ArticlePDF Available

CP110 and its network of partners coordinately regulate cilia assembly

Authors:

Abstract and Figures

Cilia are hair-like protrusions found at the surface of most eukaryotic cells. They can be divided into two types, motile and non-motile. Motile cilia are found in a restricted number of cell types, are generally present in large numbers, and beat in a coordinated fashion to generate fluid flow or locomotion. Non-motile or primary cilia, on the other hand, are detected in many different cell types, appear once per cell, and primarily function to transmit signals from the extracellular milieu to the cell nucleus. Defects in cilia formation, function, or maintenance are known to cause a bewildering set of human diseases, or ciliopathies, typified by retinal degeneration, renal failure and cystic kidneys, obesity, liver dysfunction, and neurological disorders. A common denominator between motile and primary cilia is their structural similarity, as both types of cilia are composed of an axoneme, the ciliary backbone that is made up of microtubules emanating from a mother centriole/basal body anchored to the cell membrane, surrounded by a ciliary membrane continuous with the plasma membrane. This structural similarity is indicative of a universal mechanism of cilia assembly involving a common set of molecular players and a sophisticated, highly regulated series of molecular events. In this review, we will mainly focus on recent advances in our understanding of the regulatory mechanisms underlying cilia assembly, with special attention paid to the centriolar protein, CP110, its interacting partner Cep290, and the various downstream molecular players and events leading to intraflagellar transport (IFT), a process that mediates the bidirectional movement of protein cargos along the axoneme and that is essential for cilia formation and maintenance.
Content may be subject to copyright.
R E V I E W Open Access
CP110 and its network of partners coordinately
regulate cilia assembly
William Y Tsang
1,2,3*
and Brian D Dynlacht
4
Abstract
Cilia are hair-like protrusions found at the surface of most eukaryotic cells. They can be divided into two types,
motile and non-motile. Motile cilia are found in a restricted number of cell types, are generally present in large
numbers, and beat in a coordinated fashion to generate fluid flow or locomotion. Non-motile or primary cilia, on
the other hand, are detected in many different cell types, appear once per cell, and primarily function to transmit
signals from the extracellular milieu to the cell nucleus. Defects in cilia formation, function, or maintenance are
known to cause a bewildering set of human diseases, or ciliopathies, typified by retinal degeneration, renal failure
and cystic kidneys, obesity, liver dysfunction, and neurological disorders. A common denominator between motile
and primary cilia is their structural similarity, as both types of cilia are composed of an axoneme, the ciliary
backbone that is made up of microtubules emanating from a mother centriole/basal body anchored to the cell
membrane, surrounded by a ciliary membrane continuous with the plasma membrane. This structural similarity is
indicative of a universal mechanism of cilia assembly involving a common set of molecular players and a
sophisticated, highly regulated series of molecular events. In this review, we will mainly focus on recent advances in
our understanding of the regulatory mechanisms underlying cilia assembly, with special attention paid to the
centriolar protein, CP110, its interacting partner Cep290, and the various downstream molecular players and events
leading to intraflagellar transport (IFT), a process that mediates the bidirectional movement of protein cargos along
the axoneme and that is essential for cilia formation and maintenance.
Keywords: Centrosomes, Cilia, Ciliogenesis, CP110, Cep290, BBSome, IFT, Protein network
Review
Links between cilia, centrosomes, and the cell cycle
It is well known that cilia and centrosomes share an in-
timate relationship during the cell cycle. A centrosome
consists of a pair of centrioles, termed the mother and
daughter centrioles, embedded in a poorly defined
pericentriolar matrix, from which cytoplasmic micro-
tubules emanate and grow [1-4]. The mother centriole
can be distinguished from the daughter centriole by the
presence of distal and sub-distal appendages. Distal ap-
pendages are thought to be important for the docking
of a basal body to the cell membrane and the recruit-
ment of IFT proteins prior to cilia assembly, whereas
sub-distal appendages anchor microtubules, participate
in endosome recycling, and form the basal foot, a struc-
ture essential for ciliogenesis and ciliary beating in
motile cilia [5-9]. In proliferating cells, a single centro-
some in the G1 phase undergoes duplication in the S
phase. The two centrosomes then separate, migrating
to opposite poles and establishing a bipolar spindle
in mitosis. Upon cell cycle exit, a centrosome obtains
competence for ciliogenesis, whereby the mother centriole
is converted into the basal body. Depending on the cell
type and/or cilia type, the basal body can migrate and
anchor to the cell surface or dock ciliary vesicles, which
elongate and eventually fuse with the plasma mem-
brane. In both scenarios, the basal body serves to nu-
cleate the growth of axonemal microtubules, a process
highly dependent on IFT [10-12]. IFT is bidirectional,
and this property can be explained by the existence of
biochemically and functionally distinct protein com-
plexes, IFT-B and IFT-A. While IFT-B and IFT-A are
commonly believed to direct anterograde (cell body to
* Correspondence: william.tsang@ircm.qc.ca
1
Institut de recherches cliniques de Montréal, 110 avenue des Pins Ouest,
Montréal, QC H2W 1R7, Canada
2
Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
Full list of author information is available at the end of the article
© 2013 Tsang and Dynlacht; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the
Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use,
distribution, and reproduction in any medium, provided the original work is properly cited.
Tsang and Dynlacht Cilia 2013, 2:9
http://www.ciliajournal.com/content/2/1/9
cilia) and retrograde (cilia to cell body) transport of mac-
romolecules, respectively, recent evidence indicates that
IFT-A is also involved in anterograde transport [13-16].
IFT is essential for cilium assembly and maintenance, since
the organelle lacks protein synthesis machinery [17]. When
cells re-enter the cell cycle, cilia are disassembled, and the
basal body relocates to the cell interior, assuming a
position near the nucleus. It is logical to postulate
that controls must exist to suppress the inappropriate
assembly of cilia in proliferating cells or the untimely
assembly of a bipolar spindle in non-proliferating
cells. In addition, vesicular trafficking, mother centri-
ole/basal body migration to the cell surface, basal
body anchoring to the cell membrane, and IFT must
be tightly regulated in a temporally-, spatially-, and
cell-type-specific manner to ensure the fidelity of
ciliogenesis. Indeed, a growing number of proteins, includ-
ing many that were originally identified in a proteomic
screen for novel centrosomal and ciliary components
[18-20], are known to modulate cilia assembly in a posi-
tive or negative manner [21,22], suggesting that cilia as-
sembly involves a complex circuitry controlled by the
coordinated inhibition of negative regulators and recruit-
ment and activation of positive regulators.
The CP110-Cep97 pathway
While there are many important modulators of cilio-
genesis, two distal centriolar proteins, CP110 and Cep97,
were the first proteins shown to negatively regulate cilia
assembly [23]. Loss of either protein elicits premature
inappropriate cilium formation in proliferating cells,
whereas its over-expression inhibits ciliogenesis in
non-proliferating cells. Fittingly, patients with chronic
rhinosinusitis, a respiratory disease associated with abnor-
mal or lack of motile cilia, have elevated levels of CP110
[24]. While the precise function of Cep97 awaits further
experimentation, this protein might serve as a chaperone
to stabilize CP110, allowing the co-recruitment of both
proteins to the centrosome. CP110, on the other hand, is
thought to impose a structural role at the centrosome and
forms discrete complexes critical for cell cycle regulation
and cilia assembly (Figure 1) [23,25-31]. This protein does
not have an associated enzymatic activity; rather, it was
shown to localize to the distal ends of centrioles, forming a
capabove the growing microtubules that could restrain
microtubule growth [32]. Indeed, CP110 has the ability to
control centriole length in non-ciliated human [33-35] and
insect cells [36] and to block ciliary axoneme formation in
ciliated mammalian (RPE-1 and NIH-3T3) cells [23,25].
Paradoxically, CP110 does not modulate cilia length,
suggesting that at least in ciliated cells, CP110 could switch
offthe ciliogenic program. Tellingly, CP110 is completely
extinguished from the basal body in ciliated cells (Figure 1
and [23]). The loss of CP110 effectively liberates the
mother centriole from its centrosomal role in cell division
and licensesthe transition from mother centriole to basal
body. Thus, it appears that the removal of CP110 from the
mother centriole, rather than cell cycle control per se,
could play a crucial role in the initiation of ciliogenesis.
CP110 levels and localization to the centrosome are
tightly regulated in a cell cycle dependent manner [29].
CP110 protein levels drop significantly in G2/M and
G0/G1 phases as a consequence of transcriptional con-
trols, ubiquitin-mediated proteasomal destruction, and
microRNA-mediated turnover of CP110 mRNA [37-39].
