ArticlePDF AvailableLiterature Review

The development and functions of multiciliated epithelia

Authors:

Abstract

Multiciliated cells are epithelial cells that are in contact with bodily fluids and are required for the proper function of major organs including the brain, the respiratory system and the reproductive tracts. Their multiple motile cilia beat unidirectionally to remove particles of external origin from their surface and/or drive cells or fluids into the lumen of the organs. Multiciliated cells in the brain are produced once, almost exclusively during embryonic development, whereas in respiratory tracts and oviducts they regenerate throughout life. In this Review, we provide a cell-to-organ overview of multiciliated cells and highlight recent studies that have greatly increased our understanding of the mechanisms driving the development and function of these cells in vertebrates. We discuss cell fate determination and differentiation of multiciliated cells, and provide a comprehensive account of their locations and functions in mammals.
Microorganisms commonly use efficient motile 9 + 2
cilia to move around1–3. These cilia also cover numerous
epithelia in vertebrates, in which they drive the move-
ment of extracellular particles or generate fluid flow.
These motile cilia are organized around an axoneme
consisting of nine microtubule doublets arranged in a
circle around two singlet microtubules4. The formation
of an axoneme is nucleated by a basal body, which is
composed of triplets of microtubules in a nine-fold sym-
metrical arrangement (related to the centrioles found
in the centrosome) that is docked at the plasma mem-
brane5. Dynein ATPase motor proteins cause adjacent
axonemal microtubules to slide over each other to bend
the cilium. Each beat of a cilium consists of a fast effec-
tive stroke that propels fluids and a slow, curved, recov-
ery stroke against the flow of the fluid that brings the
cilium back to its original location4. The central pair of
microtubules is suggested to be necessary for this planar
stereotypical waveform of ciliary beating6.
Generation of cilium-powered directional fluid flow
at the tissue level requires a dense array of cilia beating
unidirectionally at a high frequency (5–35 Hz). The term
multiciliated cell commonly refers to a cell that extends
multiple 9 + 2 cilia from its apical surface. In mammals,
multiciliated cells cover the lumen of the ear–nose–
throat sphere, the lungs, the brain ventricles and part
of the female and male reproductive tracts. Each multi-
ciliated cell nucleates from 30 to 300 cilia, depending
on the organ. Multiciliated precursors with only one
centrosome must therefore amplify up to 300 cen-
trioles to support cilia nucleation7–16. This amplification
occurs through the formation of intermediate structures,
deuterosomes, which are composed of centrosome-
related elements. Deuterosomes support massive cen-
triole formation downstream of a molecular cascade that
is shared with the cell cycle centrosome duplication pro-
gramme17–20. Once docked at the apical plasma membrane,
the new centrioles become basal bodies and acquire a
rotational polarity that determines the direction of planar
cilia beating. Basal bodies are distributed evenly across
the apical membrane in the airways and reproductive
organs. In the brain, however, basal bodies are clustered
towards the downstream flow of cerebrospinal fluid
(CSF), thereby defining its translational polarity21. Ventricle
epithelia are composed mainly of multiciliated cells in the
brain, whereas in the airways and oviducts, multiciliated
cells are interspersed with epithelial secretory cells.
Abnormalities in centriole formation or migration,
the orientation of cilia beating or its frequency result in
pathological conditions. The massive cellular changes
taking place in differentiating multiciliated cells have
been documented by extensive electron microscopy
studies. Recently, there has been renewed interest in
multiciliated cells owing to the development of ‘omics’
methodologies and progress in live and super-resolution
microscopy. In this Review, we provide a cell-to-organ
overview of a century of data on multiciliated cells in
verte brates. We discuss multiciliated cell fate determin-
ants and describe both the dynamics and molecular
events at play during the differentiation of a cell contain-
ing one single centrosome into a polarized cell with tens
of centriolar structures nucleating a functional motile
ciliary tuft. Last, we exhaustively discuss the locations
and functions of multiciliated cells in mammals,
École Normale Supérieure,
PSL Research University,
CNRS, Inserm, Institut de
Biologie de l’Ecole Normale
Supérieure (IBENS),
F-75005Paris, France.
nathalie.spassky@ens.fr;
alice.meunier@ens.fr
doi:10.1038/nrm.2017.21
Published online 12 Apr 2017
Apical plasma membrane
In epithelial cells, the
membrane located towards the
lumen in a body tube or cavity.
Rotational polarity
The orientation of ciliary
beating.
Tr a n s l a t i o n a l p o l a r i t y
Planar asymmetric localization
of clusters of basal bodies
onthe apical area of
multiciliated cells.
Ven tri cle ep ith eli a
Single layers of cells lining the
surface of the brain ventricles.
Secretory cells
Non-ciliated cells that secrete
mucus into the respiratory
tract, or fluids rich in nutrients
into the reproductive tracts.
The development and functions
ofmulticiliated epithelia
Nathalie Spassky and Alice Meunier
Abstract | Multiciliated cells are epithelial cells that are in contact with bodily fluids and are
required for the proper function of major organs including the brain, the respiratory system and the
reproductive tracts. Their multiple motile cilia beat unidirectionally to remove particles of external
origin from their surface and/or drive cells or fluids into the lumen of the organs. Multiciliated cells
in the brain are produced once, almost exclusively during embryonic development, whereas in
respiratory tracts and oviducts they regenerate throughout life. In this Review, we provide a
cell-to-organ overview of multiciliated cells and highlight recent studies that have greatly
increased our understanding of the mechanisms driving the development andfunction of these
cells in vertebrates. We discuss cell fate determination and differentiation of multiciliated cells,
andprovide a comprehensive account of their locations and functions inmammals.
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
1
REVIEWS
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Basal cell
Undifferentiated airway cells
localized deep in the
respiratory epithelium
andcharacterized by the
expre ssion o f p63 an d SOX2.
Coiled-coil domain
α-Helix-containing structural
domain in proteins that are
coiled together to form dimers
or trimers.
andhighlight atypical and understudied multiciliated
cells that would be of interest for further studies.
Multiciliated cell fate determination
Multiciliated cells are either mostly produced during
development and exist for years (in the brain) or con-
tinuously regenerated throughout life (in the airways
and reproductive organs). Identification of the progen-
itors of multiciliated cells is a first step towards under-
standing the mechanisms that direct multiciliated cell
fate determination. Recent studies have shown that in
different organs, multiciliogenesis shares common fate
determination mechanisms.
The progenitors of multiciliated cells. In the airways,
uterus and oviducts, multiciliated cells are regener-
ated throughout life22–24. In the airways, multiciliated
cells originate from p63 (also known as TP63)- and
SOX2-expressing progenitors. p63 and SOX2 are
transcription factors that are highly expressed in the
basal cell lineage and are required for the development
and regeneration of ciliated cells25–27. During post-
natal growth and adulthood, multiciliated cells derive
from basal cells or secretory cells, depending on tissue
identity and cellularcontext28.
In the adult mammalian brain, multiciliated ependy-
mal cells form a continuous layer that lines the walls of
the lateral, third and fourth ventricles. These cells are
formed only once during development16; their replace-
ment during ageing or after injury is minimal29,30. Cell-
fate tracing experiments demonstrated that radial glia
are the progenitors of ependymal cells in the mouse
brain16. Although radial glia are heterogeneous and
show restricted potential to produce various subtypes
of neurons in some parts of the brain31, no regional dif-
ferences among multiciliated progenitor cells have yet
been identified.
The progenitors in other multiciliated organs are yet
to be characterized.
Cell fate determination. It is likely that multiple inter-
cellular signalling pathways cooperate to specify the
transformation of progenitor cells into multiciliated
cell precursors. In many epithelial tissues, this choice
seems to be the default fate for progenitor cells. In the
zebrafish pronephros, mammalian airways and embry-
onic epidermis of the amphibian Xenopus laevis, the
inhibition of Notch signalling controls the balance
between the formation of ciliated cells and other cell
types32–38. Activation of the Notch pathway by genetic
and pharmacological approaches silences ciliation in
cells, which then continue to expand and differentiate
into secretory cells. Conversely, inhibition of Notch
signalling leads to differentiation into ciliated cells at
the expense of secretory cells34,35,37–41. In X.laevis epi-
dermis and human airways, the microRNA miR-449
promotes multiciliogenesis by directly repressing
the Notch 1 pathway, which is induced by its ligand
Delta-like protein1 (REF.33) (FIG.1). These regulatory
processes are maintained in adult airways, as ciliated
cells are produced directly from basal (progenitor) cells
expressing p63 or indirectly from Notch 2-expressing
secretory cells42–45. The Notch ligand is provided directly
by the basal cells to maintain the pool of their daughter
Notch 2-expressing secretory cells, which otherwise
are converted into ciliated cells, as shown by the inhib-
ition of the Notch ligands jagged 1 (JAG1) and JAG2
(REFS45,46). Although the upstream signals still need
to be identified, similar inhibition of the Notch pathway
is required to arrest the proliferation of progenitor cells
and to specify their differentiation into multiciliated
cells in the brain and oviducts23,47.
The bone morphogenetic protein (BMP) and Notch
pathways might interact to control cell fate, asBMP
ligands inhibit multiciliated cell specification in the
X.laevis epidermis and in mammalian airways and
embryonic stem cells48–50. Conversely, the transforming
growth factor-β (TGFβ) regulators SMAD6 (also known
as MADH6) and SMAD7 (also known as MADH7)
promote the formation of multiciliated cells from
embryonic stem cells51.
The most-upstream activators of the multiciliated
cell transcriptional programme found so far are gem-
inin coiled-coil domain-containing protein 1 (GEMC1)
and multicilin (FIG.1). These proteins have a central
coiled-coil domain and were originally identified as
regulators of DNA synthesis52,53. Multicilin can inter-
act with geminin and inhibit its function as a negative
regulator of DNA replication54, and GEMC1 interacts
with CDC45 and DNA topoisomerase 2-binding pro-
tein 1 to promote DNA replication52. It is currently
unknown whether, as regulators of DNA replication,
they are directly involved in multiciliogenesis and,
ifso, how. GEMC1 and multicilin seem to initiate simi-
lar transcriptional programmes that are required for
multiciliated cell differentiation36,55–57. Notch inhibition
leads to the activation of multicilin, which is required
for the formation of multiciliated cells in the X.laevis
Figure 1 | Mammalian multiciliated cell fate determination. Notch 1-induced
proliferation of progenitor cells is inhibited in human airways by the microRNAs miR-34
and miR-449. Notch 1 inhibition leads to the activation of the master regulators of
multiciliogenesis, geminin coiled-coil domain-containing protein 1 (GEMC1) and
multicilin, which, together with the transcription factors E2F4 or E2F5, commit
progenitor cells towards multiciliation and induce differentiation through activation of
p73 and the motile ciliogenic pathway (MYB, forkhead box protein J1 (FOXJ1), regulatory
factor X 2 (RFX2) and RFX3), which triggers basal body amplification, active remodelling
of the cytoskeleton and ciliogenesis. EDM, E2F4– or E2F5–DP1–multicilin.
Proliferation of progenitor cells
Multiciliated cell precursor
Motile
ciliogenic
pathway
Multicilin,
E2F4, E2F5,
DP1 (EDM complex)
Notch1
GEMC1
miR-34, miR-449
(in human airways)
p73
FOXJ1
RFX3
RFX2
MYB
Nature Reviews | Molecular Cell Biology
REVIEWS
2
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Procentriole
Immature centriole or basal
body in the process of growth.
epidermis and kidney, and in mouse and human air-
ways36,58,59. The expression of GEMC1 precedes that
of multicilin, and both GEMC1 and multicilin con-
trol the generation of multiciliated cells in the mouse
brain47. GEMC1 is required for multicilin expression in
zebrafish and X.laevis60. The trachea and oviducts of
GEMC1-knockout mice show highly reduced expres-
sion of multicilin and genes required for centriole
biogenesis56, suggesting that GEMC1 acts upstream
of multicilin. Mice lacking GEMC1 show impaired
growth, develop hydrocephalus and are infertile.
The trachea, oviducts and brain of these mice lack
multiciliatedcells56,57.
Multicilin promotes the transcription of key genes
that are required for multiciliogenesis through inter-
actions with the E2F4 or E2F5 transcription factors
and their cofactor, DP1 (the E2F4 or E2F5–DP1
multicilin ternary complex is referred to as the EDM
complex)55 (FIG.1). Although the interaction of GEMC1
with E2F5–DPI is controversial56,60, GEMC1 activity is
enhanced by E2F5 and is required to activate E2F5
target genes in tissues that give rise to multiciliated
cells55,43. Loss of E2F4 and E2F5 in mice impairs the
development of multiciliated cells in the airways and
the efferent ducts61,62. Interestingly, the multicilin gene
is located on chromosome 5q11.2 in humans and
chromo some 13D2.2 in mice; both loci harbour other
key regulators of multi ciliated cell formation, includ-
ing cyclinO (CCNO), CDC20B and mir-449a, mir-449b
and mir-449c33,58,63,64.
Multiciliated cell differentiation
In the mammalian brain, progenitor cells lose their
radial glial morphology and molecular characteris-
tics, and acquire multiciliated ependymal cell features
(expressing SIX3, CD24 and S100β) and a cuboidal
morphology16,65,66. This tr ansformation requ ires
active remodelling of the cytoskeleton and the local-
ization of tight junction proteins such as N-cadherin
on the cell membrane through the adaptor protein
ankyrin3 and the Hippo signalling pathway effector
Ye s - a s s o c i a t e d p r o t e i n ( YA P ) 67,68. In the airways and
in X.laevis epidermis, BMP signalling, the small Rho
GTPase RHOA and actin-based cell-autonomous activ-
ity dependent on the actin elongator formin1 contrib-
ute to the apical migration of basal cells to the outer
layer, where they intercalate among other cells and
resumedifferentiation27,48,69,70.
Massive production of centrioles. Mu lticiliated cell pre-
cursors, which contain one centrosome with a mother
and a daughter centriole, produce from 30 to 300 new
centrioles during the course of their differentiation.
These centrioles provide templates for the subsequent
growth of the corresponding number of motilecilia.
During canonical centrosome duplication in divid-
ing progenitor cells, mother and daughter centrioles
guide the formation of only one new centriole on their
walls. Centrosomal protein of 63 kDa (CEP63) recruits
CEP152, and they form a complex on each centrosomal
centriole. This complex initiates centriole biogenesis
through the activation of Polo-like kinase4 (PLK4)
and the subsequent stabilization of spindle assembly
abnormal protein 6 homologue (SAS6), which acts as
a scaffold for centriole formation71–73. During multi-
ciliated cell differentiation, this canonical duplication
programme is diverted to allow massive centriole
production. In response to expression of the EDM
complex, a paralogue of CEP63, deuterosome assem-
bly protein1 (DEUP1), is expressed55. DEUP1 can
oligomerize and form spherical electron-dense aggre-
gates called deuterosomes19. DEUP1 is also able to
recruit CEP152 and trigger the canonical centrosome
duplication cascade that drives centriole biogenesis19.
In parallel to DEUP1 expression, the components of
this cascade (including CEP152, PLK4 and SAS6) are
massively upregulated during multiciliated cell precur-
sor differentiation18,19,55,59,74–76. These elements organize
centriole amplification in multiciliated cells through:
first, a CEP63-dependent pathway that leads to the
bio genesis of centrioles directly from centrosomal
centrioles and, second, a DEUP1-dependent pathway
that leads to centriole biogenesis from deuterosome
structures19 (FIG.2). Consistent with the overall parallel
with the centriole duplication programme in cycling
cells, deuterosomes form on a centrosomal centriole
in the chick trachea and in the mouse brain14,20. In the
mouse brain, live imaging, super-resolution imag-
ing and electron microscopy show that the deutero-
somes are nucleated from the daughter centriole of the
centro some, suggesting the existence of a local micro-
environment that is conducive to the formation of
these auxiliary centrosome structures20. Whether this
immature, pre-existing centriole transmits informa-
tion to the deuterosomes77 or only provides a scaffold78
isunknown.
Centriole amplification is proposed to be organized
around three consecutive phases (FIG.2). The first is an
amplification phase during which several procentriole-
bearing deuterosomes sequentially form from the prox-
imal wall of the centrosomal daughter centriole. During
this phase, procentrioles are also growing directly from
each centrosomal centriole20. The second, a growth
phase, begins when all the deuterosomes havebeen
produced and the final number of centrioles has
beenreached; during this phase, all of the procentrioles
widen and elongate from centrosome and deuterosome
platforms14,20. The third is the disengagement phase,
during which all of the centrioles detach simultane-
ously from their centrosome and deuterosome plat-
forms to migrate to the apical membrane and nucleate
motile cilia14,19,20,79. Deuterosomes disappear at this stage
through an unknown process80, and the fate of the two
centrosomal centrioles remains unknown.
Interestingly, in the chick trachea, after a first round
of deuterosome-mediated centriole amplification from
the centrosome14, several small deuterosomes are addi-
tionally nucleated from newly formed basal bodies that
are migrating to the apical membrane15. This suggests
that additional sites and timing of deuterosome nucle-
ation can exist, maybe to increase or adjust the number
of centrioles produced.
REVIEWS
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
3
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
In addition to the multicilinE2FDEUP1 molecu-
lar cascade and the upregulation of common centro-
some duplication factors, the amplif ication phase
requires the activity of CCNO, which is a cyclin-like
prote in involved in deuterosome formati on and
function through an as yet undetermined mech-
anism76. People with pathogenic mutations in CCNO
have multiciliated cells with a reduced number of
centrioles and cilia81–83. In addition, the centriole-
associated protein coiled-coil domain-containing pro-
tein 78 (CCDC78) is required, together with DEUP1,
for CEP152 recruitment by the deuterosome and
subsequent procentriole formation18.
In contrast to the amplification process, which is
beginning to be understood, the signal that stops cen-
triole amplification and therefore regulates centriole
number is completely unknown. It seems to be corre-
lated with the onset of latent procentriole growth14,20,
suggesting that the two processes may be causally
linked. The signal that triggers the synchronous release
of newly formed centrioles by both centrosomal
centrioles and deuterosomes is unknown.
