ArticlePDF Available

Zona Pellucida Domain-Containing Protein β-Tectorin is Crucial for Zebrafish Proper Inner Ear Development

Authors:

Abstract and Figures

The zona pellucida (ZP) domain is part of many extracellular proteins with diverse functions from structural components to receptors. The mammalian β-tectorin is a protein of 336 amino acid residues containing a single ZP domain and a putative signal peptide at the N-terminus of the protein. It is 1 component of a gel-like structure called the tectorial membrane which is involved in transforming sound waves into neuronal signals and is important for normal auditory function. β-Tectorin is specifically expressed in the mammalian and avian inner ear. We identified and cloned the gene encoding zebrafish β-tectorin. Through whole-mount in situ hybridization, we demonstrated that β-tectorin messenger RNA was expressed in the otic placode and specialized sensory patch of the inner ear during zebrafish embryonic stages. Morpholino knockdown of zebrafish β-tectorin affected the position and number of otoliths in the ears of morphants. Finally, swimming behaviors of β-tectorin morphants were abnormal since the development of the inner ear was compromised. Our results reveal that zebrafish β-tectorin is specifically expressed in the zebrafish inner ear, and is important for regulating the development of the zebrafish inner ear. Lack of zebrafish β-tectorin caused severe defects in inner ear formation of otoliths and function.
Content may be subject to copyright.
Zona Pellucida Domain-Containing Protein b-Tectorin is
Crucial for Zebrafish Proper Inner Ear Development
Chung-Hsiang Yang
1,2
, Chia-Hsiung Cheng
2
, Gen-Der Chen
2
, Wei-Hao Liao
2
, Yi-Chung Chen
2
, Kai-Yun
Huang
1
, Pung-Pung Hwang
3
, Sheng-Ping L. Hwang
3
, Chang-Jen Huang
1,2
*
1Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan, 2Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan, 3Institute of Cellular
and Organismic Biology, Academia Sinica, Taipei, Taiwan
Abstract
Background:
The zona pellucida (ZP) domain is part of many extracellular proteins with diverse functions from structural
components to receptors. The mammalian b-tectorin is a protein of 336 amino acid residues containing a single ZP domain
and a putative signal peptide at the N-terminus of the protein. It is 1 component of a gel-like structure called the tectorial
membrane which is involved in transforming sound waves into neuronal signals and is important for normal auditory
function. b-Tectorin is specifically expressed in the mammalian and avian inner ear.
Methodology/Principal Findings:
We identified and cloned the gene encoding zebrafish b-tectorin. Through whole-mount
in situ hybridization, we demonstrated that b-tectorin messenger RNA was expressed in the otic placode and specialized
sensory patch of the inner ear during zebrafish embryonic stages. Morpholino knockdown of zebrafish b-tectorin affected
the position and number of otoliths in the ears of morphants. Finally, swimming behaviors of b-tectorin morphants were
abnormal since the development of the inner ear was compromised.
Conclusions/Significance:
Our results reveal that zebrafish b-tectorin is specifically expressed in the zebrafish inner ear, and
is important for regulating the development of the zebrafish inner ear. Lack of zebrafish b-tectorin caused severe defects in
inner ear formation of otoliths and function.
Citation: Yang C-H, Cheng C-H, Chen G-D, Liao W-H, Chen Y-C, et al. (2011) Zona Pellucida Domain-Containing Protein b-Tectorin is Crucial for Zebrafish Proper
Inner Ear Development. PLoS ONE 6(8): e23078. doi:10.1371/journal.pone.0023078
Editor: Bruce Riley, Texas A&M University, United States of America
Received April 5, 2011; Accepted July 5, 2011; Published August 2, 2011
Copyright: ß2011 Yang et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This study was supported by a grant (AS-99-TP-B08) from Academia Sinica and a grant (97-2313-B-001-002-MY3) from National Science Council, Taipei,
Taiwan. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: cjibc@gate.sinica.edu.tw
Introduction
The zona pellucida (ZP) was first discovered as a glycoprotein
surrounding the plasma membrane of an oocyte, and it is
important in fertilization. The ZP domain is a sequence shared
by many extracellular proteins with diverse functions from
structural components to receptors. Among these proteins are
the mammalian ZP1, ZP2, and ZP3, and non-mammalian egg-
coating proteins, Tamm-Horsfall protein (THP), glycoprotein
(GP)-2, a- and b-tectorins, transforming growth factor (TGF)-b
receptor III, endoglin, deleted in malignant brain tumor (DMBT)-
1, no-mechanoreceptor potential-A (Nomp A), Dumpy, and
cuticlin-1 [1]. Each of these ZP-containing proteins is composed
of a signal sequence driving these proteins to the endoplasmic
reticulum (ER), and each possesses a ZP domain of approximately
260 amino acids long that is comprised of 8,10 conserved
cysteine residues, a C-terminal, a hydrophobic transmembrane-
like region, and a short cytoplasmic tail [2,3]. ZP domain-
containing proteins are highly conserved among all species and are
often glycosylated [4]. They are generally modified with a variable
number of high-mannose type, N-linked oligosaccharides in the
ER. These proteins can be further modified by the addition of O-
linked oligosaccharides and by processing of high-mannose-type,
N-linked oligosaccharides to the complex type when transferred
to Golgi apparatuses. ZP domain-containing proteins are often
present in filaments and/or matrices which play important roles in
protein polymerization [1].
In the inner ear organ of Corti, the tip of hair cell stereocilium
bundles is covered by a gel-like matrix called the tectorial
membrane. The mammalian tectorial membrane is formed by 3
different collagens (types II, V, and IX) combined with 3 non-
collagenous, glycosylated polypeptides, called a-tectorin, b-
tectorin, and otogelin [1]. The tectorial membrane is a particular
structure that deflects the stereocilia of hair cells during sound-
triggered vibrations of the basilar membrane, and hair cells
facilitate the transduction of sounds into neural signals. a- and b-
tectorins belong to the ZP domain-containing protein family, and
mutations in a-tectorin or b-tectorin were reported to result in
human nonsyndromic deafness. Studies of the human a-tectorin
gene, TECTA, showed that it is related to dominant forms of
prelingual, nonprogressive deafness: DFNA8 (MIM601543) and
DFNA12 (MIM601842) [5] or a recessive form at locus DFNB21
[6]. On the other hand, TECTB, which encodes b-tectorin, also
plays an important role in maintaining the normal function of the
tectorial membrane. Previous studies on knockout mice reported
that the structure of the striated-sheet matrix is disrupted and
PLoS ONE | www.plosone.org 1 August 2011 | Volume 6 | Issue 8 | e23078
cochlear tuning is sharpened in TECTB
2/2
mice [7]. Together,
both types of tectorins are important for maintaining normal
auditory function of the inner ear.
Currently, only mammalian and chicken tectorins have been
identified and cloned [8]. Little is known about zebrafish b-
tectorin and its role during embryonic development. Zebrafish
have many benefits as a model animal for studying genes involved
in inner ear development: first, zebrafish eggs develop in vitro;
second, the zebrafish ear is transparent for the first few weeks of
life; and third, forward genetic screens and antisense technology
are well-established [9]. The zebrafish inner ear system consists of
semicircular canals, otoliths, and different sensory patches that
are formed by special type of cells, called hair cells. The
semicircular canals are attached to the sensory epithelium called
cristae which are located at the base of each canal. Cristae are
important for sensing the position of the head and angular
acceleration. Polycrystalline masses called otoliths are connected
to 2 or more macular organs, called saccules and utricles [9].
Zebrafish is considered to be a fish with excellent hearing in the
teleost family due to the function of Weberian ossicles which
connect the swim bladder to the saccule allowing sound
amplification [10,11]. Although, the structure of the zebrafish
inner ear greatly differs from that of the mammalian inner ear
due to the lack of a cochlea, the convenience in handling it and
the evolutionarily conserved molecular mechanisms of inner ear
development make zebrafish a good animal model for studying
development of the inner ear.
In order to investigate the role of ZP domain-containing
proteins in zebrafish inner ear development, we predicted and
identified a zebrafish ZP domain-containing protein, zebrafish b-
tectorin, through a bioinformatics method, and designed a
specific morpholino to knock down the expression of zebrafish
b-tectorin. We also characterized the temporal and spatial
expressions of zebrafish b-tectorin by whole-mount in situ
hybridization, and demonstrated that it is specifically expressed
in the zebrafish inner ear in the early stages of development.
Morpholino knockdown of b-tectorin expression in zebrafish
resulted in abnormal inner ear development. Otoliths of b-
tectorin morphants showed delocalization or a fused pattern.
Taken together, we propose that the zebrafish b-tectorin plays an
important role in the proper formation of zebrafish inner ear
components, and therefore a lack of it will cause serious defects in
development of the inner ear.
Results
Cloning of b-tectorin from zebrafish
The overall deduced amino acid sequences of zebrafish b-
tectorin respectively showed 49%, 50%, 50%, and 49% identities
to those of human, mouse, chicken, and Xenopus b-tectorin (Fig. 1).
The zebrafish b-tectorin protein contains a conserved ZP domain,
which has highly conserved cysteines of C1 to C8, and Cx, Cy, Ca,
and Cb. A signal peptide of 16-amino acids, MAAVGLF-
FILLPVTWA, in the NH
2
-terminal was predicted by the online
software, SignalP 3.0 (http://www.cbs.dtu.dk/services/SignalP/),
which likewise occurs in mammalian b-tectorin proteins. Mam-
malian b-tectorin proteins have a signal peptide of 17 amino acids.
