling Output. Dev Cell 34, 338–350 (2015). [DOI] [PubMed] [Google Scholar]
  • 141.Wong W & Scott JD AKAP signalling complexes: focal points in space and time. Nat Rev Mol Cell Biol 5, 959–970 (2004). [DOI] [PubMed] [Google Scholar]
  • 142.Bachmann VA et al. Gpr161 anchoring of PKA consolidates GPCR and cAMP signaling. Proc Natl Acad Sci U S A 113, 7786–7791 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Mick DU et al. Proteomics of Primary Cilia by Proximity Labeling. Dev Cell 35, 497–512 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Choi YH et al. 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 U S A 108, 10679–10684 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bishop GA, Berbari NF, Lewis J & Mykytyn K Type III adenylyl cyclase localizes to primary cilia throughout the adult mouse brain. J Comp Neurol 505, 562–571 (2007). [DOI] [PubMed] [Google Scholar]
  • 146.Vuolo L, Herrera A, Torroba B, Menendez A & Pons S Ciliary adenylyl cyclases control the Hedgehog pathway. J Cell Sci 128, 2928–2937 (2015). [DOI] [PubMed] [Google Scholar]
  • 147.Fredriksson R, Lagerstrom MC, Lundin LG & Schioth HB The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol 63, 1256–1272 (2003). [DOI] [PubMed] [Google Scholar]
  • 148.Pandy-Szekeres G et al. GPCRdb in 2018: adding GPCR structure models and ligands. Nucleic Acids Res 46, D440–D446 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hauser AS, Attwood MM, Rask-Andersen M, Schioth HB & Gloriam DE Trends in GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 16, 829–842 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Hilger D, Masureel M & Kobilka BK Structure and dynamics of GPCR signaling complexes. Nat Struct Mol Biol 25, 4–12 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Eichel K & von Zastrow M Subcellular Organization of GPCR Signaling. Trends Pharmacol Sci 39, 200–208 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Mykytyn K & Askwith C G-Protein-Coupled Receptor Signaling in Cilia. Cold Spring Harb Perspect Biol 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Schou KB, Pedersen LB & Christensen ST Ins and outs of GPCR signaling in primary cilia. EMBO Rep 16, 1099–1113 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Tabibian JH, Masyuk AI, Masyuk TV, O’Hara SP & LaRusso NF Physiology of cholangiocytes. Compr Physiol 3, 541–565 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Masyuk AI et al. Cholangiocyte primary cilia are chemosensory organelles that detect biliary nucleotides via P2Y12 purinergic receptors. Am J Physiol Gastrointest Liver Physiol 295, G725–734 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Masyuk TV, Masyuk AI & LaRusso NF TGR5 in the Cholangiociliopathies. Dig Dis 33, 420–425 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Keitel V et al. The membrane-bound bile acid receptor TGR5 is localized in the epithelium of human gallbladders. Hepatology 50, 861–870 (2009). [DOI] [PubMed] [Google Scholar]
  • 158.Masyuk AI et al. Ciliary subcellular localization of TGR5 determines the cholangiocyte functional response to bile acid signaling. Am J Physiol Gastrointest Liver Physiol 304, G1013–1024 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Cramer MT & Guay-Woodford LM Cystic kidney disease: a primer. Adv Chronic Kidney Dis 22, 297–305 (2015). [DOI] [PubMed] [Google Scholar]
  • 160.Jin X et al. Cilioplasm is a cellular compartment for calcium signaling in response to mechanical and chemical stimuli. Cell Mol Life Sci 71, 2165–2178 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Upadhyay VS et al. Roles of dopamine receptor on chemosensory and mechanosensory primary cilia in renal epithelial cells. Front Physiol 5, 72 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Raychowdhury MK et al. Vasopressin receptor-mediated functional signaling pathway in primary cilia of renal epithelial cells. Am J Physiol Renal Physiol 296, F87–97 (2009). [DOI] [PubMed] [Google Scholar]
  • 163.Torres VE et al. Tolvaptan in Later-Stage Autosomal Dominant Polycystic Kidney Disease. N Engl J Med 377, 1930–1942 (2017). [DOI] [PubMed] [Google Scholar]