1
2 3
Centriole amplification
Centriole growth
Mother centriole
Centrosomal
mother
centriole
Centrosomal
daughter
centriole
Daughter centriole
DEUP1
CEP63
CEP152
PLK4
POC5
CP110
Centrin
Glutamylated
microtubules
CEP164SAS6–STIL
Deuterosome
in formation
Procentriole Completed
deuterosome
Centriole disengagement and docking,
and cilium formation
b
a
Daughter centriole
Multiple
rounds
Centrosome
100 nm
REVIEWS
4
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Morphants
Organisms treated with
amorpholino oligomer
todecreas e gene expre ssion.
Distal appendages
Acces sory co mponen ts of
mature centrioles that are
involved in centriole docking
atthe vesicular or apical
plasma membrane.
Secretory vesicles
Cytoplasmic vesicles that
fusewith the apical surface
ofthecell.
Exocyst
A multiple-subunit complex
that is required for the
interaction of secretory vesicles
with the plasma membrane,
inpreparation for
membranefusion.
Tr a n s i t i o n z o n e
The most proximal region of
acilium, upstream of the distal
appendages of the basal body.
Centriole docking to the plasma membrane and cilium
growth. Following collective enmasse migration to the
apex of the cell, centrioles dock at the plasma membrane,
where they become basal bodies and nucleate motile
cilia. Centriole migration is actomyosin dependent84,85.
In mice, subsequent basal body anchoring requires the
assembly of an apical actin meshwork, controlled by
the actin regulators RHOA, ezrin, RAS and filaminA,
that develops downstream of the multiciliogenesis
factors miR-34, miR-449 and forkhead box proteinJ1
(FOXJ1)69,86,87. The network implicated in basal body
connection to the actin cytoskeleton has been thoroughly
investigated in the X.laevis skin model. It involves inter-
connected apical and subapical actin plates as well as
focal adhesion proteins79,88–90. Components of the planar
cell polarity (PCP) — that is, the coordinated polariza-
tion and alignment of cells — include dishevelled (DVL)
and the effectors inturned and fuzzy. These proteins also
regulate basal body docking through apical actin enrich-
ment, membrane trafficking or the interaction of basal
bodies with the cytoskeleton90–95. In addition, ciliation is
abnormal in mice with mutations in the PCP protein cad-
herin EGF LAG seven-pass G-type receptor 2 (CELSR2)
owing to defects in basal body docking96; thus, basal
body docking and polarity are, at least in part, controlled
by common regulators. Although basal body docking
does not seem to be defective in mice in which one of
the core PCP genes, vang-like 2 (Vangl 2 ), is mutated80,97,
undocked basal bodies98 have been observed in X.laevis
vangl2 morphants, suggesting that the mechanisms differ
in the X.laevis and mousemodels.
The appearance of vesicles bound to migrating basal
bodies in multiciliated cells9,10,84,99 sug gests that, a s in pri-
mary ciliated cells100, the formation of ciliary vesicles at
distal appendages during the early steps of ciliogenesis
facilitates basal body docking. Similarly, in X.laevis, Dvl
proteins were proposed to be involved in the recruitment
of secretory vesicles to basal bodies, suggesting that the
exocys t may participate in basal body migration, planar
positioning or docking to the plasma membrane92.
Inmouse airway multiciliated cells, the association of the
basal body component chibby with the distal append-
age protein CEP164 was shown to lead to the assembly
of ciliary vesicles mediated by the small GTPase RAB8
(REF.101). As in primary ciliogenesis100, the knock-
down of centrosomal protein of 110 kDa (CP110; also
known as CCP110), which is a distal centriolar protein
that controls centriole length, is required for axonemal
extension. Inmult iciliated cells, CP110 depletion is
mediated, at least in part, by the activity of miR-34 and
miR-449(REF.63).
In addition to its involvement in basal body docking,
FOXJ1 is a master regulator of the motile cilium tran-
scriptional programme. In cooperation with the regula-
tory factor X (RFX) family of transcription factors that
drive the expression of core cilium genes, FOXJ1 con-
trols the expression of motility genes102109. Both RFX2
and FOXJ1 expression can be induced by multicilin55.
Inaddition, FOXJ1 can be activated directly by other core
components of the multiciliated cell differentiation pro-
gramme, such as GEMC1 and MYB56,57,59 (FIG.1). In paral-
lel with the FOXJ1–RFX network, TGFβ was recently
shown to be involved in assembly of both primary cilia
and multicilia, probably by controlling the expression of
transition zone prote ins110. As for primary cilia, the assem-
bly and maintenance of motile cilia in multi ciliated cells
rely on the intraflagellar transport machinery that carries
ciliary components along axoneme microtubules111.
Incontrast to the peripheral microtubule doublets of
the axoneme that are templated on centriolar micro-
tubules, the central doublet, which arises from the top
of the motile cilia transition zone and is required for
different aspects of cilia motility6, does not attach to any
recognizable structure. The nucleating mechanism and
the transcriptional components that drive the growth of
this central doublet are undetermined6.
Recently, the p53 homologue p73 was discovered
to be a major regulator of centriole docking and cili-
ation112114 that lies downstream of the multicilin–E2F
network (FIG.1). Chromatin immunoprecipitation and
sequencing analysis predicted it to bind to more than
100 cilium- related genes113. Functional studies have
shown that it directs FOXJ1, RFX3 and miR-34 expres-
sion, which explains, at least in part, the docking and cili-
ation defects of p73 mutants112,113. p73 genomic binding
sites are also found in proximity to regulators of cen triole
amplification such as MYB113 but, according to func-
tional studies, p73 does not have an important role in
centrioleformation112,114.
Figure 2 | Mammalian multiciliated cell differentiation. a|Model for centriole
biogenesis in multiciliated cells. Centriole amplification (step1). About 10% of the
centrioles from which the multiple cilia will grow form in a deuterosome-independent
pathway that is initiated by centrosomal protein of 63 kDa (CEP63), whereas a
deuterosome-
dependent pathway initiated by deuterosome assembly protein 1 (DEUP1)
is responsible for the formation of around 90% of centrioles. Both pathways are active
concurrently and involve the same downstream molecular cascade, which is also shared
by the centriole duplication process in cycling cells. The CEP63-dependent pathway is
used by both centrosomal centrioles, whereas the DEUP1-dependent pathway is used
only by the centrosomal daughter centriole. During deuterosome formation,
procentrioles remain latent at an early stage of their biogenesis. Centriole growth
(step2). When the final number of centrioles is reached after several rounds of
deuterosome formation, the deuterosome formation process stops and all the latent
procentrioles grow and mature simultaneously from both centrosomal centrioles and
deuterosomes. At the molecular level, procentrioles become positive for the marker of
the late step of centriole assembly, protein of centriole 5 (POC5), and have their tubulin
polyglutamylated. Centriole disengagement and docking, and cilium formation (step3).
Centrioles disengage synchronously from the platforms on which they grow and migrate
collectively to the apical membrane. By this time, they have recruited CEP164, which
enables them to anchor on the membrane and nucleate the ciliary tuft. b|Electron
microscopy images show that both pathways of centriole biogenesis are active
concurrently. Accumulation of electron-dense material (composed at least of DEUP1
andthe deuterosome-associated protein coiled-coil domain-containing protein78
(CCDC78)) on the proximal side (that is, the lower part with respect to centriole docking)
of the centrosomal daughter centriole leads to the progressive formation of
deuterosome spheres (arrows). These spheres then extend small latent procentrioles
(arrowheads). Concomitantly, procentrioles grow directly from the proximal part of both
centrosomal centrioles (third panel from the left; note that only the daughter centriole
isshown here). Whereas procentrioles that grow directly from the two centrosomal
centrioles do not disengage, deuterosomes detach from the centriolar wall and
accumulate in the cytoplasm, allowing another round of deuterosome formation.
Formation of up to two deuterosomes can be observed at the same time on the daughter
centriole. CP110, centrosomal protein of 110 kDa; SAS6, spindle assembly abnormal
protein 6 homologue; STIL, SCL-interrupting locus protein. Part b is adapted with
permission from REF.20, Macmillan Publishers Ltd.
REVIEWS
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
5
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Microtubule organizing
centre
Major site of microtubule
nucleation and anchoring
in a cell.
Basal-foot caps
Slight swellings at the ends
ofbasal foot conical structures.
Multiciliated cell maturation
As motile cilia grow from each basal body, the cells are
polarized to generate a collective, efficient and uni-
directional fluid flow across the multiciliated epithe-
lium. The PCP requires integration of polarization
information at the subcellular, cellular and tissue levels,
which is mainly transduced to each cell through prefer-
ential binding of the four-pass transmembrane protein
complex VANGL–CELSR (Van Gogh–Flamingo in
Drosophila melano gaster) on one cell to the seven-pass
trans membrane WNT receptor complex frizzled (FZD)–
CELSR on its neighbouring cell. This is followed by the
selective recruitment of cytoplasmic PCP components
such as Prickle-like protein (PRICKLE), DVL and diversin
(alsoknown as ANKRD6), which further promote
asymmetric protein localization along the planaraxis115.
Establishment of initial polarization bias in multi-
ciliated tissues. In the X.laevis larvae epidermis and in
respiratory airways, oviduct and brain, PCP proteins are
already polarized before ciliogenesis80,96,97,98,116118. Both
mechanical and chemical cues have been proposed to
determine the onset of planar organization (FIG.3a).
Forexample, gradients of WNT ligands are thought
to act as global cues for PCP119121. In the mammal ian
brain, the early passive flow of CSF, based on pressure
differences that arise from the drainage of CSF through
theforamen of Monro, has been suggested to providethe
initial cue for the planar polarization of progenitor cells
by activating the transmembrane mechano sensory
receptors polycystin1 and poly cystin2 on the primary
cilia of radial glial cells122. However, knockout of these
proteins only partially disrupts PCP in these cells; thus,
other mechano receptors may participate inthe initial
polarization or other mechanisms may guide the axis
and direction of PCP across ciliated tissues. Interestingly,
it was recently shown in X.laevis that a mechanical force
exerted along the anterior– posterior axis can drive
global planar pattern ing of multi ciliated tissue123. It is
thus possible that a combin ation of mechanical forces
and chemical signals directs global planar patterning in
ciliated tissues.
Rotational polarity of motile cilia. The rot ationa l polar-
ity of motile cilia defines the orientation of planar ciliary
beating. As basal bodies dock at the apical membrane,
their orientation is already biased along the anterior-
to-posterior embryonic axis in X.laevis epidermis and
in the oral direction in the airway but is randomly orien-
ted in the mouse brain80,124. Basal body orientation is then
progressively refined to the correctorientationthrough
a mechanism coupling thePCP pathway and the hydro-
dynamic forces generated by the fluid flow itself80,98,124.
Interestingly, motile cilia in X.laevis epidermis are able to
change orientation even after refinement, whereas brain
ependymal cells are fixed after the refinement stage80,124.
The coiled-coil protein basal body orientation factor 1
(Bbof1) islocalized to the ciliary rootlet in X.laevis and
possibly involved inthe mainten ance of ciliary orien-
tation, although the corres ponding mechanism remains
unclear125. Cilia motility is required for axoneme orien-
tation in the X.laevis and mammal ian brain124,126,127,
butdoes not seem to be required in the airways127.
Cytoskeletal networks are involved in the rotational
polarity of motile cilia. The microtubules interact with
each basal body through their basal foot, which is
aspeci fic basal body appendage that is proposed to be a
microtubule organizing centre128,129. γ-tubulin, outer dense
fibre protein 2 (ODF2) and galectin3 are microtubule-
associated proteins that are localized on basal-foot caps.
They are associated with the minus end of micro tubules
at nucleation and anchoring sites on the basal body.
Interestingly, loss of galectin3 or ODF2 or treatment
with the microtubule-depolymerizing agent nocoda-
zole leads to failure of rotational polarization and basal
body alignment89,130132. Moreover, nocodazole treatment
disrupts the asymmetric localization of PCP proteins
such as VANGL1, VANGL2, CELSR1 and PRICKLE2
Figure 3 | Different kinds of planar cell polarity in multiciliated tissues. a|Planar
polarization of multiciliated tissues is initiated by mechanical forces —passive fluid
flow sensed by the primary cilium or strain on tissues — that polarize primary cilia or
apical microtubules. The core planar cell polarity proteins (frizzled 3 (FZD3), FZD6,
cadherin EGF LAG seven-pass G-type receptors (CELSRs), vang-like proteins (VANGLs),
pyruvate kinase 1 (PK1; also known as PKLR) and PK2 (also known as PKM)) are then
distributed asymmetrically to the front or back of the cell with respect to the final
direction of ciliary beating. Motile cilia start beating in biased or random directions
(parts b and c). b|In Xenopus laevis epider mis and in mammalian airways a nd oviduc ts,
cilia cover the entire apical surface of multiciliated cells. Rotational polarity, which is
defined by the direction of the beating, is already biased along the anteroposterior
axisin X.laevis and in the oral direction in the airway, and is then refined by the
hydrodynamic forces generated by the ciliary beating itself and/or by the planar cell
polarity pathway. c|In brai n ependym al cells, b asal bodi es dock at the apica l plasma
membrane in random orientation. Rotational polarity is set by a coupling between the
hydrodynamic forces generated by the ciliary beating itself and the planar cell polarity
pathway. Cilia are clustered on half of the apical surface in the downstream direction of
cerebrospinal fluid (CSF) flow, which is termed translational polarity. DVL, dishevelled.
Mechanical forces:
tissue strain and/or
passive CSF ow
b Rotational polarity in airways and oviduct
a Initial global planar patterning
Centrosome
c Rotational and translational polarity in cerebral ventricles
Planar cell polarity
and uid ow
Planar cell polarity
andor uid ow
Microtubules FZD3, FZD6 CELSRs Actin, DVL1, DVL2
VANGLs, PK1, PK2
Basal
body
Nature Reviews | Molecular Cell Biology
Secretory cell
REVIEWS
6
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Myosin Id
A short-tailed classI myosin; a
monomeric actin-based motor.
Metachronal
Self-organization of ciliary
beating in a wave-like pattern.
in epithelial cells97,116, suggesting that microtubule net-
works contribute to the asymmetric localization of
these proteins. Loss of centrin2, which is a core basal
bodyprotein, or sperm- associated antigen6 (SPAG6),
which is a core axoneme protein, leads to an abnormal
number of basal feet and defects in PCP, although the
mechanism remains to bedetermined133,134.
In multiciliated cells, actin networks are organized in
two pools that are found in the apical (centriolar) and
the subapical surface of the cell (just beneath centri oles).
Disruption of actin networks with low doses of cytocha-
lasin D or the absence of CELSR2 leads to disorganized
distributions of basal bodies, metachronal ciliary beating
defects and rotational polarity defects, as motile cilia fail
to undergo beating-direction refinement89,97. Similarly,
loss of myosinId, which is localized in the cortex actin net-
work, disrupts the asymmetric localization of VANGL1 in
the cerebral trachea and leads to rotational defects135. Two
cytosolic DVL proteins, DVL1 and DVL2, associate with
basal bodies in X.laevis epidermis, mammal ian trachea
and multi ciliated cells of the brain and are required for
the establishment and refinement of cili ary polariza-
tion36,80,92,97,117,136. Interestingly, ablation of the polarity
proteins VANGL2, FZD3, CELSR2 or CELSR3 affects
the orientation of cili ary beating, suggesting that these
proteins act upstream of rotational polarity establishment
associated with the basal bodies80,92,96,97,116,117(FIG.3b).
In contrast to X.laevis epidermis and mammalian
trachea and oviducts, in which multiciliated cells are
entirely covered by cilia, brain ependymal cilia are clus-
tered on half of the apical surface, in the downstream
direction of CSF flow, termed translational polarity65
(FIG.3c). This polarity is established at the precursor-cell
stage and involves primar y cilia signalling and the
transmembrane mechanoreceptors polycystin1 and
polycystin2 (REF.122). Rostral migration of basal bodies
in ependymal multiciliated cells requires an actin-based
motor protein complex called non-muscle myosin II136,
myosin Id135 and the PCP proteins VANGL2, FZD3,
CELSR2, CELSR3 and DVL97,117.
Control of ciliary beating. Assembly of 9 + 2 cilia self-
organizes their beating through hydrodynamic coupling
and the cytoskeleton, to form metachronal waves that
increase propulsion efficiency89,137,138. Inthe airways,
ciliary beat frequency can be modulated by numerous
inhaled chemicals139. For example, bitter compounds
increase the frequency of ciliary beating140. Mechanical
constraints are also involved. Increased fluid viscosity
induces entry of calcium into the cells and subsequent
activation of cilia in the oviducts, but decreases ciliary
beat amplitude in the brain and in the airways141143.
Polycystin1 and polycystin2 are localized on motile
cilia in the oviducts, brain and airways, and might regu-
late cilia beat frequency by sensing external fluid flow
and intracellular calcium flux144,145. In addition, inflam-
mation mediators such as interleukins and hormones
(for example, progesterone) modulate ciliary beat fre-
quency in the airways and oviduct139,146149. Inthe brain,
ATP d ecre ases c ilia ry b eat f requ enc y, wh ere as se ro-
tonin and melanin-concentrating hormone have the
oppositeeffect150,151.
Functions of multiciliated cells
Multiciliated cells fulfil crucial and diverse functions in
different organs. Defects in multiciliated cell develop-
ment or function lead to severe disorders such as devel-
opmental brain defects, irreversible lung failure and
subfertility (BOX1). Ciliary beating enables protective
mucus clearance in the airway, circulation of CSF in the
adult brain and ovum transport in the oviduct. Inaddi-
tion, less well studied and atypical multiciliated cells
might also regulate essential functions in other
tissues
and organs (BOXES2,3).
Box 1 | Genetic disorders of motile cilia
Primary cilia dyskinesia (PCD), previously called immotile-cilia syndrome or Kartagener
syndrome in people with left–right asymmetry defects219, is an autosomal recessive
disorder with an estimated prevalence of around 1 in 4,000 to 1 in 50,000 people220.
Almost 40 genes are known to cause cilium motility defects in individuals with PCD,
butthese explain only 50% of the cases221. The associated cellular defects range from
abnormalities of ciliary beating pattern or frequency to total immotility.