Furthermore, disappearance of CP110 from the basal
body in quiescent cells coincides with an enrichment of
a serine/threonine kinase, Ttbk2, at the same location
(Figure 2) [40]. Ttbk2, a microtubule plus-end tracking
protein, likely promotes the onset of ciliogenesis by co-
operating with end binding proteins [40-42]. Depletion
of Ttbk2 impairs both the loss of CP110 and the recruit-
ment of IFT complexes, including IFT88, a protein local-
ized to the distal appendages of the emerging basal body
and/or the transition zone [40]. Further, the loss of
Cep83, a distal appendage protein that functions in a
concerted and hierarchical manner to recruit other pro-
teins (including Cep89, SCLT1, FBF1, and Cep164), pre-
vents the recruitment of Ttbk2 to, and the release of
CP110 from, the basal body, thereby blocking basal body
anchoring to the cell membrane (Figure 2) [43]. Another
study highlighted a role for CCDC41/Cep83 in the re-
cruitment of IFT20 to the basal body and ciliary vesicle
docking to the mother centriole as important functions
of CCDC41/Cep83 during early ciliogenesis, although
Cep164 localization and abundance were not substan-
tially impacted [44]. Since Cep83 and Cep164 can recruit
IFT proteins to the basal body and/or the transition
zone, these results imply that distal appendage proteins,
Ttbk2, CP110, and IFT proteins could functionally inter-
act [43,45]. In addition to Ttbk2, the loss of a second
serine/threonine kinase, MARK4, causes mis-localization
of its interacting partner, Odf2, which is normally found
at sub-distal appendages, and likewise, inhibits cilia for-
mation by preventing the removal of CP110/Cep97 from
the basal body (Figure 2) [46-48]. In light of recent find-
ings that distal and sub-distal appendages are assembled
independently of one another [43], these intriguing
observations suggest that Ttbk2 and MARK4 activities
might be necessary to modulate the molecular frame-
work of distal and sub-distal appendages, respectively,
ultimately leading to the destruction and removal of
CP110 from the basal body. Alternatively, the two kinases
could function after the assembly of the appendages to
remove CP110 [49]. Furthermore, these studies suggest
that protein phosphorylation is crucial for the maturation
of a mother centriole into a functional basal body, and
future phospho-proteomic studies, in combination with
Tsang and Dynlacht Cilia 2013, 2:9 Page 2 of 8
http://www.ciliajournal.com/content/2/1/9
high resolution imaging, will be essential to identify key
substrates and to examine these maturation events in
greater detail.
CP110-interacting partners and its protein network
Besides Cep97, CP110 has been shown to associate with
a cadre of proteins important for ciliogenesis, suggesting
that it could assemble a multi-functional platform to in-
tegrate centriolar and basal body functions (Figure 2).
Cep104, a microtubule plus-end tracking protein identi-
fied by a proteomic screen for novel end binding-
interacting partners, interacts with CP110 and Cep97
[41]. This protein co-localizes with CP110 at the distal
ends of centrioles in proliferating cells and is similarly
absent from the basal body in quiescent cells. However,
unlike CP110 and Cep97, Cep104 is essential for cilio-
genesis, suggesting that it may regulate axonemal growth
at the onset of cilia assembly by counteracting the activ-
ities of CP110 and Cep97. In contrast to Cep104, another
protein, Kif24, appears to reinforce the role of CP110 as
a suppressor of ciliogenesis [28]. As a member of the
microtubule de-polymerizing kinesin family of proteins,
Kif24 specifically de-polymerizes and remodels centriolar
microtubules at the mother centriole/basal body, and de-
pletion of this protein promotes ciliation, whereas over-
expression inhibits cilia growth. Although Kif24 binds
CP110 and Cep97, it specifically stabilizes CP110 and
recruits it to the centrosome, suggesting that both the
de-polymerizing activity of Kif24 and its ability to re-
cruit a distal end capping protein (CP110) to centrioles
contribute to cilia suppression. In addition, CP110 has
been demonstrated to associate with a human ciliopathy
protein, Cep290, (also known as BBS14, NPHP6, JBTS5,
SLSN6, MKS4 and LCA10; [25]). Its many names
can be attributed to the diverse spectrum of clinical
manifestations, including Bardet-Biedl syndrome (BBS),
nephronophthisis, Joubert syndrome, Senior-Loken
syndrome, Meckel-Gruber syndrome, and Leber con-
genital amaurosis, associated with mutations in the
Cep290 gene [50-52]. Despite the identification of over
S
G2 M
Duplication
G1
Separation
Maturation
PM
G0
Cilia
Assembly
Cep76
CP110
Neurl4
CP110
Cyclin/cdk
Mother Centriole
Chromosome
Daughter Centriole
Pro-centriole
Cilium
CP110
Cytokinesis
Over-duplication
Centrin
CP110
CaM
Cep290
CP110 Cep104
Kif24
P
Usp33
Cyclin F
PPhosphorylation
Cep97
Figure 1 The role of CP110 in cell cycle control and ciliogenesis. CP110 and its network of partners form distinct complexes that regulate
different aspects of centrosome function, including centrosome over-duplication, centrosome separation, cytokinesis, and cilia assembly. The
localization of CP110 is also illustrated. PM denotes plasma membrane.
Tsang and Dynlacht Cilia 2013, 2:9 Page 3 of 8
http://www.ciliajournal.com/content/2/1/9
100 unique mutations, there is no clear relationship be-
tween genotype and phenotype. The loss of Cep290 abol-
ishes cilia assembly and disrupts the migration/anchoring
of centrioles to the cell cortex, suggesting that this pro-
tein functions to promote ciliogenesis at an early step
of the ciliogenic pathway [21,25,53]. This positive func-
tion of Cep290 is antagonized by CP110, and over-
expression of a CP110 mutant refractory to Cep290
binding is incapable of suppressing ciliation in non-
proliferating cells. Because the protein levels of Cep290
remain constant throughout the cell cycle, including
G0 [25], it seems plausible that CP110 restrains
Cep290 activity in proliferating cells through direct
interaction, but once cells exit the cell cycle, the loss
of CP110 protein releases Cep290 from inhibition. It is
currently not clear how Cep290 might promote centri-
ole migration/anchoring to the cell cortex, although it
is known that this protein directly interacts with an-
other ciliopathy protein NPHP5 [54], and depletion of
NPHP5 phenocopies the loss of Cep290 [55-57]. Inter-
estingly, analysis of the primary amino acid sequence of
Cep290 reveals the presence of multiple N-terminal tropo-
myosin homology domains and a C-terminal myosin-tail
homology domain, suggesting that it might have an actin-
related function, and that centriole migration/anchoring
could involve cytoskeletal re-organization and modulation
of actin dynamics [51,58]. Indeed, the role of actin cyto-
skeleton dynamics in cilia assembly has recently been illus-
trated in a high-throughput RNA interference screen,
wherein actin polymerization was shown to have an inhibi-
tory role in cilia assembly [22]. Two proteins belonging to
the gelsolin family members, GSN and AVIL, promote cili-
ation by severing actin filaments. On the other hand,
ACTR3, a protein known to mediate the formation of
branched actin networks, suppresses cilia formation. Treat-
ment of cells with drugs that inhibit actin filament
polymerization and/or affect actin dynamics, such as cyto-
chalasin D or latrunculin B, can facilitate ciliation in
addition to causing an increase in cilium length [22,55].
Notably, impaired cilia formation associated with the loss
of Cep290 or NPHP5 can be restored by the aforemen-
tioned drugs, strongly suggesting that proteins involved in
the regulation of actin dynamics could influence the
ciliogenic pathway and could be exploited as potential
therapeutic targets [55]. Besides Cep290 and NPHP5, two
other ciliopathy-associated proteins, MKS1 and MKS3, are
also required for the translocation of centrioles to the cell
surface, whereas IFT88 is not [59]. Thus, it seems likely
that a subset of centrosomal proteins is specifically dedi-
cated to basal body migration and anchoring to the cell
membrane, and it will be most interesting to identify the
complete set of factors that control this important process.
Cep290 function and its protein network
Beyond its potential contribution in basal body migra-
tion and/or anchoring to the cell membrane, Cep290
has additional functions critical to cilia assembly. An ele-
gant ultra-structural study conducted in Chlamydomonas
reinhardtii suggests that Cep290 localizes to the transi-
tion zone, a small region immediately distal to the basal
body characterized by the presence of Y-shaped fibers
that connect the axonemal microtubules to the ciliary
Figure 2 A system-wide schematic of protein interaction networks that modulate cilium assembly. Solid lines indicate known
protein-protein interactions, confirmed by immunoprecipitation, yeast two-hybrid, and/or in vitro binding experiments. Not every
protein-protein interaction indicated is direct. Dashed lines indicate known functional connections with no evidence of protein-protein
interactions to date. EB denotes end binding proteins.
Tsang and Dynlacht Cilia 2013, 2:9 Page 4 of 8
http://www.ciliajournal.com/content/2/1/9
membrane [60]. This region is thought to regulate the
entry and exit of protein and lipid cargos into and out of
the ciliary compartment. Consistent with this idea,
Cep290 is present at the transition zone of rat motile tra-
cheal cilia [61] and associates with CC2D2A and TCTN1,
both of which are known to form a large protein complex
with several other ciliopathy proteins (AHI1, MKS1,
TCTN2, TCTN3, B9D1, B9D2, TMEM216, TMEM67) at
the transition zone (Figure 2 and [62-64]). Cep290 also
binds to Cep162, an axoneme-recognition protein re-
quired for transition zone assembly (Figure 2 and [65]).
In addition, Cep290 is required for the targeting of
Rab8a, a small GTPase responsible for vesicular trafficking
into the cilium in cultured human epithelial cells [25,53],
and has a functional connection with the BBSome, a stable
multi-subunit complex known to mediate ciliary transport
(Figure 2). The BBSome is composed of seven BBS pro-
teins (BBS1, BBS2, BBS4, BBS5, BBS7, BBS8, and BBS9)
and BBIP10, a protein required for cytoplasmic micro-
tubule polymerization and acetylation. Assembly of the
BBSome follows a hierarchical order that initially involves
the stabilization of BBS7 by the chaperonin complex
(MKKS/BBS6, BBS10, BBS12 and CCT/TRiC family of
chaperonins), followed by the formation of the BBSome
core (BBS7, BBS2, BBS9) and the subsequent incorpor-
ation of the remaining BBSome subunits through a series
of protein-protein interactions [66,67]. Interestingly, two
components of the BBSome, BBS4 and BBS8, are not
properly recruited to the cilium upon Cep290 loss [68].