  • 164.Wang CY, Tsai HL, Syu JS, Chen TY & Su MT Primary Cilium-Regulated EG-VEGF Signaling Facilitates Trophoblast Invasion. J Cell Physiol 232, 1467–1477 (2017). [DOI] [PubMed] [Google Scholar]
  • 165.Guemez-Gamboa A, Coufal NG & Gleeson JG Primary cilia in the developing and mature brain. Neuron 82, 511–521 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Bishop GA, Berbari NF, Lewis JS & Mykytyn K Type III Adenylyl Cyclase Localizes to Primary Cilia throughout the Adult Mouse Brain. J Comp Neurol 505, 562–571 (2007). [DOI] [PubMed] [Google Scholar]
  • 167.Green JA et al. Recruitment of beta-Arrestin into Neuronal Cilia Modulates Somatostatin Receptor Subtype 3 Ciliary Localization. Mol Cell Biol 36, 223–235 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Domire JS et al. Dopamine receptor 1 localizes to neuronal cilia in a dynamic process that requires the Bardet-Biedl syndrome proteins. Cell Mol Life Sci 68, 2951–2960 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Sun X et al. Tubby is required for trafficking G protein-coupled receptors to neuronal cilia. Cilia 1, 21 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Marin O Interneuron dysfunction in psychiatric disorders. Nat Rev Neurosci 13, 107–120 (2012). [DOI] [PubMed] [Google Scholar]
  • 171.Guo J et al. Primary Cilia Signaling Shapes the Development of Interneuronal Connectivity. Dev Cell 42, 286–300 e284 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ye F, Nager AR & Nachury MV BBSome trains remove activated GPCRs from cilia by enabling passage through the transition zone. J Cell Biol 217, 1847–1868 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Berbari NF et al. Hippocampal and cortical primary cilia are required for aversive memory in mice. PLoS One 9, e106576 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Einstein EB et al. Somatostatin signaling in neuronal cilia is critical for object recognition memory. J Neurosci 30, 4306–4314 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Liu X et al. beta-Arrestin-biased signaling mediates memory reconsolidation. Proc Natl Acad Sci U S A 112, 4483–4488 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Wang Z, Phan T & Storm DR The type 3 adenylyl cyclase is required for novel object learning and extinction of contextual memory: role of cAMP signaling in primary cilia. J Neurosci 31, 5557–5561 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Berbari NF et al. Leptin resistance is a secondary consequence of the obesity in ciliopathy mutant mice. Proc Natl Acad Sci U S A 110, 7796–7801 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Davenport JR et al. Disruption of intraflagellar transport in adult mice leads to obesity and slow-onset cystic kidney disease. Curr Biol 17, 1586–1594 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Farooqi IS et al. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med 348, 1085–1095 (2003). [DOI] [PubMed] [Google Scholar]
  • 180.Siljee JE et al. Subcellular localization of MC4R with ADCY3 at neuronal primary cilia underlies a common pathway for genetic predisposition to obesity. Nat Genet 50, 180–185 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Loh K, Herzog H & Shi YC Regulation of energy homeostasis by the NPY system. Trends Endocrinol Metab 26, 125–135 (2015). [DOI] [PubMed] [Google Scholar]
  • 182.Marion S, Oakley RH, Kim KM, Caron MG & Barak LS A beta-arrestin binding determinant common to the second intracellular loops of rhodopsin family G protein-coupled receptors. J Biol Chem 281, 2932–2938 (2006). [DOI] [PubMed] [Google Scholar]
  • 183.Clevers H Wnt/β-Catenin Signaling in Development and Disease. Cell 127, 469–480 (2006). [DOI] [PubMed] [Google Scholar]
  • 184.Niehrs C The complex world of WNT receptor signalling. Nature Reviews Molecular Cell Biology 13, 767 (2012). [DOI] [PubMed] [Google Scholar]
  • 185.MacDonald BT & He X Frizzled and LRP5/6 Receptors for Wnt/β-Catenin Signaling. Cold Spring Harbor Perspectives in Biology 4 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Sineva GS & Pospelov VA Chapter Two - β-Catenin in Pluripotency: Adhering to Self-Renewal or Wnting to Differentiate?, in International Review of Cell and Molecular Biology, Vol. 312 (ed. Jeon KW) 53–78 (Academic Press, 2014). [DOI] [PubMed] [Google Scholar]