Individuals diagnosed with reduced generation of motile cilia (RGMC) exhibit a
totalabsence of cilia or a drastic decrease in cilia number owing to the absence or
mislocalization of ciliary basal bodies58,81–83,222–239. This condition, also called ciliary
aplasia/hypoplasia or acilia syndrome, is prevalent in 1–6% of individuals with
PCD82,222,233,235. Causal mutations were recently described in genes involved in centriole
generation58,81. PCD and RGMC share major clinical symptoms, which are described
below. Disease severity is highly variable, and the relationship between genotype,
ciliadefect and phenotype is unclear221.
Brain-associated defects. Hydrocephalus is characterized by the accumulation of
cerebrospinal fluid in the ventricular cavities and the mechanical compression
of brain parenchyma240. Numerous studies observed an association between ciliary
aplasia82,222,227,232 or dyski nesia241–248 and hydrocephalus. In two cohorts of individuals with
PCD, 2.5–3.5% of the individuals had hydrocephalus242,249. In smaller cohorts of RGMC,
theprevalence was 10–50%82,222,223,236. All of the studies indicate a higher prevalence of
hydrocephalus in PCD than in the general population, suggesting that cilium defects
increase morbidity. Further studies are needed to determine whether ciliary aplasia results
in a more severe phenotype than ciliary dyskinesia. Defects in motile cilia could also be
involved in the pathophysiology of Alzheimer disease and Huntington disease250,251,252,253,254.
Fertility. Although women with dyskinetic cilia can be fertile244,255,256, some studies
reporta correlation b etween cilia absence or dy skinesia and female steri lity257–262.
Studies in small cohorts report a higher prevalence of infertility in people with PCD
orRGMC81,82,249,263. Penetrance seems to be variable and may depend on the type of
mutation and physiological context. It is often stated that cilium defects can lead to
ectopic pregnancies but, to date, only two cases of ectopic pregnancy associated with
ciliary dyskinesia have been described262,264. Accurate assessment of fertility problems
ina large cohort of women with PCD or RGMC is needed.
A large proportion of men carrying a PCD mutation are sterile, but this is attributed
tosperm flagella dyskinesia. Fertility in people with RGMC mutations, which only affect
the number of cilia of multiciliated cells and not the sperm flagella, needs to be assessed.
Ofnote, a case of azoospermia in a male with ciliary aplasia and normal spermatozoa
hasbeen reported238.
Mucociliary clearance-associated defects. A universal feature of cilium defects is the
recurren ce of airway i nfections st arting from ear ly childhood as a result of in adequate
mucociliary clearance, mucal stasis and secondary pathogen contamination in the
Eustachian tubes, sinonasal tracts, throat and pulmonary tracts265. Recurrent infections
lead to mucus gland hypertrophy, further impairing mucociliary clearance266. Cilium
defects underlie chronic infections such as sinusitis, rhinitis and otitis media with
effusion. The most serious consequence is the occurrence of chronic lower respiratory
tract infections that can eventually lead to bronchiectasis — a permanent dilation of the
airway and thickening of the bronchial wall that is conductive to further infection. This
can, in the most severe cases, necessitate lung transplantation or lead to early death265.
REVIEWS
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
7
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Glycocalyx
The polysaccharide matrix that
surrounds the cell membrane.
Sperm capacitation
Physiological changes of
spermatozoa that give them
the ability to penetrate
andfertilize an egg.
Ears, nose, throat and lungs. To protect against co ntinu-
ous exposure to the pathogens, particles and toxic chem-
icals in inhaled air, secretory cells covering the epithelium
of the ear–nose–throat and lung sphere continually pro-
duce liquids that function as physical barriers against
inhaled elements and secrete anti microbial agents that
act as an immune defence152154. Inthe tracheo- bronchial
tree, secretory cells are interspersed with ciliated cells in
a ratio approaching 50:50, although the proportion of
cili ated cells decreases towards the distal end of the bron-
chioles155157. Together with coughs, constantly beating
cilia provide the necessary force to transport pathogen-
filled airway liquids out of the respiratory system to the
pharynx, where they are swallowed152 (FIG.4). The nasal
cavity and para nasal sinuses, together with the tym-
panic cavity and the Eustachian tubes, are covered with
a comparable mucociliary epithelium158161. Inaddition
to pressure differences during respiration and swallow-
ing and drainage via blood and lymph, acilium- driven
mucus blanket helps to keep the ear and nose sphere
sterile162164 by sweeping inhaled particles towards the
pharynx (FIG.4).
The mucociliary system consists of two compo-
nents: awatery periciliary layer that provides a favour-
able environ ment for cilia beating and a gel-like mucus
that is composed of water and glycosylated proteins
called mucins. The mucus lies above the tip of motile
cilia, where it traps particles152,154 (FIG.4). Densely
packed macro molecules, tethered to respiratory cilia,
are thought to stabilize the two-layer system by form-
ing a mesh that prevents mucus and inhaled particles
from penetrating into the interciliary space165. In addi-
tion, intermolecular repulsion prevents the osmotically
active mucus from compressing the periciliary layer142.
Mucus hydration (viscosity) and ciliary beat frequency
are linked through Ca2+ and cAMP signalling, which
enable the continu ous adaptation of the two parameters
for optimal mucusclearance142,166.
In addition to mucus clearance, a recent study inzebra-
fish suggests that motile-cilium-mediated flow in the
nasal cavity improves the sensitivity and temporal resolu-
tion of the sensory cilia of olfactory neurons167. Whether
this applies in non-aquatic vertebrates, in which the olfac-
tory region is also located in a motile-cilium-covered
epithelium159, is unknown.
Reproductive tracts. Multiciliated cells are found from
the uterine cavity to the oviduct fimbria. Owing to men-
struation, the uterine epithelium reforms at each ovar-
ian cycle. The proportion of ciliated cells peaks at 20%
in both the uterine glands and lumen at the middle of
the ovarian cycle22,168170. Although modest de-ciliation
has been described in some parts of the oviduct epi-
thelium during the secretory phase13,171, most ciliated
cells remain intact in the oviducts in the course of the
ovarian cycle13,172175. However, the proportion of cili-
ated cells varies along the length of the oviduct, from
up to 80% of the cells in the fimbrium to 30% in the
isthmus13,172,175. The percentage of ciliated cells decreases
during pregnancy and with steroid contraception13,174.
The function of cilia in the uterus is unknown, but
they are thought to transport secreted material from
glandular and luminal secretory cells. In the oviduct,
ciliary beating is the principal driver of oocyte trans-
port towards the uterine cavity176. Cilia are proposed to
interact with the ovum through a ‘crownof glycocalyx
that is present on the tip of cilia177,178. Consistent with
the role of cilia in ovum transport, the beat frequency of
cilia increases after ovulation175,179, a process that is pro-
posed to be triggered by follicular fluid180. Intercourse
up to 6days before ovulation can lead to pregnancies in
humans181, so sperm can be stored in the female repro-
ductive tract, in particular by multiciliated cells in the
oviduct. Long-term sperm storage is used by various
animals, including mammals, in which copulation and
ovulation are asynchro nous182. Although controversial183,
numerous studies found a close interaction between
sperm heads and cilia in human oviducts both invitro
and invivo184186. Inaddition, direct contact between
oviduct and sperm cells is thought to increase sperm
survival and to be involved in sperm capacitation, at least
invitro187–189. In cows and pigs, capacitated sperm cells
are less prompt to attach to multiciliated cells in the
oviduct 189,190. Tubal epithelium may thus prolong the
availability of viable sperm and, by inducing capacita-
tion, allow their release for successful fertilization. The
recent development of human fallopian tube organoids
will help to test these hypotheses23.
In the male reproductive tract, efferent ducts trans-
port fluid and spermatozoa from the testis, where they
are produced, to the epididymis. The lumen is lined by a
single epithelium composed of an increasing proportion
— from the testis to the epididymis — of multiciliated
cells191193. The main function of efferent duct epithelium
Box 2 | Under-studied multiciliated cells
Several tissues have multiciliated epithelia of unknown function in physiological or
pathological conditions.
Kidney. Although multiciliated cells are not uncommon in the renal tubules of simpler
forms of animals, they are not usually found in the renal tubules of humans. Numerous
studies, however, have detected multiciliated cells that are typical of ciliated epithelia
in the adult human kidney267–272 and urethra 273–275 in a numbe r of pathol ogies.
Thissuggests that human kidney cells have retained the capacity for multiciliation.
Thiscapacity might be exploited in pathological conditions to facilitate filtration,
asinlower species. Interestingly, the efferent duct, which possesses a ciliated
epithelium that resorbs testis fluid, originates from the embryonic kidney194.
Oesophagus. Multiciliated cells line the human fetal oesophagus. Their proportion
increases gradually and peaks at around the sixteenth gestational week, then decreases
to practically zero at birth, the time of first feeding276,277, potentially by desquamation278.
A similar transitory developmental process exists in fish, birds, amphibians and
rodents279,280. Beating cilia may drive the circulation of embryonic fluids and possibly
participate in oesophageal development228. Oesophageal and tracheal cells both
differentiate from p63‑expressing progenitors in the foregut endoderm281. As adult
p63‑knockout mice retain a ciliated oesophageal epithelium, p63 expression might
regulat e the transito ry ciliation of the fetal oeso phagus25.
Spinal canal. Multiciliated cells are present in the zebrafish, rabbit, macaque and
human spinal cord282–285 but are absent in rodents. They were proposed to be involved in
propelling cerebrospinal fluid. A recent study in zebrafish revealed that cilium-motility
mutants develop the spinal deformity idiopathic scoliosis283, and scoliosis was also
reporte d in primary ci liary dyskin esia228,286–288. Whether cilium-driven cerebrospinal fluid
flow in the spine is involved in skeletal development needs to be investigated.
REVIEWS
8
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Interstitial fluids
Extracellular fluids formed
from plasma at the capillary
walls in th e brain.
Near-wall CSF circulation
Movement of cerebrospinal
fluid (CSF) close to the
ventricular walls that is
opposed to the movement
ofCSF in the central region of
the cavity.
is to stir and resorb testis fluid, thereby concentrating
the sperm191,194,195. Fluid and sperm are transported
towards the epididymis along a gradient of pressure
that is generated by seminiferous secretions, contrac-
tions and fluid absorption by the efferent ducts196198.
Instead of transporting gametes, cilia are thought to cre-
ate a reflux196 and have been reported to beat in opposite
directions depending on the side of the lumen on which
they are located199. Mathematical modelling predicted
that this reflux would be bypassed when the viscosity
of sperm increases as a result of its concentration196,
thereby allowing only sufficiently concentrated sperm
to reach the epididymis. Additional measurements of
cilia motility in the efferent duct are needed to confirm
this hypothesis.
The nervous system. In the human adult central nervous
system, half a litre of CSF is secreted every day into the
brain ventricular system by the epithelial cells of the cho-
roid plexus (BOX3). In addition to its role as a cushion,
CSF distributes secreted trophic and metabolic signals
throughout the ventricular system and clears the brain
of toxins and waste200. Multiciliated ependymal cells
cover the lumen of most of the ventricular network and
were traditionally thought to drive CSF propulsion201,202.
However, the contribution of ciliary beating to bulk fluid
propulsion in the brain ventricles might, in fact, depend
on the species and, in particular, on the size of the brain
cavities. Indeed, in small mammals such as rodents,
ependymal ciliary beating seems to be crucial for CSF
propulsion, as evidenced by the systematic develop-
ment of hydrocephalus in cilium-defective mutant mice.
However, recent evidence in humans suggests that the
main elements driving bulk CSF circulation are: first,
the pressure gradient created by secretion of the fluid
at the back of the ventricles and outflow at the front;
second, bidirectional fluid exchanges with interstitial
fluids; andthird, changes in arterial pressure during the
heartbeat cycle203,204. Confirming that bulk flow is not
driven by cilia, flow rates, measured by phase contrast
magnetic resonance imaging (MRI) in humans, show
that movement of the CSF is pulsatile and correlated
with heartbeat; under some circumstances, net flow
can even be reversed with flow into rather than outside
thebrain203,204.
In humans, cilia are thus more likely to be important
for near-wall CSF circulation. In support of this hypothesis,
computational simulations of fluid dynamics in humans
using subject-specific parameters derived from MRI pre-
dicted that microscale near-wall flow is dominated by
the action of ependymal cilia. By contrast, macroscale
CSF dynamics in the central regions of the ventricles
are predicted to be the consequence of wall motion and
choroid plexus pulsations205. Near-wall flow circula-
tion was observed long ago in humans202 and recently
described in more detail in mouse, rat and pig samples206.
These studies show that multiciliated cells are arranged
in modules and beat in different, sometimes contrary
directions, thus creating separate flow compartments.
Computational fluid modelling based on the geometry
of the human ventricles and their connecting aque-
ducts extracted from MRI scans predicted that such
recircu lation regions exist as a result of macroscale fluid
dynamics207–209. Because cilium orientation in the brain
is determined during development in response to hydro-
dynamic forces80, macro scale fluid dynamics could set
up the orientation of cilia in separated modules.
The complex flows may result in differential distrib-
ution of signalling molecules in the ventricles that
could account for preferential routes of communication
between different brain regions206,209, but this remains
to be tested experimentally. For example, transport
of the chemo-repulsive SLIT proteins by the CSF has
Box 3 | Atypical multiciliated cells
In the nervous system, multiciliated cells with an atypical cilium structure or number
exist. These include choroid plexus multiciliated cells, biciliated cells in the brain
andthe spinal cord canal, and multiciliated olfactory neurons.
Choroid plexus multiciliated cells. The choroid plexus is a highly vascularized secretory
epithelium found in the lateral, third and fourth brain ventricles in continuity with the
ependyma. Choroid plexus epithelial cells specialize in the production of cerebrospinal
fluid (CSF)200. Each cell harbours a tuft of less than 20 cilia, which are thought to be
involved in the regulation of ion transport and CSF production289,290. The differentiation
of choroid plexus cells seems to involve the Notch, multicilin and forkhead box proteinJ1
(FOXJ1) signalling pathway, which is typically involved in the formation of multiple
motile cilia291. However, the identity of choroidal cilia is controversial. Although the first
electron microscopy studies detected a 9 + 2 axoneme in humans and other species292–294,
convincing images have since accumulated showing a 9 + 0 axonemal structure289,295–297.
The authors of the earliest studies might have been misled by the organization of
microtubule doublets in choroid plexus epithelial cells, where doublets are positioned
towards the centre of the axoneme295. Alternatively, putative differences between
species and/or the existence of a shift from a 9 + 2 motile pattern to the immotile 9 + 0
pattern during the perinatal period might account for these discrepancies298,299;
ifconfirmed, this transition in axoneme organization would be unique.
Biciliated cells in the brain and the spinal cord canal. Unusual biciliated ependymal
cellshave been observed in brain ventricles and in the central canal of the spinal cord.
In the mouse brain, they are rare in the lateral ventricle but form a distinct biciliated
epithelium extending along the ventral third into the fourth ventricle214,300. Their
function is as yet undetermined. In the central canal of the rodent spinal cord, most
ependymal cells harbour two 9 + 2 cilia (a small proportion of cells have one, three or
four 9 + 2 cilia) that project into the spinal canal. In rabbits, macaques and humans, uni-
and biciliated cells are also observed, but most spinal ependymal cells are multiciliated
(15–30 cilia)282,284,285. Biciliated cells in macaques and rodents divide and are thought to
contribute to extending the length of the central canal during growth282,301 and to the
formation of scar-forming glia and oligodendrocytes when the spinal cord is injured302.
Dividing biciliated cells have not been observed in humans282. Biciliated cells can also
help to propel the CSF and/or act as sensory organelles that monitor its composition301.
Multiciliated olfactory neurons. The olfactory epithelium is a small area located in
therespiratory epithelium in the upper region of the nasal cavities. Olfactory neurons
contact the nasal cavity through their dendritic knobs and synapses with secondary
neurons in the olfactory bulb. Each neuron projects multiple (10–30) sensory cilia, which
are essential for the conversion of external chemical stimuli into intracellular electrical
responses303. Olfactory cilia form a dense, intermingled meshwork covering the
epithelium159. Their length (1–110 mm (REFS303,304)) is correlated with their sensitivity
to odour in rodents305. These cilia contain a unique axonemal structure. Whereas their
proximal segment has a 9 + 2 axoneme and the typical cilium diameter of 0.3 mm, cilia
taper down to a diameter of0.1 mm as they extend over the epithelial surface. Their
axonemal structure thus shifts gradually to nine and then to two singlets159,306.
Mammalian olfactory cilia lack dynein arms and are therefore immotile303. Despite the
essential function of olfactory cilia307–311, the molecular mechanism of their formation is
undetermined. The absence of deuterosomes and the presence of centriolar rosettes
observed by electron microscopy on differentiating neurons suggest that centrioles are
formed through a specific centriolar pathway312–314.
REVIEWS
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
9
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
Basal process
Cell extension from the cell
body that contacts the pial
surface of the brain.
been shown, in the adult mouse, to create a gradient
in the neuro genic region lying beneath the ependy-
mal epithelium. This gradient has been proposed to be
responsible for the migration of new neurons towards
the olfactorybulb210,211.
The heartbeat, breathing and state of awareness
modify the complex flows206,208,212. Given the major
contrib ution of molecules in the CSF to brain develop-
ment and homeostasis200,213, one can hypothesize that
cilium-driven near-wall CSF circulation represents an
underestimated cell-extrinsic route of regulating brain
function and neurogenesis. In adult rodents, neural
stem cells contact the ventricle by extending a thin basal
process with a single, non-motile sensory cilium at the
centre of rosettes of ependymal cells214. Ependymal
cells secrete factors that modulate adult neurogenesis,
such as noggin215, the chemokine stromal cell-derived
factor1 (SDF1)216 and pigment epithelium-derived fac-
tor (PEDF)217,218. Together, these observations suggest the
existence of direct biochemical and/or physical regulation
of adult neurogenesis by ependymalcells.
Conclusions and open questions
Multici liated cells form a prote ctive s heath over the cav-
ities of a number of organs and also help to maintain
their homeostasis and proper functioning. Depending
on the organ, these cells may undergo rapid turnover
or last for years. The fine mechanisms of their develop-
ment and maintenance at both the cell and tissue levels
are beginning to be understood. However, a number of
questions still need to be answered to fully understand
the molecular mechanisms regulating the formation of
centrioles, the alignment of ciliary beating at cell and
tissue levels, and the renewal or maintenance of these
cells throughout life. More specifically, how are the
major regulators of centriole duplication in cycling cells
repurposed to permit the amplification of centrioles?