The lack of BBSome recruitment to the cilium could be
due to an assembly defect, as Cep290 is known to directly
interact with MKKS/BBS6, a chaperonin-like molecule
required at an early step in BBSome assembly [69]. In
addition, a Cep290 mutant in Chlamydomonas reinhardtii
possesses malformed flagella with abnormal protein com-
position, with increased amounts of IFT-B proteins and
decreased amounts of IFT-A proteins, suggesting that
retrograde and possibly anterograde IFT are impaired [60].
AlthoughneitherCep290norCP110hasbeendemon-
strated to directly interact with IFT proteins thus far, a
proteomic screen reveals IFT122 as a novel interacting
partner of NPHP5 (Figure 2 and [56]), a protein that dir-
ectly binds to, and shares a number of common features
with, Cep290 [55-57]. Further experiments will be neces-
sary to delineate the extent to which the CP110-Cep290
axis overlaps with the BBSome and/or the IFT pathway.
Other than its localization to the transition zone, Cep290
is also targeted to centriolar satellites [53,58]. Centriolar
satellites are small, electron-dense proteinaceous granules
found in the vicinity of the centrosome and have been
implicated in microtubule-dependent protein trafficking
towards the centrosome [70-72]. These structures may
be closely related to the pericentrosomal pre-ciliary com-
partment reported at the basal body during the onset of
ciliogenesis [22]. Interestingly, several satellite components,
including PCM1, BBS4, OFD1, Cep72, and Cep290 are re-
quired for cilia assembly, and the integrity of these unique
structures is highly dependent on protein-protein interac-
tions between them (Figure 2) [53,68,73]. Of note, BBS4 is
unique among satellite proteins in that it completely re-
localizes from its original satellite position to the cilium
during ciliogenesis [74]. Thus, Cep290, together with other
satellite proteins, might regulate the trafficking of BBS4 be-
tween the two different sub-cellular compartments, and
henceplayanindirectroleinBBSomeassembly.Further
studies will be needed to decipher the mechanisms
through which satellite proteins (and possibly other un-
identified associated factors) modulate the number, size,
and integrity of satellites in space and time and how such
modulation contributes to basal body function, transition
zone assembly, and ciliogenesis.
The role of the BBSome and the IFT complex
BBS is a ciliopathy characterized by renal and retinal
failure, obesity, polydactyly, diabetes, hypogenitalism,
and hypertension [75]. Seventeen causative genes have
been identified so far, and recent studies have begun to un-
ravel the role of BBS proteins in cilia homeostasis. As men-
tioned earlier, eight different proteins (BBS1, BBS2, BBS4,
BBS5, BBS7, BBS8, BBS9, and BBIP10) are required to
form a functional unit called the BBSome [74,76]. Intri-
guingly, the BBSome binds Rabin8, a GDP/GTP exchange
factor for Rab8a, and directly interacts with phospholipids,
suggesting that this complex likely mediates vesicular traf-
ficking during ciliogenesis (Figure 2) [74]. More recently,
another BBS subunit, BBS3/Arl6, an Arf-like GTPase, was
shown to be a major effector of the BBSome [77]. BBS3/
Arl6 recruits the BBSome to the membrane, where it as-
sembles a coatthat sorts proteins to the cilium. This coat
recognizes a unique ciliary localization signal found in sev-
eral ciliary membrane proteins, leading to their efficient
trafficking to the cilium [77,78]. Future biochemical and
biophysical studies will shed light on the structure of the
coatand the precise nature of the ciliary localization signal
it recognizes.
Although the BBSome is thought to play an important
role in sorting certain membrane proteins to the cilium,
neither this complex, nor its assembly factors or BBS3/
Arl6, is generally required for ciliogenesis, as depletion
or loss of some of these proteins does not severely im-
pair ciliation but rather leads to defective IFT transport
[79-82]. In addition, while BBS knockout mice (BBS1,
BBS2, BBS4, BBS6 or BBS7) display subtle phenotypes
[81,83-86], a loss of BBS7 in combination with a reduc-
tion in IFT function results in a more severe phenotype
[85], suggesting that the BBSome and the IFT complex
could function in a synergistic manner. These find-
ings have led to the speculation that the BBSome is only
Tsang and Dynlacht Cilia 2013, 2:9 Page 5 of 8
http://www.ciliajournal.com/content/2/1/9
responsible for transporting a subset of ciliary proteins,
whereas the IFT complex is more universally required for
all transport processes. Recently, an elegant study which
combines a whole-genome mutagenesis screen for mu-
tants with abnormal cilia formation, time-lapse micros-
copy, and bimolecular fluorescence complementation in
Caenorhabditis elegans showed that the BBSome acts on
the IFT complex by controlling its assembly and turn-
around in cilia [14]. The BBSome first interacts with the
IFT complex (Figure 2) and motor proteins to organize
them into a functional super-complex. This super-
complex undergoes anterograde transport to the ciliary
tip, and once there, the BBSome dissociates from the
IFT complex, unloading cargos during the process. The
BBSome then re-organizes the IFT complex and re-
loads new cargos for retrograde transport back to the
ciliary base. It remains to be determined if the role of
the BBSome in worms is mechanistically conserved in
higher eukaryotes, since subtle differences exist in the
ciliary structures, and not every BBS subunit is evolu-
tionarily conserved. Nevertheless, elucidating the mo-
lecular functions of the individual BBS and IFT
components would undoubtedly provide a better un-
derstanding of how these two complexes coordinately
promote cilia assembly.
Conclusions
Our knowledge of the architecture of the cilium and the
functions of individual ciliary components has expanded
considerably in the past 10 to 15 years. The use of
forward and reverse genetic screens, animal models,
system-wide proteomics, time-lapse microscopy, cryo-
electron microscopy, and new innovations in super-
resolution microscopy have led to rapid and unprece-
dented breakthroughs in the field, highlighted by many
landmark discoveries. Among these, CP110 and Cep290
have emerged as key players in the regulation of the cilia
assembly process. Despite our current knowledge of
their functions, important questions remain: is CP110
the protein responsible for the conversion of mother
centrioles (ciliogenesis incompetent) to basal bodies
(ciliogenesis competent), and how are the diverse func-
tions of Cep290 intertwined, if at all, in modulating cilia
assembly? We believe that the answers to these questions
lie in our ability to decipher and build upon the existing
ciliary protein interaction network (Figure 2). These stud-
ies should allow us to understand how this network con-
tributes to health and disease and to devise rational
therapeutic approaches for treating ciliopathies based on
these proteomic and genetic networks.
Abbreviations
IFT: Intraflagellar transport; BBS: Bardet-Biedl syndrome.
Competing interests
The authors declare that they have no competing interests.
Authorscontributions
WYT wrote the manuscript. WYT and BDD revised the manuscript. Both authors
read and approved the final manuscript.
Acknowledgements
We thank Sehyun Kim for critically reading the manuscript, and we apologize to
our colleagues whose findings could not be included due to space limitations.
WYT was a Canadian Institutes of Health Research New Investigator and a Fonds
de Recherché Santé Junior 1 Research Scholar. This work was supported by the
Canadian Institutes of Health Research (MOP-115033) to WYT and by an NIH
grant (1R01HD069647-01) to BDD.
Author details
1
Institut de recherches cliniques de Montréal, 110 avenue des Pins Ouest,
Montréal, QC H2W 1R7, Canada.
2
Faculté de Médecine, Université de
Montréal, Montréal, QC H3C 3J7, Canada.
3
Division of Experimental Medicine,
McGill University, Montréal, QC H3A 1A3, Canada.
4
Department of Pathology
and Cancer Institute, Smilow Research Center, New York University School of
Medicine, New York, NY 10016, USA.
Received: 29 April 2013 Accepted: 3 July 2013
References
1. Brito DA, Gouveia SM, Bettencourt-Dias M (2012) Deconstructing the
centriole: structure and number control. Curr Opin Cell Biol 24:413
2. Nigg EA, Stearns T (2011) The centrosome cycle: centriole biogenesis,
duplication and inherent asymmetries. Nat Cell Biol 13:11541160
3. Bettencourt-Dias M, Hildebrandt F, Pellman D, Woods G, Godinho SA (2011)
Centrosomes and cilia in human disease. Trends Genet 27:307315
4. Nigg EA, Raff JW (2009) Centrioles, centrosomes, and cilia in health and
disease. Cell 139:663678
5. Kobayashi T, Dynlacht BD (2011) Regulating the transition from centriole to
basal body. J Cell Biol 193:435444
6. Jana SC, Machado P, Bettencourt-Dias M (2012) A structural road map to
unveil basal body composition and assembly. EMBO J 31:519521
7. Kunimoto K, Yamazaki Y, Nishida T, Shinohara K, Ishikawa H, Hasegawa T, Okanoue
T, Hamada H, Noda T, Tamura A, Tsukita S (2012) Coordinated ciliary beating
requires Odf2-mediated polarization of basal bodies via basal feet. Cell 148:189200
8. Kodani A, Salome Sirerol-Piquer M, Seol A, Garcia-Verdugo JM, Reiter JF
(2013) Kif3a interacts with dynactin subunit p150 Glued to organize
centriole sub-distal appendages. EMBO J 32:597607
9. Hehnly H, Chen CT, Powers CM, Liu HL, Doxsey S (2012) The centrosome
regulates the Rab11-dependent recycling endosome pathway at
appendages of the mother centriole. Curr Biol 22:19441950
10. Avasthi P, Marshall WF (2012) Stages of ciliogenesis and regulation of ciliary
length. Differentiation 83:S30S42
11. Pedersen LB, Veland IR, Schroder JM, Christensen ST (2008) Assembly of
primary cilia. Dev Dyn 237:19932006
12. Reiter JF, Blacque OE, Leroux MR (2012) The base of the cilium: roles for
transition fibers and the transition zone in ciliary formation, maintenance
and compartmentalization. EMBO Rep 13:608618
13. Mukhopadhyay S, Wen X, Chih B, Nelson CD, Lane WS, Scales SJ, Jackson PK
(2010) TULP3 bridges the IFT-A complex and membrane phosphoinositides
to promote trafficking of G protein-coupled receptors into primary cilia.