  • 187.Kim W, Kim M & Jho E.-h. Wnt/β-catenin signalling: from plasma membrane to nucleus. Biochemical Journal 450, 9–21 (2013). [DOI] [PubMed] [Google Scholar]
  • 188.Green J, Nusse R & van Amerongen R The Role of Ryk and Ror Receptor Tyrosine Kinases in Wnt Signal Transduction. Cold Spring Harbor Perspectives in Biology 6 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Yang Y & Mlodzik M Wnt-Frizzled/Planar Cell Polarity Signaling: Cellular Orientation by Facing the Wind (Wnt). Annual Review of Cell and Developmental Biology 31, 623–646 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Berger H, Wodarz A & Borchers A PTK7 Faces the Wnt in Development and Disease. Frontiers in Cell and Developmental Biology 5 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Nishita M et al. Ror2/Frizzled Complex Mediates Wnt5a-Induced AP-1 Activation by Regulating Dishevelled Polymerization. Molecular and Cellular Biology 30, 3610–3619 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Witte F et al. Negative regulation of Wnt signaling mediated by CK1-phosphorylated Dishevelled via Ror2. The FASEB Journal 24, 2417–2426 (2010). [DOI] [PubMed] [Google Scholar]
  • 193.Corbit KC et al. Kif3a constrains beta-catenin-dependent Wnt signalling through dual ciliary and non-ciliary mechanisms. Nat Cell Biol 10, 70–76 (2008). [DOI] [PubMed] [Google Scholar]
  • 194.Zhang B et al. GSK3β-Dzip1-Rab8 Cascade Regulates Ciliogenesis after Mitosis. PLOS Biology 13, e1002129 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Chen Y et al. Sonic Hedgehog Dependent Phosphorylation by CK1α and GRK2 Is Required for Ciliary Accumulation and Activation of Smoothened. PLOS Biology 9, e1001083 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Veland IR et al. Inversin/Nephrocystin-2 Is Required for Fibroblast Polarity and Directional Cell Migration. PloS one 8, e60193 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Marion V et al. Transient ciliogenesis involving Bardet-Biedl syndrome proteins is a fundamental characteristic of adipogenic differentiation. Proceedings of the National Academy of Sciences 106, 1820–1825 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Simons M et al. Inversin, the gene product mutated in nephronophthisis type II, functions as a molecular switch between Wnt signaling pathways. Nat Genet 37, 537–543 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Lienkamp S et al. Inversin relays Frizzled-8 signals to promote proximal pronephros development. Proceedings of the National Academy of Sciences 107, 20388–20393 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Ocbina PJR, Tuson M & Anderson KV Primary cilia are not required for normal canonical Wnt signaling in the mouse embryo. PloS one 4, e6839–e6839 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Kim M et al. KIF3A binds to β-arrestin for suppressing Wnt/β-catenin signalling independently of primary cilia in lung cancer. Scientific reports 6, 32770–32770 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Vuong LT, Mukhopadhyay B & Choi K-W Kinesin-II recruits Armadillo and Dishevelled for Wingless signaling in Drosophila. Development 141, 3222–3232 (2014). [DOI] [PubMed] [Google Scholar]
  • 203.Huang P & Schier AF Dampened Hedgehog signaling but normal Wnt signaling in zebrafish without cilia. Development (Cambridge, England) 136, 3089–3098 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Oh EC & Katsanis N Context-Dependent Regulation of Wnt Signaling through the Primary Cilium. Journal of the American Society of Nephrology 24, 10–18 (2013). [DOI] [PubMed] [Google Scholar]