A hydrophobic C-terminus characteristic of proteins that are
membrane bound via a putative GPI-anchor was reported in avian
and mammalian b-tectorins [12,13]. Moreover, human b-tectorin
is a glycoprotein and contains 4 N-linked sugar chains on Asn80,
Asn104, Asn116, and Asn145. Similarly, the zebrafish b-tectorin
protein contains 4 N-linked glycosylation sites of
77
NHS,
104
NDS,
116
NYT, and
145
NGS (Fig. 1).
Genomic structure of the zebrafish b-tectorin gene
We then used the 1542 bp of zebrafish b-tectorin cDNA (with
accession no. FJ374270) to perform an online BLAST search of
the GenBank database. The zebrafish b-tectorin cDNA matched
10 non-contiguous regions of a 92,355-bp zebrafish BAC clone,
CH73-92E20 (GenBank accession no. CU462848). Subsequently,
a BLAST 2-sequence comparison of BAC CH73-92E20 with the
zebrafish b-tectorin cDNA indicated that b-tectorin cDNA
contained 10 putative exons and 9 introns spanning at least
8.6 kb (Fig. 2). Using these putative exons as a model, a sequence
alignment was produced such that each intron concurred with the
GT/AG intron donor/acceptor site rule [14]. Exon 1 contained
the 59-UTR, while exon 2 contained the putative translation
initiation site. Exon 2 contained 9 bp of the 59-UTR and 66 bp of
the first coding sequences of b-tectorin cDNA. Exon 10 contained
the last 69 bp of the coding sequences and 362 bp of 39-UTR. The
size of the introns considerably varied, ranging 81 (intron 8) to
2419 bp (intron 9).
Comparison of the exon-intron organization of zebrafish and
mouse b-tectorin genes indicated that their genomic structures were
similar with 10 exons and 9 introns. The mouse b-tectorin gene
spanned approximately 15.4 kb. In addition, the average intron
size of the mouse b-tectorin gene (1411 bp) was larger than that of
the zebrafish b-tectorin gene (799 bp) (Fig. 2).
Expression profiles of zebrafish b-tectorin messenger
(m)RNA in adult tissues and embryos at different
developmental stages
Expression levels of zebrafish b-tectorin transcripts in adult tissues
and embryos from different developmental stages were examined
by an RT-PCR analysis. A pair of primers was used to amplify a
DNA fragment that spanned exons 2 to 4 to avoid genomic DNA
interference in the PCRs. The amplified product of this pair of
primers was about 1208 bp long. As shown in Fig. 3A, a high level
of b-tectorin expression was detected in the brain, with moderate
expression in the kidneys and less in the intestines.
During embryogenesis, b-tectorin transcripts were not detected at
1 day post-fertilization (dpf), and their expression levels constantly
increased thereafter; however, a significant decrease in the
expression level was observed at 5 dpf and thereafter (Fig. 3A).
Spatial and temporal expression patterns of zebrafish b-tectorin
were further analyzed by whole-mount in situ hybridization.
During different stages of development, expression of the b-tectorin
transcript was specifically detected in the anterior and posterior
maculae on both sides of the zebrafish from 48,120 h post-
fertilization (hpf) (Fig. 3B, panels a,l). It was interesting to note
that the expression of b-tectorin mRNA in the anterior macula was
much weaker than that of the posterior macula in 48-hpf embryos.
The signals of b-tectorin in situ hybridization are restricted to the
macula of the inner ear, no signals in other parts of the embryos
can be detected in various stages (Fig. 3B, panels c, f, i and l).
Through longitudal section of the zebrafish inner ear at 72 hpf, b-
tectorin is both expressed in the hair cells and supporting cells of
macula, and the expression pattern resembles the early expression
of pax5 in the macula [15]. The overall expression pattern of
zebrafish b-tectorin in the inner ear was quite similar to that of the
Starmaker gene, which is also expressed in the anterior and posterior
maculae on both sides of the inner ear [16].
Abnormal otolith formation in b-tectorin morphants
Morpholino (MO)-mediated knockdown of genes in zebrafish
embryos has become a routine and efficient method to provide
information about gene function in vivo [17]. To examine the
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 2 August 2011 | Volume 6 | Issue 8 | e23078
function of b-tectorin in vivo, we designed zebrafish b-tectorin MOs,
which targeted the sequence located at the 59-UTR of b-tectorin
mRNA, to specifically knock down the translation of endogenous
b-tectorin mRNA.
To determine the specificity of the MO used, a control
approach was used. The 25-bp target sequence of the b-tectorin
MO was cloned upstream of the green fluorescent protein (GFP)
ORF into a pCMV backbone expression vector (bTec-GFP). As
for the control, a target sequence containing 5 mismatches was
used (MM b-Tec-GFP). The MM b-Tec-GFP or bTec-GFP
plasmid was injected into zebrafish embryos in the absence or
presence of a 9-ng b-tectorin MO. Co-injection of bTec-GFP RNA
and the b-tectorin MO completely blocked GFP expression
(n= 22/24; Fig. S1A). Conversely, GFP expression was not
affected when 5 bp of the target sequence was exchanged (MM b-
Tec-GFP) (n= 36/36; Fig. S1B) indicating the spe cificity of the b-
tectorin MO.
After injecting 4 ng/embryo of b-tectorin MO, we observed
abnormal otolith morphology in embryos at 120 hpf. There are 2
otoliths on each side in wild-type (WT) zebrafish embryos; the
one in the anterior has a flattened-oval shape, whereas the other
one in the posterior, is larger, and has a round shape (Fig. 4A,
panel a). b-Tectorin morphants displayed 2 different phenotypes
including the fusion of 2 otoliths (Fig. 4A, panel b, n= 50/168,
29%; Fig. 4B) as well as a single otolith (Fig. 4A, panel c, n=5/
168, 2.9%; Fig. 4B). Changes in morphology of the otoliths were
correlated with the irregular formation of the vestibular system in
the inner ear; development of semicircular canals seemed to be
affected in b-tectorin morphants when observed at 72,120 hpf. b-
Tectorin morphants with either fused or single otoliths were
Figure 1. Zebrafish b-tectorin amino acid sequence alignment with those of other species. Deduced amino acid sequences of zebrafish
b-tectorin were aligned with those of human, mouse, chicken, and Xenopus. All b-tectorin proteins contained a conserved zona pellucida (ZP) domain
of approximately 260 amino acids, with 12 highly conserved cysteine residues (indicated by arrows). Identical residues in 4 or 5 proteins are
highlighted. Signal peptide and putative GPI-anchored domains are heavily overlined. The putative N-linked glycosylation sites are indicated by dots
(). The accession numbers of each b-tectorin from different species are listed below: human (XM_521604), mouse (X99806), chicken (AAA92461),
and Xenopus (CAJ82963).
doi:10.1371/journal.pone.0023078.g001
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 3 August 2011 | Volume 6 | Issue 8 | e23078
observed with abnormal semicircular canal outgrowth (Fig. 4A,
panels e, h, f, and i, arrows), whereas the control showed 2
normal otoliths and outgrowth of the semicircular canals (Fig. 4A,
panels d and g).
We also generated splice MOs to block splicings of the b-
tectorin sequence within exon 2 and exon 3 (supplementatry
Fig. 2A). The splice MO 1 which blocks a splicing donor site
locates at the boundry of Exon 2 and intron 2 while the splice
MO 2 targets the acceptor site at the boundry of intron 2 and
Exon 3. Each of the splice MOs was used individually,
producing abnormal phenotypes similar to those of the ATG
MOs, differing only in efficiency (data not shown). The
combination of the two splice MOs is more efficient in
generating abnormal phenotypes that include both the fused
and single otolith phenotype like the ATG MO. The efficiency of
the splice MO mixture was analyzed by RT-PCR (supplemen-
tary Fig. 2B). After injecting a mixture of splice MOs 2 ng each,
we observed the appearance of the fusion of 2 otoliths (n = 113/
287, 39.37% ; Fig. 4B) and single otolith (n = 7/287, 2.43%;
Fig. 4B) in the resulting morphants. There were no other severe
morphological defects in the ATG MO (supplementary Fig. 3).
Gradient increases in the amount of ATG MO injected into the
embryos also showed increases in the abnormal phenotypes in a
dosage-dependent manner (Fig. 4B).
To rule out the possibilities that the phenotypes of these
morphants are the results of off-target effect of the morpholino
used [18], the p53 MO was used in co-injecting with ATG MO
into embryos and the phenotypes of these embryos were observed.
The percentage of abnormalities in the b-Tectorin morphants
coinjected with p53 MO was approximately equal to that of b-
Tectorin morphants (Fig. 4B), suggesting that the phenotypes of
these morphants were not related to off-target effect of the
morpholino.
To further confirm the specificity of gene knockdowns by the b-
tectorin MO, mRNA rescue was performed. Full length b-Tectorin
mRNAs, which were synthesized in vitro and injected into
embryos in one- or two- cell stage, were used to investigate the
rescue of b-Tectorin morphants. Misexpressions of the b-Tectorin
mRNA have no effect on morphologies of the control embryos
without MO injection. Coinjection of 8 ng ATG MO with
,100 pg b-Tectorin mRNA into each embryo resulted in a
reduction in the percentage of the b-Tectorin morphant with
abnormal ear phenotypes, fused otoliths (n = 12/35, 34%) and
single otolith (n = 3/35, 8.5%), whereas injection of 8 ng ATG
MO alone showed a percentage of as high as 84% abnormalities in
the inner ear, fused otoliths (n = 35/45, 78%), single otolith (n = 3/
45, 6%) (Fig. 4B). These results demonstrated that the b-Tectorin
mRNA can rescue defects in b-Tectorin morphants, validating the
specificity of b-Tectorin MO.