How do multiciliated cells orient and coordinate their
ciliary beating at the tissue level to produce efficient fluid
flow in the cavities of each organ? How are these cells
replaced throughout life? Recent advances in live imag-
ing and the development of cultured organoids should
enable us to address these fascinatingquestions.
Figure 4 | Locations and proposed functions of multiciliated cells in the human body. In the mucociliary system
(topright), the convergence of cilium-powered mucus transport (arrows) from the middle ear, the bronchial tree and the
paranasal sinuses towards the pharynx helps to keep the entire earnosethroatbronchial sphere free from airborne
hazards. The mucociliary system consists of a two-component gel-on-a-brush system: a watery periciliary layer of motile
cilia and a gel-like mucus, which is composed of water and glycosylated proteins called mucins and lies above the tip of
motile cilia. Densely packed macromolecules that are tethered to respiratory cilia are thought to stabilize the two-layer
system by forming a mesh that prevents mucus and inhaled particles from penetrating into the interciliary space.
In the female reproductive tract (bottom right), ovum pick-up by ciliated cells is needed for efficient transport towards the
uterine cavity. In human efferent ducts (bottom left), multiciliated cells resorb testis fluid, and cilia are thought to create
areflux that allows only sufficiently concentrated sperm to pass. Whereas the function of multiciliated cells in the
spinalcanal is unknown (top left), in the lateral, third and fourth brain ventricles, ependymal cilia contribute to near-wall
cerebrospinal fluid (CSF) propulsion. The functions of multiciliated cells in the fetal oesophagus and in pathological
kidneyconditions areunknown.
Brain Middle ear
Eustachian tubes
Nasal cavities
Paranasal sinuses
Trachea
Lung
Oviduct
Ampulla Isthmus
Fimbria
Ovary
Mucus clearance
Ovum transport
Naso-pharynx
Pharynx = swallowed
Larynx
Gel-on-a-brush
Mucus
layer
Periciliary
layer
Spinal canal
Fetal oesophagus
Pathological
kidney conditions
Male eerent ducts
Concentrating the sperm
y uid reasorption
Unknown function
CSF near-wall circulation
Lateral
Uterus
Third
Fourth
REVIEWS
10
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
1. Tyler,S. Development of cilia in embryos of the
turbellarian Macrostomum. Hydrobiologia 84,
231–239 (1981).
2. Tamm,S.L. & Tamm,S. Development of macrociliary
cells in Beroe. J.Cell Sci. 89, 81–95 (1988).
3. Machemer,H. Ciliary activity and the origin of
metachrony in Paramecium: effects of increased
viscosity. J.Exp. Biol. 57, 239–259 (1972).
4. Ishikawa,T. Axoneme structure from motile cilia.
ColdSpring Harb. Perspect. Biol. 9, a028076 (2017).
5. Ver ti i, A ., Hun g, H. -F., Hehnly,H. & Doxsey,S. Human
basal body basics. Cilia 5, 13 (2016).
6. Loreng,T.D. & Smith,E.F. The central apparatus of
cilia and eukaryotic flagella. Cold Spring Harb. Perspect.
Biol. 9, a028118 (2016).
7. Stockinger,L. & Cireli,E. An up-to-now unknown way
ofcentriole propagation. Z.Zellforsch. Mikrosk. Anat.
68, 733–740 (in German) (1965).
8. Steinman,R.M. An electron microscopic study
ofciliogenesis in developing epidermis and trachea
inthe embryo of Xen opus la evis. Am. J.Anat . 122,
19–55 (1968).
9. Sorokin,S.P. Reconstructions of centriole formation
and ciliogenesis in mammalian lungs. J.Cell Sci. 3,
207–230 (1968).
10. Dirksen,E.R. Centriole morphogenesis in developing
ciliated epithelium of the mouse oviduct. J.Cell Biol.
51, 286–302 (1971).
11. Anderson,R. & Brenner,R. The formation of basal
bodies (centrioles) in the rhesus monkey oviduct. J.Cell
Biol. 50, 10–34 (1971).
12. Verhage,H.G. & Brenner,R.M. Estradiol-induced
differentiation of the oviductal epithelium in
ovariectom ized cats. Biol. Reprod. 13, 104–111 (1975).
13. Verha ge,H. G ., Bar eit her, M. L. , J aff e,R .C. & Akb ar,M.
Cyclic changes in ciliation, secretion and cell height of
the oviductal epithelium in women. Am. J.Anat. 156,
505–521 (1979).
14. Kalnins,V.I. & Porter,K.R. Centriole replication
duringciliogenesis in the chick tracheal epithelium.
Z.Zellforsch. Mikrosk. Anat. 100, 1–30 (1969).
15. Kalnins,V.I., Chung,C.K. & Turnbull,C. Procentrioles
in ciliating and ciliated cells of chick trachea.
Z.Zellforsch. Mikrosk. Anat. 135, 461–471 (1972).
16. Spassky,N. etal. Adult ependymal cells are postmitotic
and are derived from radial glial cells during
embryogenesis. J.Neurosci. 25, 10–18 (2005).
References 7–16 describe centriole amplification
inprogenitors of multiciliated cells, mainly by using
transmission electron microscopy.
17. Vladar,E.K. & Stearns,T. Molecular characterization of
centriole assembly in ciliated epithelial cells. J.Cell Biol.
178, 31–42 (2007).
18. Klos Dehring,D.A. etal. Deuterosome-mediated
centriole biogenesis. Dev. Cell 27, 103–112 (2013).
19. Zhao,H. etal. The Cep63 paralogue Deup1 enables
massive denovo centriole biogenesis for vertebrate
multiciliogenesis. Nat. Cell Biol. 15, 1434–1444 (2013).
20. Al Jord,A. etal. Centriole amplification by mother and
daughter centrioles differs in multiciliated cells. Nature
516, 104–107 (2014).
References 17–20 show that a comparable
signalling pathway is used for centriole amplification
in progenitors of multiciliated cells and for
centrosome duplication during the cell cycle.
21. Mirzadeh,Z., Han,Y.-G., Soriano-Navarro,M.,
García-Verd ug o,J . M . & Alv are z-B uyl la, A. Ci lia
organize ependymal planar polarity. J.Neurosci. 30,
2600–2610 (2010).
22. Masterton,R., Armstrong,E.M. & More,I.A.
Thecyclical variation in the percentage of ciliated
cellsin the normal human endometrium. J.Reprod.
Ferti l. 42, 537–540 (1974).
23. Kessler,M. etal. The Notch and Wnt pathways regulate
stemness and differentiation in human fallopian tube
organoids. Nat. Commun. 6, 8989 (2015).
24. Rock,J.R. etal. Basal cells as stem cells of the mouse
trachea and human airway epithelium. Proc. Natl Acad.
Sci. USA 106, 12771–12775 (2009).
25. Daniely,Y. etal. Critical role of p63 in the development
of a normal esophageal and tracheobronchial
epithelium. Am. J.Physiol. Cell Physiol. 287,
C171–C181 (2004).
26. Que,J., Luo,X., Schwartz,R.J. & Hogan,B.L.M.
Multiple roles for Sox2 in the developing and adult
mouse trachea. Development 136, 1899–1907
(2009).
27. Hogan,B.L.M. etal. Repair and regeneration
of the respiratory system: complexity, plasticity, and
mechanisms of lung stem cell function. Cell Stem Cell
15, 123–138 (2014).
28. Rock,J.R. & Hogan,B.L.M. Epithelial progenitor
cells in lung development, maintenance, repair,
anddisease. Annu. Rev. Cell Dev. Biol. 27, 493–512
(2011).
29. Luo,J., Shook,B.A., Daniels,S.B. & Conover,J.C.
Subventricular zone-mediated ependyma repair in the
adult mammalian brain. J.Neurosci. 28, 3804–3813
(2008).
30. Luo,Y. etal. Single-cell transcriptome analyses reveal
signals to activate dormant neural stem cells. Cell 161,
117 5 –11 8 8 (2 01 5 ).
31. Fuentealba,L.C. etal. Embryonic origin of postnatal
neural stem cells. Cell 161, 1644–1655 (2015).
32. Liu,Y., Pathak,N., Kramer-Zucker,A.
&Drummond,I.A. Notch signaling controls the
differentiation of transporting epithelia and
multiciliated cells in the zebrafish pronephros.
Development 134, 1111–1122 (2007).
33. Marcet,B. etal. Con trol of ver tebrate mu lticiliog enesis
by miR-449 through direct repression of the Delta/
Notch pathway. Nat. Cell Biol. 13, 693–699 (2011).
34. Morimoto,M. etal. Canonical Notch signaling in the
developing lung is required for determination of arterial
smooth muscle cells and selection of Clara versus
ciliated cell fate. J.Cell Sci. 123, 213–224 (2010).
35. Mori,M. etal. Notch3-Jagged signaling controls
thepool of undifferentiated airway progenitors.
Development 142, 258–267 (2015).
36. Stubbs,J.L., Vladar,E.K., Axelrod,J.D. & Kintner,C.
Multicilin promotes centriole assembly and ciliogenesis
during multiciliate cell differentiation. Nat. Cell Biol. 14,
140–147 (2012).
37. Morimoto,M., Nishinakamura,R., Saga,Y. & Kopan,R.
Different assemblies of Notch receptors coordinate the
distribution of the major bronchial Clara, ciliated and
neuroendocrine cells. Development 139, 4365–4373
(2012).
38. Deblandre,G.A., Wettstein,D.A., Koyano-Nakagawa,N.
& Kintner,C. A two-step mechanism generates the
spacing pattern of the ciliated cells in the skin of Xenopus
embryos. Development 126, 4715–4728 (1999).
39. Guseh,J.S. etal. Notch signaling promotes airway
mucous metaplasia and inhibits alveolar development.
Development 136, 1751–1759 (2009).
40. Ts a o , P. -N. etal. Notch signaling controls the balance
ofciliated and secretory cell fates in developing airways.
Development 136, 2297–2307 (2009).
41. Gomi,K., Arbelaez,V., Crystal,R.G. & Walters,M.S.
Activation of NOTCH1 or NOTCH3 signaling skews
human airway basal cell differentiation toward a
secretory pathway. PLoS ONE 10, e0116507 (2015).
42. Rawlins,E.L. etal. The role of Scgb1a1+ Clara cells
inthe long-term maintenance and repair of lung airway,
but not alveolar, epithelium. Cell Stem Cell 4, 525–534
(2009).
43. Watson ,J .K. etal. Clonal dynamics reveal two distinct
populations of basal cells in slow-turnover airway
epithelium. Cell Rep. 12, 90–101 (2015).
44. Pard o- Sagan ta,A . etal. Injury induces direct lineage
segregation of functionally distinct airway basal stem/
progenitor cell subpopulations. Cell Stem Cell 16,
184–197 (2015).
45. Pard o- Sagan ta,A . etal. Parent stem cells can serve as
niches for their daughter cells. Nature 523, 597–601
(2015).
46. Lafkas,D. etal. Therapeutic antibodies reveal Notch
control of transdifferentiation in the adult lung. Nature
528, 127–131 (2015).
47. Kyrou si,C. etal. Mcidas and GemC1/Lynkeas are key
regulators for the generation of multiciliated ependymal
cells in the adult neurogenic niche. Development 44,
3661–3674 (2015).
References 23, 32–41 and 47 show that inhibition
of Notch controls the balance between specification
into ciliated cells or into other cell types in different
organs and species.
48. Cibois,M. etal. BMP signalling controls the
construction of vertebrate mucociliary epithelia.
Development 142, 2352–2363 (2015).
49. Ta d o k o r o , T. etal. IL -6/STAT3 promotes regeneration
ofairway ciliated cells from basal stem cells. Proc. Natl
Acad. Sci. USA 111, E3641–E3699 (2014).
50. Nishimura,Y. etal. Ciliated cells differentiated
frommouse embryonic stem cells. Stem Cells 24,
1381–1388 (2006).
51. Nishimura,Y. etal. Inhibitory Smad proteins promote
the differentiation of mouse embryonic stem cells into
ependymal-like ciliated cells. Biochem. Biophys. Res.
Commun. 401, 1–6 (2010).
52. Balestrini,A., Cosentino,C., Errico,A., Garner,E.
&Costanzo,V. GEMC1 is a TopBP1-interacting protein
required for chro mosomal DNA replic ation. Nat. Cell
Biol. 12, 484–491 (2010).
53. Pefa ni ,D .E . etal. Idas, a novel phylogenetically
conserved geminin-related protein, binds to geminin
and is required for cell cycle progression. J.Biol. Chem.
286, 23234–23246 (2011).
54. Caillat,C. etal. The Geminin and Idas coiled coils
preferentially form a heterodimer that inhibits Geminin
function in DNA replication licensing. J.Biol. Chem.
288, 31624–31634 (2013).
55. Ma,L., Quigley,I., Omran,H. & Kintner,C. Multicilin
drives centriole biogenesis via E2f proteins. Genes Dev.
28, 1461–1471 (2014).
56. Te r r é, B . etal. GEMC 1 is a critical regulator of
multiciliated cell differentiation. EMBO J. 35,
942–960 (2016).
57. Arbi,M. etal. GemC1 co ntrols mul ticiliog enesis in t he
airway epithelium. EMBO Rep. 17, 400–413 (2016).
58. Boon,M. etal. MCIDAS mutations result in a
mucociliary clearance disorder with reduced generation
of multiple motile cilia. Nat. Commun. 5, 4418 (2014).
References 36, 47, 55–57 and 60 show that
GEMC1 and multicilin are the most upstream
activators of the multiciliated cell programme.
59. Ta n , F. E . etal. Myb promotes centriole amplification
and later steps of the multiciliogenesis program.
Development 140, 4277–4286 (2013).
60. Zhou,F. etal. Gmnc is a maste r regulator of the
multiciliated cell differentiation program. Curr. Biol. 25,
3267–3273 (2015).
61. Danielian,P.S. etal. E2f4 is required for normal
development of the airway epithelium. Dev. Biol. 305,
564–576 (2007).
62. Danielian,P.S., Hess,R.A. & Lees,J.A. E2f4 and E2f5
are essential for the development of the male
reproductive system. Cell Cycle 15, 250–260 (2016).
63. Song,R. etal. miR-34/449 mi RNAs are required for
motile ciliogenesis by repressing cp110. Nature 510,
115 12 0 ( 20 14 ) .
64. Wu,J . etal. Two miR NA cluste rs, miR-34b/c and
miR-449, are essential for normal brain development,
motile ciliogenesis, and spermatogenesis. Proc. Natl
Acad. Sci. USA 111, E2851–E2857 (2014).
65. Mirzadeh,Z., Doetsch,F., Sawamoto,K., Wichterle,H.
& Alvarez-Buylla,A. The subventricular zone en-face:
wholemount staining and ependymal flow. J.Vis. Exp.
http://dx.doi.org/10.3791/1938 (2 010).
66. Lavado,A. & Oliver,G. Six3 is required for ependymal
cell maturation. Development 138, 5291–5300 (2011).
67. Paez-g on zal ez ,P. etal. Art icle Ank3 -dependent SVZ
niche assembly is required for the continued production
of new neurons. Neuron 71, 61–75 (2011).
68. Park, R. etal. Yap is required for ependymal integrity
and is suppressed in LPA-induced hydrocephalus.
Nat.Commun. 7, 10329 (2016).
69. Pan,J., You,Y., Huang,T. & Brody,S.L. RhoA-mediated
apical actin enrichment is required for ciliogenesis
andpromoted by Foxj1. J.Cell Sci. 120, 1868–1876
(2007).
70. Sedzinski,J., Hannezo,E., Tu,F., Biro,M.
&Wallingford,J.B. Emergence of an apical epithelial
cell surface invivo. Dev. Cell 36, 24–35 (2016).
References 48, 69 and 70 identify the mechanisms
required for morphological changes in the
progenitors of multiciliated cells.
71. Brown,N.J., Marjanovic,M., Lüders,J., Stracker,T.H.
& Costanzo,V. Cep63 and Cep152 cooperate to ensure
centriole duplication. PLoS ONE 8, e69986 (2013).
72. Lukinavicˇius,G. etal. Selective chemical crosslinking
reveals a Cep57-Cep63-Cep152 centrosomal complex.
Curr. Biol. 23, 265–270 (2013).
73. Sir,J.-H. etal. A primary microcephaly protein complex
forms a ring around parental cent rioles. Nat. Genet. 43,
114 7 –11 5 3 (2 011 ) .
74. Hoh,R.A., Stowe,T.R., Turk,E. & Stearns,T.
Transcriptional program of ciliated epithelial cells
reveals new cilium and centrosome components and
links to human disease. PLoS ONE 7, e52166 (2012).
75. Wang, L. etal. miR-34b regulates multiciliogenesis
during organ formation in zebrafish. Development 140,
2755–2764 (2013).
76. Funk,M.C. etal. Cyclin O (Ccno) functions during
deuterosome-mediated centriole amplification of
multiciliated cells. EMBO Rep. 34, 1078–1089 (2015).
77. Meunier,A. & Spassky,N. Centriole continuity:
out with the new, in with the old. Curr. Opin. Cell Biol.
38, 60–67 (2016).
78. Klein,H.C.R., Guichard,P., Hamel,V., Gönczy,P.
&Schwarz,U.S. Computational support for a
scaffolding mechanism of centriole assembly.
Sci.Rep.6, 27075 (2016).
REVIEWS
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
11
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
79. Ioannou,A., Santama,N. & Skourides,P.A.
Xenop uslaevis nucleotide binding protein 1 (xNubp1)
is important for convergent extension movements
andcontrols ciliogenesis via regulation of the actin
cytoskeleton. Dev. Biol. 380, 243–258 (2013).
80. Guirao,B. etal. Coupling between hydrodynamic forces
and planar cell polarity orients mammalian motile cilia.
Nat. Cell Biol. 12, 341–350 (2010).