  • 205.Lancaster MA et al. Impaired Wnt–β-catenin signaling disrupts adult renal homeostasis and leads to cystic kidney ciliopathy. Nature Medicine 15, 1046 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Lancaster MA, Schroth J & Gleeson JG Subcellular spatial regulation of canonical Wnt signalling at the primary cilium. Nature Cell Biology 13, 700 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Lancaster MA et al. Defective Wnt-dependent cerebellar midline fusion in a mouse model of Joubert syndrome. Nature Medicine 17, 726 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Abdelhamed ZA et al. The Meckel-Gruber syndrome protein TMEM67 controls basal body positioning and epithelial branching morphogenesis in mice via the non-canonical Wnt pathway. Disease Models & Mechanisms 8, 527 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Bergmann C et al. Loss of Nephrocystin-3 Function Can Cause Embryonic Lethality, Meckel-Gruber-like Syndrome, Situs Inversus, and Renal-Hepatic-Pancreatic Dysplasia. The American Journal of Human Genetics 82, 959–970 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Burcklé C et al. Control of the Wnt pathways by nephrocystin-4 is required for morphogenesis of the zebrafish pronephros. Human Molecular Genetics 20, 2611–2627 (2011). [DOI] [PubMed] [Google Scholar]
  • 211.Gerhardt C et al. The transition zone protein Rpgrip1l regulates proteasomal activity at the primary cilium. The Journal of Cell Biology 210, 1027 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Patnaik SR et al. RPGR protein complex regulates proteasome activity and mediates store-operated calcium entry. Oncotarget 9, 23183–23197 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Borgal L et al. The Ciliary Protein Nephrocystin-4 Translocates the Canonical Wnt Regulator Jade-1 to the Nucleus to Negatively Regulate β-Catenin Signaling. The Journal of Biological Chemistry 287, 25370–25380 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Chitalia VC et al. Jade-1 inhibits Wnt signaling by ubiquitinating β-catenin and mediates Wnt pathway inhibition by pVHL. Nature cell biology 10, 1208–1216 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Mahuzier A et al. Dishevelled stabilization by the ciliopathy protein Rpgrip1l is essential for planar cell polarity. The Journal of Cell Biology 198, 927–940 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Gerdes JM et al. Disruption of the basal body compromises proteasomal function and perturbs intracellular Wnt response. Nat Genet 39, 1350–1360 (2007). [DOI] [PubMed] [Google Scholar]
  • 217.Gerhardt C, Leu T, Lier JM & Rüther U The cilia-regulated proteasome and its role in the development of ciliopathies and cancer. Cilia 5, 14 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Liu YP et al. Ciliopathy proteins regulate paracrine signaling by modulating proteasomal degradation of mediators. The Journal of clinical investigation 124, 2059–2070 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Hua K & Ferland RJ Primary cilia proteins: ciliary and extraciliary sites and functions. Cellular and Molecular Life Sciences 75, 1521–1540 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Lemmon MA & Schlessinger J Cell signaling by receptor-tyrosine kinases. Cell 141, 1117–1134 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Crudden C et al. Chapter One - Blurring Boundaries: Receptor Tyrosine Kinases as functional G Protein-Coupled Receptors, in International Review of Cell and Molecular Biology, Vol. 339 (ed. Shukla AK) 1–40 (Academic Press, 2018). [DOI] [PubMed] [Google Scholar]
  • 222.Christensen ST, Clement CA, Satir P & Pedersen LB Primary cilia and coordination of receptor tyrosine kinase (RTK) signalling. The Journal of pathology 226, 172–184 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Christensen ST, Morthorst SK, Mogensen JB & Pedersen LB Primary Cilia and Coordination of Receptor Tyrosine Kinase (RTK) and Transforming Growth Factor beta (TGF-beta) Signaling. Cold Spring Harb Perspect Biol 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Ma R et al. PKD2 Functions as an Epidermal Growth Factor-Activated Plasma Membrane Channel. Molecular and Cellular Biology 25, 8285–8298 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Danilov AI et al. Ultrastructural and antigenic properties of neural stem cells and their progeny in adult rat subventricular zone. Glia 57, 136–152 (2009). [DOI] [PubMed] [Google Scholar]
  • 226.Martin L et al. Constitutively-active FGFR3 disrupts primary cilium length and IFT20 trafficking in various chondrocyte models of achondroplasia. Human Molecular Genetics 27, 1–13 (2018). [DOI] [PubMed] [Google Scholar]