Genes Dev 24:21802193
14. Wei Q, Zhang Y, Li Y, Zhang Q, Ling K, Hu J (2012) The BBSome controls IFT
assembly and turnaround in cilia. Nat Cell Biol 14:950957
15. Pedersen LB, Christensen ST (2012) Regulating intraflagellar transport.
Nat Cell Biol 14:904906
16. Liem KF Jr, Ashe A, He M, Satir P, Moran J, Beier D, Wicking C, Anderson KV
(2012) The IFT-A complex regulates Shh signaling through cilia structure
and membrane protein trafficking. J Cell Biol 197:789800
17. Ishikawa H, Marshall WF (2011) Ciliogenesis: building the cells antenna.
Nat Rev Mol Cell Biol 12:222234
18. Andersen JS, Wilkinson CJ, Mayor T, Mortensen P, Nigg EA, Mann M (2003)
Proteomic characterization of the human centrosome by protein correlation
profiling. Nature 426:570574
Tsang and Dynlacht Cilia 2013, 2:9 Page 6 of 8
http://www.ciliajournal.com/content/2/1/9
Published: 2 July 20136
19. Ostrowski LE, Blackburn K, Radde KM, Moyer MB, Schlatzer DM, Moseley A,
Boucher RC (2002) A proteomic analysis of human cilia: identification of
novel components. Mol Cell Proteomics 1:451465
20. Li JB, Gerdes JM, Haycraft CJ, Fan Y, Teslovich TM, May-Simera H, Li H,
Blacque OE, Li L, Leitch CC et al (2004) Comparative genomics identifies a
flagellar and basal body proteome that includes the BBS5 human disease
gene. Cell 117:541552
21. Graser S, Stierhof YD, Lavoie SB, Gassner OS, Lamla S, Le Clech M, Nigg EA
(2007) Cep164, a novel centriole appendage protein required for primary
cilium formation. J Cell Biol 179:321330
22. Kim J, Lee JE, Heynen-Genel S, Suyama E, Ono K, Lee K, Ideker T, Aza-Blanc P,
Gleeson JG (2010) Functional genomic screen for modulators of ciliogenesis
and cilium length. Nature 464:10481051
23. Spektor A, Tsang WY, Khoo D, Dynlacht BD (2007) Cep97 and CP110
suppress a cilia assembly program. Cell 130:678690
24. Lai Y, Chen B, Shi J, Palmer JN, Kennedy DW, Cohen NA (2011) Inflammation-
mediated upregulation of centrosomal protein 110, a negative modulator of
ciliogenesis, in patients with chronic rhinosinusitis. J Allergy Clin Immunol
128:12071215, e1201
25. Tsang WY, Bossard C, Khanna H, Peranen J, Swaroop A, Malhotra V,
Dynlacht BD (2008) CP110 suppresses primary cilia formation through its
interaction with CEP290, a protein deficient in human ciliary disease. Dev
Cell 15:187197
26. Tsang WY, Spektor A, Luciano DJ, Indjeian VB, Chen Z, Salisbury JL, Sanchez I,
Dynlacht BD (2006) CP110 cooperates with two calcium-binding proteins to
regulate cytokinesis and genome stability. Mol Biol Cell 17:34233434
27. Tsang WY, Spektor A, Vijayakumar S, Bista BR, Li J, Sanchez I, Duensing S,
Dynlacht BD (2009) Cep76, a centrosomal protein that specifically restrains
centriole reduplication. Dev Cell 16:649660
28. Kobayashi T, Tsang WY, Li J, Lane W, Dynlacht BD (2011) Centriolar kinesin
Kif24 interacts with CP110 to remodel microtubules and regulate
ciliogenesis. Cell 145:914925
29. Chen Z, Indjeian VB, McManus M, Wang L, Dynlacht BD (2002) CP110, a cell
cycle-dependent CDK substrate, regulates centrosome duplication in
human cells. Dev Cell 3:339350
30. Li J, Kim S, Kobayashi T, Liang FX, Korzeniewski N, Duensing S, Dynlacht BD
(2012) Neurl4, a novel daughter centriole protein, prevents formation of
ectopic microtubule organizing centers. EMBO Rep 13:547553
31. Al-Hakim AK, Bashkurov M, Gingras AC, Durocher D, Pelletier L (2012)
InteractionproteomicsidentifyNEURL4andtheHECTE3ligaseHERC2
as novel modulators of centrosome architecture. Mol Cell Proteomics
11:M111 014233
32. Kleylein-Sohn J, Westendorf J, Le Clech M, Habedanck R, Stierhof YD,
Nigg EA (2007) Plk4-induced centriole biogenesis in human cells. Dev
Cell 13:190202
33. Schmidt TI, Kleylein-Sohn J, Westendorf J, Le Clech M, Lavoie SB, Stierhof
YD, Nigg EA (2009) Control of centriole length by CPAP and CP110. Curr
Biol 19:10051011
34. Kohlmaier G, Loncarek J, Meng X, McEwen BF, Mogensen MM, Spektor A,
Dynlacht BD, Khodjakov A, Gonczy P (2009) Overly long centrioles and
defective cell division upon excess of the SAS-4-related protein CPAP. Curr
Biol 19:10121018
35. Tang CJ, Fu RH, Wu KS, Hsu WB, Tang TK (2009) CPAP is a cell-cycle
regulated protein that controls centriole length. Nat Cell Biol 11:825831
36. Delgehyr N, Rangone H, Fu J, Mao G, Tom B, Riparbelli MG, Callaini G, Glover
DM (2012) Klp10A, a microtubule-depolymerizing kinesin-13, cooperates with
CP110 to control Drosophila centriole length. Curr Biol 22:502509
37. Li J, DAngiolella V, Seeley ES, Kim S, Kobayashi T, Fu W, Campos EI, Pagano M,
Dynlacht BD (2013) USP33 regulates centrosome biogenesis via
deubiquitination of the centriolar protein CP110. Nature 495:255259
38. DAngiolella V, Donato V, Vijayakumar S, Saraf A, Florens L, Washburn MP,
Dynlacht B, Pagano M (2010) SCF (Cyc lin F) controls centrosome homeostasis
and mitotic fidelity through CP110 degradation. Nature 466:138142
39. Cao J, Shen Y, Zhu L, Xu Y, Zhou Y, Wu Z, Li Y, Yan X, Zhu X (2012)
miR-129-3p controls cilia assembly by regulating CP110 and actin
dynamics. Nat Cell Biol 14:697706
40. Goetz SC, Liem KF Jr, Anderson KV (2012) The spinocerebellar ataxia-
associated gene Tau tubulin kinase 2 controls the initiation of ciliogenesis.