Development of the inner ear was affected in the
b-tectorin MO morphants as shown by whole-mount in
situ hybridization
To further analyze inner ear defects observed in morphants,
whole-mount in situ hybridization was first performed with
Starmaker (stm), which was reported to regulate the growth, shape,
and crystal lattice of otoliths [15]. In control MO-injected
zebrafish, the stm transcript was expressed in anterior and
posterior maculae on both sides at 96 hpf (Fig. 5B, lateral, panel
a, dorsal panel a9). However, the stm transcript was less expressed
in the anterior macula of b-tectorin morphants either with fused
otoliths or a single otolith compared to control MO-injected
zebrafish (Fig. 5B, panels b, b9c, and c9; arrows). Otolith matrix
protein 1 (omp-1) is important for otolith growth and correct
anchoring of otoliths to the maculae [19]. The expression pattern
of omp-1 in b-tectorin morphants was also reduced in the anterior
macula compared to the control, and its distribution seemed
to differ from that of control MO-injected zebrafish (Fig. 5C).
On the other hand, another gene marker, zona pellucida-like
domain-containing protein-like 1 (zpDL1) (Genbank accession
no. XM_00192195), was used to label anterior, lateral, and
posterior cristae of otoliths of control MO-injected zebrafish at 96
hpf (Fig. 5D, panels a and b). In b-tectorin morphants with fused
otoliths, the zpDL1 signal was detected only in the anterior and
posterior cristae of the inner ear, and the signal in the lateral
crista was lost (Fig. 5D, arrow).
FM1-43, a fluorescent dye, is known for labeling hair cells of the
inner ear by entering the mechanotransduction channels [20].
This function of FM1-43 allows us to monitor the formation of
active hair cells in the morphants. For this purpose, zebrafish at
diffenerent stages were injected with FM1-43 dyes specifically in
Figure 2. Genomic organization of zebrafish and mouse
b-tectorin
genes. Coding regions are shown as filled boxes numbered from 1 to 10
in both zebrafish and mouse b-tectorin genes. The 59- and 39-untranslated regions are shown as open boxes, while introns and 59- and 39-flanking
regions are indicated by solid lines.
doi:10.1371/journal.pone.0023078.g002
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 4 August 2011 | Volume 6 | Issue 8 | e23078
lumen of the otic vesicle through an injection tube. FM1-43 dyes,
which were taken up by macula and cristae, could be easily
observed under a confocal fluorescence microscopy [20]. ATG
MO-injected zebrafish embryos were further injected with FM1-
43 dyes at 72 hpf to investigate whether the functions of these hair
cells are affected. b-tectorin morphants with fused otoliths of 72
hpf had lost its lateral crista as compared to control zebrafish.
These results are consistent with the data gained from whole-
mount in situ hybridization, as signals of zpDL1 lost in the lateral
crista in the b-tectorin morphants.
Taken together, the altered expression patterns observed in ear-
marker genes indicated that b-tectorin may play an important role
in both otolith and inner ear formation during zebrafish
development.
Figure 3. Expression profiles of zebrafish
b-tectorin
mRNAs by RT-PCR and whole-mount
in situ
hybridization. (A) RT-PCR of the
b-tectorin transcript was performed using a pair of primers to produce a DNA fragment of 1208 bp. b-Actin bands were also used to normalize the
amount of cDNA prepared from different tissues and embryos at different developmental stages. (B) Whole-mount in situ hybridization with
antisense b-tectorin at different developmental stages was performed. The images were taken from the dorsal (a, d, g, j) and the lateral view (b, e, h,
k), and complete lateral view (c, f, i, l) with the anterior to the left and dorsal to the top. Longitudinal sections of the embryo were at 72 hpf with
anterior to the left and dorsal to the top (panel m). The straight line in panel n represents the region of sections in panel m. hc, hair cell; sc,
supporting cell.
doi:10.1371/journal.pone.0023078.g003
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 5 August 2011 | Volume 6 | Issue 8 | e23078
Figure 4. Abnormal otolith phenotypes in
b-tectorin
morphants. (A) The otolith phenotypes of b-tectorin morphants are classified into normal
(normal, panel a), fused (fused, panel b) and single otoliths (single, panel c). Abnormal development of the vestibular system is shown by arrows in
b-tectorin morphants from 72 to 120 hpf (panels d to i). (B) The percentage of abnormal otolith phenotypes in zebrafish embryos injected with
different b-tectorin MOs or combined with b-tectorin mRNAs or p53 MOs. All samples are observed at 72 hpf. Bars, 50 mm.
doi:10.1371/journal.pone.0023078.g004
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 6 August 2011 | Volume 6 | Issue 8 | e23078
Behavioral defects in b-tectorin morphants
Altered swimming behaviors and a lack of balance are indices of
abnormal ear function, for example, swimming in a corkscrew or
circular path [21]. Those b-tectorin morphants with either fused
otoliths or a single otolith were further tested for their ability to
maintain balance and swim after stimulation. At 5 dpf, about 40%
of noninjected embryos (WT) and control MO-injected embryos
(N = 70) displayed floating in an upright position and sometimes
swam spontaneously in random directions. Another 45% of
control MO-injected embryos remained lying down on the bottom
Figure 5. Characterization of ear defects in
b-tectorin
morphants. The expression levels of the following inner ear marker genes, such as
starmaker (stm) (A), otolith matrix protein 1 (omp-1) (B), and zona pellucida-like domain-containing protein-like 1 (zpDL1) (C), were examined by
whole-mount in situ hybridization in b-tectorin morphants. Stm signals in the anterior macula (am) of b-tectorin morphants decreased or even
disappeared in fishes with either fused (panels b and b9) or single otoliths (panels c and c9), as indicated by arrows. zpDL1 signals in the lateral crista
(lc) of b-tectorin morphants vanished. Black bars, 100 mm (D) Confocal microscopy image analysis of b-tectorin morphants injected with FM1-43 dyes
into the otic vesicle at 72 hpf. After injection, hair cells in anterior crista (ac), lateral crista (lc), macula (m) and posterior crista (pc) of control MO-
injected embryos can take up FM1-43 dyes. White bar, 50 mm.
doi:10.1371/journal.pone.0023078.g005
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 7 August 2011 | Volume 6 | Issue 8 | e23078
of the Petri dish. In order to test whether these 5-dpf zebrafish
could respond to vibration, several short vibrations were created
by deploying an ultrasonic processor in the water, and all zebrafish
swam away immediately (Video S1). The remaining 15% of
control MO-injected embryos were further stimulated using a glass
tube to touch the head of a zebrafish, and they swam away in a
straight manner (Video S3). On the other hand, b-tectorin
morphants with single or fused otoliths showed a failure to
maintain balance, and all stayed at the bottom of the Petri dish.
About 10% of those b-tectorin morphants responded to short
vibrations created by the ultrasonic processor in the water with
short irregular movements (n= 70) (Video S2). The remaining b-
tectorin morphants with fused otoliths were stimulated on the head
with a glass tube about 5 times, and they either did not respond to
the stimuli or swam a very short range in a circular path (Fig. 6, B1
to B4) (Videos S4 and S5). b-Tectorin morphants with a single
otolith had similar behavioral defects as those with fused otoliths
described above. Some swam in a corkscrew path which implied
profound defects (Fig. 6, A1 to A4) (Videos S6 and S7).
Discussion
In this study, the zebrafish b-tectorin gene and its cDNA were
cloned and characterized. The cDNA encodes a protein of 336
amino acids, which displays 49% and 50% identities to human
and chick b-tectorins. RT-PCR analyses showed that zebrafish b-
tectorin mRNA was primarily expressed in the brain with moderate
expression in the kidneys. Whole-mount in situ hybridization
showed that expression of the b-tectorin transcript was specifically
found in the anterior and posterior maculae of the ear. Similar to
human b-tectorin, zebrafish b-tectorin contains 4 N-linked
glycosylation sites (Fig. 1). Knockdown of zebrafish b-tectorin
expression caused the fusion of 2 otoliths or there was only a single
otolith, both of which led to severe malfunction of the inner ear.
The predicted amino acid sequence of zebrafish b-tectorin
exhibited overall identities of 49%, 50%, 50%, and 50% to b-
tectorins from the human, mouse, chick, and Xenopus (Fig. 1).
However, chick and mouse b-tectorins were homologous with 75%
identity at the amino acid level [13,22]. The higher similarity in
identity of chick b-tectorin to the human and mouse compared to
zebrafish may be due to differences in habitat, terrestrial and
aquatic, respectively. This suggests that the environment may
influence the evolution of this molecule. If fish b-tectorins from
different species were compared, these fish b-tectorins might display
higher identities to each other. Indeed, zebrafish b-tectorin showed
higher identities of 74%, 76%, and 74%, with 85% similarity, to b-
tectorin from Tetraodon, fugu, and medaka (Fig. S4).
In this study, zebrafish b-tectorin contained a highly conserved
ZP domain, which was a sequence of approximately 260 amino
acid residues with 8 or 10 cysteine residues and was located at the
C-terminus (Fig. 1). Many ZP domain-containing proteins with
various functions were found in vertebrates [1]. Some of those
proteins constitute the extracellular coat of animal eggs, such as
ZP1, ZP2 and ZP3. They are responsible for egg/sperm
recognition as well as for blocking polyspermy [23]. Other
proteins like a- and b-tectorins are 2 major components of the
tectorial membrane, which is an extracellular matrix covering the
sensory epithelia of the cochlea of the inner ear [24]. In transgenic
mice with a specific mutation in a-tectorin, the structure of the
tectorial membrane is disrupted leading to hearing loss [25].