  • 227.Leitch CC & Zaghloul NA BBS4 Is Necessary for Ciliary Localization of TrkB Receptor and Activation by BDNF. PloS one 9, e98687 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Teilmann SC & Christensen ST Localization of the angiopoietin receptors Tie-1 and Tie-2 on the primary cilia in the female reproductive organs. Cell Biology International 29, 340–346 (2005). [DOI] [PubMed] [Google Scholar]
  • 229.Kunova Bosakova M et al. Regulation of ciliary function by fibroblast growth factor signaling identifies FGFR3-related disorders achondroplasia and thanatophoric dysplasia as ciliopathies. Human Molecular Genetics 27, 1093–1105 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Zhu D, Shi S, Wang H & Liao K Growth arrest induces primary-cilium formation and sensitizes IGF-1-receptor signaling during differentiation induction of 3T3-L1 preadipocytes. Journal of Cell Science 122, 2760–2768 (2009). [DOI] [PubMed] [Google Scholar]
  • 231.Dalbay MT, Thorpe SD, Connelly JT, Chapple JP & Knight MM Adipogenic Differentiation of hMSCs is Mediated by Recruitment of IGF‐1r Onto the Primary Cilium Associated With Cilia Elongation. Stem Cells (Dayton, Ohio) 33, 1952–1961 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Yeh C et al. IGF-1 activates a cilium-localized non-canonical Gβγ signaling pathway that regulates cell cycle progression. Developmental cell 26, 358–368 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Gabriel E et al. CPAP promotes timely cilium disassembly to maintain neural progenitor pool. The EMBO Journal 35, 803–819 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Wang H et al. Hsp90α forms a stable complex at the cilium neck for the interaction of signalling molecules in IGF-1 receptor signalling. Journal of Cell Science 128, 100–108 (2015). [DOI] [PubMed] [Google Scholar]
  • 235.Gerdes JM et al. Ciliary dysfunction impairs beta-cell insulin secretion and promotes development of type 2 diabetes in rodents. Nature Communications 5, 5308 (2014). [DOI] [PubMed] [Google Scholar]
  • 236.Volta F & Gerdes JM The role of primary cilia in obesity and diabetes. Annals of the New York Academy of Sciences 1391, 71–84 (2017). [DOI] [PubMed] [Google Scholar]
  • 237.Song DK, Choi JH & Kim M-S Primary Cilia as a Signaling Platform for Control of Energy Metabolism. Diabetes & Metabolism Journal 42, 117–127 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Leibiger B et al. Selective Insulin Signaling through A and B Insulin Receptors Regulates Transcription of Insulin and Glucokinase Genes in Pancreatic β Cells . Molecular cell 7, 559–570 (2001). [DOI] [PubMed] [Google Scholar]
  • 239.Heldin C-H, Lennartsson J & Westermark B Involvement of platelet-derived growth factor ligands and receptors in tumorigenesis. Journal of Internal Medicine 283, 16–44 (2018). [DOI] [PubMed] [Google Scholar]
  • 240.Schneider L et al. PDGFRαα Signaling Is Regulated through the Primary Cilium in Fibroblasts. Current Biology 15, 1861–1866 (2005). [DOI] [PubMed] [Google Scholar]
  • 241.Vestergaard ML, Awan A, Warzecha CB, Christensen ST & Andersen CY Immunofluorescence Microscopy and mRNA Analysis of Human Embryonic Stem Cells (hESCs) Including Primary Cilia Associated Signaling Pathways, in Human Embryonic Stem Cell Protocols. (ed. Turksen K) 123–140 (Springer; New York, New York, NY; 2016). [DOI] [PubMed] [Google Scholar]
  • 242.Noda K, Kitami M, Kitami K, Kaku M & Komatsu Y Canonical and noncanonical intraflagellar transport regulates craniofacial skeletal development. Proceedings of the National Academy of Sciences of the United States of America 113, E2589–E2597 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Gerhardt C, Lier JM, Kuschel S & Rüther U The Ciliary Protein Ftm Is Required for Ventricular Wall and Septal Development. PloS one 8, e57545 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Kopinke D, Roberson EC & Reiter JF Ciliary Hedgehog signaling restricts injury-induced adipogenesis. Cell 170, 340–351.e312 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Falcón-Urrutia P, Carrasco CM, Lois P, Palma V & Roth AD Shh Signaling through the Primary Cilium Modulates Rat Oligodendrocyte Differentiation. PloS one 10, e0133567 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Nielsen BS et al. PDGFRβ and oncogenic mutant PDGFRα D842V promote disassembly of primary cilia through a PLCγ- and AURKA-dependent mechanism. Journal of Cell Science 128, 3543–3549 (2015). [DOI] [PubMed] [Google Scholar]