81. Wal lmeie r,J . etal. Mutations in CCNO result in congenital
mucociliary clearance disorder with reduced generation
ofmultiple motile cilia. Nat. Genet. 46, 646–651 (2014).
82. Amirav,I. etal. Systematic analysis of CCNO variants in
a defined population: implications for clinical phenotype
and differential diagnosis. Hum. Mutat. 37, 396–405
(2016).
83. Casey,J.P. etal. Unexpected genetic heterogeneity
forprimary ciliary dyskinesia in the Irish Traveller
population. Eur. J.Hum. Genet. 23, 210–217 (2015).
84. Lemullois,M., Boisvieux-Ulrich,E., Laine,M.C.,
Chailley,B. & Sandoz,D. Development and functions
ofthe cytoskeleton during ciliogenesis in metazoa.
Biol.Cell 63, 195–208 (1988).
85. Boisvieux-Ulrich,E., Lainé,M.C. & Sandoz,D.
Cytochalasin D inhibits basal body migration and ciliary
elongation in quail oviduct epithelium. Cell Tissue Res.
259, 443–454 (1990).
86. Gomperts,B.N., Gong-Cooper,X. & Hackett,B.P.
Foxj1regulates basal body anchoring to the
cytoskeleton of ciliated pulmonary epithelial cells.
J.Cell Sci. 117 , 1329–1337 (2004).
87. Chevalier,B. etal. miR-34/449 control apical actin
network formation during multiciliogenesis through
small GTPase pathways. Nat. Commun. 6, 8386 (2015).
88. Antoniades,I., Stylianou,P. & Skourides,P.A. Making
the connection: ciliary adhesion complexes anchor
basal bodies to the actin cytoskeleton. Dev. Cell 28,
70–80 (2014).
89. Werner,M. E. etal. Actin and microtubules drive
differential aspects of planar cell polarity in multiciliated
cells. J.Cell Biol. 195, 19–26 (2011).
90. Miyatake,K., Kusakabe,M., Takahashi,C. & Nishida,E.
ERK7 regulates ciliogenesis by phosphorylating the
actin regulator CapZIP in cooperation with Dishevelled.
Nat. Commun. 6, 6666 (2015).
References 88–90 show the roles of the actin
cytoskeleton in the apical migration and
organization of basal bodies.
91. Park,T.J., Haigo,S.L. & Wallingford,J.B. Ciliogenesis
defects in embryos lacking inturned or fuzzy function
are associated with failure of planar cell polarity and
Hedgehog signaling. Nat. Genet. 38, 303–311 (2006).
92. Park, T.J., Mitc hell ,B.J ., Ab itua ,P.B., Kint ner,C .
&Wallingford,J.B. Dishevelled controls apical docking
and planar polarization of basal bodies in ciliated
epithelial cells. Nat. Genet. 40, 871–879 (2008).
93. Gegg,M. etal. Flattop regulates basal body docking
and positioning in mono- and multiciliated cells. eLife 3,
e03842 (2014).
94. Gray,R.S. etal. The planar cell polarity effector Fuz
isessential for targeted membrane trafficking,
ciliogenesis, and mouse embryonic development.
Nat.Cell Biol. 11, 1225–1232 (2009).
95. Yasu n a g a ,T. etal. The polarity protein inturned links
NPHP4 to daam1 to control the subapical actin
network in multiciliated cells. J.Cell Biol. 211,
963–973 (2015).
96. Tissir,F. etal. Lack of cadherins Celsr2 and Celsr3
impairs ependymal ciliogenesis, leading to fatal
hydrocephalus. Nat. Neurosci. 13, 700–707 (2010).
97. Boutin,C. etal. A dual role for planar cell polarity
genes in ciliated cells. Proc. Natl Acad. Sci. USA 111 ,
E3129–E3138 (2014).
98. Mitchell,B. etal. The PCP pathway instructs the planar
orientation of ciliated cells in the Xenopus larval skin.
Curr. Biol. 19, 924–929 (2009).
99. Boisvieux-Ulrich,E., Laine,M.C. & Sandoz,D.
Invitroeffect s of taxol on c iliogene sis in qua il oviduct .
J.Cell Sci. 92, 9–20 (1989).
100. Wei,Q., Ling,K. & Hu,J. The essential roles of
transition fibers in the context of cilia. Curr. Opin. Cell
Biol. 35, 98–105 (2015).
101. Burke,M.C. etal. Chibby promotes ciliary vesicle
formation and basal body docking during airway cell
differentiation. J.Cell Biol. 207, 123–137 (2014).
102. El Zein,L. etal. RFX3 governs growth and beating
efficiency of motile cilia in mouse and controls the
expre ssion of genes i nvolved in human ciliop athies.
J.Cell Sci. 122, 3180–3189 (2009).
103. Chung,M.I. etal. RFX2 is broadly required for
ciliogenesis during vertebrate development. Dev. Biol.
363, 155–165 (2012).
104. Chung,M.I. etal. Coordi nated gen omic cont rol
ofciliogenesis and cell movement by RFX2. eLife 3,
e01439 (2014).
105. Choksi,S.P., Lauter,G., Swoboda,P. & Roy,S.
Switching on cilia: transcriptional networks regulating
ciliogenesis. Development 141, 1427–1441 (2014).
106. Chen,J., Knowles,H.J., Hebert,J.L. & Hackett,B.P.
Mutation of the mouse hepatocyte nuclear factor/
forkhead homologue 4 gene results in an absence of
cilia and random left-right asymmetry. J.Clin. Invest.
102, 1077–1082 (1998).
107. Brody,S.L., Yan,X.H., Wuerffel,M.K., Song,S.
&Shapiro,S.D. Ciliogenesis and left–right axis defects
in forkhead factor HFH-4 – null mice (aka FOXJ1 mutant
mice). Am. J.Respir. Cell Mol. Biol. 23, 45–51 (2000).
108. Yu,X., Ng,C.P., Habacher,H. & Roy,S. Foxj1
transcription factors are master regulators of the motile
ciliogenic program. Nat. Genet. 40, 1445–1453
(2008).
109. Didon,L. etal. RFX3 modulation of FOXJ1 regulation
ofcilia genes in the human epithelium. Respir. Res. 14,
70 (201 3).
110 . Tözs er, J . etal. TGF-β signaling regulates the
differentiation of motile cilia. Cell Rep. 11 , 1000–1007
(2015).
111. Lechtreck,K.F. IFT-cargo interactions and protein
transport in cilia. Tre n d s B i o c he m. S c i. 40, 765–778
(2015).
112 . Nemajerova,A. etal. TAp73 is a central transcriptional
regulator of airway mul ticiliogenesis. Genes Dev. 30,
1300–1312 (2016).
113 . Marshall,C.B. etal. P73 is required for
multiciliogenesis and regulates the Foxj1-associated
gene network. Cell Rep. 14, 2289–2300 (2016).
114 . Gonzalez-Cano,L. etal. p73 is required for ependymal
cell maturation and neurogenic SVZ cytoarchitecture
Abbreviated. Dev. Neurobiol. 76, 730–747 (2015).
115 . Singh,J. & Mlodzik,M. Planar cell polarity signaling:
coordination of cellular orientation across tissues.
WileyInterdiscip. Rev. Dev. Biol. 1, 479–499 (2012).
116 . Vladar,E.K., Bayly,R.D., Sangoram,A.M., Scott,M.P.
& Axelrod,J.D. Microtubules enable the planar cell
polarity of airway cilia. Curr. Biol. 22, 2203–2212
(2012).
117 . Ohata,S. etal. Loss of dishevelleds disrupts planar
polarity in ependymal motile cilia and results in
hydrocephalus. Neuron 83, 558–571 (2014).
118 . Shi,D. etal. Dynamics of planar cell polarity protein
Van gl2 in th e mo us e ov idu ct epi th eli um. Mech. Dev.
141, 78–89 (2016).
References 80, 88–98 and 116–118 show the roles
of PCP genes in the planar orientation of
multiciliated cells.
119 . Qian,D. etal. Wnt5a functions in planar cell polarity
regulation in mice. Dev. Biol. 306, 121–133 (2007).
120. Gros,J., Serralbo,O. & Marcelle,C. WNT11 acts
as a directional cue to organize the elongation
ofearlymuscle fibres. Nature 457, 589–593 (2009).
121. Gao,B. etal. Wnt signaling gradients establish planar
cell polarity by inducing Vangl2 phosphorylation
through Ror2. Dev. Cell 20, 163–176 (2011).
122. Ohata,S. etal. Mechanosensory genes Pkd1 and Pkd2
contribute to the planar polarization of brain ventricular
epithelium. J.Neurosci. 35, 11153–11168 (2015).
123. Chien,Y.H., Keller,R., Kintner,C. & Shook,D.R.
Mechanical strain determines the axis of planar polarity
in ciliated epithelia. Curr. Biol. 25, 2774–2784 (2015).
124. Mitchell,B., Jacobs,R., Li,J., Chien,S. & Kintner,C.
Apositive feedback mechanism governs the polarity
andmotion of motile cilia. Nature 447, 97–101 (2007).
125. Chien,Y.-H. etal. Bbof1 is required to maintain cilia
orientation. Development 140, 3468–3477 (2013).
126. Lechtreck,K.-F., Delmotte,P., Robinson,M.L.,
Sanderson,M.J. & Witman,G.B. Mutations in
Hydinimpair ciliary motility in mice. J.Cell Biol. 180,
633–643 (2008).
127. Matsuo,M., Shimada,A., Koshida,S., Saga,Y.
&Takeda,H. The establishment of rotational polarity
inthe airway and ependymal cilia: analysis with a novel
cilium motility mutant mouse. Am. J.Physiol. Lung Cell.
Mol. Physiol. 304, L736–L745 (2013).
128. Sandoz,D. etal. Organization and functions of
cytoskeleton in metazoan ciliated cells. Biol. Cell 63,
183–193 (1988).
129. Hagiwara,H., Kano,A., Aoki,T., Ohwada,N.
&Takata,K. Localization of gamma-tubulin to the basal
foot associated with the basal body extending a cilium.
Histochem. J. 32, 669–671 (2000).
130. Kunimoto,K. etal. Coordinated ciliary beating requires
Odf2-mediated polarization of basal bodies via basal
feet. Cell 148, 189–200 (2012).
131. Herawati,E. etal. Mu lticilia ted cell b asal bodie s align
instereotypical patterns coordinated by the apical
cytoskeleton. J.Cell Biol. 214 , 571–586 (2016).
132. Clare,D.K. etal. Basal foot MTOC organizes pillar MTs
required for coo rdination of beating cilia. Nat. Commun.
5, 4888 (2014).
133. Ying,G. etal. Centrin 2 is required for mouse olfactory
ciliary trafficking and development of ependymal cilia
planar polarity. J.Neurosci. 34, 6377–6388 (2014).
134. Te v e s, M . E . etal. Sperm-associated antigen 6 (SPAG6)
deficiency and defects in ciliogenesis and cilia function:
polarity, density, and beat. PLoS ONE 9, e107271
(2014).
135. Hegan,P.S., Ostertag,E., Geurts,A.M.
&Mooseker,M.S. Myosin Id is required for planar cell
polarity in ciliated tracheal and ependymal epithelial
cells. Cytoskeleton (Hoboken) 72, 503–516 (2015).
136. Hirota,Y. etal. Planar polarity of multiciliated
ependymal cells involves the anterior migration of basal
bodies regulated by non-muscle myosin II. Development
137, 3037–3046 (2010).
137. Guirao,B. & Joanny,J.-F. Spontaneous creation of
macroscopic flow and metachronal waves in an array
ofcilia. Biophys. J. 92, 1900–1917 (2007).
138. Elgeti,J. & Gompper,G. Emergence of metachronal
waves in cilia arrays. Proc. Natl Acad. Sci. USA 110 ,
4470–4475 (2013).
139. Workman,A.D. & Cohen,N.A. The effect of drugs
andother compounds on the ciliary beat frequency
ofhuman respiratory epithelium. Am. J.Rhinol. Allergy
28, 454–464 (2014).
140. Shah,A.S., Ben-Shahar,Y., Moninger,T.O., Kline,J.N.
& Welsh,M.J. Motile cilia of human airway epithelia
are chemosensory. Science 325, 1131–1134 (2009).
141. Andrade,Y.N. etal. TRPV4 channel is involved in the
coupling of fluid viscosity changes to epithelial ciliary
activity. J.Cell Biol. 168, 869–874 (2005).
142. Button,B.M. & Button,B. Structure and function of the
mucus clearance system of the lung. Cold Spring Harb.
Pers pect . Med . 3, a009720 (2013).
143. O’Callaghan,C.L., Sikand,K., Rutman,A. & Hirst,R.A.
The effect of viscous loading on brain ependymal cilia.
Neurosci. Lett. 439, 56–60 (2008).
144. Wodarc zyk, C. etal. A novel mouse model reveals that
polycystin-1 deficiency in ependyma and choroid plexus
results in dysfunctional cilia and hydrocephalus.
PLoSONE 4, e7137 (2009).
145. Jain,R. etal. Sensory function s of motile cilia and
implication for bronchiectasis. Front. Biosci. (Schol. Ed.)
4, 1088–1098 (2012).
146. Gudis,D., Zhao,K.Q. & Cohen,N.A. Acquired cilia
dysfunction in chronic rhinosinusitis. Am. J.Rhinol.
Allergy 26, 1–6 (2012).
147. Laoukili,J. etal. IL-13 alters mucociliary differentiation
and ciliary beating of human respiratory epithelial cells.
J.Clin. Invest. 108,
1817–1824 (2001).
148. Papathanasiou,A., Djahanbakhch,O., Saridogan,E.
&Lyons,R.A. The effect of interleukin-6 on ciliary beat
frequency in the human fallopian tube. Fertil . Steril . 90,
391–394 (2008).
149. Bylander,A. etal. Rapid effects of progesterone on
ciliary beat frequency in the mouse fallopian tube.
Reprod. Biol. Endocrinol. 8, 48 (2010).
150. Nguyen,T., Chin,W.C., O’Brien,J.A., Verdugo,P.
&Berger,A.J. Intracellular pathways regulating ciliary
beating of rat brain ependymal cells. J.Physiol. 531,
131–140 (2001).
151. Conductier,G. etal. Melanin-concentrating hormone
regulate s beat frequency of ependymal cil ia and
ventricu lar volume. Nat. Neurosci. 16, 845–847 (2013).
152. Fahy,J.V. & Dickey,B.F. Airway mucus function and
dysfunction. N.Engl. J.Med. 363, 2233–2247 (2010).
153. Ganz,T. Antimicrobial polypeptides in host defense of
the respiratory tract. J.Clin. Invest. 109, 693–697
(2002).
154. Knowles,M.R. & Boucher,R.C. Mucus clearance
as a primary innate defense mechanism for mammalian
airways. J.Clin. Invest. 109, 571–577 (2002).
155. Tremble,E. Further observations of nasal cilia.
Laryngoscope 63, 619–631 (1953).
156. Toskala,E., Smiley-Jewell,S.M., Wong,V.J., King,D.
&Plopper,C.G. Temporal and spatial distribution
ofciliogenesis in the tracheobronchial airways of mice.
Am. J.Physiol. Lung Cell. Mol. Physiol. 289,
L454–L459 (2005).
157. Mercer,R.R., Russell,M.L., Roggli,V.L. & Crapo,J.D.
Cell number and distribution in human and rat airways.
Am. J.Respir. Cell Mol. Biol. 10, 613–624 (1994).
158. Jones,N. The nose and paranasal sinuses. Adv. Drug
Deliv. Rev. 51, 5–19 (2001).
REVIEWS
12
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
159. Morrison,E. & Costanzo,R. Morphology of the human
olfactory epithelium. J.Comp. Neurol. 297, 1–13
(1990).
160. Hentzer,E. Ultrastructure of the middle ear mucosa.
Acta O tolaryn gol. Sup pl. 414 , 19–27 (1984).
161. Lim,D.J. Functional morphology of the
tubotympanum. An overview. Acta Otolaryngol. Suppl.
414, 13–18 (1984).
162. Sade,J. Ciliary activity and middle ear clearance.
Arch.Otolaryngol. 86, 128–135 (1967).
163. Li,X. etal. Otitis media in sperm-associated antigen 6
(Spag6)-deficient mice. PLoS ONE 9, 2–9 (2014).
164. Morgan,L.C. & Birman,C.S. The impact of primary
ciliary dyskinesia on the upper respiratory tract. 18,
33–38 (2016).
165. Button,B. etal. A periciliary brush promotes the lung
health by separating the mucus layer from airway
epithelia. Science 337, 937–941 (2012).
166. Button,B., Okada,S.F., Frederick,C.B., Thelin,W.R.
&Boucher,R.C. Mechanosensitive ATP release
maintains proper mucus hydration of airways.
Sci.Signal. 6, ra46 (2013).
167. Reiten,I. etal. Motile-cilia-mediated flow improves
sensitivity and temporal resolution of olfactory
computations. Curr. Biol. 27, 166–174 (2016).
168. Hafez,E.S., Ludwig,H. & Metzger,H. Human
endometrial fluid kinetics as observed by scanning
electron microscopy. Am. J.Obstet. Gynecol. 122,
929–938 (1975).
169. Amso,N.N., Crow,J., Lewin,J. & Shaw,R.W.
Acomparative morphological and ultrastructural study
of endometrial gland and fallopian tube epithelia
atdifferent stages of the menstrual cycle and the
menopause. Hum. Reprod. 9, 2234–2241 (1994).
170. Ludwig,H. & Metzger,H. The re-epithelization of
endometrium after menstrual desquamation.
Arch.Gynakol. 221, 51–60 (1976).
171. Donnez,J., Casanas-Roux,F., Caprasse,J., Ferin,J.
&Thomas,K. Cyclic changes in ciliation, cell height,
andmitotic activity in human tubal epithelium during
reproductive life. Fer til. Ste ril. 43, 554–559 (1985).
172. Crow,J., Amso,N.N., Lewin,J. & Shaw,R.W.
Morphology and infrastructure of Fallopian tube
epithelium at different stages of the menstrual cycle
and menopause. Hum. Reprod. 9, 2224–2233
(1994).
173. Patek,E., Nilsson,L. & Johannisson,E. Scanning
electron microscopic study of the human fallopian tube.