Similarly, mice lacking b-tectorin have sharpened cochlear tuning
leading to low-frequency hearing loss [7]. Interestingly, zebrafish
b-tectorin is not expressed as a tectorial membrane in the cochlea;
instead, it is expressed in anterior and posterior maculae of the
zebrafish ear (Fig. 3B), which is similar to the expression of
Starmaker mRNA [16]. MO knockdown of b-tectorin expression
affected otolith formation in zebrafish larvae (Fig. 4A). These b-
tectorin morphants showed a failure to maintain balance and float.
Only a few b-tectorin morphants (10%) were able to respond to a
vibration created by an ultrasonic processor in the water, while
most of them continued to lie on the bottom of the Petri dish
(Video S2). In addition to their responses to short vibrations, b-
tectorin morphants with either fused or single otoliths also showed
abnormal swimming patterns after tactile stimulation (Videos S5
and S7). These phenotypes suggest that zebrafish b-tectorin has
crucial roles in the development and function of the zebrafish
inner ear.
Some extracellular matrix proteins are reported to play
important roles in the development of the zebrafish inner ear.
For instance, MO knockdown of specific genes like omp-1 and
otolin-1, which respectively encode otolith matrix protein 1 and a
collagen-like protein [19], also showed abnormal otolith formation
and impaired swimming behaviors. The omp-1 MO resulted in a
reduced otolith size, while otolin-1 MO caused fusion of the 2
otoliths. Therefore, omp-1 was proposed to play important roles in
normal otolith growth, while otolin-1 is involved in stabilizing the
otolith matrix. In this study, b-tectorin morphants also showed
similar fused otoliths, but the zebrafish b-tectorin is not a collagen-
like protein. Therefore, both otolin-1 and b-tectorin may interact
with each other and polymerize into the otolith matrix.
In addition, the expression patterns of starmaker and b-tectorin
mRNAs were similar in the inner ear and in anterior and posterior
maculae of the ear as shown in Figs. 3B and 5A. The zebrafish
Starmaker protein is a 66-kD protein that is enriched in strongly
acidic amino acid residues and 35% proteins, and is also extremely
hydrophilic [16]. During zebrafish development, the Starmaker
protein is required for otolith biomineralization, and starmaker
morphants showed starry or chunky otoliths with improper
balance in freely swimming zebrafish larvae. Expression of the
starmaker transcript was slightly reduced in the anterior macula in
zebrafish b-tectorin morphants with fused otoliths. However, in
zebrafish b-tectorin morphants with a single otolith, a large portion
of the starmaker signal was lost in the anterior macula. Interactions
among omp-1, b-tectorin, and Starmaker proteins might be the
foundation of proper otolith formation.
We also studied the expression of zpDL1 mRNA in zebrafish b-
tectorin morphants. ZpDL1 can be used as a marker to label the 3
sensory cristae of zebrafish embryos at 4 dpf. As shown in Fig. 5C,
zpDL1 signals were lost in lateral cristae of zebrafish b-tectorin
morphants. These data suggest that zebrafish b-tectorin not only
regulates anterior macula formation but is also involved in the
morphogenesis of cristae. However, the underlying mechanisms
require further investigation.
Materials and Methods
Zebrafish care
Zebrafish embryos were raised at 28.5uC, and different
developmental stages were determined based on criteria described
in the Zebrafish Book [26]. All animal procedures were approved by
the Animal Use and Care Committee of Academia Sinica
(protocol #10-12-114).
Total RNA isolation and reverse-transcription polymerase
chain reaction (RT-PCR) analysis of zebrafish b-tectorin
mRNA
Total RNA was isolated from different developmental stages
and various tissues of adult zebrafish, using the RNAzol reagent
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 8 August 2011 | Volume 6 | Issue 8 | e23078
(Tel-Test, Friendswood, TX, USA) according to the instructions of
the manufacturer. After treatment with RQ1 RNase-Free DNaseI
(Promega, Madison, WI, USA), 50,100 mg of total RNA was
subjected to the first-strand cDNA synthesis. PCR amplifications
were performed with the following zebrafish b-tectorin RT-PCR
primers (b-tectorin-RT-F, 59- GCT GCT GAA GAC CTA CAC
AGG AAC-39and b-tectorin-RT-R, 59-TGG ATG TAT GCA
TGC ATG CGT GTC-39). Zebrafish b-actin primers (zACT-F,
59-GTG CTA GAC TCT GGT GAT GGT GTG-39and zACT-
R, 59-GGT GAT GAC CTG ACC GTC AGG AAG-39) were
used for the internal control to amplify a DNA fragment using
cDNA as a template. Primers for examining the efficiency of the
splice MOs are as follow (P1, 59- GCT GCT GAA GAC CTA
CAC AGG AAC- 39; P2, 59-GGC TAA ACA CGG CGT TGT
TGA CCA- 39.)
Cloning of the full-length cDNA encoding zebrafish b-
tectorin
To identify zebrafish complementary (c)DNA related to the
human b-tectorin gene, we used the coding region of human b-
tectorin (accession no. XM_521604) to search GenBank for related
expression sequence tags (ESTs) using the tBLAST program and
Figure 6. Abnormal swimming behaviors of
b-tectorin
morphants. b-Tectorin morphants were examined for their abilities to remain balance
and react to a stimulus. Tactile stimulation was created by poking a zebrafish on the head with a glass tube: b-tectorin morphants with a single (panel
A) and a fused otoliths (panel B), and a control with normal otoliths (panel C). Swimming behaviors of b-tectorin morphants at 5 days post-fertilization
under stimulation were recorded with a digital video camera. b-Tectorin morphants with either single or fused otoliths failed to maintain their
balance, tended to remain leaning on one side, remained on the bottom (panel A, B), and tended to swim in a corkscrew (panel A, A1 to A4) or
circular manner (panel B, B1 to B4). Control zebrafish maintained their balance, had immediate responses to stimulation, and swam in a straight line
(panel C, C1 to C4). The trails of the zebrafish movement were illustrated by dark arrows.
doi:10.1371/journal.pone.0023078.g006
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 9 August 2011 | Volume 6 | Issue 8 | e23078
found some zebrafish EST clones (CN315850 and EG585664)
related to human b-tectorin. Using 59- and 39-RACE to obtain
the 59- and 39-untranslated regions (UTRs), we assembled all
sequences to obtain 1542-bp cDNA with an open reading frame
(ORF) of 1011 bp encoding a protein of 336 amino acid residues.
The complete sequence was deposited in GenBank with the
accession number of FJ374270.
The full-length cDNA encoding zebrafish b-tectorin was isolated
by PCR amplification using gene-specific primers with linkers (b-
tectorin-BamH1-F, 59-CC GGA TCC ATG GCA GCT GTT
GGC CTT-39, and b-tectorin-EcoR1-R, 59-GG GAA TTC AAA
AGT AAA GTA TCC TAA-39) according to the sequence
submitted with GenBank accession no. FJ374270. Full-length
zebrafish b-tectorin was subcloned into the BamH1 and EcoR1 sites
of pcDNA3.1-myc to generate pcDNA3.1-b-tectorin- myc. Full
length b-tectorin was then further subcloned to T7TS plasmid using
BamH1 and Xba1 sites.
Rescue of defects in b-Tectorin morphant by injecting
b-Tectorin RNA
Full length b-Tectorin was cloned into T7TS plasmid and
synthesized in vitro. T7TS-b-Tectorin was linearized to synthesize
capping mRNA by using mMESSAGE mMACHINE T7 Kit
(Ambion, Foster City, CA, USA). ,100 pg of b-Tectorin RNA was
injected into embryos at the one- to two-cell stage.
Morpholino oligonucleotide (MO) injection
Antisense MOs were obtained from Gene Tools (Philomath,
OR, USA), and the sequence of zebrafish b-tectorin MO was as
follows: 59-GTG GCA GAA TCC AGA AGA AAT GTT G-39.
The sequence of the two splice MOs used were as follow: splice
MO 1 : 59- AAC CCA TCA AAC ATC TTA CCT CAG A-39
and splice MO 2 : 59-CCT CCT ACA TAC TGA AAA GAA
GGT A-39. The morpholinos were resolved to 24 mg/ml injection
stock, and stored in a 220uC refrigerator. The diluted morpholino
was injected into wild-type (WT) zebrafish embryos at the 1,2-cell
stage using a microinjection system consisting of an SZX9
stereomicroscope (Olympus, Tokoyo, Japan) and an IM300
Microinjector (Narishige, Tokoyo, Japan). The sequence of p53
MO was as follow (p53 MO: 59-AAA ATG TCT GTA CTA TCT
CCA TCC G-39) [27].
To confirm the specificities of the b-tectorin morpholino, several
pCMV-GFP reporter plasmids were created. The morpholino
targeted a 25-bp sequence of the PCR by the following primer pairs
for the perfect match, bTec-GFP (bTec-GFP-F, 59- GAT CCC
AAC ATT TCT TCT GGA TTC TGC CAC G-39and bTec-
GFP-R, 59-AAT TCG TGG CAG AAT CCA GAA GAA ATG
TTG G-39). For a 5-base exchanged mismatch, MM-b-Tec-GFP
was used (MM-b-Tec-GFP-F, 59-GAT CCG AAG ATT ACT
TCT GCA TTC TGG CAC G-39and MM-b-Tec-GFP-R,59-AAT
TCG TGC CAG AAT GCA GAA GTA ATC TTC G-39). The 59
region of the zebrafish b-tectorin mRNA was fused to the N-terminal
of the GFP protein. Either construct bTec-GFP or MM-b-Tec-GFP
was co-injected with zebrafish b-tectorin morpholino, and the
fluorescence was analyzed by a fluorescent microscope at 48 hpf.