  • 247.Schneider L et al. Directional Cell Migration and Chemotaxis in Wound Healing Response to PDGF-AA are Coordinated by the Primary Cilium in Fibroblasts. Cellular Physiology and Biochemistry 25, 279–292 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Schneider L et al. The Na(+)/H(+) exchanger NHE1 is required for directional migration stimulated via PDGFR-α in the primary cilium. The Journal of Cell Biology 185, 163–176 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Clement DL et al. PDGFRα signaling in the primary cilium regulates NHE1-dependent fibroblast migration via coordinated differential activity of MEK1/2–ERK1/2–p90(RSK) and AKT signaling pathways. Journal of Cell Science 126, 953–965 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Umberger NL & Caspary T Ciliary transport regulates PDGF-AA/αα signaling via elevated mammalian target of rapamycin signaling and diminished PP2A activity. Molecular biology of the cell 26, 350–358 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Suizu F et al. Phosphorylation‐dependent Akt–Inversin interaction at the basal body of primary cilia. The EMBO Journal 35, 1346–1363 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.O’Driscoll M, Ruiz-Perez VL, Woods CG, Jeggo PA & Goodship JA A splicing mutation affecting expression of ataxia–telangiectasia and Rad3–related protein (ATR) results in Seckel syndrome. Nature Genetics 33, 497 (2003). [DOI] [PubMed] [Google Scholar]
  • 253.Stiff T, Casar Tena T, O’Driscoll M, Jeggo PA & Philipp M ATR promotes cilia signalling: links to developmental impacts. Human Molecular Genetics 25, 1574–1587 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Vierkotten J, Dildrop R, Peters T, Wang B & Rüther U Ftm is a novel basal body protein of cilia involved in Shh signalling. Development 134, 2569–2577 (2007). [DOI] [PubMed] [Google Scholar]
  • 255.Koefoed K, Veland IR, Pedersen LB, Larsen LA & Christensen ST Cilia and coordination of signaling networks during heart development. Organogenesis 10, 108–125 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Mohapatra B et al. Protein tyrosine kinase regulation by ubiquitination: Critical roles of Cbl-family ubiquitin ligases. Biochimica et biophysica acta 1833, 122–139 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Liyasova MS, Ma K & Lipkowitz S Molecular Pathways: Cbl Proteins in Tumorigenesis and Antitumor Immunity—Opportunities for Cancer Treatment. Clinical cancer research : an official journal of the American Association for Cancer Research 21, 1789–1794 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Schmid FM et al. IFT20 modulates ciliary PDGFRalpha signaling by regulating the stability of Cbl E3 ubiquitin ligases. J Cell Biol 217, 151–161 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Szucs Z et al. Molecular subtypes of gastrointestinal stromal tumors and their prognostic and therapeutic implications. Future Oncology 13, 93–107 (2017). [DOI] [PubMed] [Google Scholar]
  • 260.Mohapatra B et al. An essential role of CBL and CBL-B ubiquitin ligases in mammary stem cell maintenance. Development (Cambridge, England) 144, 1072–1086 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Bielas SL et al. Mutations in the inositol polyphosphate-5-phosphatase E gene link phosphatidyl inositol signaling to the ciliopathies. Nature genetics 41, 1032–1036 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Kisseleva MV, Cao L & Majerus PW Phosphoinositide-specific Inositol Polyphosphate 5-Phosphatase IV Inhibits Akt/Protein Kinase B Phosphorylation and Leads to Apoptotic Cell Death. Journal of Biological Chemistry 277, 6266–6272 (2002). [DOI] [PubMed] [Google Scholar]
  • 263.Jacoby M et al. INPP5E mutations cause primary cilium signaling defects, ciliary instability and ciliopathies in human and mouse. Nature Genetics 41, 1027 (2009). [DOI] [PubMed] [Google Scholar]
  • 264.Nickel J, ten Dijke P & Mueller TD TGF-β family co-receptor function and signaling. Acta Biochimica et Biophysica Sinica 50, 12–36 (2018). [DOI] [PubMed] [Google Scholar]