Cell 151:847858
41. Jiang K, Toedt G, Montenegro Gouveia S, Davey NE, Hua S, van der Vaart B,
Grigoriev I, Larsen J, Pedersen LB, Bezstarosti K et al (2012) A Proteome-
wide screen for mammalian SxIP motif-containing microtubule plus-end
tracking proteins. Curr Biol 22:18001807
42. Schroder JM, Larsen J, Komarova Y, Akhmanova A, Thorsteinsson RI,
Grigoriev I, Manguso R, Christensen ST, Pedersen SF, Geimer S, Pedersen LB
(2011) EB1 and EB3 promote cilia biogenesis by several centrosome-related
mechanisms. J Cell Sci 124:25392551
43. Tanos BE, Yang HJ, Soni R, Wang WJ, Macaluso FP, Asara JM, Tsou MF (2013)
Centriole distal appendages promote membrane docking, leading to cilia
initiation. Genes Dev 27:163168
44. Joo K, Kim CG, Lee MS, Moon HY, Lee SH, Kim MJ, Kweon HS, Park WY,
Kim CH, Gleeson JG, Kim J (2013) CCDC41 is required for ciliary vesicle
docking to the mother centriole. Proc Natl Acad Sci U S A 110:59875992
45. Schmidt KN, Kuhns S, Neuner A, Hub B, Zentgraf H, Pereira G (2012) Cep164
mediates vesicular docking to the mother centriole during early steps of
ciliogenesis. J Cell Biol 199:10831101
46. Kuhns S, Schmidt KN, Reymann J, Gilbert DF, Neuner A, Hub B, Carvalho R,
Wiedemann P, Zentgraf H, Erfle H et al (2013) The microtubule affinity
regulating kinase MARK4 promotes axoneme extension during early
ciliogenesis. J Cell Biol 200:505522
47. Yoshimura S, Egerer J, Fuchs E, Haas AK, Barr FA (2007) Functional dissection
of Rab GTPases involved in primary cilium formation. J Cell Biol 178:363369
48. Chang J, Seo SG, Lee KH, Nagashima K, Bang JK, Kim BY, Erikson RL, Lee KW,
Lee HJ, Park JE, Lee KS (2013) Essential role of Cenexin1, but not Odf2, in
ciliogenesis. Cell Cycle 12:655662
49. Sonnen KF, Schermelleh L, Leonhardt H, Nigg EA (2012) 3D-structured
illumination microscopy provides novel insight into architecture of human
centrosomes. Biol Open 1:965976
50. Coppieters F, Lefever S, Leroy BP, De Baere E (2010) CEP290, a gene with
many faces: mutation overview and presentation of CEP290base. Hum
Mutat 31:10971108
51. Sayer JA, Otto EA, OToole JF, Nurnberg G, Kennedy MA, Becker C, Hennies HC,
Helou J, Attanasio M, Fausett BV et al (2006) The centrosomal protein
nephrocystin-6 is mutated in Joubert syndrome and activates transcription
factor ATF4. Nat Genet 38:674681
52. Valente EM, Silhavy JL, Brancati F, Barrano G, Krishnaswami SR, Castori M,
Lancaster MA, Boltshauser E, Boccone L, Al-Gazali L et al (2006) Mutations in
CEP290, which encodes a centrosomal protein, cause pleiotropic forms of
Joubert syndrome. Nat Genet 38:623625
53. Kim J, Krishnaswami SR, Gleeson JG (2008) CEP290 interacts with the
centriolar satellite component PCM-1 and is required for Rab8 localization
to the primary cilium. Hum Mol Genet 17:37963805
54. Otto EA, Loeys B, Khanna H, Hellemans J, Sudbrak R, Fan S, Muerb U,
OToole JF, Helou J, Attanasio M et al (2005) Nephrocystin-5, a ciliary IQ
domain protein, is mutated in Senior-Loken syndrome and interacts with
RPGR and calmodulin. Nat Genet 37:282288
55. Barbelanne M, Song J, Ahmadzai M, Tsang WY (2013) Pathogenic NPHP5
mutations impair protein interaction with Cep290, a prerequisite for
ciliogenesis. Hum Mol Genet 22:24822494
56. Sang L, Miller JJ, Corbit KC, Giles RH, Brauer MJ, Otto EA, Baye LM, Wen X,
Scales SJ, Kwong M et al (2011) Mapping the NPHP-JBTS-MKS protein
network reveals ciliopathy disease genes and pathways. Cell 145:513528
57. Schafer T, Putz M, Lienkamp S, Ganner A, Bergbreiter A, Ramachandran H,
Gieloff V, Gerner M, Mattonet C, Czarnecki PG et al (2008) Genetic and
physical interaction between the NPHP5 and NPHP6 gene products. Hum Mol
Genet 17:36553662
58. Chang B, Khanna H, Hawes N, Jimeno D, He S, Lillo C, Parapuram SK,
Cheng H, Scott A, Hurd RE et al (2006) In-frame deletion in a novel
centrosomal/ciliary protein CEP290/NPHP6 perturbs its interaction with
RPGR and results in early-onset retinal degeneration in the rd16 mouse.
Hum Mol Genet 15:18471857
59. Dawe HR, Smith UM, Cullinane AR, Gerrelli D, Cox P, Badano JL, Blair-Reid S,
Sriram N, Katsanis N, Attie-Bitach T et al (2007) The Meckel-Gruber Syndrome
proteins MKS1 and meckelin interact and are required for primary cilium
formation. Hum Mol Genet 16:173186
60. Craige B, Tsao CC, Diener DR, Hou Y, Lechtreck KF, Rosenbaum JL,
Witman GB (2010) CEP290 tethers flagellar transition zone microtubules
to the membrane and regulates flagellar protein content. J Cell Biol
190:927940
61. Kee HL, Dishinger JF, Blasius TL, Liu CJ, Margolis B, Verhey KJ (2012) A size-
exclusion permeability barrier and nucleoporins characterize a ciliary pore
complex that regulates transport into cilia. Nat Cell Biol 14:431437
Tsang and Dynlacht Cilia 2013, 2:9 Page 7 of 8
http://www.ciliajournal.com/content/2/1/9
62. Garcia-Gonzalo FR, Corbit KC, Sirerol-Piquer MS, Ramaswami G, Otto EA,
Noriega TR, Seol AD, Robinson JF, Bennett CL, Josifova DJ et al (2011) A
transition zone complex regulates mammalian ciliogenesis and ciliary
membrane composition. Nat Genet 43:776784
63. Chih B, Liu P, Chinn Y, Chalouni C, Komuves LG, Hass PE, Sandoval W,
Peterson AS (2012) A ciliopathy complex at the transition zone protects the
cilia as a privileged membrane domain. Nat Cell Biol 14:6172
64. Gorden NT, Arts HH, Parisi MA, Coene KL, Letteboer SJ, van Beersum SE,
Mans DA, Hikida A, Eckert M, Knutzen D et al (2008) CC2D2A is mutated in
Joubert syndrome and interacts with the ciliopathy-associated basal body
protein CEP290. Am J Hum Genet 83:559571
65. Wang WJ, Tay HG, Soni R, Perumal GS, Goll MG, Macaluso FP, Asara JM,
Amack JD, Bryan Tsou MF (2013) CEP162 is an axoneme-recognition protein
promoting ciliary transition zone assembly at the cilia base. Nat Cell Biol
15:591601
66. Zhang Q, Yu D, Seo S, Stone EM, Sheffield VC (2012) Intrinsic protein-protein
interaction-mediated and chaperonin-assisted sequential assembly of stable
Bardet-Biedl syndrome protein complex, the BBSome. J Biol Chem
287:2062520635
67. Seo S, Baye LM, Schulz NP, Beck JS, Zhang Q, Slusarski DC, Sheffield VC
(2010) BBS6, BBS10, and BBS12 form a complex with CCT/TRiC family
chaperonins and mediate BBSome assembly. Proc Natl Acad Sci U S A
107:14881493
68. Stowe TR, Wilkinson CJ, Iqbal A, Stearns T (2012) The centriolar satellite
proteins Cep72 and Cep290 interact and are required for recruitment of BBS
proteins to the cilium. Mol Biol Cell 23:33223335
69. Rachel RA, May-Simera HL, Veleri S, Gotoh N, Choi BY, Murga-Zamalloa C,
McIntyre JC, Marek J, Lopez I, Hackett AN et al (2012) Combining Cep290
and Mkks ciliopathy alleles in mice rescues sensory defects and restores
ciliogenesis. J Clin Invest 122:12331245
70. Barenz F, Mayilo D, Gruss OJ (2011) Centriolar satellites: busy orbits around
the centrosome. Eur J Cell Biol 90:983989
71. Kubo A, Sasaki H, Yuba-Kubo A, Tsukita S, Shiina N (1999) Centriolar
satellites: molecular characterization, ATP-dependent movement
toward centrioles and possible involvement in ciliogenesis. J Cell Biol
147:969980
72. Dammermann A, Merdes A (2002) Assembly of centrosomal proteins and
microtubule organization depends on PCM-1. J Cell Biol 159:255266
73. Lopes CA, Prosser SL, Romio L, Hirst RA, OCallaghan C, Woolf AS, Fry AM
(2011) Centriolar satellites are assembly points for proteins implicated in
human ciliopathies, including oral-facial-digital syndrome 1. J Cell Sci
124:600612
74. Nachury MV, Loktev AV, Zhang Q, Westlake CJ, Peranen J, Merdes A,
Slusarski DC, Scheller RH, Bazan JF, Sheffield VC, Jackson PK (2007) A core
complex of BBS proteins cooperates with the GTPase Rab8 to promote
ciliary membrane biogenesis. Cell 129:12011213
75. Sheffield VC (2010) The blind leading the obese: the molecular pathophysiology
of a human obesity syndrome. Trans Am Clin Climatol Assoc 121:172181,
discussion 181172
76. Loktev AV, Zhang Q, Beck JS, Searby CC, Scheetz TE, Bazan JF, Slusarski DC,
Sheffield VC, Jackson PK, Nachury MV (2008) A BBSome subunit links
ciliogenesis, microtubule stability, and acetylation. Dev Cell 15:854865
77. Jin H, White SR, Shida T, Schulz S, Aguiar M, Gygi SP, Bazan JF, Nachury MV
(2010) The conserved Bardet-Biedl syndrome proteins assemble a coat that
traffics membrane proteins to cilia. Cell 141:12081219
78. Berbari NF, Johnson AD, Lewis JS, Askwith CC, Mykytyn K (2008)
Identification of ciliary localization sequences within the third intracellular
loop of G protein-coupled receptors. Mol Biol Cell 19:15401547
79. Lechtreck KF, Johnson EC, Sakai T, Cochran D, Ballif BA, Rush J, Pazour GJ,
Ikebe M, Witman GB (2009) The Chlamydomonas reinhardtii BBSome is an
IFT cargo required for export of specific signaling proteins from flagella.