Whole-mount in situ hybridization
Digoxigenin-labeled RNA probes (Roche, Penzberg, Germany)
were generated by in vitro transcription using the linearized
pGEM-T-easy plasmids (Promega, Madison, WI, USA) carrying
the 39-UTR of the following zebrafish genes. Whole-mount in situ
hybridization was performed following a previously described
protocol [28]. Specific primers for stm (stm-F, 59- GAA TCA ACT
GAG ACA GTC AAG ATA ACC -39and stm-R, 59- TGA GAG
TGG AGA GCG GGA ATT ATC TGC - 39), zpDL1 ( zpDL1-F,
59-GCG GGA CAT CAG TGT GTA TTG TGG AGT TCA -39
and zpDL1-R, 59- GCA AGC TGT GTG TTG TTG ACC AGG
TAT TCC -39), and omp-1 (zomp-1-F, 59- CAC ACT ACA GTC
TTT GAC AAC ATG - 39and zomp-1-R, 59- CAT CAG ATC
AAC ACA AAC CTT CAC - 39) were used to amplify the 39-
UTR of each gene. Primers used in the RT-PCR of zebrafish b-
tectorin were also used to make the zebrafish b-tectorin probe.
FM1-43 labeling of hair cells
Labeling the hair cells in the inner ear with 40 mM of FM1-43
(N-(3-triethylammoniumpropyl)-4(4-(dibutylamino)styryl) pyridi-
nium dibromide), (Invitrogen, Carlsbad, CA, USA) dissolved in
the extracellular solution. An injection tube was used to inject
FM1-43 into the otic vesicle following the protocols described
previously [29]. The formula of extracellular solution is described
as follow, 134 mM NaCl, 2.9 mM KCl, 1.2 mM MgCl
2
, 2.1 mM
CaCl
2
, 10 mM HEPES, and 10 mM glucose, and was adjusted to
pH 7.8.
Video recording of the swimming behavior of zebrafish
b-tectorin morphants
Zebrafish b-tectorin morphants were recorded with a digital
video camera (Sony DCR-PC120 digital camera, Tokyo, Japan) at
5 dpf to examine their reactions to short vibrations and tactile
stimulation. Short vibrations were created by a Hielscher Up50H
ultrasonic processor (Hielscher, Teltow, Germany), at 30 kHz and
50 W with an amplitude of 30% and 0.5 s per cycle. Tactile
stimulation was created using a glass tube to touch the head of a
zebrafish, and instantaneous responses were recorded with a
digital camera. Zebrafish b-tectorin morphants and control MO-
injected zebrafish were touched on the head at least 5 times for
each test. The swimming behavior of the zebrafish was observed
and defined by whether the fish swam in a straight or circular
manner.
Supporting Information
Figure S1 Control experiments for morpholino speci-
ficity. To determine the specificities of the morpholinos used,
pCMV-GFP reporter plasmids containing a perfect (bTec-GFP) or
mismatched (MM-b-Tec-GFP) MO target sequence were em-
ployed. Both bTec-GFP (A) and MM-b-Tec-GFP were co-injected
with the b-tectorin MO. All images were taken from zebrafish
embryos at 48 h post-fertilization.
(TIF)
Figure S2 The splice MO targeting and RT-PCR anal-
ysis of b-tectorin mRNAs of embryos injected with splice
MOs. (A) The exon-intron genomic structure from exons 1–4 was
shown. Splice MO 1 and MO 2 target the donor and acceptor
sites, respectively. (B) Total RNAs were extracted from control
MO (C) and splice MO1/MO2-injected (MO) embryos at 72 hpf,
then RT-PCR was performed. Primers (P1/P2) flanking the region
resulted in a single 500 bp band in the case of control embryos.
On the other hand, in the case of morphants, the level of this band
was strongly reduced and a second 1800 bp band was visible. The
second band resulted from the use of an alternative splice donor.
b-Actin bands were used to normalize the amount of cDNA
prepared from both embyos.
(TIF)
Figure S3 The morphology of b-tectorin morphants. The
ATG MO injected zebrafish embryos with fused (a, b), and single
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 10 August 2011 | Volume 6 | Issue 8 | e23078
otoliths (c, d) appeared to be normal without obvious defects. All
photographs were taken at 72 hpf.
(TIF)
Figure S4 Zebrafish b-tectorin amino acid sequence
alignment with other fish species. The deduced amino acid
sequences of zebrafish b-tectorin were aligned with those from
Tetraodon, fugu, and medaka. Identical residues in 3 or 4 proteins
are highlighted. The accession numbers of each b-tectorin from
different fish species are listed below: Tetraodon (GenBank, accession
no: CAG06543), fugu (ensembl no: ENSTRUP00000021095), and
medaka (ensembl no: ENSORLP00000014650).
(TIF)
Video S1 Control MO-injected zebrafish responded to
vibrations made with an ultrasonic processor. Control
MO-injected zebrafish at 5 days post-fertilization (dpf) with normal
otoliths were placed in a Petri dish, and a transient vibration was
generated with a Hielscher Up50H ultrasonic processor, at 30 kHz
and 50 W with an amplitude of 30% and 0.5 s per cycle.
(WMV)
Video S2 The b-tectorin morphant responded to the
vibration made with an ultrasonic processor. The b-
tectorin morphant at 5 days post-fertilization with either single or
fused otoliths were placed in a Petri dish, and a transient vibration
was generated with a Hielscher Up50H ultrasonic processor.
(WMV)
Video S3 Response of control MO-injected zebrafish to
touch with a glass tube. Control MO-injected zebrafish at 5
days post-fertilization with normal otoliths was touched on the
head with a glass tube at least 5 times, and its swimming behavior
was observed. The bright-field videomicrograph was taken with an
Olympus IX70-FLA inverted fluorescence microscope equipped
with a Sony DCR-PC120 digital video camera.
(WMV)
Video S4 Response of a b-tectorin morphant with fused
otoliths to touch with a glass tube. The b-tectorin morphant
at 5 days post-fertilization with fused otoliths was touched on
the head with a glass tube at least 5 times. The bright-field
videomicrograph was taken with an Olympus IX70-FLA inverted
fluorescence microscope equipped with a Sony DCR-PC120
digital video camera.
(WMV)
Video S5 Swimming behavior of a b-tectorin morphant
with fused otoliths. The b-tectorin morphant at 5 days post-
fertilization (dpf) with fused otoliths was touched on the head with a
glass tube at least 5 times, and its swimming behavior was observed.
The video was taken with a Sony DCR-PC120 digital video camera.
(WMV)
Video S6 Response of a b-tectorin morphant with a
single otolith to touch with a glass tube. The b-tectorin
morphant at 5 days post-fertilization with a single otolith was
touched on the head with a glass tube at least 5 times. The bright-
field videomicrograph was taken with an Olympus IX70-FLA
inverted fluorescence microscope equipped with a Sony DCR-
PC120 digital video camera.
(WMV)
Video S7 Swimming behavior of a b-tectorin morphant
with a single otolith. The b-tectorin morphant at 5 days post-
fertilization with a single otolith was touched on the head with a
glass tube at least 5 times, and its swimming behavior was observed.
The video was taken with a Sony DCR-PC120 digital video camera.
(WMV)
Author Contributions
Conceived and designed the experiments: C-HY C-JH. Performed the
experiments: C-HY C-HC G-DC W-HL Y-CC K-YH. Analyzed the data:
C-HY C-HC G-DC Y-CC C-JH. Contributed reagents/materials/analysis
tools: C-HC G-DC P-PH S-PLH. Wrote the paper: C-HY C-JH.
References
1. Jovine L, Darie CC, Litscher ES, Wassarman PM (2005) Zona pellucida domain
proteins. Annu Rev Biochem. 2005/06/15 ed. pp 83–114.
2. Jovine L, Qi H, Williams Z, Litscher E, Wassarman PM (2002) The ZP domain
is a conserved module for polymerization of extracellular proteins. Nat Cell Biol
4: 457–461.
3. Wassarman PM, Jovine L, Litscher ES (2001) A profile of fertilization in
mammals. Nat Cell Biol 3: E59–64.
4. Bork P, Sander C (1992) A large domain common to spe rm receptors (Zp2 and
Zp3) and TGF-beta type III receptor. FEBS Lett 300: 237–240.
5. Verhoeven K, Van Laer L, Kirschhofer K, Legan PK, Hughes DC, et al. (1998)
Mutations in the human alpha-tectorin gene cause autosomal dominant non-
syndromic hearing impairment. Nat Genet 19: 60–62.
6. Meyer NC, Alasti F, Nishimura CJ, Imanirad P, Kahrizi K, et al. (2007)
Identification of three novel TECTA mutations in Iranian families with
autosomal recessive nonsyndromic hearing impairment at the DFNB21 locus.
Am J Med Genet A 143A: 1623–1629.
7. Russell IJ, Legan PK, Lukashkina VA, Lukashkin AN, Goodyear RJ, et al.
(2007) Sharpened cochlear tuning in a mouse with a genetically modified
tectorial membrane. Nat Neurosci 10: 215–223.
8. Coutinho P, Goodyear R, Legan PK, Richardson GP (1999) Chick alpha-
tectorin: molecular cloning and expression during embryogenesis. Hear Res 130:
62–74.
9. Nicolson T (2005) The genetics of hearing and balance in zebrafish. Annu Rev
Genet 39: 9–22.
10. Whitfield TT, Riley BB, Chiang MY, Phillips B (2002) Development of the
zebrafish inner ear. Dev Dyn 223: 427–458.
11. Popper AN, Fay RR (1993) Sound detection and processing by fish: critical
review and major research questions. Brain Behav Evol 41: 14–38.
12. Killick R, Legan PK, Malenczak C, Richardson GP (1995) Molecular cloning of
chick beta-tectorin, an extracellular matrix molecule of the inner ear. J Cell Biol
129: 535–547.