  • 265.Heldin C-H & Moustakas A Signaling Receptors for TGF-β Family Members. Cold Spring Harbor Perspectives in Biology 8, a022053 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Luo K Signaling Cross Talk between TGF-β/Smad and Other Signaling Pathways. Cold Spring Harbor Perspectives in Biology 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Bakkebø M et al. SARA is dispensable for functional TGF-β signaling. FEBS Letters 586, 3367–3372 (2012). [DOI] [PubMed] [Google Scholar]
  • 268.Clement CA et al. TGF-beta signaling is associated with endocytosis at the pocket region of the primary cilium. Cell reports 3, 1806–1814 (2013). [DOI] [PubMed] [Google Scholar]
  • 269.Xie Y-F et al. Pulsed electromagnetic fields stimulate osteogenic differentiation and maturation of osteoblasts by upregulating the expression of BMPRII localized at the base of primary cilium. Bone 93, 22–32 (2016). [DOI] [PubMed] [Google Scholar]
  • 270.Labour M-N, Riffault M, Christensen ST & Hoey DA TGFβ1 – induced recruitment of human bone mesenchymal stem cells is mediated by the primary cilium in a SMAD3-dependent manner. Scientific Reports 6, 35542 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Zhang J et al. Topography of calcium phosphate ceramics regulates primary cilia length and TGF receptor recruitment associated with osteogenesis. Acta Biomaterialia 57, 487–497 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Gencer S et al. TGF-β receptor I/II trafficking and signaling at primary cilia are inhibited by ceramide to attenuate cell migration and tumor metastasis. Science signaling 10, eaam7464 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Koefoed K et al. The E3 ubiquitin ligase SMURF1 regulates cell-fate specification and outflow tract septation during mammalian heart development. Scientific Reports 8, 9542 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Yao G et al. Disruption of polycystin-L causes hippocampal and thalamocortical hyperexcitability. Hum Mol Genet 25, 448–458 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Arrighi N et al. The primary cilium is necessary for the differentiation and the maintenance of human adipose progenitors into myofibroblasts. Scientific Reports 7, 15248 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Goetz Jacky G. et al. Endothelial Cilia Mediate Low Flow Sensing during Zebrafish Vascular Development. Cell reports 6, 799–808 (2014). [DOI] [PubMed] [Google Scholar]
  • 277.Iomini C, Tejada K, Mo W, Vaananen H & Piperno G Primary cilia of human endothelial cells disassemble under laminar shear stress. The Journal of Cell Biology 164, 811–817 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Kallakuri S et al. Endothelial Cilia Are Essential for Developmental Vascular Integrity in Zebrafish. Journal of the American Society of Nephrology : JASN 26, 864–875 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Hierck BP et al. Primary cilia sensitize endothelial cells for fluid shear stress. Developmental dynamics : an official publication of the American Association of Anatomists 237, 725–735 (2008). [DOI] [PubMed] [Google Scholar]
  • 280.Egorova AD et al. Lack of primary cilia primes shear-induced Endothelial-to-Mesenchymal Transition. Circulation research 108, 1093–1101 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Vion A-C et al. Primary cilia sensitize endothelial cells to BMP and prevent excessive vascular regression. The Journal of Cell Biology (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Kawasaki M et al. TGF-β Suppresses Ift88 Expression in Chondrocytic ATDC5 Cells. Journal of Cellular Physiology 230, 2788–2795 (2015). [DOI] [PubMed] [Google Scholar]
  • 283.Ehnert S et al. TGF-β1 impairs mechanosensation of human osteoblasts via HDAC6-mediated shortening and distortion of primary cilia. Journal of Molecular Medicine 95, 653–663 (2017). [DOI] [PubMed] [Google Scholar]
  • 284.Han SJ et al. Deficiency of primary cilia in kidney epithelial cells induces epithelial to mesenchymal transition. Biochemical and Biophysical Research Communications 496, 450–454 (2018). [DOI] [PubMed] [Google Scholar]