Report,I. The proliferative and secretory stages.
Ferti l.S teril. 23, 549–565 (1972).
174. Brosens,I.A. & Vasquez,G. Fimbrial microbiopsy.
J.Reprod. Med. 16, 171–178 (1976).
175. Critoph,F.N. & Dennis,K.J. Ciliary activity in the
human oviduct. Br. J.Obstet. Gynaecol. 84, 216–218
(1977).
176. Halbert,S.A., Tam,P.Y. & Blandau,R.J. Egg transport
in the rabbit oviduct: the roles of cilia and muscle.
Science 191, 1052–1053 (1976).
Reference 176 shows that ciliary beating is required
for oocyte transpor t in the ovid uct.
177. Norwood,J.T. & Anderson,R.G. Evidence that
adhesive sites on the tips of oviduct cilia membranes
are required for ovum pickup insitu. Biol. Reprod. 23,
788–791 (1980).
178. Dirksen,E.R. & Satir,P. Ciliary activity in the mouse
oviduct as studied by transmission and scanning
electron microscopy. Tissue Cell 4, 389–404 (1972).
179. Lyons,R.A. etal. Fallopian tube ciliary beat frequency
in relation to the stage of menstrual cycle and
anatomical site. Hum. Reprod. 17, 584–588 (2002).
180. Lyons,R.A., Saridogan,E. & Djahanbakh ch,O.
Theeffect of ovarian follicular fluid and peritoneal fluid
on Fallopian tube ciliary beat frequency. Hum. Reprod.
21, 52–56 (2006).
181. Wilcox,A.J., Weinberg,C.R. & Baird,D.B. Timing
ofsexual intercourse in relation to ovulation. N.Engl.
J.Med. 333, 7–11 (1995).
182. Holt,W.V. & Fazeli,A. Sperm storage in the female
reproductive tract. Annu. Rev. Anim. Biosci. 4,
291–310 (2016).
183. Williams,M. etal. Sperm numbers and distribution
within the human fallopian tube around ovulation.
Hum. Reprod. 8, 2019–2026 (1993).
184. Motta,P. & Van Blerkom,J. A scanning electron
microscopic study of rabbit spermatozoa in the female
reproductive tract following coitus. Cell Tissue Res.
163, 29–44 (1975).
185. Pacey,A .A . etal. Androl ogy: the i nteractio n invitro
ofhuman spermatozoa with epithelial cells from the
human uterine (Fallopian) tube. Hum. Reprod 10,
360–366 (1995).
186. Vigil,P., Salgado,A.M. & Cortés,M.E. Ultrastructural
interaction between spermatozoon and human oviductal
cells invitro. J.Electron. Microsc. (Tokyo) 61, 123–126
(2012).
187. Kervancioglu,E.M., Saridogan,E., John Aitken,R.
&Djahanbakhch,O. Importance of sperm-to-epithelial
cell contact for the capacitation of human spermatozoa
in fallopian tube epithelial cell cocultures. Fertil. Steril.
74, 780–784 (2000).
188. Morales,P., Palma,V., Salgado,A.M. & Villalon,M.
Sperm interaction with human oviductal cells invitro.
Hum. Reprod. 11, 1504–1509 (1996).
189. Fazeli,A., Duncan,A.E., Watson,P.F. & Holt,W.V.
Sperm-oviduct interaction: Induction of capacitation
andpreferential binding of uncapacitated spermatozoa
to oviductal epithelial cells in porcine species.
Biol.Reprod. 60, 879–886 (1999).
190. Lefebvre,R. etal. Characterization of the oviductal
sperm reservoir in cattle. Biol. Reprod. 53,
1066 –1074 (199 5).
191. Ilio,K.Y. & Hess,R.A. Structure and function of
theductuli efferentes: a review. Microsc. Res. Tech. 29,
432–467 (1994).
192. Ford,J.J., Carnes,K. & Hess,R.A. Ductuli efferentes
ofthe male Golden Syrian hamster reproductive tract.
Andrology 2, 510–520 (2014).
193. Yeung,C.H., Cooper,T.G., Bergmann,M. & Schulze,H.
Organization of tubules in the human caput
epididymidis and the ultrastructure of their epithelia.
Am. J.Anat. 191, 261–279 (1991).
194. Hess,R. in The Epididymis: From Molecules to Clinical
Practice (eds Robaire,B. & Hinton,B.T.) 49–80 (2002).
195. Sullivan,R. & Mieusset,R. The human epididymis:
itsfunction in sperm maturation. Hum. Reprod. Update
22, 574–587 (2016).
196. Winet,H. On the mechanism for flow in the efferent
ducts fluid. J.Androl. 1, 304–311 (1980).
197. Mason,K. & Shaver,S. Some functions of the caput
epididymis. Ann. NY Acad. Sci. 55, 585–593 (1952).
198. Hargrove,J.L., MacIndoe,J.H. & Ellis,L.C. Testicular
contractile cells and sperm transport. Fert il. Ste ril. 28,
114 6 –11 5 7 ( 19 7 7) .
199. Ta l o , A . In-vitro spontaneous electrical activity of rat
efferent ductules. J.Reprod. Fertil. 63, 17–20 (1981).
200. Lun,M.P., Monuki,E.S. & Lehtinen,M.K.
Development and functions of the choroid plexus–
cerebrospinal fluid system. Nat. Rev. Neurosci. 16,
445–457 (2015).
201. Worthington,W.C. & Cathcart,R.S. Ependymal cilia:
distribution and activity in the adult human brain.
Science 139, 221–222 (1963).
202. Worthington,W.C. & Cathcart,R.S. Ciliary currents
onependymal surfaces. Ann. NY Acad. Sci. 130,
944–950 (1966).
203. Brinker,T., Stopa,E., Morrison,J. & Klinge,P. A new
look at cerebrospinal fluid circulation. Fluids Barriers
CNS 11, 10 (2014).
204. Hladky,S.B. & Barrand,M.A. Mechanisms of fluid
movement into, through and out of the brain: evaluation
of the evidence. Fluids Barriers CNS 11, 26 (2014).
205. Siyahhan,B. etal. Fl ow induced by ependym al cilia
dominates near-wall cerebrospinal fluid dynamics in
thelateral ventricles. J.R.Soc. Interface 11, 20131189
(2014).
206. Faubel,R., Westendorf,C., Bodenschatz,E. & Eichele,G.
Cilia-based flow network in the brain ventricles. Science
353, 176–178 (2016).
207. Kurtcuoglu,V., Poulikakos,D. & Ventikos,Y.
Computational modeling of the mechanical behavior
ofthe cerebrospinal fluid system. J.Biomech. Eng. 127,
264–269 (2005).
208. Kurtcuog lu,V. etal. Computational investigation of
subject-specific cerebrospinal fluid flow in the third
ventricl e and aqueduct of Sy lvius. J.Biomech. 40,
1235–1245 (2007).
209. Kurtcuoglu,V., Soellinger,M., Summers,P.,
Poulikakos,D. & Boesiger,P. Mixing and modes of mass
transfer in the third cerebral ventricle: a computational
analysis. J.Biomech. Eng. 129,
695–702 (2007).
References 202 and 205–209 predict and/or show
the complex macro- and microscale flow patterns
that exist in brain ventricles and the involvement
ofependymal ciliary beating in the formation of
these patterns.
210. Sawamoto,K. etal. New neu rons follow the flow
ofcerebrospinal fluid in the adult brain. Science 311 ,
629–632 (2006).
211. Kaneko,N . etal. New neurons clear the path
ofastrocytic processes for their rapid migration
intheadult brain. Neuron 5, 213–223 (2010).
212. Yamad a , S. etal. Influence of respiration on
cerebrospinal fluid movement using magnetic resonance
spin labeling. Fluids Barriers CNS 10, 36 (2013).
213. Lehtinen,M.K. etal. The cerebrospinal fluid provides
aproliferative niche for neural progenitor cells. Neuron
69, 893–905 (2011).
214. Mirzadeh,Z., Merkle,F.T., Soriano-Navarro,M.,
Garcia-Verdugo,J.M. & Alvarez-Buylla,A.
Neural stem cells confer unique pinwheel architecture
to the ventricular surface in neurogenic regions of
theadult brain. Cell Stem Cell 3, 265–278 (2008).
215. Lim,D.A. etal. Noggin antagonizes BMP signaling
tocreate a ni che for adult neurogenesis. Neuron 28,
713– 726 (200 0).
216. Kokovay,E. etal. Adult SVZ lineage cells home to
andleave the vascular niche via differential responses
to SDF1/CXCR 4 signaling. Cell Stem Cell 7, 163–173
(2010).
217. Ramírez-Castillejo,C. etal. Pigme nt epithe lium-der ived
factor is a ni che signal for neural stem cell renewal.
Nat.Neurosci. 9, 331–339 (2006).
218. Andreu-Agulló,C., Morante-Redolat,J.M.,
Delgado,A.C. & Fariñas,I. Vascular niche factor PEDF
modulates Notch-dependent stemness in the adult
subependymal zone. Nat. Neurosci. 12, 1514–1523
(2009).
219. Sleight,M. Primary cilia dyskinesia. Lancet 318, 476
(1981).
220. Fliegauf,M., Benzing,T. & Omran,H. When cilia go
bad: cilia defects and ciliopathies. Nat. Rev. Mol. Cell
Biol. 8, 880–893 (2007).
221. Horani,A., Ferkol,T.W., Dutcher,S.K. & Brody,S.L.
Genetics and biology of primary ciliary dyskinesia.
Paedi atr. Respi r. R ev. 18, 18–24 (2016).
222. Barlocco,E.G. etal. Ultrastructural ciliary defects
inchildren with recurrent infections of the lower
respiratory tract. Pe diat r. Pul mono l. 10, 11–17 (1991).
223. Berlucchi,M. etal. Ciliary aplasia a ssociated with
hydrocephalus: an extremely rare occurrence. Eur. Arch.
Otorhinolaryngol. 269, 2295–2299 (2012).
224. Busquets,R.M., Caballero-Rabasco,M.A.,
Velasco,M., Lloreta,J. & García-Algar,Ó. Primary
ciliary dyskinesia: clinical criteria indicating
ultrastructural studies. Arch. Bronconeumol. 49,
99–104 (2013).
225. Carlén,B. & Stenram,U. Primary ciliary dyskinesia:
areview. Ultrastruct. Pathol. 29, 217–220 (2005).
226. de Santi,M.M. etal. Cilia-lacking respiratory cells
inciliary aplasia. Biol. Cell 64, 67–70 (1988).
227. DeBoeck,K. etal. Aplasia of respiratory tract cilia.
Pedi atr. P ulmo nol. 13, 259–265 (1992).
228. Engesaeth,V.G., Warner,J.O. & Bush,M.D.
Newassociations of primary ciliary dyskinesia
syndrome. Ped iatr. Pulm onol . 16, 9–12 (1993).
229. Gordon,R.E. & Kattan,M. Absence of cilia and basal
bodies with predominance of brush cells in the
respiratory mucosa from a patient with immotile
ciliasyndrome. Ultrastruct. Pathol. 6, 45–49 (1984).
230. Gotz,M. & Stockinger,L. Aplasia of respiratory tract
cilia. Lancet 321, 1283 (1983).
231. Lungarella,G., de Santi,M.M., Palestri,R. & Tosi,P.
Ultrastructural observations on basal apparatus
ofrespiratory cilia in immotile cilia syndrome. Eur. J.
Respir. Dis. 66, 165–172 (1985).
232. De Santi,M.M., Magni,A., Valletta,E., Gardi,C.
&Lungarella,G. Hydrocephalus, bronchiectasis, and
ciliary aplasia. Arch. Dis. Child. 65, 543–544 (1990).
233. Maiti,A. etal. No deleterious mutations in the FOXJ1
(aliasHFH-4) gene in patients with primary ciliary
dyskinesia (PCD). Cytogenet. Cell Genet. 90, 119–122
(2000).
234. Richard,S. etal. Congenital ciliary aplasia in two
siblings. Pathol. Res. Pra ct. 185, 181–183 (1989).
235. Rodriguez Gil,Y., González,M.A.M. & Orradre,J.L.
Ciliary hypoplasia: a rare cause of ciliary dyskinesia.
Ultrastruct. Pathol. 30, 401–402 (2006).
236. Wessel s, M.W. etal. Candidate gene analysis in three
families with acilia syndrome. Am. J.Med. Genet. A
146, 1765–1767 (2008).
237. Babin,R.W. & Kavanagh,K.T. Familial nasal acilia
syndrome. South. Med. J. 78, 737–739 (1985).
238. Matwijiw,I., Thliveris,J.A. & Faiman,C. Aplasia of
nasal cilia with situs inversus, azoospermia and normal
sperm flagella: a unique variant of the immotile cilia
syndrome. J.Urol. 137, 522–524 (1987).
239. Cerezo,L. & Price,G. Absence of cilia and basal bodies
with predominance of brush cells in the respiratory
mucosa from a patient with immotile cilia syndrome.
Ultrastruct. Pathol. 8, 381–382 (1985).
References 58, 81–83 and 222–239 highlight cases
of the under-studied ciliary aplasia phenotype.
REVIEWS
NATURE REVIEWS
|
MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION
|
13
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
240. Del Bigio,M.R. Ependymal cells: biology and
pathology. Acta Neuro pathol. 119 , 55–73 (2010).
241. Greenstone,M.A., Jones,R.W., Dewar,A.,
Neville,B.G. & Cole,P.J. Hydrocephalus and primary
ciliary dyskinesia. Arch. Dis. Child. 59, 481–482
(1984).
242. Ibañez-Tallon,I. etal. Dysfu nction of a xonemal dy nein
heavy chain Mdnah5 inhibits ependymal flow and
reveals a novel mechanism for hydrocephalus
formation. Hum. Mol. Genet. 13, 2133–2141 (2004).
243. Kosaki,K . etal. Absent inner dynein arms in a fetus
with familial hydrocephalus-situs abnormality.
Am.J.Med. Genet. A 129A, 308–311 (2004).
244. Rott,H.D. Kartagener’s syndrome and the syndrome
ofimmotile cilia. Hum. Genet. 46, 249–261 (1979).
245. Vieira,J.P., Lopes,P. & Silva,R. Primary ciliary
dyskinesia and hydrocephalus with aqueductal stenosis.
J.Child Neurol. 27, 938–941 (2012).
246. Picco,P. etal. Immotile cilia syndrome associated with
hydrocephalus and precocious puberty: a case report.
Eur. J.Pediatr. Surg. 3 (Sup pl. 1), 20–21 (1993).
247. Jabourian,Z. etal. Hydrocephalus in Kartagener’s
syndrome. Ear Nose Throat J. 65, 468–472 (1986).
248. Wessels,M.W., den Hollander,N.S. & Willems,P.J.
Mild fetal cerebral ventriculomegaly as a prenatal
sonographic marker for Kartagener syndrome.
Prenat.Diagn. 23, 239–242 (2003).
249. Greenstone,M., Rutman,A., Dewar,A., Mackay,I.
&Cole,P.J. Primary ciliary dyskinesia: cytological
andclinical features. Q.J.Med. 67, 405–423 (1988).
250. Silverberg,G.D., Mayo,M., Saul,T., Rubenstein,E.
&McGuire,D. Alzheimer’s disease, normal-pressure
hydrocephalus, and senescent changes in CSF
circulatory physiology: a hypothesis. Lancet Neurol. 2,
506–511 (2003).
251. Johanson,C.E. etal. Multipli city of ce rebrospina l
fluidfunctions: new challenges in health and disease.
Cerebrospinal Fluid Res. 5, 10 (2008).
252. Cruchaga,C. etal. GWAS of cerebrospinal fluid tau
levels identifies risk variants for Alzheimer’s disease.
Neuron 78, 256–268 (2013).
253. Lambert,J.C. etal. Meta-analysis of 74,046 individuals
identifies 11 new susceptibility loci for Alzheimer’s
disease. Nat. Genet. 45, 1452–1458 (2013).
254. Kerye r,G. etal. Ciliogenesis is regulated by a
huntingtin- HAP1-PCM1 pathway and is altered in
Huntington disease. J.Clin. Invest.121, 4372–4382
(2011).
255. Bleau,G., Richer,C.L. & Bousquet,D. Absence of
dynein arms in cilia of endocervical cells in a fertile
woman. Fer til. St eril. 30, 362–363 (1978).
256. Jean,Y., Langlais,J., Roberts,K.D., Chapdelaine,A.
&Bleau,G. Fertility of a woman with nonfunctional
ciliated cells in the fallopian tubes. Ferti l. Steri l. 31,
349–350 (1979).
257. Abu-Musa,A., Nassar,A. & Usta,I. In vitro fertilization
in two patients with Kartagener’s syndrome and
infertility. Gynecol. Obstet. Invest. 65, 29–31 (2008).
258. Halbert,S.A., Patton,D.L., Zarutskie,P.W.
&Soules,M.R. Function and structure of cilia in the
fallopian tube of an infertile woman with Kartagener’s
syndrome. Hum. Reprod. 12, 55–58 (1997).
259. Lurie,M. etal. Ciliary ultrastructure of respiratory
andfallopian tube epithelium in a sterile woman with
Kartagener’s syndrome. A quantitative estimation.
Chest 95, 578–581 (1989).
260. McLean,L. & Claman,P. Chronic cough and infertility:
areport of two cases. Ferti l. Ster il. 74, 1251–1253
(2000).
261. Peders en,H . Abs ence of d ynei n ar ms in end ometr ial
cilia: cause of infertility? Acta Obstet. Gynecol. Scand.
62, 625–627 (1983).
262. Lin,T.K. etal. A succe ssful preg nancy wit h invitro
fertilization and embryo transfer in an infertile woman
with Kartagener’s syndrome: a case report. J.Assist.
Reprod. Genet. 15, 625–627 (1998).
263. Afzelius,B.A. & Eliasson,R. Male and female infertility
problems in the immotile-cilia syndrome. Eur. J.Respir.
Dis. Suppl. 127, 144–147 (1983).
264. Blyth,M. & Wellesley,D. Ectopic pregnancy in primary
ciliary dyskinesia. J.Obstet. Gynaecol. 28, 358 (2008).