13. Legan PK, Rau A, Keen JN, Richardson GP (1997) The mouse tectorins.
Modular matrix proteins of the inner ear homologous to components of the
sperm-egg adhesion system. J Biol Chem 272: 8791–8801.
14. Breathnach R, Benoist C, O’Hare K, Gannon F, Chambon P (1978) Ovalbumin
gene: evidence for a leader sequence in mRNA and DNA sequences at the exon-
intron boundaries. Proc Natl Acad Sci U S A 75: 4853–4857.
15. Kwak SJ, Vemaraju S, Moorman SJ, Zeddies D, Popper AN, et al. (2006)
Zebrafish pax5 regulates development of the utricular macula and vestibular
function. Dev Dyn 235: 3026–3038.
16. Sollner C, Burghammer M, Busch-Nentwich E, Berger J, Schwarz H, et al.
(2003) Control of crystal size and lattice formation by starmaker in otolith
biomineralization. Science 302: 282–286.
17. Nasevicius A, Ekker SC (2000) Effective targeted gene ‘knockdown’ in zebrafish.
Nat Genet 26: 216–220.
18. Robu ME, Larson JD, Nasevicius A, Beiraghi S, Brenner C, et al. (2007) p53
activation by knockdown technologies. PLoS Genet 3: e78.
19. Murayama E, Herbomel P, Kawakami A, Takeda H, Nagasawa H (2005)
Otolith matrix proteins OMP-1 and Otolin-1 are necessary for normal otolith
growth and their correct anchoring onto the sensory maculae. Mech Dev 122:
791–803.
20. Meyers JR, MacDonald RB, Duggan A, Lenzi D, Standaert DG, et al. (2003)
Lighting up the senses: FM1-43 loading of sensory cells through nonselective ion
channels. J Neurosci 23: 4054–4065.
21. Granato M, van Eeden FJ, Schach U, Trowe T, Brand M, et al. (1996) Genes
controlling and mediating locomotion behavior of the zebrafish embryo and
larva. Development 123: 399–413.
22. Goodyear RJ, Richardson GP (2002) Extracellular matrices associated with the
apical surfaces of sensory epithelia in the inner ear: molecular and structural
diversity. J Neurobiol 53: 212–227.
23. Wassarman PM (1999) Mammalian fertilization: molecular aspects of gamete
adhesion, exocytosis, and fusion. Cell 96: 175–183.
24. Rau A, Legan PK, Richardson GP (1999) Tectorin mRNA expressio n is
spatially and temporally restricted during mouse inner ear development. J Comp
Neurol 405: 271–280.
25. Legan PK, Lukashkina VA, Goodyear RJ, Lukashkin AN, Verhoeven K, et al.
(2005) A deafness mutation isolates a second role for the tectorial membrane in
hearing. Nat Neurosci 8: 1035–1042.
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 11 August 2011 | Volume 6 | Issue 8 | e23078
26. Westerfield M (2007) THE ZEBRAFISH BOOK: A guide for the laboratory use
of zebrafish (Danio rerio), 5th Edition. Eugene: University of Oregon Press.
27. Chen J, Ruan H, Ng SM, Gao C, Soo HM, et al. (2005) Loss of function of def
selectively up-regulates Delta113p53 expression to arrest expansion growth of
digestive organs in zebrafish. Genes Dev 19: 2900–2911.
28. Chen YC, Cheng CH, Chen GD, Hung CC, Yang CH, et al. (2009)
Recapitulation of zebrafish sncga expression pattern and labeling the habenular
complex in transgenic zebrafish using green fluorescent protein reporter gene.
Dev Dyn 238: 746–754.
29. Tanimoto M, Ota Y, Inoue M, Oda Y (2011) Origin of inner ear hair cells:
morphological and functional differentiation from ciliary cells into hair cells in
zebrafish inner ear. J Neurosci 31: 3784–3794.
b-Tectorin in Zebrafish Inner Ear Development
PLoS ONE | www.plosone.org 12 August 2011 | Volume 6 | Issue 8 | e23078
... Expressed in the cochlea and vestibular organs (mainly the utricle and saccule) (Weigele et al., 2015) Maintains normal otolith growth and is directly related to otolith shape (Murayama et al., 2005) Associated with movement disorders (Weigele et al., 2015) Cochlin Expressed in the cochlea and vestibule (mainly in spiral ligament, spiral bone and spiral rim of the inner ear) (Rhyu et al., 2020) Main otolith constituent, determinant of calcium carbonate crystalline formation (Leventea et al., 2021) Its overexpression is associated with Ménière's disease (Calzada et al., 2012); associated with conductive hearing loss and vestibular dysfunction (Leventea et al., 2021;Verdoodt et al., 2021); diagnosis of exolymphatic fistula (Xiong et al., 2020); diagnosis of exolymphatic fistula Otogelin Expression in the cochlea and vestibule (Avan et al., 2019) Anchoring of the otolith (Schraders et al., 2012;Whitfield, 2020); stabilization of the cochlear covering membrane (Avan et al., 2019) Deficiency leading to otolith detachment and instability of the cochlear covering membrane; OTOG mutation leading to DFNB18B hereditary deafness (Avan et al., 2019) α-tectorin and β-tectorin Non-collagenous glycoprotein component of the Cochlear Covering Membrane (TM) (Andrade et al., 2016) Adherence of TM to the spiral rim and to the stereocilia of the outer hair cells of the cochlea (Andrade et al., 2016) Determinants of otolith formation (Asgharzade et al., 2011) Deficiency can lead to varying degrees of hearing loss (Asgharzade et al., 2017) β-tectorin deficiency can lead to imbalance (Zou et al., 2006) Otopetrin1 and Otopetrin-2 Expression in the cochlea and vestibule (Kim et al., 2010;Tu et al., 2018;Khan et al., 2019;Lopez et al., 2019) Maintains nucleus formation, and growth of otoliths (Kim et al., 2010); a proton-selective channel involved in gravity sensing in the vestibular system Lack of otopetrin-1 knockout in mice leads to vestibular dysfunction (Khan et al., 2019). ...
... In humans, mutations in the TECTA gene, which encodes alpha-tectorin, cause moderate to severe hearing loss (Asgharzade et al., 2017). Studies have shown that β-tectorin knockouts in zebrafish show balance defect due to disrupted otolith formation and result in severe defects in otolith formation and function in the inner ear, thus suggesting that β-tectorin can affect otolith formation (Asgharzade et al., 2011). However, the underlying molecular mechanisms need to be further investigated. ...
Article
Full-text available
Otoliths are biological crystals formed by a layer of calcium carbonate crystal that adhere to the ciliary surface of the utricular and saccular receptors in the vestibule of all vertebrates inner ear, enabling the utricle and saccule to better perceive the changes in linear and gravitational acceleration. However, the molecular etiology of otolith related diseases is still unclear. In this review, we have summarized the recent findings and provided an overview of the proteins that play important roles in otolith formation and maintenance (Otoconin-90, Otolin-1, Otolith Matrix Protein-1, Cochlin, Otogelin, α-Tectorin, β-Tectorin, Otopetrin-1, and Otopetrin-2, PMCA2, etc.), providing new insight for the prevention and management of benign paroxysmal positional vertigo (BPPV) with basis for otolith-related proteins as potential biomarkers of vestibular disease.
... The tectb gene encodes Tectorin beta, a component of the tectorial membrane associated with cochlear hair cells in mammals (Goodyear et al., 2017), and a component of otoliths in zebrafish (Kalka et al., 2019). The zpld1a gene, encoding Zona-pellucida-like domain containing protein 1a, is expressed in the cristae in fish (Dernedde et al., 2014;Yang et al., 2011) and mouse (Vijayakumar et al., 2019). Using fluorescent in situ hybridization, we find that tectb is expressed in the macular organs but is absent from cristae ( Figure 5C). ...
Preprint
Full-text available
A major cause of human deafness and vestibular dysfunction is permanent loss of the mechanosensory hair cells of the inner ear. In non-mammalian vertebrates such as zebrafish, regeneration of missing hair cells can occur throughout life. While a comparative approach has the potential to reveal the basis of such differential regenerative ability, the degree to which the inner ears of fish and mammals share common hair cells and supporting cell types remains unresolved. Here we perform single-cell RNA sequencing of the zebrafish inner ear at embryonic through adult stages to catalog the diversity of hair cell and non-sensory supporting cells. We identify a putative progenitor population for hair cells and supporting cells, as well as distinct hair cells and supporting cell types in the maculae versus cristae. The hair cell and supporting cell types differ from those described for the lateral line system, a distributed mechanosensory organ in zebrafish in which most studies of hair cell regeneration have been conducted. In the maculae, we identify two subtypes of hair cells that share gene expression with mammalian striolar or extrastriolar hair cells. In situ hybridization reveals that these hair cell subtypes occupy distinct spatial domains within the two major macular organs, the utricle and saccule, consistent with the reported distinct electrophysiological properties of hair cells within these domains. These findings suggest that primitive specialization of spatially distinct striolar and extrastriolar hair cells likely arose in the last common ancestor of fish and mammals. The similarities of inner ear cell type composition between fish and mammals also support using zebrafish as a relevant model for understanding inner ear-specific hair cell function and regeneration.
... As shown in Fig. 3G, the cells with high tectb gene expression were mainly gathering in the cluster 5, and the tectb gene was proved to be expressed specifically in the macula hair cells, including the utricular hair cells and saccular hair cells, by WISH. The zpld1a gene was mainly expressed in the crista hair cells, which were thought to be located in the cluster 12 according to our analysis (Fig. 3H), and this is consistent with the previous studies [21]. The cells from the cluster 0 and 7 were considered as neuromast hair cells because of the expression patterns of their marker genes, such as calm1b (Fig. 3I). ...