J Cell Biol 187:11171132
80. Yen HJ, Tayeh MK, Mullins RF, Stone EM, Sheffield VC, Slusarski DC (2006)
Bardet-Biedl syndrome genes are important in retrograde intracellular
trafficking and Kupffers vesicle cilia function. Hum Mol Genet 15:667677
81. Mykytyn K, Mullins RF, Andrews M, Chiang AP, Swiderski RE, Yang B, Braun T,
Casavant T, Stone EM, Sheffield VC (2004) Bardet-Biedl syndrome type 4
(BBS4)-null mice implicate Bbs4 in flagella formation but not global cilia
assembly. Proc Natl Acad Sci U S A 101:86648669
82. Shah AS, Farmen SL, Moninger TO, Businga TR, Andrews MP, Bugge K,
Searby CC, Nishimura D, Brogden KA, Kline JN et al (2008) Loss of Bardet-Biedl
syndrome proteins alters the morphology and function of motile cilia in airway
epithelia. Proc Natl Acad Sci U S A 105: 33803385
83. Davis RE, Swiderski RE, Rahmouni K, Nishimura DY, Mullins RF, Agassandian K,
Philp AR, Searby CC, Andrews MP, Thompson S et al (2007) A knockin mouse
model of the Bardet-Biedl syndrome 1 M390R mutation has cilia defects,
ventriculomegaly, retinopathy, and obesity. Proc Natl Acad Sci USA
104:1942219427
84. Nishimura DY, Fath M, Mullins RF, Searby C, Andrews M, Davis R, Andorf JL,
Mykytyn K, Swiderski RE, Yang B et al (2004) Bbs2-null mice have
neurosensory deficits, a defect in social dominance, and retinopathy
associated with mislocalization of rhodopsin. Proc Natl Acad Sci USA
101:1658816593
85. Zhang Q, Seo S, Bugge K, Stone EM, Sheffield VC (2012) BBS proteins
interact genetically with the IFT pathway to influence SHH-related
phenotypes. Hum Mol Genet 21:19451953
86. Fath MA, Mullins RF, Searby C, Nishimura DY, Wei J, Rahmouni K, Davis RE,
Tayeh MK, Andrews M, Yang B et al (2005) Mkks-null mice have a phenotype
resembling Bardet-Biedl syndrome. Hum Mol Genet 14:11091118
doi:10.1186/2046-2530-2-9
Cite this article as: Tsang and Dynlacht: CP110 and its network of
partners coordinately regulate cilia assembly. Cilia 2013 2:9.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Tsang and Dynlacht Cilia 2013, 2:9 Page 8 of 8
http://www.ciliajournal.com/content/2/1/9
... Immediately after ciliary vesicle docking, or concurrent with it, the ciliary axoneme is formed by extension from the distal end of the mother centriole/basal body [31]. Axoneme extension requires the elimination of the centriolar coiled coil protein of 110 kDa, in short CP110, and its interacting partner CEP97 from the distal end of the mother centriole [16,32,33]. CP110 and its associated partners form a cap at the distal ends of the two centrioles, which prevents microtubule elongation and axoneme extension [34,35]. ...
... Furthermore, additional proteins and CP110 binding partners are involved in the regulation of cilium assembly [32]. The removal of CP110 and ciliogenesis requires the serine/threonine kinase TTBK2 [43][44][45][46]. ...
... In addition, the inhibitory cap at the distal ends of the centrioles consisting of CP110 and other proteins must be removed, which is achieved by degrading CP110 [16,32,34,35,70]. CP110 is mainly degraded by the ubiquitin-dependent proteasome pathway in which the binding to the NEURL4-HERC2 complex plays an essential role [39,40,42,71]. ...
Article
Full-text available
Primary cilia are essential sensory organelles that develop when an inhibitory cap consisting of CP110 and other proteins is eliminated. The degradation of CP110 by the ubiquitin-dependent proteasome pathway mediated by NEURL4 and HYLS1 removes the inhibitory cap. Here, we investigated the suitability of rapamycin-mediated dimerization for centriolar recruitment and asked whether the induced recruitment of NEURL4 or HYLS1 to the centriole promotes primary cilia development and CP110 degradation. We used rapamycin-mediated dimerization with ODF2 to induce their targeted recruitment to the centriole. We found decreased CP110 levels in the transfected cells, but independent of rapamycin-mediated dimerization. By knocking down ODF2, we showed that ODF2 controls CP110 levels. The overexpression of ODF2 is not sufficient to promote the formation of primary cilia, but the overexpression of NEURL4 or HYLS1 is. The co-expression of ODF2 and HYLS1 resulted in the formation of tube-like structures, indicating an interaction. Thus, ODF2 controls primary cilia formation by negatively regulating the concentration of CP110 levels. Our data suggest that ODF2 most likely acts as a scaffold for the binding of proteins such as NEURL4 or HYLS1 to mediate CP110 degradation.
... Immediately after ciliary vesicle docking, or concurrently with it, the ciliary axoneme is formed by extension from the distal end of the mother centriole/basal body (Sánchez and Dynlacht, 2016). Axoneme extension requires the elimination of the Centriolar Coiled Coil Protein of 110 kDa, in short CP110, and its interacting partner CEP97 from the distal end of the mother centriole (Avidor-Reiss and Gopalakrishnan, 2013;Tsang and Dynlacht, 2013;Nagai et al., 2018). CP110 and its associated partners form a cap at the distal ends of the two centrioles, which prevents microtubule elongation and axoneme extension (Spektor et al., 2007;Schmidt et al., 2009). ...
... Furthermore, additional proteins and CP110 binding partners are involved in the regulation of cilium assembly (Tsang and Dynlacht, 2013). Removal of CP110 and ciliogenesis requires the serine/threonine kinase TTBK2 (Goetz et al., 2012;Cajanek and Nigg, 2014;Oda et al., 2014;Lo et al., 2019). ...
... In addition, the inhibitory cap at the distal ends of centrioles consisting of CP110 and other proteins must be removed, which is achieved by degrading CP110 (Spektor et al., 2007;Tsang et al., 2008;Schmidt et al., 2009;Avidor-Reiss and Gopalakrishnan, 2013;Tsang and Dynlacht, 2013). CP110 is mainly degraded by the ubiquitin-dependent proteasome pathway in which the binding to the NEURL4-HERC2 complex plays an essential role (D'Angiolella et al., 2010;Al-Hakim et al., 2012;Li et al., 2013;Gonçalves et al., 2021). ...
Preprint
Full-text available
Primary cilia are essential sensory organelles that develop when an inhibitory cap consisting of CP110 and other proteins is eliminated. Degradation of CP110 by the ubiquitin-dependent proteasome pathway mediated by NEURL4 and HYLS1 removes the inhibitory cap. Here, we investigated the suitability of rapamycin-mediated dimerization for centriolar recruitment and asked whether the induced recruitment of NEURL4 or HYLS1 to the centriole promotes primary cilia development and CP110 degradation. We used rapamycin-mediated dimerization with ODF2 to induce their targeted recruitment to the centriole. We found decreased CP110 levels in transfected cells, but independent of rapamycin-mediated dimerization. By knocking down ODF2, we show that ODF2 controls CP110 levels. Overexpression of ODF2 is not sufficient to promote the formation of primary cilia, but overexpression of NEURL4 or HYLS1 is. Co-expression of ODF2 and HYLS1 resulted in the formation of tube-like structures, indicating an interaction. Thus, ODF2 controls primary cilia formation by negatively regulating the concentration of CP110 levels. Our data suggest that ODF2 most likely acts as a scaffold for the binding of proteins such as NEURL4 or HYLS1 to mediate CP110 degradation. Summary NEURL4 and HYLS1 mediate the degradation of CP110 to allow cilium formation. We used rapamycin-mediated dimerization with ODF2 to recruit NEURL4 and HYLS1 to the centriole and show that ODF2 controls CP110 levels.
... Centriolar coiled-coil protein 110 (CP110), originally characterized as a cyclin-dependent kinase substrate, caps the distal ends of both mother and daughter centrioles, and is essential for centriole duplication and ciliogenesis initiation in mammalian cells. 7,[17][18][19][20][21][22][23] The removal of CP110 from mother centrioles is prerequisite for enabling axoneme outgrowth and appears to be one of the earliest steps in initiating ciliogenesis. 7,19-23 CP110 antagonizes the action of centrosomal protein of 290 kDa (CEP290) to suppress the recruitment of small GTPase Rab8a, resulting in the inhibition of cilia formation. ...
... 29,30 A number of studies have shown that CP110 removal from mother centrioles is required for ciliogenesis initiation after serum starvation. 7,[19][20][21][22][23] Here, we verified that serum deprivation induced CP110 removal from mother centrioles, promoted ciliogenesis, and decreased the protein level of CP110 in mouse embryonic fibroblast (MEF) cells (Supplementary information, Fig. S1). There are two major degradation pathways for protein turnover: the ubiquitin-proteasome pathway and autophagylysosome pathway. ...
... We then tested whether LC3 is associated with CP110 at mother centrioles, and detected the localization of LC3 with CP110, which decorates daughter centrioles under serum starvation. 7,[19][20][21][22][23] Our data revealed that few LC3 was detected at mother or daughter centrioles in most cells cultured with serum, while LC3 was clearly localized at only daughter centrioles with CP110 after serum deprivation (Fig. 2a). Inhibition of serum deprivation-induced autophagy with CQ led to the co-localization of LC3 with CP110 at both the mother and daughter centrioles (Fig. 2a). ...
Article
Full-text available
Primary cilia extending from mother centrioles are essential for vertebrate development and homeostasis maintenance. Centriolar coiled-coil protein 110 (CP110) has been reported to suppress ciliogenesis initiation by capping the distal ends of mother centrioles. However, the mechanism underlying the specific degradation of mother centriole-capping CP110 to promote cilia initiation remains unknown. Here, we find that autophagy is crucial for CP110 degradation at mother centrioles after serum starvation in MEF cells. We further identify NudC-like protein 2 (NudCL2) as a novel selective autophagy receptor at mother centrioles, which contains an LC3-interacting region (LIR) motif mediating the association of CP110 and the autophagosome marker LC3. Knockout of NudCL2 induces defects in the removal of CP110 from mother centrioles and ciliogenesis, which are rescued by wild-type NudCL2 but not its LIR motif mutant. Knockdown of CP110 significantly attenuates ciliogenesis defects in NudCL2 -deficient cells. In addition, NudCL2 morphants exhibit ciliation-related phenotypes in zebrafish, which are reversed by wild-type NudCL2, but not its LIR motif mutant. Importantly, CP110 depletion significantly reverses these ciliary phenotypes in NudCL2 morphants. Taken together, our data suggest that NudCL2 functions as an autophagy receptor mediating the selective degradation of mother centriole-capping CP110 to promote ciliogenesis, which is indispensable for embryo development in vertebrates.
... During cell cycle progression, CP110 is localized at the cap of mother and daughter centrioles and inhibits ciliary axoneme elongation. For proper primary cilium formation, CP110 must be removed from the cap of the mother centriole to initiate elongation of the ciliary axonemes (Tsang et al., 2008;Tsang and Dynlacht, 2013). In addition, BBS4, which is a component of the BBSome complex, is localized at the PCM and interacts with PCM-1, dynein, and intraflagellar transport protein (IFT) during the delivery of ciliary building blocks to ciliary axonemes (Figure 6-figure supplement 1A; Uytingco et al., 2019;Nachury et al., 2007). ...
... Rab8 is localized in the primary cilium and regulates ciliary membrane extension (Nachury et al., 2007;Blacque et al., 2018). Previous studies have also shown that Rab8 interacts with CP110 and BBS4 (Tsang et al., 2008;Tsang and Dynlacht, 2013;Nachury et al., 2007). Rab11 is enriched at the basal part of the primary cilium and recruits Rab8 to the basal body by interacting with Rabin8 (Westlake et al., 2011;Blacque et al., 2018). ...
Article
Full-text available
The gap junction complex functions as a transport channel across the membrane. Among gap junction subunits, gap junction protein α1 (GJA1) is the most commonly expressed subunit. A recent study showed that GJA1 is necessary for the maintenance of motile cilia; however, the molecular mechanism and function of GJA1 in ciliogenesis remain unknown. Here, we examined the functions of GJA1 during ciliogenesis in human retinal pigment epithelium-1 and Xenopus laevis embryonic multiciliated-cells. GJA1 localizes to the motile ciliary axonemes or pericentriolar regions beneath the primary cilium. GJA1 depletion caused malformation of both the primary cilium and motile cilia. Further study revealed that GJA1 depletion affected several ciliary proteins such as BBS4, CP110, and Rab11 in the pericentriolar region and basal body. Interestingly, CP110 removal from the mother centriole was significantly reduced by GJA1 depletion. Importantly, Rab11, a key regulator during ciliogenesis, was immunoprecipitated with GJA1, and GJA1 knockdown caused the mislocalization of Rab11. These findings suggest that GJA1 regulates ciliogenesis by interacting with the Rab11-Rab8 ciliary trafficking pathway.
... CP110 localizes at the distal tips of both mother and daughter centrioles ( Figure 6E in Kleylein-Sohn et al. 2007) (Yadav et al. 2016). CP110 acts as a suppressor of cilia formation by forming a cap over the centriole microtubule tips, restraining the microtubules from growing the cilia/axoneme (Reviewed in Tsang and Dynlacht 2013). As a result, CP110 must be removed from the mother centriole tip for the primary cilium to form (Shen et al. 2022). ...
Article
Full-text available
Centrosomes play an important role in the microtubule organization of a cell. The sperm's specialized centrosome consists of the canonical barrel-shaped proximal centriole, the funnel-shaped distal centriole, and the pericentriolar material known as striated columns (or segmented columns). Here, we examined the localization of the centriole proteins CEP135 and CP110 in cattle and human spermatozoa. In canonical centrioles, CP110 is a centriole tip protein that controls cilia formation, while CEP135 is a structural protein essential for constructing the centriole. In contrast, we found antibodies recognizing CEP135 and CP110 label near the proximal and distal centrioles at the expected location of the striated columns and capitulum in cattle and humans in an antibody and species-specific way. These findings provide a pathway to understanding the unique functions of spermatozoan centrosome.
... Cells that withdraw into G0 phase maintain a more stable cilium (Ho and Tucker, 1989;Tucker et al., 1979a). Ciliogenesis starts with MC to BB morphogenesis and involves recruitment of structural and regulatory proteins (Čajánek and Nigg, 2014;Graser et al., 2007;Mönnich et al., 2018;Tsang and Dynlacht, 2013). DC proteins regulate centriole duplication in proliferating cells (Mahjoub et al., 2010;Zou et al., 2005), but in quiescent and differentiated cells they promote ciliogenesis from the MC (Betleja et al., 2018;Gottardo et al., 2015;Ogungbenro et al., 2018). ...
Article
Full-text available
The centrosome is an evolutionarily conserved, ancient organelle whose role in cell division was first described over a century ago. The structure and function of the centrosome as a microtubule-organizing center, and of its extracellular extension - the primary cilium - as a sensory antenna, have since been extensively studied, but the role of the cilium-centrosome axis in cell fate is still emerging. In this Opinion piece, we view cellular quiescence and tissue homeostasis from the vantage point of the cilium-centrosome axis. We focus on a less explored role in the choice between distinct forms of mitotic arrest - reversible quiescence and terminal differentiation, which play distinct roles in tissue homeostasis. We outline evidence implicating the centrosome-basal body switch in stem cell function, including how the cilium-centrosome complex regulates reversible versus irreversible arrest in adult skeletal muscle progenitors. We then highlight exciting new findings in other quiescent cell types that suggest signal-dependent coupling of nuclear and cytoplasmic events to the centrosome-basal body switch. Finally, we propose a framework for involvement of this axis in mitotically inactive cells and identify future avenues for understanding how the cilium-centrosome axis impacts central decisions in tissue homeostasis.
... The early steps of ciliogenesis, termed licensing, can be induced in cell culture via serum starvation. This initiates the recruitment of CEP164 to the mother centriole followed by recruitment of TTBK2 and later "uncapping" or loss of CP110 (Tsang and Dynlacht, 2013;Yadav et al., 2016). The release of CP110 is followed by ciliary vesicle docking and axoneme extension (Yadav et al., 2016). ...
Article
Full-text available
ELMODs are a family of three mammalian paralogs that display GTPase activating protein (GAP) activity towards a uniquely broad array of ADP-ribosylation factor (ARF) family GTPases that includes ARF-like (ARL) proteins. ELMODs are ubiquitously expressed in mammalian tissues, highly conserved across eukaryotes, and ancient in origin, being present in the last eukaryotic common ancestor. We described functions of ELMOD2 in immortalized mouse embryonic fibroblasts (MEFs) in the regulation of cell division, microtubules, ciliogenesis, and mitochondrial fusion. Here, using similar strategies with the paralogs ELMOD1 and ELMOD3, we identify novel functions and locations of these cell regulators and compare them to those of ELMOD2, allowing determination of functional redundancy among the family members. We found strong similarities in phenotypes resulting from deletion of either Elmod1 or Elmod3 and marked differences from those arising in Elmod2 deletion lines. Deletion of either Elmod1 or Elmod3 results in the decreased ability of cells to form primary cilia, loss of a subset of proteins from cilia, and accumulation of some ciliary proteins at the Golgi, predicted to result from compromised traffic from the Golgi to cilia. These phenotypes are reversed upon activating mutant expression of either ARL3 or ARL16, linking their roles to ELMOD1/3 actions.
Article
Full-text available
Multiciliated cells (MCCS) form bundles of cilia and their activities are essential for the proper development and physiology of many organ systems. Not surprisingly, defects in MCCs have profound consequences and are associated with numerous disease states. Here, we discuss the current understanding of MCC formation, with a special focus on the genetic and molecular mechanisms of MCC fate choice and differentiation. Furthermore, we cast a spotlight on the use of zebrafish to study MCC ontogeny and several recent advances made in understanding MCCs using this vertebrate model to delineate mechanisms of MCC emergence in the developing kidney.
Article
LUZP1 (leucine zipper protein 1) was first described as being important for embryonic development. Luzp1 null mice present defective neural tube closure and cardiovascular problems, which cause perinatal death. Since then, LUZP1 has also been implicated in the etiology of diseases like the 1p36 and the Townes-Brocks syndromes, and the molecular mechanisms involving this protein started being uncovered. Proteomics studies placed LUZP1 in the interactomes of the centrosome-cilium interface, centriolar satellites, and midbody. Concordantly, LUZP1 is an actin and microtubule-associated protein, which localizes to the centrosome, the basal body of primary cilia, the midbody, actin filaments and cellular junctions. LUZP1, like its interactor EPLIN, is an actin-stabilizing protein and a negative regulator of primary cilia formation. Moreover, through the regulation of actin, LUZP1 has been implicated in the regulation of cell cycle progression, cell migration and epithelial cell apical constriction. This review discusses the latest findings concerning LUZP1 molecular functions and implications in disease development.
Article
Full-text available
Despite the importance of cilia in cell signaling and motility, the molecular mechanisms regulating cilium formation remain incompletely understood. Herein, we characterize enkurin domain-containing protein 1 (ENKD1) as a novel centrosomal protein that mediates the removal of centriolar coiled-coil protein 110 (CP110) from the mother centriole to promote ciliogenesis. We show that Enkd1 knockout mice possess ciliogenesis defects in multiple organs. Super-resolution microscopy reveals that ENKD1 is a stable component of the centrosome throughout the ciliogenesis process. Simultaneous knockdown of ENKD1 and CP110 significantly reverses the ciliogenesis defects induced by ENKD1 depletion. Protein interaction analysis shows that ENKD1 competes with centrosomal protein 97 (CEP97) in binding to CP110. Depletion of ENKD1 enhances the CP110-CEP97 interaction and detains CP110 at the mother centriole. These findings thus identify ENKD1 as a centrosomal protein and uncover a novel mechanism controlling CP110 removal from the mother centriole for the initiation of ciliogenesis.
Article
Full-text available
The transition zone is a specialized compartment found at the base of cilia, adjacent to the centriole distal end, where axonemal microtubules are heavily crosslinked to the surrounding membrane to form a barrier that gates the ciliary compartment. A number of ciliopathy molecules have been found to associate with the transition zone, but factors that directly recognize axonemal microtubules to specify transition zone assembly at the cilia base remain unclear. Here, through quantitative centrosome proteomics, we identify an axoneme-associated protein, CEP162 (KIAA1009), tethered specifically at centriole distal ends to promote transition zone assembly. CEP162 interacts with core transition zone components, and mediates their association with microtubules. Loss of CEP162 arrests ciliogenesis at the stage of transition zone assembly. Abolishing its centriolar tethering, however, allows CEP162 to stay on the growing end of the axoneme and ectopically assemble transition zone components at cilia tips. This generates extra-long cilia with strikingly swollen tips that actively release ciliary contents into the extracellular environment. CEP162 is thus an axoneme-recognition protein pre-tethered at centriole distal ends before ciliogenesis to promote and restrict transition zone formation specifically at the cilia base.
Article
Full-text available
We identified Xenopus pericentriolar material-1 (PCM-1), which had been reported to constitute pericentriolar material, cloned its cDNA, and generated a specific pAb against this molecule. Immunolabeling revealed that PCM-1 was not a pericentriolar material protein, but a specific component of centriolar satellites, morphologically characterized as electron-dense granules, ∼70–100 nm in diameter, scattered around centrosomes. Using a GFP fusion protein with PCM-1, we found that PCM-1–containing centriolar satellites moved along microtubules toward their minus ends, i.e., toward centrosomes, in live cells, as well as in vitro reconstituted asters. These findings defined centriolar satellites at the molecular level, and explained their pericentriolar localization. Next, to understand the relationship between centriolar satellites and centriolar replication, we examined the expression and subcellular localization of PCM-1 in ciliated epithelial cells during ciliogenesis. When ciliogenesis was induced in mouse nasal respiratory epithelial cells, PCM-1 immunofluorescence was markedly elevated at the apical cytoplasm. At the electron microscopic level, anti–PCM-1 pAb exclusively labeled fibrous granules, but not deuterosomes, both of which have been suggested to play central roles in centriolar replication in ciliogenesis. These findings suggested that centriolar satellites and fibrous granules are identical novel nonmembranous organelles containing PCM-1, which may play some important role(s) in centriolar replication.
Article
Full-text available
The initiation of primary cilium assembly entails the docking of ciliary vesicles presumably derived from the Golgi complex to the distal end of the mother centriole. Distal appendages, which anchor the mother centriole to the plasma membrane, are thought to be involved in the docking process. However, little is known about the molecular players and mechanisms that mediate the vesicle-centriole association. Here we report that coiled-coil domain containing 41 (CCDC41) is required for the docking of ciliary vesicles. CCDC41 specifically localizes to the distal end of the mother centriole and interacts with centrosomal protein 164 (Cep164), a distal appendage component. In addition, a pool of CCDC41 colocalizes with intraflagellar transport protein 20 (IFT20) subunit of the intraflagellar transport particle at the Golgi complex. Remarkably, knockdown of CCDC41 inhibits the recruitment of IFT20 to the centrosome. Moreover, depletion of CCDC41 or IFT20 inhibits ciliogenesis at the ciliary vesicle docking step, whereas intraflagellar transport protein 88 (IFT88) depletion interferes with later cilium elongation steps. Our results suggest that CCDC41 collaborates with IFT20 to support the vesicle-centriole association at the onset of ciliogenesis.
Article
Full-text available
Centrosome duplication is critical for cell division, and genome instability can result if duplication is not restricted to a single round per cell cycle. Centrosome duplication is controlled in part by CP110, a centriolar protein that positively regulates centriole duplication while restricting centriole elongation and ciliogenesis. Maintenance of normal CP110 levels is essential, as excessive CP110 drives centrosome over-duplication and suppresses ciliogenesis, whereas its depletion inhibits centriole amplification and leads to highly elongated centrioles and aberrant assembly of cilia in growing cells. CP110 levels are tightly controlled, partly through ubiquitination by the ubiquitin ligase complex SCF(cyclin F) during G2 and M phases of the cell cycle. Here, using human cells, we report a new mechanism for the regulation of centrosome duplication that requires USP33, a deubiquitinating enzyme that is able to regulate CP110 levels. USP33 interacts with CP110 and localizes to centrioles primarily in S and G2/M phases, the periods during which centrioles duplicate and elongate. USP33 potently and specifically deubiquitinates CP110, but not other cyclin-F substrates. USP33 activity antagonizes SCF(cyclin F)-mediated ubiquitination and promotes the generation of supernumerary centriolar foci, whereas ablation of USP33 destabilizes CP110 and thereby inhibits centrosome amplification and mitotic defects. To our knowledge, we have identified the first centriolar deubiquitinating enzyme whose expression regulates centrosome homeostasis by countering cyclin-F-mediated destruction of a key substrate. Our results point towards potential therapeutic strategies for inhibiting tumorigenesis associated with centrosome amplification.
Article
Full-text available
Mutations in the human NPHP5 gene cause retinal and renal disease, but the precise mechanism by which NPHP5 functions is not understood. We report that NPHP5 is a centriolar protein whose depletion inhibits an early step of ciliogenesis, a phenotype reminiscent of Cep290 loss and contrary to IFT88 loss. Functional dissection of NPHP5 interactions with Cep290 and CaM reveals a requirement of the former for ciliogenesis, while the latter prevents NPHP5 self-aggregation. Disease-causing mutations lead to truncated products unable to bind Cep290 and localize to centrosomes, thereby compromising cilia formation. In contrast, a modifier mutation cripples CaM binding but has no overt effect on ciliogenesis. Drugs that antagonize negative regulators of the ciliogenic pathway can rescue ciliogenesis in cells depleted of NPHP5, with response profiles similar to those of Cep290- but not IFT88-depleted cells. Our results uncover the underlying molecular basis of disease and provide novel insights into mitigating NPHP5 deficiency.
Article
Full-text available
Despite the critical contributions of cilia to embryonic development and human health, key regulators of cilia formation await identification. In this paper, a functional RNA interference-based screen linked 30 novel protein kinases with ciliogenesis. Of them, we have studied the role of the microtubule (MT)-associated protein/MT affinity regulating kinase 4 (MARK4) in depth. MARK4 associated with the basal body and ciliary axoneme in human and murine cell lines. Ultrastructural and functional analyses established that MARK4 kinase activity was required for initiation of axoneme extension. We identified the mother centriolar protein ODF2 as an interaction partner of MARK4 and showed that ODF2 localization to the centriole partially depended on MARK4. Our data indicated that, upon MARK4 or ODF2 knockdown, the ciliary program arrested before the complete removal of the CP110-Cep97 inhibitory complex from the mother centriole, suggesting that these proteins act at this level of axonemal extension. We propose that MARK4 is a critical positive regulator of early steps in ciliogenesis.
Article
Full-text available
Formation of cilia, microtubule-based structures that function in propulsion and sensation, requires Kif3a, a subunit of Kinesin II essential for intraflagellar transport (IFT). We have found that, Kif3a is also required to organize centrioles. In the absence of Kif3a, the subdistal appendages of centrioles are disorganized and lack p150(Glued) and Ninein. Consequently, microtubule anchoring, centriole cohesion and basal foot formation are abrogated by loss of Kif3a. Kif3a localizes to the mother centriole and interacts with the Dynactin subunit p150(Glued). Depletion of p150(Glued) phenocopies the effects of loss of Kif3a, indicating that Kif3a recruitment of p150(Glued) is critical for subdistal appendage formation. The transport functions of Kif3a are dispensable for subdistal appendage organization as mutant forms of Kif3a lacking motor activity or the motor domain can restore p150(Glued) localization. Comparison to cells lacking Ift88 reveals that the centriolar functions of Kif3a are independent of IFT. Thus, in addition to its ciliogenic roles, Kif3a recruits p150(Glued) to the subdistal appendages of mother centrioles, critical for centrosomes to function as microtubule-organizing centres.
Article
Nephronophthisis (NPHP), Joubert (JBTS), and Meckel-Gruber (MKS) syndromes are autosomal-recessive ciliopathies presenting with cystic kidneys, retinal degeneration, and cerebellar/neural tube malformation. Whether defects in kidney, retinal, or neural disease primarily involve ciliary, Hedgehog, or cell polarity pathways remains unclear. Using high-confidence proteomics, we identified 850 interactors copurifying with nine NPHP/JBTS/MKS proteins and discovered three connected modules: "NPHP1-4-8" functioning at the apical surface, "NPHP5-6" at centrosomes, and "MKS" linked to Hedgehog signaling. Assays for ciliogenesis and epithelial morphogenesis in 3D renal cultures link renal cystic disease to apical organization defects, whereas ciliary and Hedgehog pathway defects lead to retinal or neural deficits. Using 38 interactors as candidates, linkage and sequencing analysis of 250 patients identified ATXN10 and TCTN2 as new NPHP-JBTS genes, and our Tctn2 mouse knockout shows neural tube and Hedgehog signaling defects. Our study further illustrates the power of linking proteomic networks and human genetics to uncover critical disease pathways.