265. Knowles,M., Daniels,L., Davis,S., Zariwala,M.
&Leigh,M. Primary ciliary dyskinesia. Recent advances
in diagnostics, genetics, and characterization of clinical
disease. Am. J.Respir. Crit. Care Med. 188, 913–922
(2013).
266. Munkholm,M. & Mortensen,J. Mucociliary clearance:
pathophysiological aspects. Clin. Physiol. Funct.
Imaging 34, 171–177 (2014).
267. Katz,S.M. & M organ,J.J. Cilia i n the hu man kidn ey.
Ultrastruct. Pathol. 6, 285–294 (1984).
268. Duffy,J.L. & Suzuki,Y. Ciliated human renal proximal
tubular cells. Observations in three cases of
hypercalcemia. Am. J.Pathol. 53, 609–616 (1968).
269. Ong,A.C.M. & Wagner,B. Detection of proximal
tubular motile cilia in a patient with renal sarcoidosis
associated with hypercalcemia. Am. J.Kidney Dis. 45,
1096 –1099 (20 05).
270. Datsis,S.A. & Boman,I.A. Ciliated renal tubular
epithelium in congenital nephrosis. Beitr. Pathol. 151,
297–303 (1974).
271. Larsen,T.E. & Ghadially,F.N. Cilia in lupus nephritis.
J.Pathol. 114 , 69–73 (1974).
272. Lungarella,G., de Santi,M.M. & Tosi,P.
Ultrastructuralstudy of the ciliated cells from
renaltubular epithelium in acute progressive
glomerulonephritis. Ultrastruct. Pathol. 6, 1–7 (1984).
References 267–272 show that multiciliated cells
develop in the human kidney tubules in pathological
situations.
273. Zecchi-Orlandini,S., Gulisano,M., Orlandini,G.E.
&Holstein,A.F. Scanning electron microscopic
observations on the epithelium of the human spongy
urethra. Andrologia 20, 132–137 (1988).
274. Orlandini,G.E., Orlandini,S.Z., Holstein,A.F.,
Evangelisti,R. & Ponchietti,R. Scanning electron
microscopic observations on the epithelium of the
human prostatic urethra. Andrologia 19, 315–321
(1987).
275. Che,M., Ro,J.Y., Ordonez,N.G., Miller,R.W. &
Ayala,A.G. Ciliated epithelia in the urethra: case report
and literature review. Pathol . Int. 51 , 892–895 (2001).
276. Johns,BA. etal. Developmental changes in the
oesophageal epithelium in man. J.Anat. 86, 431–442.4
(1952).
277. Botha,G. Organogenesis and growth of the
gastroesophageal region in man. Anat. Rec. 133,
219– 239 (195 9).
278. Ménard,D. & Arsenault,P. Maturation of human fetal
esophagus maintained in organ culture. Anat. Rec.
217, 348–354 (1987).
279. Ivey,W.D. & Edgar,S.A. The histogenesis of the
esophagus and crop of the chicken, turkey, guinea fowl
and pigeaon, with special reference to ciliated
epithelium. Anat. Rec. 114, 189–211 (1952).
280. Raymond,C., Anne,V. & Millane,G. Development of
esophageal epithelium in the fetal and neonatal mouse.
Anat. Rec. 230, 225–234 (1991).
References 276–280 reveal the existence of a
transient multiciliated epithelium in the fetal
oesophagus of vertebrates.
281. Wells,J.M. & Melton,D.A. Vertebrate endoderm
development. Annu. Rev. Cell Dev. Biol. 15, 393–410
(1999).
282. Alfaro-Cervello,C. etal. Th e adult ma caque spi nal cord
central canal zone contains proliferative cells and
closely resembles the human. J.Comp. Neurol. 522,
1800–1817 (2014).
283. Grimes,D.T. etal. Zebrafish models of idiopathic
scoliosis link cerebrospinal fluid flow defects to spine
curvature. Science 352, 1341–1344 (2016).
284. Ohtsuki,K. Scanning electron microscopic studies
onrabbit’s spinal cord by resin cracking method.
Arch.Histol. Jpn 34, 405–415 (1972).
285. Nakayama,Y. & Kohno,K. Number and polarity of
theependymal cilia in the central canal of some
vertebrates. J.Neurocytol. 3, 449–458 (1974).
286. Ta n a k a , K . etal. Ciliary ultrastructure in two sisters
withKartagener’s syndrome. Med. Mol. Morphol. 40,
34–39 (2007).
287. Juncos,C. etal. Situs inversus totalis — 2 case reports.
Rev. Chil. Pediatr. 85, 344–350 (in Spanish) (2014).
288. Evander,E., Arborelius,M., Jonson,B.,
Simonsson,B.G. & Svensson,G. Lung function and
bronchial reactivity in six patients with immotile cilia
syndrome. Eur. J.Respir. Dis. Suppl. 127, 137–143
(1983).
289. Narita,K., Kawate,T., Kakinuma,N. & Takeda,S.
Multiple primary cilia modulate the fluid transcytosis in
choroid plexus epithelium. Tr a f f i c 11 , 287–301 (2010).
290. Banizs,B. etal. Dysfunctional cilia lead to altered
ependyma and choroid plexus function, and result
inthe formation of hydrocephalus. Development 132,
5329–5339 (2005).
291. Li,L. etal. Sonic Hedgehog promotes proliferation of
Notch-dependent monociliated choroid plexus tumour
cells. Nat. Cell Biol. 18, 418–430 (2016).
292. Dohrmann,G.J. & Bucy,P.C. Human choroid plexus:
alight and electron microscopic study. J.Neurosurg.
33, 506–516 (1970).
293. Davis,D.A., Lloyd,B.J.Jr & Milhorat,T.H.
Acomparative ultrastructural study of the choroid
plexuses of the immature pig. Anat. Rec. 176,
443–454 (1973).
294. Doolin,P.F. & Birge,W.J. Ultrastructural organization
of cilia and basal bodies of the epithelium of the choroid
plexus in the chick embryo. J.Cell Biol. 29, 333–345
(1966).
295. Peters,A. & Swan,R.C. The choroid plexus of the
mature and aging rat: the choroidal epithelium.
Anat.Rec. 194, 325–353 (1979).
296. Santolaya,R. & Rodriguez Echandia,E. The surface of
the choroid plexus cell under normal and experimental
conditions. Z.Zellforsch. Mikrosk. Anat. 92, 43–51
(1968).
297. Swiderski,R.E. etal. Structura l defects i n cilia of
thechoroid plexus, subfornical organ and ventricular
ependyma are associated with ventriculomegaly.
FluidsBarriers CNS 9, 22 (2012).
298. Narita,K. etal. Proteomic analysis of multiple primary
cilia reveals a novel mode of ciliary development in
mammals. Biol. Open 1, 815–825 (2012).
299. Nonami,Y., Narita,K., Nakamura,H., Inoue,T.
&Takeda,S. Developmental changes in ciliary motility
on choroid plexus epithelial cells during the perinatal
period. Cytoskeleton (Hoboken) 70 , 797–803 (2013).
300. Mirzadeh,Z. etal. Bi and uniciliated ependymal cells
define continuous floor-plate-derived tanycytic
territories. Nat. Commun. 8, 13759 (2017).
301. Alfaro-Cervello,C., Soriano-Navarro,M., Mirzadeh,Z.,
Alvarez-Buylla,A. & Garcia-Verdugo,J.M. Biciliated
ependymal cell proliferation contributes to spinal cord
growth. J.Comp. Neurol. 520, 3528–3552 (2012).
302. Meletis,K. etal. Spinal co rd injury reveals multiline age
differentiation of ependymal cells. PLoS Biol. 6,
1494–1507 (2008).
303. McEwen,D.P., Jenkins,P.M. & Martens,J.R. Olfactory
cilia: our direct neuronal connection to the external
world. Curr. Top. Dev. Biol. 85, 333–370 (2008).
304. Williams,C.L. etal. Di rect eviden ce for BBSo me-
associated intraflagellar transport reveals distinct
properties of native mammalian cilia. Nat. Commun. 5,
5813 (2014).
305. Challis,R.C. etal. An olfactory cilia pattern in the
mammalian nose ensures high sensitivity to odors.
Curr.Biol. 25, 2503–2512 (2015).
306. Moran,D.T., Rowley,J.C., Jafek,B.W. & Lovell,M.A.
The fine structure of the olfactory mucosa in man.
J.Neurocytol. 11, 721–746 (1982).
307. Ta d e n e v, A . L . D . etal. Loss of Bardet-Biedl syndrome
protein-8 (BBS8) perturbs olfactory function, protein
localization, and axon targeting. Proc. Natl Acad.
Sci.USA 108, 10320–10325 (2011).
308. Kulaga, H.M . etal. Loss of BBS proteins causes anosmia
in humans and defects in olfactory cilia structure and
function in the mouse. Nat. Genet. 36, 994–998 (2004).
309. McEwen,D.P. etal. Hypomorphic CEP290/NPHP6
mutations result in anosmia caused by the selective
lossof G proteins in cilia of olfactory sensory neurons.
Proc.Natl Acad. Sci. USA 104, 15917–15922 (2007).
310. McIntyre,J.C. etal. Gene therapy rescues cilia defects
and restores olfactory function in a mammalian
ciliopathy model. Nat. Med. 18, 1423–1428 (2012).
311. Kaneko-Goto,T. etal. Goofy coordinates the acuity
ofolfactory signaling. J.Neurosci. 33, 12987–12996
(2013).
312. Mulvaney,B.D. & Heist,H.E. Centriole migration during
regeneration and normal development of olfactory
epithelium. J.Ultrastruct. Res. 35, 274–281 (1971).
313. Menco,B.P. & Farbman,A.I. Genesis of cilia and
microvilli of rat nasal epithelia during pre-natal
development. I.Olfactory epithelium, qualitative
studies. J.Cell Sci. 78, 283–310 (1985).
314. Cuschieri,A. & Bannister,L.H. The development of
theolfactory mucosa in the mouse: electron microscopy.
J.Anat. 119 , 471–498 (1975).
Ackno wle dgem ent s
Research in the Spassky laboratory was financed by the
Institut National pour la Santé Et la Recherche Médicale
(INSERM), the Centre National de la Recherche Scientifique
(CNRS), the Ecole Normale Supérieure (ENS), the Agence
Nationale de la Recherche (ANR-12-BSV4-0006andANRJC
JC-15-CE13-0005-01), the European Research Council (ERC
Consolidator grant 647466), the Fondation pour la
Recherche Médicale (FRM20140329547), the Cancéropôle
Ile-de-France (2014-1-PL BIO-11-INSERM 12–1) and the
Fondation Pierre-Gilles de Gennes (FPGG03). The authors are
grateful to the members of their laboratory for insightful and
stimulating discussions.
Competing interests statement
The authors declare no competing interests.
REVIEWS
14
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrm
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
ǟɥƐƎƏƗɥ!,(++-ɥ4 +(2'#12ɥ(,(3#"Ʀɥ/13ɥ.$ɥ/1(-%#1ɥ341#ƥɥ++ɥ1(%'32ɥ1#2#15#"ƥ
... This modified self-assembling system, composed of the 73 developing epidermal cells, was selected for its well characterized cell types and diverse 74 uses in self-organization, cell polarity, stem-cell differentiation, wound healing, human 75 pulmonary disease, and biomaterial science [40][41][42][43][44][45][46][47]. To show an explicit example of 76 tracking whole tissue-level behavior using these approaches, our primary focus is on how 77 cellular networks respond to perturbation by inducing a mechanical puncture wound. 78 Using techniques developed for medical imaging, we stabilize videos of recorded 79 May 9, 2024 3/23 organoids and track intracellular Ca 2+ over time. ...
... After healing for 1 hour, the embryos were 374 washed twice in 0.1x MMR, pH 7.8, to remove the Ficoll solution, and any damaged 375 embryos were discarded before moving the dish to a 14 • C incubator. Two mRNA's were 376 co-injected in the reported work; GCaMP6s, a reporter of calcium activity [73,74], and 377 the intracellular domain of Notch (Notch ICD), which is known to inhibit multiciliated 378 cell induction in developing frog epidermis [50,75,76]. Multiciliated cells were 379 molecularly inhibited in the current study as the presence of these motile structures 380 causes the mucociliary organoid to move during observation, complicating image 381 analysis [77][78][79]. ...
Preprint
Full-text available
A central challenge in the progression of a variety of open questions in biology, such as morphogenesis, wound healing, and development, is learning from empirical data how information is integrated to support tissue-level function and behavior. Information-theoretic approaches provide a quantitative framework for extracting patterns from data, but so far have been predominantly applied to neuronal systems at the tissue-level. Here, we demonstrate how time series of Ca ²⁺ dynamics can be used to identify the structure and information dynamics of other biological tissues. To this end, we expressed the calcium reporter GCaMP6s in an organoid system of explanted amphibian epidermis derived from the African clawed frog Xenopus laevis , and imaged calcium activity pre- and post- a puncture injury, for six replicate organoids. We constructed functional connectivity networks by computing mutual information between cells from time series derived using medical imaging techniques to track intracellular Ca ²⁺ . We analyzed network properties including degree distribution, spatial embedding, and modular structure. We find organoid networks exhibit more connectivity than null models, with high degree hubs and mesoscale community structure with spatial clustering. Utilizing functional connectivity networks, we show the tissue retains non-random features after injury, displays long range correlations and structure, and non-trivial clustering that is not necessarily spatially dependent. Our results suggest increased integration after injury, possible cellular coordination in response to injury, and some type of generative structure of the anatomy. While we study Ca ²⁺ in Xenopus epidermal cells, our computational approach and analyses highlight how methods developed to analyze functional connectivity in neuronal tissues can be generalized to any tissue and fluorescent signal type. Our framework therefore provides a bridge between neuroscience and more basal modes of information processing. Author summary A central challenge in understanding several diverse processes in biology, including morphogenesis, wound healing, and development, is learning from empirical data how information is integrated to support tissue-level function and behavior. Significant progress in understanding information integration has occurred in neuroscience via the use of observable live calcium reporters throughout neural tissues. However, these same techniques have seen limited use in the non-neural tissues of multicellular organisms despite similarities in tissue communication. Here we utilize methods designed for neural tissues and modify them to work on any tissue type, demonstrating how non-neural tissues also contain non-random and potentially meaningful structures to be gleaned from information theoretic approaches. In the case of epidermal tissue derived from developing amphibians, we find non-trivial informational structure over greater spatial and temporal scales than those found in neural tissue. This hints at how more exploration into information structures within these tissue types could provide a deeper understanding into information processing within living systems beyond the nervous system.
... For example, lateral inhibition by Notch and Delta ligands in p63-positive basal stem cells results in the expression of geminin coiled-coil domain-containing protein 1 (GEMC1) and multicilin (MCI) [33][34][35]. MCI leads to the activation of the motile ciliogenesis program through the activation of transcription factors like RFX2/3, C-myb, and FoxJ1, which promote the expression of core cilium and motile cilia genes [36][37][38][39]. Collectively, this transcriptional cascade results in the differentiation into MCCs that contain hundreds of motile cilia that beat in a metachronal synchronization on the epithelial surface [40]. ...
Article
Full-text available
Lung epithelial development relies on the proper balance of cell proliferation and differentiation to maintain homeostasis. When this balance is disturbed, it can lead to diseases like cancer, where cells undergo hyperproliferation and then can undergo migration and metastasis. Lung cancer is one of the deadliest cancers, and even though there are a variety of therapeutic approaches, there are cases where treatment remains elusive. The rho-associated protein kinase (ROCK) has been thought to be an ideal molecular target due to its role in activating oncogenic signaling pathways. However, in a variety of cases, inhibition of ROCK has been shown to have the opposite outcome. Here, we show that ROCK inhibition with y-27632 causes abnormal epithelial tissue development in Xenopus laevis embryonic skin, which is an ideal model for studying lung cancer development. We found that treatment with y-27632 caused an increase in proliferation and the formation of ciliated epithelial outgrowths along the tail edge. Our results suggest that, in certain cases, ROCK inhibition can disturb tissue homeostasis. We anticipate that these findings could provide insight into possible mechanisms to overcome instances when ROCK inhibition results in heightened proliferation. Also, these findings are significant because y-27632 is a common pharmacological inhibitor used to study ROCK signaling, so it is important to know that in certain in vivo developmental models and conditions, this treatment can enhance proliferation rather than lead to cell cycle suppression.
... The passage of the embryo within the oviduct is believed to be facilitated by three different mechanisms, the production of oviductal fluid, the beating of cilia, and the peristaltic contraction of smooth muscle [10] (Figure 1). The contribution of each factor to embryo descent is still matter of debate [100][101][102], and ciliary beating has typically been regarded as the primary propulsive force involved [101,[103][104][105][106]. However, while all factors may exert a biophysical influence on the developing embryo, recent studies have underscored the primary role of muscle contraction in embryo transport. ...
Article
Full-text available
Preimplantation embryo culture, pivotal in assisted reproductive technology (ART), has lagged in innovation compared to embryo selection advancements. This review examines the persisting gap between in vivo and in vitro embryo development, emphasizing the need for improved culture conditions. While in humans this gap is hardly estimated, animal models, particularly bovines, reveal clear disparities in developmental competence, cryotolerance, pregnancy and live birth rates between in vitro-produced (IVP) and in vivo-derived (IVD) embryos. Molecular analyses unveil distinct differences in morphology, metabolism, and genomic stability, underscoring the need for refining culture conditions for better ART outcomes. To this end, a deeper comprehension of oviduct physiology and embryo transport is crucial for grasping embryo–maternal interactions’ mechanisms. Research on autocrine and paracrine factors, and extracellular vesicles in embryo–maternal tract interactions, elucidates vital communication networks for successful implantation and pregnancy. In vitro, confinement, and embryo density are key factors to boost embryo development. Advanced dynamic culture systems mimicking fluid mechanical stimulation in the oviduct, through vibration, tilting, and microfluidic methods, and the use of innovative softer substrates, hold promise for optimizing in vitro embryo development.
... During multiciliated epithelial cell differentiation, an essential step involves parental centriole and deuterosome-mediated amplification of centrioles, which subsequently serve as basal bodies to form motile cilia [51,52]. During this phase, PCM1 forms fibrous granules that first appear as porous condensates, measuring up to a few micrometers in diameter ( Fig. 3E) [6,7,30,53,54]. ...
Article
Full-text available
Centriolar satellites are ubiquitous membrane‐less organelles that play critical roles in numerous cellular and organismal processes. They were initially discovered through electron microscopy as cytoplasmic granules surrounding centrosomes in vertebrate cells. These structures remained enigmatic until the identification of pericentriolar material 1 protein (PCM1) as their molecular marker, which has enabled their in‐depth characterization. Recently, centriolar satellites have come into the spotlight due to their links to developmental and neurodegenerative disorders. This review presents a comprehensive summary of the major advances in centriolar satellite biology, with a focus on studies that investigated their biology associated with the essential scaffolding protein PCM1. We begin by exploring the molecular, cellular, and biochemical properties of centriolar satellites, laying the groundwork for a deeper understanding of their functions and mechanisms at both cellular and organismal levels. We then examine the implications of their dysregulation in various diseases, particularly highlighting their emerging roles in neurodegenerative and developmental disorders, as revealed by organismal models of PCM1. We conclude by discussing the current state of knowledge and posing questions about the adaptable nature of these organelles, thereby setting the stage for future research.
... Популяции структурных клеток, включая эпителиальные, эндотелиальные и мезенхимальные, образуют непрерывный физический барьер, охватывающий все дыхательные пути и ткани легких [8]. Реснитчатые эпителиальные клетки продуцируют факторы роста, являются важными сенсорами инфекции и повреждения тканей, секретируют антимикробные факторы, включая антимикробные пептиды (АМП), комплемент, муцины, интерфероны и цитокины [9]. Внутри паренхимы легких эпителиальные клетки формируют структуру альвеол и опосредуют газообмен с кровью, наряду с эндотелиальными клетками. ...
Chapter
В настоящем издании, составленном с учетом актуальных клинических рекомендаций, рассматриваются вопросы клиники, диагностики и лечения основных заболеваний респираторной системы. Руководство предназначено для повышения квалификации широкого круга врачей: терапевтов, пульмонологов, фтизиатров, аллергологов и иммунологов, онкологов, хирургов, оториноларингологов, педиатров, инфекционистов, патологоанатомов и представителей смежных специальностей. Также издание адресовано студентам старших курсов медицинских вузов, ординаторам, аспирантам и может быть использовано в качестве учебника для подготовки к практическим занятиям и итоговой государственной аттестации.
... Supporting this "fail-safe" hypothesis, several medical reports reveal the existence of MCC in cysts that have been found in a wide variety of normally non multiciliatedorgans (e.g. in (37)(38)(39)(40)). Such potential for multiciliation in a progenitor cell could also underpin the observed formation of MCC along the lumen of non-MCC fluid producing tissues (kidney, urethra) in pathological situations where they could contribute to restore fluid flow (41)(42)(43). In addition to providing a detailed prism for the study of multiciliation mechanisms, the close similarity of multiciliated differentiation mechanisms to those of the cell cycle revealed in this study expands the putative functions of multiciliated cells, sheds new light on the link between . ...
Preprint
Full-text available
A complex and conserved regulatory network drives the cell cycle. Individual components of this network are sometimes used in differentiated cells, i.e. to control organelle destruction in mammalian lens cells or light response in land plants. Some differentiated cells co-opt cell-cycle regulators more largely, to increase their ploidy using a cell cycle variant named endoreplication. Using single-cell RNA-seq profiling and functional assays in differentiating multiciliated cells, we identified a novel type of cell cycle variant that supports cytoplasmic organelle, rather than nuclear content amplification. This variant operates in post-mitotic, centriole-amplifying differentiating multiciliated cells and is characterized by (i) a circular trajectory of the transcriptome, (ii) sequential expression of more than 70% of the genes involved in S, G2 and M-like progression along this trajectory, and (iii) successive waves of cyclins. This cell cycle variant is tailored by the expression of the non-canonical cyclins O and A1 – which replace the transcriptionally silent cyclins E2 and A2 – and by the silencing of the APC/C inhibitor Emi1, two switches also detected in male meiosis, another variant of the canonical cell cycle where centriole and DNA replications are uncoupled. Re-expressing Cyclin E2, cyclin A2 or Emi1 is sufficient to induce partial replication and mitosis, suggesting that change in the regulation of expression of a few cell cycle key players drives a qualitative and quantitative tuning of Cdk activity, allowing the diversion of the cell cycle in the multiciliation variant. We also propose that this new cell cycle variant relies on the existence of a cytoplasmic – or centriolar – Cdk threshold, lower than the S-phase threshold, which affects only the cytoplasmic reorganization. One-Sentence Summary MCC progenitors undergo a final, tailored iteration of the cell cycle during differentiation, to drive centriole amplification without DNA replication or mitosis.
... Cilia are microtubule-based organelles that project from most cell types in the body, including those in the nervous system (Lee and Gleeson, 2011). In the brain, motile cilia generate cerebrospinal flow (Spassky and Meunier, 2017) and neuronal primary cilia form synapses (Sheu et al., 2022). Ciliopathies, disorders of cilia, comprise a diverse group of diseases ranging from single-organ dysfunction (e.g., polycystic kidney disease and retinitis pigmentosa) to congenital malformations and developmental defects that affect multiple organ systems including the brain (e.g., Joubert syndrome and Bardet-Biedl syndrome) (Mill et al., 2023). ...
Article
Full-text available
Ciliopathies are often caused by defects in the ciliary microtubule core. Glutamylation is abundant in cilia, and its dysregulation may contribute to ciliopathies and neurodegeneration. Mutation of the deglutamylase CCP1 causes infantile-onset neurodegeneration. In C. elegans, ccpp-1 loss causes age-related ciliary degradation that is suppressed by a mutation in the conserved NEK10 homolog nekl-4. NEKL-4 is absent from cilia, yet it negatively regulates ciliary stability via an unknown, glutamylation-independent mechanism. We show that NEKL-4 was mitochondria-associated. Additionally, nekl-4 mutants had longer mitochondria, a higher baseline mitochondrial oxidation state, and suppressed ccpp-1∆ mutant lifespan extension in response to oxidative stress. A kinase-dead nekl-4(KD) mutant ectopically localized to ccpp-1∆ cilia and rescued degenerating microtubule doublet B-tubules. A nondegradable nekl-4(PEST∆) mutant resembled the ccpp-1∆ mutant with dye-filling defects and B-tubule breaks. The nekl-4(PEST∆) Dyf phenotype was suppressed by mutation in the depolymerizing kinesin-8 KLP-13/KIF19A. We conclude that NEKL-4 influences ciliary stability by activating ciliary kinesins and promoting mitochondrial homeostasis.
... The epithelium is composed of goblet cells, which secrete protective mucus, and ciliated cells, which occupy more than half of the epithelium. Ciliated cells move the mucus layer into the laryngopharynx by rhythmically beating, ultimately allowing it to be swallowed (26). This mechanism of mucus clearance prevents the accumulation of particles and mucus in the lungs. ...
Article
Full-text available
Novel respiratory viruses can cause a pandemic and then evolve to coexist with humans. The Omicron strain of severe acute respiratory syndrome coronavirus 2 has spread worldwide since its emergence in late 2021, and its sub-lineages are now established in human society. Compared to previous strains, Omicron is markedly less invasive in the lungs and causes less severe disease. One reason for this is that humans are acquiring immunity through previous infection and vaccination, but the nature of the virus itself is also changing. Using our newly established low-volume inoculation system, which reflects natural human infection, we show that the Omicron strain spreads less efficiently into the lungs of hamsters compared with an earlier Wuhan strain. Furthermore, by characterizing chimeric viruses with the Omicron gene in the Wuhan strain genetic background and vice versa, we found that viral genes downstream of ORF3a, but not the S gene, were responsible for the limited spread of the Omicron strain in the lower airways of the virus-infected hamsters. Moreover, molecular evolutionary analysis of SARS-CoV-2 revealed a positive selection of genes downstream of ORF3a (M and E genes). Our findings provide insight into the adaptive evolution of the virus in humans during the pandemic convergence phase. IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has spread worldwide since its emergence in late 2021, and its sub-lineages are established in human society. Compared to previous strains, the Omicron strain is less invasive in the lower respiratory tract, including the lungs, and causes less severe disease; however, the mechanistic basis for its restricted replication in the lower airways is poorly understood. In this study, using a newly established low-volume inoculation system that reflects natural human infection, we demonstrated that the Omicron strain spreads less efficiently into the lungs of hamsters compared with an earlier Wuhan strain and found that viral genes downstream of ORF3a are responsible for replication restriction in the lower respiratory tract of Omicron-infected hamsters. Furthermore, we detected a positive selection of genes downstream of ORF3a (especially the M and E genes) in SARS-CoV-2, suggesting that these genes may undergo adaptive changes in humans.
Preprint
Full-text available
The ventricular zone (VZ) is made up of adult neural stem cells (NSCs) and multi-ciliated ependymal cells (EPCs). Both NSCs and EPCs are derived from radial glial cells (RGCs). To date, the transcriptomic dynamics and the molecular mechanisms guiding the cell fate commitment during the differentiation remain poorly understood. In this study, we analysed the developing VZ at the single-cell resolution and identified three distinct cellular states of RGCs: bipotent glial progenitor cells (bGPCs), neonatal NSC-neuroblasts (nNSC-NBs) and neonatal EPCs (nEPCs). The differentiation from bGPCs to nNSC-NBs and nEPCs forms a continuous bifurcating trajectory. Further molecular analysis along the NSC branch unveiled a novel intermediate state of cells expressing oligodendrocyte precursor cell (OPC) and neuroblast (NB) marker genes. Several transcription factors (TFs) were proved to be essential for the EPC-lineage differentiation, with TFEB emerging as a key regulator dictating the bGPC fate. The activation of TFEB promoted differentiation towards NSC-NBs while restrained the trajectory towards EPCs. Our findings offer detailed insights into further understanding VZ development and lay the groundwork for investigating potential therapeutic strategies against VZ-related disorders, such as hydrocephalus and neurodegenerative diseases (NDDs).
Article
Full-text available
The canonical mitotic cell cycle coordinates DNA replication, centriole duplication and cytokinesis to generate two cells from one¹. Some cells, such as mammalian trophoblast giant cells, use cell cycle variants like the endocycle to bypass mitosis². Differentiating multiciliated cells, found in the mammalian airway, brain ventricles and reproductive tract, are post-mitotic but generate hundreds of centrioles, each of which matures into a basal body and nucleates a motile cilium3,4. Several cell cycle regulators have previously been implicated in specific steps of multiciliated cell differentiation5,6. Here we show that differentiating multiciliated cells integrate cell cycle regulators into a new alternative cell cycle, which we refer to as the multiciliation cycle. The multiciliation cycle redeploys many canonical cell cycle regulators, including cyclin-dependent kinases (CDKs) and their cognate cyclins. For example, cyclin D1, CDK4 and CDK6, which are regulators of mitotic G1-to-S progression, are required to initiate multiciliated cell differentiation. The multiciliation cycle amplifies some aspects of the canonical cell cycle, such as centriole synthesis, and blocks others, such as DNA replication. E2F7, a transcriptional regulator of canonical S-to-G2 progression, is expressed at high levels during the multiciliation cycle. In the multiciliation cycle, E2F7 directly dampens the expression of genes encoding DNA replication machinery and terminates the S phase-like gene expression program. Loss of E2F7 causes aberrant acquisition of DNA synthesis in multiciliated cells and dysregulation of multiciliation cycle progression, which disrupts centriole maturation and ciliogenesis. We conclude that multiciliated cells use an alternative cell cycle that orchestrates differentiation instead of controlling proliferation.
Article
Full-text available
Multiciliated ependymal (E1) cells line the brain ventricles and are essential for brain homeostasis. We previously identified in the lateral ventricles a rare ependymal subpopulation (E2) with only two cilia and unique basal bodies. Here we show that E2 cells form a distinct biciliated epithelium extending along the ventral third into the fourth ventricle. In the third ventricle floor, apical profiles with only primary cilia define an additional uniciliated (E3) epithelium. E2 and E3 cells' ultrastructure, marker expression and basal processes indicate that they correspond to subtypes of tanycytes. Using sonic hedgehog lineage tracing, we show that the third and fourth ventricle E2 and E3 epithelia originate from the anterior floor plate. E2 and E3 cells complete their differentiation 2-3 weeks after birth, suggesting a link to postnatal maturation. These data reveal discrete bands of E2 and E3 cells that may relay information from the CSF to underlying neural circuits along the ventral midline.
Article
Full-text available
Multiciliated cells (MCCs) promote fluid flow through coordinated ciliary beating, which requires properly organized basal bodies (BBs). Airway MCCs have large numbers of BBs, which are uniformly oriented and, as we show here, align linearly. The mechanism for BB alignment is unexplored. To study this mechanism, we developed a long-term and high-resolution live-imaging system and used it to observe green fluorescent protein–centrin2–labeled BBs in cultured mouse tracheal MCCs. During MCC differentiation, the BB array adopted four stereotypical patterns, from a clustering “floret” pattern to the linear “alignment.” This alignment process was correlated with BB orientations, revealed by double immunostaining for BBs and their asymmetrically associated basal feet (BF). The BB alignment was disrupted by disturbing apical microtubules with nocodazole and by a BF-depleting Odf2 mutation. We constructed a theoretical model, which indicated that the apical cytoskeleton, acting like a viscoelastic fluid, provides a self-organizing mechanism in tracheal MCCs to align BBs linearly for mucociliary transport (J. Cell Biol. Vol. 214 No. 5 571–586; August 29, 2016).
Article
Histologic studies of recent years have definitely concluded that the epithelium of the middle ear is a modified respiratory epithelium with ciliated and secretory cells, including goblet cells. These cells, like the ciliated cells, are developd from the basal cell and are just ordinary secretory cells completely filled with secretory granules. For different reasons it is postulated, that the dark secretory granules are prestages of the actual secretory product presenting as light granules and that dark and light granules do not represent different secretory capacities of the cell. The subepithelial layer which consists of loose connective tissue is a structure of just as great importance as the epithelial layer.
Article
Motile cilia are actively beating hair-like structures that cover the surface of multiple epithelia. The flow that ciliary beating generates is utilized for diverse functions and depends on the spatial location and biophysical properties of cilia. Here we show that the motile cilia in the nose of aquatic vertebrates are spatially organized and stably beat with an asymmetric pattern, resulting in a robust and stereotypical flow around the nose. Our results demonstrate that these flow fields attract odors to the nose pit and facilitate detection of odors by the olfactory system in stagnant environments. Moreover, we show that ciliary beating quickly exchanges the content of the nose, thereby improving the temporal resolution of the olfactory system for detecting dynamic changes of odor plumes in turbulent environments. Altogether, our work unravels a central function of ciliary beating for generating flow fields that increase the sensitivity and the temporal resolution of olfactory computations in the vertebrate brain.
Article
The motile cilium is a complex organelle that is typically comprised of a 9+2 microtubule skeleton; nine doublet microtubules surrounding a pair of central singlet microtubules. Like the doublet microtubules, the central microtubules form a scaffold for the assembly of protein complexes forming an intricate network of interconnected projections. The central microtubules and associated structures are collectively referred to as the central apparatus (CA). Studies using a variety of experimental approaches and model organisms have led to the discovery of a number of highly conserved protein complexes, unprecedented high-resolution views of projection structure, and new insights into regulation of dynein-driven microtubule sliding. Here, we review recent progress in defining mechanisms for the assembly and function of the CA and include possible implications for the importance of the CA in human health.
Article
The axoneme is the main extracellular part of cilia and flagella in eukaryotes. It consists of a microtubule cytoskeleton, which normally comprises nine doublets. In motile cilia, dynein ATPase motor proteins generate sliding motions between adjacent microtubules, which are integrated into a well-orchestrated beating or rotational motion. In primary cilia, there are a number of sensory proteins functioning on membranes surrounding the axoneme. In both cases, as the study of proteomics has elucidated, hundreds of proteins exist in this compartmentalized biomolecular system. In this article, we review the recent progress of structural studies of the axoneme and its components using electron microscopy and X-ray crystallography, mainly focusing on motile cilia. Structural biology presents snapshots (but not live imaging) of dynamic structural change and gives insights into the force generation mechanism of dynein, ciliary bending mechanism, ciliogenesis, and evolution of the axoneme.
Article
Going with the flow The interstitial spaces of the brain are filled with cerebrospinal fluid (CSF). Faubel et al. studied fluid transport in the third ventricle of the brain of mice, rats, and pigs. Sophisticated, state-of-the-art fluid dynamic studies revealed a complex pattern of cilia beating that leads to an intricate network of “highways” of CSF flow. This flow rapidly and efficiently transports and partitions CSF. Science , this issue p. 176
Article
In many sensory organs, specialized receptors are strategically arranged to enhance detection sensitivity and acuity. It is unclear whether the olfactory system utilizes a similar organizational scheme to facilitate odor detection. Curiously, olfactory sensory neurons (OSNs) in the mouse nose are differentially stimulated depending on the cell location. We therefore asked whether OSNs in different locations evolve unique structural and/or functional features to optimize odor detection and discrimination. Using immunohistochemistry, computational fluid dynamics modeling, and patch clamp recording, we discovered that OSNs situated in highly stimulated regions have much longer cilia and are more sensitive to odorants than those in weakly stimulated regions. Surprisingly, reduction in neuronal excitability or ablation of the olfactory G protein in OSNs does not alter the cilia length pattern, indicating that neither spontaneous nor odor-evoked activity is required for its establishment. Furthermore, the pattern is evident at birth, maintained into adulthood, and restored following pharmacologically induced degeneration of the olfactory epithelium, suggesting that it is intrinsically programmed. Intriguingly, type III adenylyl cyclase (ACIII), a key protein in olfactory signal transduction and ubiquitous marker for primary cilia, exhibits location-dependent gene expression levels. Moreover, genetic ablation or reduction of ACIII levels dramatically alters the cilia pattern. These findings reveal an intrinsically programmed, activity-independent configuration in the nose to ensure high sensitivity to odors and a novel role of ACIII in mediating olfactory cilia length. Together, this work has broad implications for how sensory receptors optimize detection sensitivity in various physiological contexts and offers new insights into the regulation of cilia morphology and function.