Article
Full-text available
Hair cells play key roles in hearing and balance, and hair cell loss would result in hearing loss or vestibular dysfunction. Cellular and molecular research in hair cell biology provides us a better understanding of hearing and deafness. Zebrafish, owing to their hair cell-enriched organs, have been widely applied in hair cell-related research worldwide. Similar to mammals, zebrafish have inner ear hair cells. In addition, they also have lateral line neuromast hair cells. These different types of hair cells vary in morphology and function. However, systematic analysis of their molecular characteristics remains lacking. In this study, we analyzed the GFP+ cells isolated from Tg(Brn3c:mGFP) larvae with GFP expression in all hair cells using single-cell RNA-sequencing (scRNA-seq). Three subtypes of hair cells, namely macula hair cell (MHC), crista hair cell (CHC), and neuromast hair cell (NHC), were characterized and validated by whole-mount in situ hybridization analysis of marker genes. The hair cell scRNA-seq data revealed hair cell-specific genes, including hearing loss genes that have been identified in humans and novel genes potentially involved in hair cell formation and function. Two novel genes were discovered to specifically function in NHCs and MHCs, corresponding to their specific expression in NHCs and MHCs. This study allows us to understand the specific genes in hair cell subpopulations of zebrafish, which will shed light on the genetics of both human vestibular and cochlear hair cell function.
... As shown in Figure 3G, the cells with high tectb gene expression were mainly gathering in the cluster 5, and the tectb gene was proved to be expressed speci cally in the macula hair cells, including the utricular hair cells and saccular hair cells, by WISH. The zpld1a gene was mainly expressed in the crista hair cells, which were thought to be located in the cluster 12 according to our analysis ( Figure 3H), and this is consistent with previous studies [22] . The cells from the cluster 0 and 7 were considered as neuromast hair cells because of the expression patterns of their marker genes, such as calm1b ( Figure 3I). ...
Preprint
Full-text available
Hair cells play key roles in hearing and balance, and hair cell loss would result in hearing loss or vestibular dysfunction. Cellular and molecular research in hair cell biology provide us a better understanding of hearing and deafness. Zebrafish, owing to their hair cell-enriched organs, have been widely applied in hair cell-related research worldwide. Similar to mammals, zebrafish have inner ear hair cells. In addition, they also have lateral line neuromast hair cells. These different hair cells vary in morphology and function. However, systematic analysis of their molecular characteristics remains lacking. In this study, we analyzed the GFP+ cells isolated from Tg(Brn3c:mGFP) larvae using single cell RNA-sequencing (scRNA-seq). Three subtypes of hair cells, namely macula hair cell (MHC), crista hair cell (CHC) and neuromast hair cell (NHC) were characterized and validated by whole-mount in situ hybridization analysis of marker genes. Furthermore, we carried out the morphological comparison of these hair cells. Based on the hair cell scRNA-seq data, we obtained hair cell-enriched genes pool, including hearing loss genes that have been identified in humans and novel genes potentially involved in hair cell formation and function. Two novel genes were discovered to specifically function in NHCs and MHCs, corresponding to their specific expression in NHCs and MHCs. This study allows us to understand the specific genes in hair cell subpopulations of zebrafish, which will shed light on the genetics of both human vestibular and cochlear hair cell function.
... Other important factors for proper tethering include α-Tectorin and β-Tectorin, which each encode non-collagenous glycoproteins that are expressed in the macular sensory epithelia (Legan et al, 1997;Rau et al., 1999). In mouse and zebrafish studies, null mutants of α-Tectorin have scattered otoconia and reduced otolithic membrane, whereas β-Tectorin have fewer or fused otoliths (Legan et al., 2000;Yang, Cheng, et al., 2011). Interestingly, analysis of the acellular membrane overlying the auditory sensory epithelium of the inner ear, the tectorial membrane, demonstrates that it also contains α-Tectorin and β-Tectorin (Goodyear & Richardson, 2002). ...
Article
The vertebrate inner ear is a labyrinthine sensory organ responsible for perceiving sound and body motion. While a great deal of research has been invested in understanding the auditory system, a growing body of work has begun to delineate the complex developmental program behind the apparatuses of the inner ear involved with vestibular function. These animal studies have helped identify genes involved in inner ear development and model syndromes known to include vestibular dysfunction, paving the way for generating treatments for people suffering from these disorders. This review will provide an overview of known inner ear anatomy and function and summarize the exciting discoveries behind inner ear development and the evolution of its vestibular apparatuses.
Article
Full-text available
Otoliths are calcium carbonate components of the stato-acoustical organ responsible for hearing and maintenance of the body balance in teleost fish. During their formation, control over, e.g., morphology and carbonate polymorph is influenced by complex insoluble collagen-like protein and soluble non-collagenous protein assemblages; many of these proteins are incorporated into their aragonite crystal structure. However, in the fossil record these proteins are considered lost through diagenetic processes, hampering studies of past biomineralization mechanisms. Here we report the presence of 11 fish-specific proteins (and several isoforms) in Miocene (ca. 14.8–14.6 Ma) phycid hake otoliths. These fossil otoliths were preserved in water-impermeable clays and exhibit microscopic and crystallographic features indistinguishable from modern representatives, consistent with an exceptionally pristine state of preservation. Indeed, these fossil otoliths retain ca. 10% of the proteins sequenced from modern counterparts, including proteins specific to inner ear development, such as otolin-1-like proteins involved in the arrangement of the otoliths into the sensory epithelium and otogelin/otogelin-like proteins that are located in the acellular membranes of the inner ear in modern fish. The specificity of these proteins excludes the possibility of external contamination. Identification of a fraction of identical proteins in modern and fossil phycid hake otoliths implies a highly conserved inner ear biomineralization process through time.
Article
Full-text available
A major cause of human deafness and vestibular dysfunction is permanent loss of the mechanosensory hair cells of the inner ear. In non-mammalian vertebrates such as zebrafish, regeneration of missing hair cells can occur throughout life. While a comparative approach has the potential to reveal the basis of such differential regenerative ability, the degree to which the inner ears of fish and mammals share common hair cells and supporting cell types remains unresolved. Here we perform single-cell RNA sequencing of the zebrafish inner ear at embryonic through adult stages to catalog the diversity of hair cells and non-sensory supporting cells. We identify a putative progenitor population for hair cells and supporting cells, as well as distinct hair and supporting cell types in the maculae versus cristae. The hair cell and supporting cell types differ from those described for the lateral line system, a distributed mechanosensory organ in zebrafish in which most studies of hair cell regeneration have been conducted. In the maculae, we identify two subtypes of hair cells that share gene expression with mammalian striolar or extrastriolar hair cells. In situ hybridization reveals that these hair cell subtypes occupy distinct spatial domains within the three macular organs, the utricle, saccule, and lagena, consistent with the reported distinct electrophysiological properties of hair cells within these domains. These findings suggest that primitive specialization of spatially distinct striolar and extrastriolar hair cells likely arose in the last common ancestor of fish and mammals. The similarities of inner ear cell type composition between fish and mammals validate zebrafish as a relevant model for understanding inner ear-specific hair cell function and regeneration.
Article
Pinewood releases ethanol and other volatile compounds after Bursaphelenchus xylophilus infection. In the current study, we examined the influence of different ethanol concentrations on B. xylophilus reproduction. Low-concentrations of ethanol (8.5, 17, and 34 mM) increased egg production in B. xylophilus, whereas higher-concentrations (156 and 312 mM) reduced egg production. Transcriptome analysis was conducted to explore the molecular response of a low concentration of ethanol on the nematodes. The results suggest that the nematodes use ethanol as an energy source, which may promote survival. Ethanol induced changes in the expression of genes involved in the biosynthesis and metabolism of fatty acids and amino acids. Furthermore, ethanol promoted the expression of detoxification-related, cell wall-degrading, and reproduction-related genes. Such responses might contribute to the pathogenicity of B. xylophilus.
Article
Mandarin fish (Siniperca chuatsi) is a popular inland farmed fish species with high an economic value. Molecular selective breeding programs are urgently required to improve the growth traits of mandarin fish for farming industry. In this study, using 160 mandarin fish individuals, specific locus-amplified fragment sequencing and SNP genotyping, we performed a genome-wide association study (GWAS) to identify genomic regions and candidate genes that are potentially associated with various growth traits of mandarin fish, such as body weight (BW), body thickness (BT), body height (BH) total length (TL), slaughter weight (SW), and body length (BL). Through genotyping, 5205 high-quality SNPs were found located on 24 chromosomes. A total of 11 significant and 37 suggestive SNPs related to growth traits were obtained, among which five SNPs significantly related to BW were located in the linkage group (LG) 8, 17, 21, and 23, two SNPs significantly related to TL were located in LG 7 and 17, two SNPs significantly related to BH were located on LG 17 and 23, two SNPs significantly related to BT were located on LG 14 and 19, respectively. Furthermore, LG17_937,734 and LG23_23,856,613 were growth-related traits overlap. Within 300 kb upstream and downstream of these significant SNPs, 65 candidate genes were identified within the associated QTLs, many of which were related to bone differentiation, growth and development, cell division and neurogenesis. In particular, asns, tg, mkk6, htra1, rnf213, ttn, tgfbr2, and nck2, were shown to be closely associated with growth, fat, muscle and bone formation. To our knowledge, this is the first GWAS focusing on growth traits of mandarin fish. Our findings establish a foundation for molecular marker-assisted selecting, genomic selecting, and functional analyzing the growth traits of mandarin fish.
Article
Copper is one of the most ubiquitous environmental pollutants worldwide. Previous studies have focused on the toxicology of high copper exposure, while there has been comparatively less research on the biological effects of low copper exposure. Low concentrations of copper often exist in freshwater ecosystems, and its impact on the fish is unclear. Both Xenocypris microlepis and Xenocypris davidi are bottom-feeding fishes widely distributed in freshwater ecosystems of China, and they are more likely to be contaminated by low concentrations of copper. Low copper exposure may have effects on molecular regulation at the level of gene expression in the two Xenocypris species. To investigate gene expression differences involved in the response to low copper concentrations between X. microlepis and X. davidi, we established the responses to low copper exposure of 0.01 mg/L for 14 days at the transcriptional level, and RNA-Seq was used to perform a comparative transcriptomic analysis of the liver. A total of 74,135 and 60,894 unigenes from X. microlepis and X. davidi were assembled by transcriptome profiling, respectively. Among these, 84 genes of X. microlepis and 165 genes of X. davidi were identified as differentially expressed genes (DEGs). There were 60 and 135 up-regulated, 24 and 30 down-regulated genes in the two species, respectively. Comparative transcriptome analyses identified five differentially co-expressed genes (DCGs) related to low copper exposure from the DEGs of the two Xenocypris species. The five DCGs were related to the fishes’ growth, antioxidant system, immune system and heavy metal tolerance. The results could help us to understand the molecular mechanisms of the response to low copper exposure, and the data should provide a valuable transcriptomic resource for the genus Xenocypris.
Article
Full-text available
Morpholino phosphorodiamidate antisense oligonucleotides (MOs) and short interfering RNAs (siRNAs) are commonly used platforms to study gene function by sequence-specific knockdown. Both technologies, however, can elicit undesirable off-target effects. We have used several model genes to study these effects in detail in the zebrafish, Danio rerio. Using the zebrafish embryo as a template, correct and mistargeting effects are readily discernible through direct comparison of MO-injected animals with well-studied mutants. We show here indistinguishable off-targeting effects for both maternal and zygotic mRNAs and for both translational and splice-site targeting MOs. The major off-targeting effect is mediated through p53 activation, as detected through the transferase-mediated dUTP nick end labeling assay, acridine orange, and p21 transcriptional activation assays. Concurrent knockdown of p53 specifically ameliorates the cell death induced by MO off-targeting. Importantly, reversal of p53-dependent cell death by p53 knockdown does not affect specific loss of gene function, such as the cell death caused by loss of function of chordin. Interestingly, quantitative reverse-transcriptase PCR, microarrays and whole-mount in situ hybridization assays show that MO off-targeting effects are accompanied by diagnostic transcription of an N-terminal truncated p53 isoform that uses a recently recognized internal p53 promoter. We show here that MO off-targeting results in induction of a p53-dependent cell death pathway. p53 activation has also recently been shown to be an unspecified off-target effect of siRNAs. Both commonly used knockdown technologies can thus induce secondary but sequence-specific p53 activation. p53 inhibition could potentially be applicable to other systems to suppress off-target effects caused by other knockdown technologies.
Article
Full-text available
Auditory and vestibular functions in vertebrates depend on the transduction of sound vibration or head acceleration into electrical responses in inner ear hair cells. Mechanoelectrical transduction occurs at the tip of stereocilia, which are polarized to form an orientational arrangement that determines directional sensitivity. It remains to be clarified when and how premature hair cells acquire their specialized structure and function in living animals. The developmental origin of inner ear hair cells has been studied in vivo in zebrafish embryos. Tether cells, a small number of ciliated cells associated with an "ear stone" (or otolith) in the embryonic zebrafish inner ear, are believed to be precocious hair cells. However, whether or not tether cells acquire hair bundles and mechanosensitivity remains unknown. In the present study, we investigated the morphological and functional development of tether cells. Immunohistochemical examination revealed that stereocilia appeared on the tether cell apex in a polarized arrangement at 22 h postfertilization (hpf). Labeling with FM1-43, a marker of functional mechanotransduction channels, and the in vivo electrophysiological recording of mechanotransducer responses in the developing inner ear demonstrated that tether cells acquired direction-selective mechanosensitivity at 23 hpf. These results revealed that tether cells begin to function as hair cells within an hour after the appearance of a polarized array of stereociliary bundles. Thus, the ciliary cells morphologically and functionally differentiate into the first sensory hair cells in the inner ear of the zebrafish.
Article
Full-text available
Selected regions of cloned EcoRI fragments of the chicken ovalbumin gene have been sequenced. The positions where the sequences coding for ovalbumin mRNA (ov-mRNA) are interrupted in the genome have been determined, and a previously unreported interruption in the DNA sequences coding for the 5' nontranslated region of the messenger has been discovered. Because directly repeated sequences are found at exon-intron boundaries, the nucleotide sequence alone cannot define unique excision-ligation points for the processing of a possible ov-mRNA precursor. However, the sequences in these boundary regions share common features; this leads to the proposal that there are, in fact, unique excision-ligation points common to all boundaries.
Article
The cDNA and derived amino acid sequences for the two major non-collagenous proteins of the mouse tectorial membrane, alpha- and beta-tectorin, are presented. The cDNA for alpha-tectorin predicts a protein of 239,034 Da with 33 potential N-glycosylation sites, and that of beta-tectorin a smaller protein of 36,074 Da with 4 consensus N-glycosylation sites. Southern and Northern blot analysis indicate alpha and beta-tectorin are single copy genes only expressed in the inner ear, and in situ hybridization shows they are expressed by cells both in and surrounding the mechanosensory epithelia. Both sequences terminate with a hydrophobic COOH terminus preceded by a potential endoproteinase cleavage site suggesting the tectorins are synthesized as glycosylphosphatidylinositol-linked, membrane bound precursors, targeted to the apical surface of the inner ear epithelia by the lipid and proteolytically released into the extracellular compartment. The mouse beta-tectorin sequence contains a single zona pellucida domain, whereas alpha-tectorin is composed of three distinct modules: an NH2-terminal region similar to part of the entactin G1 domain, a large central segment with three full and two partial von Willebrand factor type D repeats, and a carboxyl-terminal region which, like beta-tectorin, contains a single zona pellucida domain. The central, high molecular mass region of alpha-tectorin containing the von Willebrand factor type D repeats has homology with zonadhesin, a sperm membrane protein that binds to the zona pellucida. These results indicate the two major non-collagenous proteins of the tectorial membrane are similar to components of the sperm-egg adhesion system, and, as such may interact in the same manner.
Article
The tectorial membrane is an extracellular matrix lying over the apical surface of the auditory epithelium. Immunofluorescence studies have suggested that some proteins of the avian tectorial membrane, the tectorins, may be unique to the inner ear (Killick, R., C. Malenczak, and G. P. Richardson. 1992. Hearing Res. 64:21-38). The cDNA and deduced amino acid sequences for chick beta-tectorin are presented. The cDNA encodes a protein of 36,902.6 D with a putative signal sequence, four potential N-glycosylation sites, 13 cysteines, and a hydrophobic COOH terminus. Western blots of two-dimensional gels using antibodies to a synthetic peptide confirm the identity of the cDNA. Southern and Northern analysis suggests that beta-tectorin is a single-copy gene only expressed in the inner ear. The predicted COOH terminus is similar to that of glycosylphosphatidylinositol-linked proteins, and antisera raised to this region react with in vitro translation products of the cDNA clone but not with mature beta-tectorin. These data suggest beta-tectorin is synthesized as a glycosylphosphatidyl-inositol-linked precursor, targeted to the apical surface of the sensory epithelium by the lipid moiety, and then further processed. Sequence analysis indicates the predicted protein possesses a zona pellucida domain, a sequence that is common to a limited number of other matrix-forming proteins and may be involved in the formation of filaments. In the cochlear duct, beta-tectorin is expressed in the basilar papilla, in the clear cells and the cuboidal cells, as well as in the striolar region of the lagena macula. The expression of beta-tectorin is associated with hair cells that have an apical cell surface specialization known as the 275-kD hair cell antigen restricted to the basal region of the hair bundle, suggesting that matrices containing beta-tectorin are required to drive this hair cell type.
Article
The literature on fish hearing has increased significantly since our last critical review in 1973. The purpose of the current paper is to review the more recent literature and to identify those questions that need to be asked to develop a fuller understanding of the auditory capabilities and processing mechanisms of fishes. We conclude that while our understanding of fish hearing has increased substantially in the past years, there are still major gaps in what we know. In particular, the comparative functional literature is extremely limited, and we do not yet know whether different species, and particularly hearing specialists as compared to hearing nonspecialists, have fundamentally different auditory capabilities and mechanisms.Copyright © 1993 S. Karger AG, Basel
Article
Human synuclein family consists of alpha-, beta-, and gamma-synucleins. Here, we cloned three genes, sncb, sncga and sncgb from zebrafish. They encode beta-, gamma1-, and gamma2-synucleins, respectively. The zSyn-beta, zSyn-gamma1, and zSyn-gamma2 proteins display 69%, 47%, and 50% identity to human beta-synuclein and gamma-synuclein, respectively. By reverse transcriptase-polymerase chain reaction, we demonstrated that sncb and sncga mRNA were abundant in brain and eye, while sncgb expression was moderate in brain, kidney, ovary and testis. The 1.8-kb 5'-upstream/promoter region of the sncga gene was sufficient to direct green fluorescent protein (GFP) expression in the central nervous system and cranial ganglions. A transgenic line, Tg(sncga:GFP), was generated and its GFP expression is similar to that of endogenous sncga mRNA. Moreover, this line also labels the habenular complex and the domain of GFP expression is larger in the left than in the right habenula. Thus, this line can be used to study sncga gene regulation and for left-right asymmetry study in zebrafish brain.