  • 285.Westlake CJ 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 U S A 108, 2759–2764 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Mitchell H, Choudhury A, Pagano RE & Leof EB Ligand-dependent and -independent Transforming Growth Factor-β Receptor Recycling Regulated by Clathrin-mediated Endocytosis and Rab11. Molecular biology of the cell 15, 4166–4178 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Monnich M et al. CEP128 Localizes to the Subdistal Appendages of the Mother Centriole and Regulates TGF-beta/BMP Signaling at the Primary Cilium. Cell reports 22, 2584–2592 (2018). [DOI] [PubMed] [Google Scholar]
  • 288.Miyazawa K & Miyazono K Regulation of TGF-β Family Signaling by Inhibitory Smads. Cold Spring Harbor Perspectives in Biology 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Rosengren T, Larsen LJ, Pedersen LB, Christensen ST & Møller LB TSC1 and TSC2 regulate cilia length and canonical Hedgehog signaling via different mechanisms. Cellular and Molecular Life Sciences 75, 2663–2680 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Pedersen LB, Mogensen JB & Christensen ST Endocytic Control of Cellular Signaling at the Primary Cilium. Trends in biochemical sciences 41, 784–797 (2016). [DOI] [PubMed] [Google Scholar]
  • 291.Wheway G, Nazlamova L & Hancock JT Signaling through the Primary Cilium. Frontiers in Cell and Developmental Biology 6 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Seeger-Nukpezah T & Golemis EA The extracellular matrix and ciliary signaling. Current opinion in cell biology 24, 652–661 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Wood CR & Rosenbaum JL Ciliary ectosomes: transmissions from the cell’s antenna. Trends Cell Biol 25, 276–285 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Lee KH et al. Identification of a novel Wnt5a-CK1varepsilon-Dvl2-Plk1-mediated primary cilia disassembly pathway. Embo j 31, 3104–3117 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Keitel V, Ullmer C & Haussinger D The membrane-bound bile acid receptor TGR5 (Gpbar-1) is localized in the primary cilium of cholangiocytes. Biol Chem 391, 785–789 (2010). [DOI] [PubMed] [Google Scholar]
  • 296.Abdul-Majeed S & Nauli SM Dopamine receptor type 5 in the primary cilia has dual chemo- and mechano-sensory roles. Hypertension 58, 325–331 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Singh J, Wen X & Scales SJ The Orphan G Protein-coupled Receptor Gpr175 (Tpra40) Enhances Hedgehog Signaling by Modulating cAMP Levels. The Journal of Biological Chemistry 290, 29663–29675 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Koemeter-Cox AI et al. Primary cilia enhance kisspeptin receptor signaling on gonadotropin-releasing hormone neurons. Proc Natl Acad Sci U S A 111, 10335–10340 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.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 19, 1540–1547 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Jiang Y, Li YR, Tian H, Ma M & Matsunami H Muscarinic acetylcholine receptor M3 modulates odorant receptor activity via inhibition of beta-arrestin-2 recruitment. Nat Commun 6, 6448 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Zheng L et al. Ciliary parathyroid hormone signaling activates transforming growth factor-beta to maintain intervertebral disc homeostasis during aging. Bone Res 6, 21 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Omori Y et al. Identification of G Protein-Coupled Receptors (GPCRs) in Primary Cilia and Their Possible Involvement in Body Weight Control. PLoS One 10, e0128422 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Jin D et al. Prostaglandin signalling regulates ciliogenesis by modulating intraflagellar transport. Nat Cell Biol 16, 841–851 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Brailov I et al. Localization of 5-HT(6) receptors at the plasma membrane of neuronal cilia in the rat brain. Brain Res 872, 271–275 (2000). [DOI] [PubMed] [Google Scholar]
  • 305.Handel M et al. Selective targeting of somatostatin receptor 3 to neuronal cilia. Neuroscience 89, 909–926 (1999). [DOI] [PubMed] [Google Scholar]
  • 306.Szumska J et al. Trace Amine-Associated Receptor 1 Localization at the Apical Plasma Membrane Domain of Fisher Rat Thyroid Epithelial Cells Is Confined to Cilia. Eur Thyroid J 4, 30–41 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES