Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Human Molecular Genetics, 2006, Vol. 15, No.

17 2569–2587
doi:10.1093/hmg/ddl184
Advance Access published on July 18, 2006

Pathogenetic role of the deafness-related M34T


mutation of Cx26
Massimiliano Bicego1,{, Martina Beltramello2,{, Salvatore Melchionda3, Massimo Carella3,
Valeria Piazza2, Leopoldo Zelante3, Feliksas F. Bukauskas4, Edoardo Arslan5, Elona Cama5,
Sergio Pantano2,7, Roberto Bruzzone6,*, Paola D’Andrea1,{ and Fabio Mammano2,7,8,{
1
Dipartimento di Biochimica, Biofisica e Chimica delle Macromolecole, University of Trieste, 34127 Trieste, Italy,
2
Istituto Veneto di Medicina Molecolare (VIMM), Fondazione per la Ricerca Biomedica Avanzata, 35129 Padova, Italy,
3
Servizio di Genetica Medica, IRCCS-Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy,
4
Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA, 5Servizio di Audiologia e
Foniatria, University of Padova, 35128 Padova, Italy, 6Département de Neuroscience, Institut Pasteur, 75015 Paris,
France and 7Consorzio Nazionale Interuniversitario per le Scienze Fisiche della Materia (CNISM) and 8Dipartimento di
Fisica ‘G.Galilei’, Università di Padova, 35131 Padova, Italy

Received April 21, 2006; Revised July 5, 2006; Accepted July 12, 2006

Mutations in the GJB2 gene, which encodes the gap junction protein connexin26 (Cx26), are the major cause
of genetic non-syndromic hearing loss. The role of the allelic variant M34T in causing hereditary deafness
remains controversial. By combining genetic, clinical, biochemical, electrophysiological and structural mod-
eling studies, we have re-assessed the pathogenetic role of the M34T mutation. Genetic and audiological data
indicate that the majority of heterozygous carriers and all five compound heterozygotes exhibited an
impaired auditory function. Functional expression in transiently transfected HeLa cells showed that,
although M34T was correctly synthesized and targeted to the plasma membrane, it inefficiently formed inter-
cellular channels that displayed an abnormal electrical behavior and retained only 11% of the unitary conduc-
tance of the wild-type protein (HCx26wt). Moreover, M34T channels failed to support the intercellular
diffusion of Lucifer Yellow and the spreading of mechanically induced intercellular Ca21 waves. When
co-expressed together with HCx26wt, M34T exerted dominant-negative effects on cell – cell coupling.
Our findings are consistent with a structural model, predicting that the mutation leads to a constriction of
the channel pore. These data support the view that M34T is a pathological variant of Cx26 associated with
hearing impairment.

INTRODUCTION association of mutations in several human connexins with a


variety of genetic diseases and by the specific phenotypes
Connexins comprise a multi-gene family of proteins that revealed by targeted connexin gene deletion in mice (3 – 5).
form the intercellular channels clustered at gap junctions, In the inner ear, three connexin isoforms have been found
through which ions, small metabolites and second messen- to be expressed in overlapping patterns (6) and their
gers are exchanged between connected cells (1). Intercellular crucial role in organ physiology has been revealed by their
channels are formed by two half-channels, or connexons, implication in different forms of hereditary deafness (7).
consisting of six connexin subunits that span the plasma Thus, mutations in human Cx26 (HCx26), HCx30 and
membranes of communicating cells and dock in the intercel- HCx31 (GJB2, GJB6 and GJB3 genes, respectively) have
lular space (2). The specific role of connexin channels in the been linked to both syndromic and non-syndromic forms of
homeostasis of different organs has been illustrated by the hearing loss (reviewed in 8).

*To whom correspondence should be addressed at: Département de Neuroscience, Institut Pasteur, 25, rue du Dr Roux, 75015 Paris, France.
Tel: þ33 140613436; Fax: þ33 140613421; Email: bruzzone@pasteur.fr
{
These authors have contributed equally.

# The Author 2006. Published by Oxford University Press. All rights reserved.
For Permissions, please email: journals.permissions@oxfordjournals.org
2570 Human Molecular Genetics, 2006, Vol. 15, No. 17

The arrangement of gap junctions between supporting cells (as found in heterozygous individuals) did not significantly
of the organ of Corti appears to be the structural basis for the reduce wild-type coupling.
re-circulation of cochlear Kþ ions that flow from the endo- Given the apparent contradictions of these reports, we
lymph, where their concentrations are 150 mM , into hair sought to re-assess the pathogenetic role of the M34T
cells upon sound stimulation (9). In this scheme, Kþ ions variant by combining genetic, auditory, biochemical, electro-
flow into the hair cells through mechanically gated channels physiological and structural modeling studies. We provide
by the combined effect of the positive endolymphatic potential here genetic and audiological evidence that although heterozy-
(þ80 mV) and the negative intracellular potential and gous carriers are found among normal hearing subjects, the
depolarize them. Kþ ions exit hair cells reaching the intersti- majority of them (62.5%) exhibited a mild to moderate
tial space of the organ of Corti, where they are partly taken hearing impairment. Furthermore, all compound heterozygotes
up by cochlear supporting cells (10). The excess Kþ concen- were affected by hearing loss, supporting the view that M34T
tration that results from auditory signals or sustained deleter- is a pathological variant. We also show that M34T is correctly
ious stimuli, such as ischemia or prolonged exposure to high synthesized and targeted to HeLa cells membrane, although it
sound pressure levels (reviewed in 11), is thought to be forms intercellular channels that retain an abnormal electrical
removed via an intercellular pathway delineated by intercellu- activity. Interestingly, these channels fail to sustain the inter-
lar channels composed of Cx26 and Cx30 that provide a cellular diffusion of Lucifer Yellow (LY) and largely inhibit
spatial buffering network similar to that of astrocytic gap junc- the spreading of intercellular Ca2þ waves. Finally, when
tions in the brain (12). Genetic studies have demonstrated that co-expressed with HCx26wt at a 1:1 protein ratio, M34T
a specific single-base deletion in Cx26, named 35delG, is the invariably shows dominant-negative effects on cell – cell com-
most common mutation worldwide, although its prevalence munication. These results fit well with our structural model,
varies with ethnical groups (8). In addition, several missense predicting that the mutation induces a constriction of the
mutations responsible for either recessive or dominant forms channel pore.
of the disease have been identified (for an updated list, see
Rabionet et al. (2002) http://www.iro.es/cx26deaf.html). The
first deafness-related Cx26 mutation, which was detected in a RESULTS
dominant pedigree of non-syndromic hearing loss, was a
methionine-to-threonine substitution at position 34 (M34T) (13). Genetic and audiological features
The pathogenetic role of M34T was initially supported by We have studied seven families in which one or more
the results of its functional expression in Xenopus oocytes, hearing-impaired members had the M34T mutation. Besides
which showed a dominant-negative effect of the M34T carrying out the routine genetic tests, we examined the audio-
mutant (14). Further studies on the family first described by logical features of all members of the families to disclose
Kelsell et al. (13), however, uncovered the association of der- even mild auditory defects potentially associated with the
matological signs in deaf patients and identified another domi- mutation. Cx26 mutations in heterozygous or compound het-
nant mutation in GJB2 segregating with the disease, casting erozygous condition were found in 21 patients, without any
doubts on the significance of the M34T variant. Subsequently, instance of homozygosity (Fig. 1). The M34T mutation was
the same M34T substitution was also reported to be respon- identified in 16 individuals, either as heterozygotes or com-
sible for a recessive form of hearing loss (15), whereas pound heterozygotes. Moreover, in 14 of them, we found
several authors described normal hearing in heterozygous the presence of a 10 bp deletion in the 50 -untranslated
carriers of M34T associated with either 35delG or other region of the gene (2493del10), in cis with M34T. This del-
Cx26 recessive mutations (16 – 19) and, therefore, considered etion was absent in all other individuals enrolled in this study.
it a benign polymorphism. Patients were divided into three groups: group I, consisting of
More recent in vitro data have shown that in transfected patients negative for GJB2/GJB6 mutations; group II, which
HeLa cells, the M34T variant disrupts either channel for- included patients carrying mutations different from the
mation or its oligomerization (20,21). In contrast, Thonnissen M34T and group III, with patients carrying at least the
et al. (22) observed low levels of dye transfer between HeLa M34T mutation.
cells expressing M34T, providing the first evidence that this In group I, hearing loss was present only in one patient
mutant could traffic to the cell membrane and form intercellu- (Fig. 1; family 5, subject I.2), whose hearing threshold was
lar channels, although with reduced efficiency. More recently, higher than that of controls. The other six exhibited normal
Oshima et al. (23) reported that assembly of M34T in HeLa audiological features and their hearing thresholds were well
and Sf9 cells resembles that of wild-type human Cx26 in line with those of age- and sex-matched controls. A flat
(HCx26wt) and that dye transfer in these cells is close to audiogram was recorded in four subjects, whereas high-
normal. Finally, electrophysiological studies performed in frequency sloping and low-frequency ascending curves were
paired Xenopus oocytes concluded that M34T was capable observed in the other two (Fig. 2).
of forming with Cx32 functional heterotypic channels with In group II, three subjects (all heterozygotes) had mild
abnormal gating properties, which allowed to suggest that hearing loss (delGJB6-D13S1830/wt, L90P/wt, 35delG/wt).
the channels are not fully open at rest but are activated In two of them, males having 64 and 67 years (family 2,
when positive transjunctional voltages are applied to the subject I.2 and family 3, subject I.2; Fig. 1), hearing impair-
M34T side (24). In addition, co-injection of M34T and ment was similar to that of the appropriate control group,
HCx26wt in oocytes supported a recessive role for the whereas in the third one (delGJB6-D13S1830/wt), hearing
M34T allele, as a 1:1 ratio of mutant to wild-type RNA threshold was higher than expected across all frequencies.
Human Molecular Genetics, 2006, Vol. 15, No. 17 2571

Figure 1. Pedigrees and genotypes of the seven families studied. Individuals with hearing loss are shown in black, whereas those with normal audiological
parameters are in white. Gray symbols indicate individuals in which at least one frequency threshold was impaired; asterisks denote individuals with a
degree of hearing loss that matches the one expected by age and sex.

Finally, the two remaining subjects (35delG/wt) exhibited subject II.1). In all M34T/wt hearing impaired patients,
normal hearing. Flat audiograms were found in two patients, thresholds across all frequencies were greater than that
whereas the other three were of the high-frequency steeply expected for an age- and sex-matched control population
sloping type. In one of the patients with a mild phenotype (Fig. 2). Even among normal hearing [PTA ,20 dB hearing
(delGJB6-D13S1830/wt; family 1, subject I.1), hearing loss level (HL)] M34T heterozygotes (six cases), thresholds were
was asymmetrical. greater than that in control subjects for at least one frequency
In group III, five M34T/wt and five compound heterozy- in three patients (family 3, subject I.2; family 6, subject I.2;
gotes were hearing impaired. Mild hearing loss was diagnosed family 7, subject I.1). Five patients had flat audiograms and
in four patients (one M34T/wt, one M34T/35delG, one M34T/ 10 showed high-frequency sloping curves (six cases gently
delGJB6-D13S1830 and one M34T/L90P). Moderate hearing sloping and four steeply sloping). A low-frequency ascending
loss was present in five patients (four M34T/wt genotypes audiogram was observed in only one patient (family 5, subject
and one M34T/35delG), whereas severe hearing loss, in the II.1). Among the 16 patients carrying the M34T mutation, six
only hearing ear, was observed in one patient with an (five of which were compound heterozygotes) had asymmetri-
M34T/delGJB6-D13S1830 genotype (Fig. 1; family 1, cal hearing loss.
2572 Human Molecular Genetics, 2006, Vol. 15, No. 17

Figure 2. Audiometric analysis of the seven families studied. Air conduction thresholds were measured across the indicated frequencies, and the degree of
hearing impairment was classified by the pure-tone average applied to the better ear. Among M34T-carrying patients, five had flat audiograms, all of which
showed the M34T/wt genotype, (families 2, 4, 6) and 10 showed high-frequency sloping curves (six gently sloping: family 1, M34T/delGJB6-D13S1830 geno-
type with the poorer threshold; family 2, M34T/L90P genotype; family 3, M34T/wt; families 6 and 7, with one and two members showing M34T/wt genotype,
respectively; four steeply sloping: family 1, the other subject with M34T/delGJB6-D13S1830 genotype and the subject M34T/wt; families 3 and 4, males with
the M34T/35delG genotype). For each individual, audiograms of the better ear are shown.

Protein expression and localization (Fig. 3A). We chose these two variants because previous
studies indicated that both mutated proteins form plaques in
To analyze the functional impact of the M34T mutation, which the junctional membrane (20,22,26). To examine to which
is positioned in the first transmembrane domain (TM1) of Cx26, extent these mutations affected the level of protein expression
we have compared its channel-forming ability with that of the and their cellular localization, HCx26wt and these two variants
recessive R127H (25), which maps to the cytoplasmic loop were transiently transfected in HeLa cells, which were
Human Molecular Genetics, 2006, Vol. 15, No. 17 2573

protein (GFP) positive cells, a dashed, membrane-associated


staining typical of gap-junctional plaques, indicating that it
was correctly targeted to the plasma membrane (Fig. 3C),
whereas in cells transfected with vector alone, no staining
was detected (20). As previously shown (20,22,23), the M34T
mutant exhibited not only a strong staining at the plasma mem-
brane, but also the presence of cytoplasmic fluorescence, which
was suggestive of partial intracellular retention (Fig. 3C). The
immunolocalization of the R127H mutation gave results
similar to HCx26wt, with junctional plaques present at zones
of cell-to-cell apposition (Fig. 3C).

Biophysical properties
To compare the electrophysiological behavior of HCx26wt
and the M34T mutant, experiments were initially carried out
on cultures in which the two cDNAs were expressed through
the pIRES construct (see Materials and Methods). Cell pairs
expressing HCx26wt were held at a transjunctional voltage
(Vj) of 295 mV, under uncoupling conditions in a medium
saturated with 100% CO2 (27). Once junctional current (Ij)
was reduced to below-detectable levels, the superfusion
medium was switched back to normal extracellular solution
(ECS) to allow Ij to recover, and a series of discrete events
of similar amplitude, reflecting the opening of junctional chan-
nels, was observed (Fig. 4A). Off-line analysis yielded a
unitary conductance (g) of 114 pS for the fully open state.
In contrast, when the M34T mutant was challenged under
the same uncoupling conditions with a sustained Vj of
2100 mV, gap junction channels tended to open more
rapidly, which hampered the detection of discrete events.
However, in some cases, small current transitions were suffi-
ciently well resolved to define them as unitary gating events
of 13 pS in magnitude (Fig. 4B). Therefore, single-channel
Figure 3. Topology, expression and localization of HCx26wt and of the two
conductance of M34T is substantially (9-fold) smaller than
variants, M34T and R127H. (A) Schematic topology of Cx26 relative to the that of HCx26wt (Fig. 4A and B). Although such small
plasma membrane, with the approximate distribution of the mutations signals were at the limit of resolution for our dual whole-
studied. (B) Western blot analysis of the expression of wild-type and cell recording conditions, this analysis indicates that the
mutated Cx26 proteins in HeLa cells. Cells were transfected with the specified M34T variant retained partial activity and that functional
constructs, lysed in SDS sample buffer 24 h post-transfection and further pro-
cessed for immunoblotting, as described in the Materials and Methods section. homotypic/homomeric channels were assembled.
A representative blot shows that the levels of expression of the mutated pro- Interactions between connexins may lead to the formation
teins were comparable with those of HCx26wt. (C) Immunolocalization of of heterotypic channels assembled by docking of two homo-
HCx26wt and deafness-associated mutations. HeLa cells were transfected meric hemichannels formed from different connexins (1). To
with the specified constructs, fixed and labeled with an anti-Cx26 antibody
24 h post-transfection, as described in the Materials and Methods section. A
study the behavior of heterotypic channels, we used cells
secondary antibody conjugated with tetramethylrhodamine isothiocyanate expressing chimeric proteins in which color variants of GFP
was used for visualization. GFP: Expression of GFP in transfected HeLa were fused at the C-terminus of the specified constructs.
cells shows a diffuse cytoplasmic staining. Cx26: Immunolocalization of When HeLa cell pairs transfected with HCx26wt-Venus/L1
Cx26 in GFP-expressing cells shows a characteristic punctate staining, but recovered from CO2-induced uncoupling with a sustained Vj
also the presence of cytoplasmic fluorescence in the case of M34T.
Bar ¼ 10 mm. of 295 mV, unitary gating events indicative of the opening
of single-junctional channels yielded a g of 115 pS for the full-
open state (Fig. 5A). This initial characterization indicates that
the electrophysiological behavior of the tagged protein was
processed for western blot and immunofluorescence analysis virtually indistinguishable from that of HCx26wt.
24 h post-transfection. Western blots performed on total cellu- Because Cx26 and Cx30 are co-localized in gap junctions of
lar lysates demonstrated that proteins of the predicted molecular the inner ear (28,29), we next determined the biophysical
mass were detected in each experimental condition (Fig. 3B). properties of HCx30wt homotypic and HCx26wt/HCx30wt
Levels of R127H appeared comparable with those of wild-type heterotypic channels. Pairs of HeLa cells transiently trans-
protein, whereas M34T showed slightly increased expression, fected with HCx30wt-CFP/L1 displayed single-channel open-
thus confirming our previous quantitative analysis (20). ings when subjected to a Vj of 90 mV (Fig. 5B). Transitions
Transfection with HCx26wt produced, in green fluorescent occurred among the closed state, the main conductance state
2574 Human Molecular Genetics, 2006, Vol. 15, No. 17

Figure 4. Electrical properties of HCx26wt and M34T mutant. (A) Conduc-


tance changes (gj, top trace) and corresponding discrete current transitions
(Ij, bottom trace) due to gating of homotypic channels formed by HCx26wt.
Several opening and closing of gap junction channels were recorded with
a transjunctional voltage (Vj) of 295 mV (V1 ¼ 25 mV; V2 ¼ 2100 mV;
n ¼ 4). The calculated unitary conductance (g) was 114 pS. (B) Conductance
changes (gj, top trace) and corresponding rapid current transitions (Ij, bottom
trace) due to opening of homotypic channels formed by M34T mutant
recorded under a sustained Vj of 2100 mV (V1 ¼ 0 mV; V2 ¼ 2100 mV;
n ¼ 2). The estimated g was 13 pS.

with gmain ¼ 160 pS and a sub-conductance state with


gresidual ¼ 27 pS. Similar results were obtained when unitary
currents (middle trace) were elicited by applying voltage
ramps from þ95 to 2100 mV (bottom trace). Ij (top trace)
was essentially linear with Vj, as expected for a normal homo-
typic channel, with gmain ¼ 160 pS, as well as gresidual ¼ 27 pS
(Fig. 5C). These observations are consistent with the findings Figure 5. Electrical properties of tagged HCx26wt and HCx30wt. (A) Con-
ductance changes (gj, top trace) and corresponding discrete current transitions
reported for mouse Cx30 by Valiunas et al. (30). (Ij, bottom trace) due to gating of homotypic channels formed by HCx26wt–
Mouse Cx26 and Cx30 have been shown to assemble Venus/L1 (Cx26, n ¼ 8). Ij was recorded with a transjunctional voltage (Vj)
heterotypic channels in functional expression systems (31). of 295 mV (V1 ¼ 25 mV; V2 ¼ 2100 mV). (B) Conductance changes
To study the behavior of HCx26wt/HCx30wt heterotypic (gj, top trace) and corresponding discrete transitions of junctional current
(Ij, bottom trace) due to gating of a single homotypic channel formed by
channels, we used co-culture of cells expressing HCx26wt- HCx30wt-CFP/L1 (Cx30, n ¼ 4). Vj (bottom trace) was initially set at
Venus/L1 and HCx30wt-CFP/L1. Pairs of cells that displayed 290 mV and stepped to zero by the end of the recording period, confirming
at least one bi-color fluorescent plaque at the appositional the complete closure of the channel. Conductance of the residual state, gresidual
membrane were examined for functional coupling and was 27 pS (tilted arrow); conductance of the main state, gmain was 160 pS.
voltage gating. Junctional currents (Ij) were recorded from (C) Shown are two Vj ramps from þ95 to 2100 mV, 2 s in duration (Vj,
bottom trace), applied to a cell pair expressing homotypic HCx30wt-CFP/L1
the cell expressing HCx26wt-Venus/L1 while alternate nega- channels (Cx30, n ¼ 3). The Ij /Vj relationship is linear. Estimated unitary con-
tive and positive voltage steps (Vj) were applied to the cell ductance (gj, upper trace) shows both main and residual (tilted arrow) states, as
expressing HCx30wt-CFP/L1. We assume that at both the in (B).
Human Molecular Genetics, 2006, Vol. 15, No. 17 2575

beginning and the end of the negative Vj step, two channels dependence of the open state with a main conductance of
were open and that, during the pulse, one channel exhibited 120 pS and no residual state (Fig. 7A). Results obtained
opening and closing transitions with a calculated unitary con- by delivering voltage commands in the range from 2100 to
ductance (gmain) of 82 pS (Fig. 6A). Ij recordings during a þ100 mV for 10 s (not shown) were consistent with those
positive Vj step showed two closing events from open to generated with ramp protocols. As expected on the basis of
residual states of 130 pS in magnitude and one closing event the properties of HCx26wt/HCx30wt heterotypic junctions,
of 30 pS in magnitude (gresidual), presumably from the sub- HCx30wt/V84L channels exhibited rectification and single-
conductance to the closed state (Fig. 6A). The different channel conductance increased from 70 to 150 pS when
values of gmain at positive and negative Vjs, indicative of rec- voltage ramps from 2100 to þ100 mV were delivered to
tification, were also measured by delivering voltage ramps the cell expressing V84L while recording Ij from the cell
from þ95 to 295 mV (Fig. 6B). In both experimental proto- expressing HCx30wt (Fig. 7B). Thus, in contrast to what
cols, single-channel conductance was smaller when the observed with M34T, the electrical characteristics of
HCx26wt side was made relatively positive and vice versa. HCx30wt/V84L channels did not differ appreciably from
The fitting curve (solid line of the gj – Vj plot in Fig. 6B) those of HCx26wt/HCx30wt channels.
showed that the conductance of the main state at a
Vj ¼ 0 mV was 110 pS. This value is reasonably close to the
expected gmain value of 130 pS [geq ¼ (1/g26 þ 1/g30)]21 cal-
culated from the arrangement in series of one homotypic Intercellular dye transfer
HCx26wt connexon (g26 ¼ 230 pS) with one homotypic To assess the permeability of mutated gap junction channels,
HCx30wt connexon (g30 ¼ 320 pS), thus supporting the we examined the cell-to-cell transfer of LY (MWionic
notion that these heterotypic channels retain the properties of form ¼ 443; charge 22) by loading one cell of a pair with a
composing hemichannels. patch pipette. We did not detect LY diffusion across M34T
To obtain heterotypic channels containing one mutated con- channels despite the presence of sizeable junctional conduc-
nexon, HeLa cells were transiently transfected with M34T, tance (gj ¼ 4.5 + 1.3 nS, n ¼ 5), measured by dual whole-cell
expressed through the pIRES vector, HCx26wt-Venus/L1 or patch-clamp (Fig. 8). It should be noted that this level of gj
HCx30wt-Venus/L1 (Fig. 6C and D). Separate cultures were corresponds to approximately 450 open channels (4.5/
eventually mixed and pairs for dual whole-cell patch clamp 0.013 nS) at any given time and that robust LY transfer is con-
analysis were identified by the simultaneous presence of dif- sistently detected with a similar number of functional
fused GFP fluorescence in the cytosol of one cell, indicating HCx26wt homotypic channels (34). LY diffusion was then
positive M34T transfectants, and punctate Venus fluorescence assayed in cultures of parental and transiently transfected
on the other side of the junction, reflecting expression of HeLa cells expressing either the wild-type or the mutated con-
tagged forms of either HCx26wt or HCx30wt. Our data nexins (Table 1). Dye coupling was scored as positive if the
show that M34T hemichannels were able to dock with either probe diffused to at least two neighboring cells. As expected,
HCx26wt or HCx30wt, leading to the formation of functional non-transfected HeLa cells did not display intercellular dye
heterotypic gap junction channels. In both cases Ij recordings diffusion (not shown). Microinjection of LY into an individual
obtained by applying voltage ramps showed a rectification that cell of an HCx26wt-positive cluster resulted in intercellular
was clearly more pronounced for the M34T/HCx26wt combi- coupling in all instances with 100% of GFP-positive cells
nation (Fig. 6C) than for M34T/HCx30wt (Fig. 6D). filled with the probe. In contrast, no dye transfer was observed
Steady-state Gj – Vj dependence was analyzed by applying pro- between cells expressing the M34T mutation, whereas R127H
longed voltage steps (see Materials and Methods), and pooled showed some incidence of dye transfer, although drastically
data were obtained by normalizing the steady-state conduc- reduced in comparison with HCx26wt, with only one or two
tance, measured just prior to command offset, to the conduc- cells of the R127H cluster receiving LY (Table 1).
tance values at command onset (Fig. 6E). These data show The original report of the heterozygous M34T mutation in a
that M34T homotypic junctions were relatively insensitive to family with dominant deafness (13) suggested that the mutant
Vj, similarly to HCx26wt channels (32), a feature maintained allele would act as a dominant-negative subunit of connexin
in both heterotypic configurations for relative positivity of channels, and functional expression studies performed on
the cytoplasmic side of the M34T connexon (Fig. 6E). Thus, pairs of Xenopus oocytes (14) initially supported this view.
although M34T channels were endowed with a residual func- To determine whether this mutation could interfere with the
tionality, their properties were significantly altered in both function of wild-type channels, we examined dye coupling
homotypic and heterotypic configurations. in HeLa cells co-transfected with the same amount of
As a control, we examined heterotypic junctions formed by HCx26wt and M34T plasmids and found that LY transfer
pairing HCx26wt or HCx30wt with V84L, a deafness-related was abolished under these experimental conditions
recessive mutant of Cx26 that retains the ability to sustain (Table 1). In contrast, the R127H mutation, segregating with
electrical communication (32,33). Homotypic V84L channels autosomal recessive deafness (25), failed to affect the ability
exhibited a unitary conductance of 110 pS, very close to the of HCx26wt to form functional channels when the two
value measured for HCx26wt channels (33). Heterotypic constructs were co-transfected at an equal ratio (Table 1).
combinations (HCx26wt/V84L and HCx30wt/V84L) were These results demonstrate that the M34T mutant, but not the
obtained by mixing transiently transfected HeLa cells, as recessive R127H mutant, significantly inhibited the functional
described previously. In contrast to what observed with activity of HCx26wt when expressed in a manner mimicking
M34T, HCx26wt/V84L channels showed a linear Ij – Vj the heterozygous genotype.
2576 Human Molecular Genetics, 2006, Vol. 15, No. 17

Figure 6. Gating behavior of heterotypic channels. (A) A transjunctional voltage (Vj ¼ VCx30 2 VCx26, bottom trace) was imposed by stepping the HCx30wt
(Cx30)-expressing cell, and junctional current (Ij, middle trace) was recorded from the HCx26wt cell (Cx26, n ¼ 4). Top trace: junctional conductance trace
(gj). (B) Junctional current (Ij, top left) was recorded from the Cx30 cell during five consecutive Vj (VCx26 2 VCx30) ramps from þ95 to 295 mV, 3 s in duration
(bottom left, n ¼ 4). The right-hand panel shows rectification in the junctional conductance gj ¼ Ij /Vj, derived from the ramp responses and plotted as a function
of Vj. (C) Junctional current (Ij, middle trace) was recorded from the cell expressing Cx26 in response to four consecutive Vj ramps (VM34T 2 VCx26, bottom trace)
from þ100 to 2100 mV, 3 s in duration (n ¼ 4). The calculated junctional conductance (gj, upper trace) of heterotypic M34T/HCx26wt channels shows pro-
nounced rectification (n ¼ 4). (D) Same protocol as in (C) shows milder rectification for heterotypic M34T/HCx30wt channels (Vj ¼ VM34T 2 VCx30). Ij (middle
trace) was recorded from the cell expressing Cx30 (n ¼ 4). (E) Plot of the relationship of Vj to steady-state junctional conductance (Gjss, calculated as the ratio of
steady-state to instantaneous junctional conductance measured at the offset and onset of voltage steps, respectively) obtained in cell pairs expressing homotypic
M34T channels (filled circles), or heterotypic M34T/HCx26wt (open circles) and M34T/HCx30wt (filled squares) combinations (pooled data from five indepen-
dent experiments). In heterotypic channels, Vj is defined as positive for depolarization and negative for hyperpolarization of the side expressing the wild-type
connexins cell relative to the M34T cell. Lines through data are least square fits with quadratic functions to aid data trend visualization. Heterotypic channels
display asymmetric Gj 2 Vj relationships because M34T is poorly voltage dependent, whereas HCx26wt and HCx30wt close at positive voltages.
Human Molecular Genetics, 2006, Vol. 15, No. 17 2577

Figure 8. Permeability to LY of M34T mutant channels. Top left: bright field


image of one representative pair of HeLa cells with superimposed regions of
interest (ROIs) used to compute average pixel fluorescence. Top right: image
of the same field illuminated at lex ¼ 480 nm to visualize EGFP positive cells
(lem ¼ 535 nm). Bottom left: fluorescence image obtained at the end of the
experiment by exciting the field at lex ¼ 425 nm to quantify LY spread
(lem ¼ 535 nm). Cell 1 was contacted by a patch pipette containing 4% LY.
Intracellular delivery of LY started 20 s after the onset of the recording.
Bottom right: normalized fluorescence from the corresponding ROIs (traces
numbered 1 and 2) measured concurrently with junctional conductance (gj,
lowermost trace). The arrowhead marks the time at which the LY fluorescence
image was acquired. Results are representative of five independent exper-
iments. Scale bar: 10 mm.

Table 1. Qualitative comparison of dye diffusion (LY; MW ¼ 457 Da;


charge 22) between HeLa cells transfected with either HCx26wt or the
specified mutations

Transfection Percentage of positive injections n

HCx26wt 100 135


M34T 0 67
R127H 14 95
Figure 7. Gating behavior of heterotypic channels containing the V84L
HCx26wt þ M34T 0 102
mutation. (A) Conductance changes (gj, top trace) and corresponding discrete
HCx26wt þ R127H 100 47
current transitions (Ij, middle trace) due to channel opening during transjunc-
tional voltage ramps (Vj ¼ VV84L 2 VCx26, bottom trace) from þ100 to 2100 mV
(n ¼ 3) (B) Two junctional voltage ramps (Vj ¼ VV84L 2 VCx30, bottom trace) The incidence of cell coupling (second column) was scored by recording
from þ100 to 2100 mV, 3 s in duration, are shown (n ¼ 3). In both panels, the number of instances in which the dye diffused from the injected cell to
junctional current (Ij, middle trace) shows an ohmic behavior (viz. a linear at least two neighboring ones. In the third column, the total numbers of
dependence) on Vj. injections (n) is indicated.

Intercellular calcium waves largely confined to the stimulated cell, and in the few instances
in which the signal propagated to neighboring cells, it reached
In many cell types, connexins are involved in the radial propa- only one cell of the first order (not shown). As expected, in
gation of intercellular calcium waves induced by mechanical monolayers expressing HCx26wt, the [Ca2þ]i response of a
stimulation of a single cell in a monolayer (reviewed in 35). mechanically stimulated cell was consistently followed by
We performed calcium-imaging experiments to determine the intercellular spreading of a calcium wave (Fig. 9A;
whether the M34T variant could interfere with the intercellular n ¼ 33) that involved all first-order cells (91% of the obser-
diffusion of second messengers such as calcium ions and ino- vations), and in 21% of the cases, cells of the second order.
sitol 1,4,5-trisphosphate (InsP3). To minimize the contribution The propagation of calcium waves was completely inhibited
of ATP release to the propagation of calcium signals (36), in by the gap junction blocker carbenoxolone (CBX, 100 mM )
all experiments, the extracellular medium was supplemented in all experiments (Fig. 9B; n ¼ 13). In M34T-expressing
with 40 U/ml apyrase grade VII. cells, calcium spreading was largely blocked, being observed
In control cells transfected with the empty vector, mechan- only in 27% of the cases (Fig. 9C; n ¼ 37), with only one
ical stimulation induced a [Ca2þ]i increase that remained cell of the first order being recruited. Similar results (blockade
2578 Human Molecular Genetics, 2006, Vol. 15, No. 17

Figure 9. Mechanically-induced intercellular calcium waves in transfected HeLa cells. HeLa cells expressing the specified constructs were analyzed by calcium
imaging 24 h post-transfection. Monolayers loaded with fura-2/AM were subjected to mechanical stimulation of single cells (cell 1 in all panels) by briefly
deforming the cell surface with a micropipette, as described in the Materials and Methods section. Apyrase (40 U/ml) was added in all experiments. For all
conditions, intercellular calcium waves shown are representative of at least 12 experiments from four independent transfections. (A) HCx26wt transfected
cells (n ¼ 32). (B) HCx26wt transfectants incubated in the presence of the gap junction blocker CBX (100 mM , n ¼ 18). (C) M34T transfected cells
(n ¼ 45). (D) HCx26 wt þ M34T co-transfected cells (n ¼ 12). In all panels, insets show the clusters of EGFP-positive cells included in the experiment with
the cell numbers corresponding to the individual fluorescence traces (see color-coding below the abscissa). Scale bar: 10 mm.

in 70% of the experiments) were also obtained when cells entirely excluded, and on the other hand, the recruitment of
were co-transfected with a 1:1 ratio of HCx26wt and M34T stretch-activated channels and downstream pathways could
plasmids (Fig. 9D; n ¼ 12). also account for the occasional calcium response of
The evidence that in a minority of cases a calcium wave was the cell(s) tightly connected to the stimulated one. To
observed in non-transfected and in M34T-expressing cells was exclude an involvement of connexin-unrelated mechanisms
not entirely surprising, given the complexity of the molecular in the propagation of calcium waves, we refined our investi-
mechanisms governing this phenomenon. On the one hand, the gation by restricting the analysis to neighboring cells involved
contribution of paracrine signals other than ATP could not be in the wave. In M34T transfectants, the [Ca2þ]i elevation in
Human Molecular Genetics, 2006, Vol. 15, No. 17 2579

the cell(s) of the first order relative to the stimulated one


(D[Ca2þ]Io/D[Ca2þ]s) decreased slightly with respect to that
of HCx26wt (Fig. 10A). In addition, the rate of the rising
phase in the first-order cells (D[Ca2þ]Io/Dt) was reduced to
less than 50% compared with that of HCx26wt pairs and
similar results were obtained in control cells transfected with
the empty vector (Fig. 10A). The delay in the calcium
response of the first-order cell(s), measured as the time
required to reach the half maximal change (EC50) in
fluorescence ratio with respect to that of the stimulated cell
(tIo 2 ts) (37), was also altered, being significantly reduced
in M34T transfectants with respect to that observed in
HCx26wt-expressing cells (Fig. 10B). Finally, to complete
this quantitative analysis, we derived a coupling index
among different transfectants by multiplying the percentage
of successful stimulations (those inducing an intercellular
Ca2þ wave) by the number of cells displaying an increase of
cytosolic Ca2þ. The value calculated for M34T-expressing
cells was similar to that of controls and five times lower
than that of HCx26wt transfected cells (Fig. 10C).

Structural model of the M34T mutant


These genetic and functional data indicate that the M34
residue in Cx26 is critical for channel function. Fleishman
et al. (38) have recently published a structural model of the
transmembrane (TM) region of Cx32 (PDB code 1TXH) on
the basis of a combination of theoretical methods and low-
resolution electron cryo-microscopy map of Cx43 (39).
Given the high conservation of the TM helices among the con-
nexin family, these data are expected to serve as a general
template for the TM regions of other connexins. In this
model, M34 mediates putative contacts between two consecu- Figure 10. Quantitative analysis of critical parameters of calcium wave propa-
tive connexins (38). However, because only the positions of gation. HeLa cells expressing the specified constructs were analyzed by
calcium imaging in response to mechanical stimulation, as described in the
the Ca atoms were determined, the evaluation of the possible Materials and Methods section. Apyrase (40 U/ml) was added in all exper-
consequences of point mutations is not immediate. To gain iments. Changes in intracellular Ca2þ concentration were estimated by calcu-
further structural insights into the consequence of the M34T lating the excitation ratio of fura-2 fluorescence at 340 and 380 nm (see
mutation on channel structure, we undertook the theoretical Materials and Methods). (A) D[Ca2þ]Io/D[Ca2þ]s describes the calcium con-
centration change in cells of the first order relative to the stimulated cell.
construction of all-atoms models of HCx26wt and M34T D[Ca2þ]Io/Dt calculates the rise rate of the calcium signal in cells of the
using the Ca structure of Cx32 as a reference template. first order relative to the stimulated cell. (B) tIo 2 ts indicates the delay in
In this atomic model, the side chains of Tyr152 within TM3, the calcium response of first order cells, measured as the time required to
which is considered to be the main pore-lining helix (2), deter- reach the half maximal (EC50) fluorescence ratio change with respect to that
mine the maximum constriction region of the pore of HCx26wt of the stimulated cell. (C) The coupling index of different transfectants is
derived by multiplying the percentage of positive experiments (i.e. the exper-
channels near the extracellular side (Fig. 11A). M34 does not iments in which intercellular calcium spreading was detected) by the number
face the pore lumen, but is partially exposed to the lipid face, of coupled cells. Results are shown as means + SD of the number of
where it also establishes hydrophobic contacts with residues experiments indicated in the legend of Figure 9. Statistical significance was
of the adjacent connexin chain (Fig. 11B, inset). assessed by the unpaired Student’s t-test.  P , 0.05;  P ¼ 0.05; ns, not
significant.
Both HCx26wt and M34T models showed alike root mean
square deviation (RMSD) behaviors, reaching a stabilization
plateau after nearly 250 ps, thereafter oscillating around a
value of 0.3 nm (Fig. 11C). The average RMSD of each con- at the extracellular side, where the backbone extremities of
nexin was 0.15 nm, suggesting that, although the helical the TM3 helices remained 0.2 nm closer in M34T
conformation was fairly well maintained, the relative orien- (Fig. 11D). Thus, the inter-helical angle between opposite
tation of the chains suffered significant changes upon relax- TM3 helices of HCx26wt decreased to almost 50 degrees,
ation. Comparison of the average distances between the whereas in the M34T mutant, it increased up to 65
backbone extremities of two opposite TM3 helices at the intra- degrees (Fig. 11E). This implies a sort of angular bending
cellular sides of both variants indicated that HCx26wt suffered between the principal angles of the helices (Fig. 11F)
a decrease of 0.2 nm, whereas M34T showed an opposite that leads, as a consequence of this movement, to a higher
behavior increasing the internal diameter by 0.1 nm constriction of the pore in the M34T mutant. Hence,
(Fig. 11D). A similarly opposite behavior was also retrieved the average distance between couples of OH atoms in
2580 Human Molecular Genetics, 2006, Vol. 15, No. 17

Figure 11. Structural models of HCx26 wt and M34T channels. (A) Atomic model of HCx26wt as seen from the intracellular side after full EM. The different
colors of the ribbons depict the six subunits forming the connexon. The Y152 residue corresponding to the minimum diameter of the pore lumen is represented in
space-filling spheres. Hydrogen atoms are not shown for visual clarity. (B) Lateral view of the transmembrane part of the HCx26wt connexon obtained rotating
the structure in (A) by 90 degrees. The position of M34 is indicated with its corresponding space-filling representation. Inset: a representative close-up of the
M34 neighborhood. Residues within 0.5 nm are shown in sticks. M34 may establish hydrophobic interactions with the lipid phase and also with residues belong-
ing to the second transmembrane helix (TM2) from the nearby connexin. (C) Root mean square deviation (RMSD) calculated over the Ca atoms plotted versus
simulated time for Cx26 wild-type (blue) and M34T (red) connexons. (D) Top panel: averaged distance between the center of the mass of the last four Ca atoms
of couples of opposite TM3 helices (see Materials and Methods). Middle panel: averaged distance between the center of the mass of the first four Ca atoms
of couples of opposite TM3 helices. Bottom panel: average distance between OH atoms belonging to opposite Y152 for HCx26wt (dark blue) and M34T
(red). (E) Trajectory of the inter-helical angle between opposite TM3 helices within the connexon. (F) The central scheme illustrates the conformational
change produced by the M34T substitution (red cylinders) with respect to wild-type (blue cylinders).

opposite Tyr152 residues remained nearly within its initial deafness. This conclusion is supported by comprehensive bio-
value of 2 nm for HCx26wt, whereas a more pronounced physical and functional studies showing that, although M34T
reduction to 1.7 nm was observed for the M34T mutant retains partial functionality, the mutant channels are defective
(Fig. 11D). with respect to wild-type in both electrical and metabolic
couplings.

DISCUSSION Genotype – phenotype correlation


Our data demonstrate that the presence of the M34T variant Although M34T has been described both as an autosomal
of HCx26 is indeed associated with mild-to-severe forms of dominant (13) and recessive mutation (15,40 – 42), this
Human Molecular Genetics, 2006, Vol. 15, No. 17 2581

interpretation has remained controversial due to the frequent pedigrees of our families suggest that environmental factors
findings of normal hearing in individuals heterozygous for alone cannot explain the hearing deficits of M34T heterozy-
the M34T variant (16 – 19) and even in compound heterozy- gotes (see, for example, family 7 in Fig. 1).
gotes with 35delG (15). Thus, it has been suggested that the
pathological effect of the M34T allele might depend on the
mutations segregating in the opposing allele, such as
167delT (43,44) or 35delG (45). However, in a recent multi- Impact of M34T on channel function
center study, all patients with a 35delG/M34T genotype By combining electrophysiology, dye transfer and dynamic
were found to have mild-to-moderate hearing impairment calcium imaging, our data demonstrate that M34T mutant
(46). These discrepancies may reflect a reduced penetrance channels, although correctly synthesized and targeted to the
of M34T or be the consequence of a diagnostic bias toward plasma membrane, are functionally defective. This conclusion
more severe forms of hearing impairment, as persons with is based on several lines of evidence. First, unitary
mild deficits are less likely to undergo audiological or conductance of M34T was 10-fold lower than that of
genetic testing (46). HCx26wt gap junction channels. Secondly, heterotypic
The audiometric features of the seven families reported here HCx26wt/M34T channels exhibited a strong Ij –Vj rectification
revealed that hearing loss was present in 14 subjects. Consid- that can have functional implications for communicating cells
ering heterozygous subjects carrying the M34T mutation, 10/ residing at different transmembrane potentials. Thirdly,
16 (62.5%) were mildly to severely impaired with respect to intercellular transfer of LY was prevented, and finally,
the audiometric parameters of age- and sex-matched controls. M34T channels failed to sustain the propagation of inter-
Of note, none of the five M34T compound heterozygotes dis- cellular Ca2þ waves. These results demonstrate that the
played normal hearing, being classified as mildly to severely mutation alters significantly the molecular cut-off size of the
impaired. The most compromised frequencies were in the pore and therefore should be considered as a pathological
range of 2000 – 8000 Hz, which resulted in an audiogram con- variant of Cx26.
figuration of the sloping type. This observation may account In addition, we found that M34T exerted a dominant-
for the discrepancy with many previous studies in which the negative inhibition of metabolic coupling when expressed
degree of hearing impairment was assessed by pure tone in a manner mimicking a heterozygous genotype, an effect
average of only three frequencies (500, 1000, 2000 Hz). A not displayed by the recessive R127H mutation. Our data
clinical re-assessment of those subjects with a wider range support the conclusion reached by previous functional
of frequencies may help uncover mild cases of hearing loss studies performed in paired Xenopus oocytes expressing
associated with M34T mutation. In fact, the only other M34T (14). Although this dominant-negative effect was
report that examined air conduction thresholds across the later attributed to an artifact because of the lack of
same range of frequencies, as in this study, found that the control of the expression levels of mutant and wild-type
two subjects with M34T/wt genotype displayed RNA in the exogenous system (40), a dominant
mild-to-moderate hearing loss (47). A moderate hearing inhibition of channel function has been confirmed in a
loss affected the heterozygous M34T child (48); in this recent study conducted on Cx26 hemichannels in single
study, the authors included only deaf children with hearing oocytes (52).
thresholds .56 dB HL. Together, these observations These observations are difficult to reconcile with a recent
indicate a pathogenetic role of the M34T mutation and are study demonstrating that M34T, although not capable of
consistent with most recent surveys associating M34T com- forming functional gap junction channels on its own in
pound heterozygosis to mild-to-moderate hearing loss Xenopus oocytes, did not exhibit a dominant-negative effect
(15,46,47,49,50). on cell – cell coupling when co-expressed at a 1:1 ratio with
The finding that 60% of patients carrying only the M34T HCx26wt (24). Such a discrepancy cannot be explained
mutation were also hearing impaired, thus suggesting the easily. It should be noted, however, that White et al. (14)
involvement of this variant even in dominant forms of deaf- and we have tested the dominant-negative action of M34T
ness, is not easily explained. It has been noted, however, in cells that were all programmed to express this variant
that M34T segregates with a 10 bp deletion in the together with HCx26wt, whereas Skerrett et al. (24) have
50 -untranslated regions of the Cx26 gene (15). It is not paired oocytes co-injected with HCx26wt and M34T in hetero-
known whether this deletion is of any significance, but its typic fashion, with oocytes expressing only the wild-type
association with M34T may account for the variability in the protein. Thus, these studies are not directly comparable. An
phenotype of M34T heterozygous, as it could alter the additional difference in the observed functional properties of
expression level of the M34T allele in vivo. Of note, this del- M34T may stem from the relative expression levels of the
etion was present, in cis, in 14 out of the 16 M34T subjects, wild-type and mutant proteins, a parameter that cannot be
whereas it was absent in all other individuals enrolled in the fully controlled in cells co-expressing the two HCx26
study. Hence, this study and functional studies (14) suggest sequences, as there are no antibodies that can distinguish
that M34T could be regarded as a dominant allele, but its between them. As found from this study and from the work
dominant-negative activity may be affected by modifying of D’Andrea et al. (20), although the amount of M34T
factors such as the level of RNA expression, as recently expression was found to be comparable with that of HCx26wt
suggested (51), thus explaining the observed phenotypic in either single transfectants or in oocytes injected with the
variability. Although a population-based study may also cognate RNA (14), it is possible that changes in the protein
give information on the penetrance of the M34T allele, the ratios may also account for the differences between
2582 Human Molecular Genetics, 2006, Vol. 15, No. 17

these three studies. Indeed, increasing the ratio of mutated to pathway by the M34T mutation may interfere with re-cycling
wild-type RNA injection (M34T:HCx26wt ¼ 2:1) led to a of Kþ ions back into the endolymph, leading to accumulation
considerable decrease of intercellular coupling (24). of Kþ ions and excitotoxic death of hair and supporting
Differences in protein levels are an inherent drawback of in cells in vivo, which would eventually result in hearing
vitro functional expression systems. Thus, the residual impairment (62). Our data are consistent with a
permeability of the M34T variant to fluorescent tracers, as mechanistic explanation of the pathogenesis of deafness
previously reported (22,23), is likely to depend on the over- which postulates that InsP3 permeability between the epi-
expression of the low conductance channel, as suggested by thelial cell network of the cochlea is crucial for normal
the same authors. hearing (33).

A structural model of M34T channels MATERIALS AND METHODS


The stringency of the requirements of distinct connexin chan- Reagents
nels in different tissues strongly suggests that altering such
composition in vivo would result in the development of func- LY, apyrase grade VII, aprotinin, chymostatin and leupeptin
tional abnormalities (53). Indeed, targeted ablation of Cx26 in were obtained from Sigma (St Louis, MO). The polyclonal
the epithelial network of the organ of Corti has elegantly anti-rat Cx26 antibody employed was from Zymed Laboratories
illustrated its essential role for cochlear function and cell sur- (San Francisco, CA). For immunoblot detection, the secondary
vival (54). Our functional analysis demonstrates that a meth- antibody was a goat anti-rabbit IgG conjugated with alkaline
ionine residue at position 34 in Cx26 substantially influences phosphatase from Calbiochem (San Diego, CA). For immuno-
homotypic, as well as heterotypic and heteromeric channels fluorescence, the secondary antibody was a tetramethylrhoda-
formed by association with both HCx26wt and HCx30wt and, mine isothiocyanate (TRITC)-conjugated affinity purified
together with the genetic and clinical data, further suggests goat anti-rabbit IgG from Jackson ImmunoResearch
that the consequences of this single-amino acid substitution Laboratories (West Grove, PA). The chemiluminescence
cannot be compensated by other connexin co-expressed in the substrate (CSPD) and enhancer (Nitro-Block) for alkaline
cochlea. phosphatase were from Tropix (Bedford, MA). Restriction
M34 is located in the TM1 domain of connexins, which is enzymes were from New England Biolabs (Beverly, MA).
likely to be one of the pore-lining helices (38,55,56) and Nitrocellulose was from Schleicher & Schuell (Dassel,
interacts with other transmembrane helices, stabilizing the Germany). All other reagents were from Sigma, unless
TM1 structure and ensuring the open-channel state (23). otherwise specified.
Our molecular dynamic simulations demonstrate that the
M34T substitution would cause a reduction of the side
chain size at this position, leading to a partially closed DNA analysis
channel, consistent with decreased channel permeability DNA was extracted from peripheral blood according to stan-
and unitary conductance. Caution should be exerted, dard protocols after obtaining informed consent. The whole
however, in the interpretation of these models, which are Cx26 coding region was amplified by polymerase chain reac-
affected by several approximations whose consequences are tion (PCR) using the following forward (CX26ORF-F:
difficult to estimate because of the lack of input data at TGCTTACCCAGACTCAGAGAA) and reverse
atomic resolution (38). Nonetheless, the results of these (CX26ORF-R: GACTGAGCCTTGACAGCTGAG) primers.
simulations are in good agreement with our own, as well PCR reaction was performed in a total volume of 50 ml con-
as previously published experimental data, suggesting that taining 15 pmol of each primer, 100 mM dNTP, 5 ml 10 reac-
this model captures at least the gross determinants of the tion buffer (100 mM Tris pH 8.3, 500 mM KCl, 15 mM MgCl2,
M34T mutation. 0.01% gelatin), 2.5 U Ampli Taq Gold Polymerase (Perkin
Elmer, Foster City, CA) and 100 ng of DNA template, using
an automated Thermal Cycler 9700 (Perkin Elmer). The
Calcium wave propagation and M34T pathogenicity
cycling profile consisted of an initial denaturation at 948C
Intercellular calcium waves propagating in the network of sup- for 12 min, which was followed by 35 cycles of 948C 
porting cells have been observed in the organ of Corti in 45 s, 608C  45 s, 728C  80 s and a final extension step at
response to mechanical stimulation of single-hair cells (57). 728C for 10 min. Exon I and flanking splice sites were ampli-
This phenomenon depends on two underlying mechanisms: fied using oligonucleotides EX1CX26F (tcaaaggaactagga-
(i) the release of an extracellular messenger, believed to be gatcgg) and EX1CX26R (aaggacgtgtgttggtccag) as forward
ATP because purinergic receptor antagonists and apyrase, and reverse primers, respectively.
which hydrolyzes ATP, block the propagation of Ca2þ When genetic analysis was either negative for GJB2 mutations
waves in cochlear organotypic cultures (57); and (ii) the or a heterozygous condition was found, samples were further
diffusion of inositol trisphosphate (InsP3) through gap junc- tested for GJB6 mutations. The D(GJB6-D13S1830) deletion
tions (35,58), because agents that interfere with InsP3 signal- of the GJB6 gene was detected by PCR amplification as
ing or gap junction communication effectively inhibit wave suggested by del Castillo et al. (63). Amplicons were directly
propagation between many cell types (59 –61), including sequenced with the same PCR primers, using an ABI-PRISM
supporting cells of the organ of Corti (33). We speculate, big-dye terminator cycle reaction kit (Perkin Elmer), and
therefore, that the disruption of this intercellular signaling samples were then analyzed on an automated sequencer (ABI
Human Molecular Genetics, 2006, Vol. 15, No. 17 2583

3100, Perkin Elmer). Patients were eventually divided into three 20 dB. Hearing thresholds were compared with those of
groups: group I, negative for GJB2/GJB6 mutations; group II, age- and sex-matched controls (ISO 7029:2000). Differences
carrying mutations different from M34T and group III, carrying in audiological parameters between groups were determined
at least the M34T mutation. by Fisher’s exact test and P-values ,0.05 were considered to
be significant.
Patients
Molecular cloning
We have studied eight probands with mild-to-severe sensori-
neural hearing loss, in which genetic analysis had identified The coding region of HCx30 (PubMed-NCBI accession
the presence of the M34T mutation as a heterozygous or a number HSA005585), HCx26wt (PubMed-NCBI accession
compound heterozygous condition and their respective number AF281280) or HCx26 mutations was amplified by
families (seven in total). A total of 28 subjects, aged PCR from genomic DNA of normal hearing subjects and
between 5 and 69 years (median ¼ 37 years and 5 months) patients with different forms of sensorineural deafness. Ampli-
were studied. The follow-up period ranged from a minimum cons were subcloned into the bicistronic vector pIRES-EGFP
of 18 months to a maximum of 12 years, and in all patients, (Clontech, Palo Alto, CA), which permits an efficient selection
serial audiograms were available. None reported a relevant of transfected cells for functional studies by monitoring the
history of otological disease, noise exposure or assumption expression of the enhanced form of Aequorea victoria GFP,
of ototoxic drugs. as previously described (32). For electrophysiological record-
A detailed history was collected from all patients, followed ings, we employed HCx26 proteins fused to either EYFP
by otoscopic and audiological examination comprehensive of (HCx26wt –EYFP) or the circularly permuted yellow mutant
pure tone audiometry (or conditioned infantile audiometry in of the GFP, called Venus (65) (HCx26wt –Venus), by two
one patient), tympanometry, stapedial reflex threshold and linkers of different lengths. For the generation of fusion pro-
auditory brainstem evoked potentials (ABR). Otoacoustic teins, the open reading frame (ORF) of the specified constructs
emissions were not available in all patients. In all cases, the was amplified by PCR using oligonucleotide primers that
audiological study revealed cochlear site of lesion, and there introduced the desired restriction enzyme sites at both ends
were no clinical features suggestive of a syndromic deafness. of the coding sequence and deleted the stop codon. The fol-
None of them accused vertigo or dizziness, so vestibular func- lowing primers, which introduced EcoRI –BamHI sites, were
tion was not tested. Audiological features of hearing loss were used for Linker L1 (six amino acids: DPPVAT):
then analyzed according to the GenDeaf study group rec- 26forward: 50 -CGGAATTCAGATGGATTGGGGCACG-30 ;
ommendations (64). Air conduction thresholds were measured 26reverse: 50 -CGGGATCCACTGGCTTTTTTGACTT-30 ; 30-
across the following frequencies: 125, 250, 500, 1000, 2000, forward: 50 -CGGAATTCCCAGCGCAATGGATTGG-30 ; 30-
4000 and 8000 Hz, whereas for bone conduction, the range reverse: 50 -CGGGATCCCTTGGGAAACCTGTGAT-30 . For
was between 250 and 4000 Hz. The dB values relate to linker L2, which introduced EcoRI–XhoI sites and comprised
dB HL. The degree of hearing impairment was classified 17 residues (RILQSTVPRARDPPVAT), the primers used
by the pure-tone average (PTA) applied to the better ear were 26forward: 50 -GGTCCTCGAGATGGATTGGGGCACG
at 500, 1000, 2000 and 4000 Hz. In the case of CTGC-30 ; 26reverse: 50 -CCCGAATTCGAACTGGCTTTTTT
normal hearing, PTA is 20 dB; in mild hearing loss, PTA GACTTCCC-30 .
is .20  40 dB; in moderate hearing loss, PTA is The resulting PCR products were digested with the two
.40  70 dB; severe hearing loss is characterized by a PTA enzymes and ligated into the respective sites of pEGFP-N1
.70  95 dB, and hearing loss is defined as profound when (Clontech). All constructs were sequenced using the Dye ter-
PTA .95 dB. minator (Perkin-Elmer), as recommended by the manufac-
Hearing impairment is progressive when a deterioration turer, to verify that PCR amplification did not introduce
of 15 dB in the PTA occurs within 10 years. Depending on unwanted mutations. The length of the linker did not interfere
its configuration, the audiometric curve is classified as flat, if with the electrophysiological profile of the recombinant gap
the difference between 125 and 8000 Hz is ,15 dB; junction channels, as assessed by measuring their unitary con-
mid-frequency U-shaped, when difference is 15 dB between ductance with a dual whole-cell patch clamp technique.
the poorest thresholds in the mid-frequencies
(.500  2000 Hz) and those at higher (.2000  8000 Hz) Cell culture and transfection
and lower (500 Hz) frequencies; low frequency-ascending, A clone of HeLa cells essentially devoid of connexins (66)
when there are 15 dB between the poorer low frequency was kindly provided by Professor Klaus Willecke (University
thresholds and the higher frequencies; high frequency-gently of Bonn, Germany) and cultured according to standard
sloping, if the difference between the mean of 500 and procedures. Twenty-four hours after plating, cells were trans-
1000 Hz and the mean of 4000 and 8000 Hz is within fected with the expression vectors described previously, using
15 –29 dB; high frequency-steeply sloping, with 30 dB lipofectamine (Gibco/Invitrogen, Leek, The Netherlands).
difference between the latter frequency pairs. Hearing impair- Experiments were performed the following day.
ment is classified as (i) asymmetrical, if .10 dB difference
occurs between the ears in at least two frequencies, with the
Protein immunoblot analysis
PTA in the better ear exceeding 20 dB HL and (ii) unilateral,
when one ear has either a PTA .20 dB or one frequency Proteins were dissolved in lysis buffer (50 mM Tris pH 6.8, 2%
threshold exceeding 50 dB while the other ear has a PTA SDS, 10 mg/ml each of aprotinin, chymostatin and leupeptin)
2584 Human Molecular Genetics, 2006, Vol. 15, No. 17

from cells grown to confluence. Prior to electrophoresis, instantaneous conductance measured at the earliest time resol-
protein concentration was measured by the bicinchoninic ution of Ij after the imposition of a voltage step), pairs coupled
acid method (Pierce, Rockford, IL) with bovine serum by conductances larger than 10 nS were discarded, thereby
albumin as the standard. For each sample, 20 mg of total pro- limiting the effect of series resistance on these measurements.
teins were separated on 13% SDS gel and then transferred to To block gap junction channels, cells were transiently super-
nitrocellulose. Coomassie blue staining of identical gels run fused in ECS saturated with 100% CO2 to produce carbonic
in parallel confirmed the equal loading of the samples. Nitro- acid (H2CO3), which, in its non-dissociated form, is membrane
cellulose filters were blocked with 1% bovine serum albumin permeable and causes a rapid closure of the gap junction
in Tween-added PBS (TPBS: 137 mM NaCl, 2.7 mM KCl, channels. The current flowing through single-gap junction
8.1 mM Na2PO4, 1.5 mM KH2PO4, pH 7.4, supplemented channels was detected as discrete step-like events while the
with 0.1% Tween 20) and incubated overnight with the cells recovered from cytoplasm acidification during washout
primary anti-Cx26 antibody under continuous shaking. Blots of the CO2.
were further incubated for 1 – 2 h with the alkaline phospha-
tase (AP)-conjugated secondary antibody, processed for
protein visualization with ECL and exposed to CL-Xposure Intracellular delivery of LY and fluorescence imaging
Films (Pierce) for 10 or 20 min. For dye transfer assays, cell 1 (donor) was patch-clamped
using patch pipettes filled with a 4% LY solution in ICS-2,
containing (in mM ) 60 LiCl and 10 HEPES (adjusted to
Immunofluorescence
pH 7.2 with KOH; 320 mOsm). Illumination wavelength
Cells grown onto glass coverslips were fixed with 2% parafor- was set at 425 nm by a fast switching monochromator (Poly-
maldehyde and incubated with the primary anti-Cx26 antibody chrome IV, TILL Photonics, Martinsried, Germany). At
for 1 h at 48C. Following a 30 min incubation at 48C in the image recording onset, cell 1 was maintained in the
presence of a secondary antibody, coverslips were mounted cell-attached configuration for a few seconds to establish a
onto glass slides and visualized under a Leica DMLS fluor- baseline. Subsequently, the patch of membrane under the
escence microscope (Leica Microsystems, Wetzlar, Germany). pipette was ruptured, allowing LY to fill the cell while
leaving the seal intact (whole-cell recording conditions), and
fluorescence emission (F) was selected around 535 nm using
Dual-patch clamp recordings a D535/30 m filter (Chroma, Rockingham, VT). Images were
For electrophysiological recordings, cells were grown on thin formed on a scientific grade CCD camera (SensiCam, PCO
(no. 0) coverslips and transferred to an experimental chamber Computer Optics, Kelheim, Germany) using either 40 or
mounted on the stage of an upright microscope equipped with 60 water-immersion objectives (LumPlan FL, 0.8
an infinity-corrected water-immersion objectives 60, and 0.9 NA, respectively; Olympus) on a motorized upright
0.90 numeric aperture (NA); LUMPlanFl, Olympus, Tokyo, microscope (BX61, Olympus) and displayed as (F 2 Fbck)/
Japan. Cells were constantly superfused with ECS containing (Fmax 2 Fbck), where Fmax is the asymptotic maximal value
(in mM ) 150 NaCl, 4 KCl, 1 MgCl2, 5 Hepes, 2 CaCl2, reached in the injected cell and Fbck is background fluor-
2 Pyruvate, 5 Glucose, 2 CsCl and 1 BaCl2 (pH 7.4, escence. At the end of image recording (3 – 6 min), cell 2
330 mOsm). Glass capillaries for patch clamp recordings (acceptor) was contacted by a second pipette containing an
were formed on a vertical puller (PP-83, Narishige, Japan) identical intracellular solution. The whole-cell configuration
from 1.5 mm outer diameter soda glass (Harvard Apparatus, was achieved in cell 2 also, and the responses to a sequence
Edenbridge, UK) and filled with an intracellular solution of 13 mV voltage pulses, fed to the patch clamp amplifier con-
(ICS-1) containing (in mM ) 130 KCl, 10 Na – Aspartate, 0.26 nected to cell 1, were used to measure junctional conductance.
CaCl2, 5 TEA –Cl, 1 MgCl2, 5 Hepes, 2 EGTA and 3 ATP – Data were analyzed offline using the Matlab 7.0 software
Mg (pH 7.2). Current and voltage were sampled at rates package (The MathWorks, Natick, MA).
between 1 and 20 kHz. In the whole-cell configuration, the
patch electrodes acquired an access resistance of 10 –
Intercellular dye transfer
12 MOhm, on average, whereas the input resistance of
untransfected HeLa cells was .0.5 GOhm. To measure junc- Glass capillaries were prepared with a dual-step puller (Nar-
tional conductance, each cell of an isolated pair was voltage- ishige, Tokyo, Japan) and filled with a 5% solution of LY (dis-
clamped independently with one of two List EPC-7 amplifiers solved in 0.33 M lithium chloride). Individual cells in
and kept at the same holding potential (Vh). By stepping the GFP-expressing clusters were injected with a pneumatic
voltage in one cell (cell 1) while keeping the potential of PLI-100 pico-injector (Medical Systems Corp., Greenvale,
cell 2 at Vh, thus establishing a transient transjunctional NY) and mounted onto a Zeiss-inverted Axiovert 35 TV
voltage Vj ; V1 2 V2 ¼ DV1, junctional current (Ij) was microscope (Carl Zeiss, Jena, Germany). For both GFP and
measured directly as the current change in the unstepped LY, the filter set was Zeiss 09 (BP450-490/LP520). Fluor-
cell (i.e. Ij ¼ 2DI2). Values of junctional conductance (gj) escence images were collected through an oil immersion
were calculated by dividing Ij/Vj, corrected for the error due objective (40, 1.8 NA), captured by a low light level CCD
to the access resistance of both pipettes. In the study of the camera (Hamamatsu Photonics, Tokyo, Japan) and fed into a
voltage dependence of normalized conductance (Gjss ¼ gjss/ digital image processor developed in the laboratory. Intercel-
gj(0), where gjss is the steady-state conductance measured lular LY diffusion was revealed by capturing three different
at the end of a prolonged voltage step and gj(0) is the images for each experiment: image 1 (GFP), showing
Human Molecular Genetics, 2006, Vol. 15, No. 17 2585

GFP-expressing cells before injection; image 2 (GFP þ LY), fragments corresponded in HCx26 to: Ser19 –Glu42 (TM1),
capturing the same field of cells 3 –5 min after LY injection Arg75– Ala96 (TM2), Leu132 –Val153 (TM3) and Glu187 –
and containing the fluorescence of both GFP and LY; image Leu209 (TM4). Charged residues found immediately next to
3 (LY), resulting from the subtraction of image 1 from those segments were also added in order to consider eventual
image 2, thus displaying LY fluorescence alone. Subsequent electrostatic interactions. Mapping of the Ca coordinates from
processing for false color display was performed with Corel the original mouse Cx32 model generated a rough Ca model
Photo-Paint software (Corel Corporation, Ottawa, Canada). of HCx26. Subsequently, all the remaining atoms of the
amino acids were added using the canonical conformation
with the XLEAP module of AMBER8.0 software suite (68).
Ca21 imaging Additionally, all helices were capped with acetyl and
Transfected HeLa cells grown onto coverslips were incubated N-methyl groups in order to avoid the spurious effects of term-
in modified Krebs solution (KRH), containing (in mM) inal zwitterionic charges.
25 Hepes/NaOH buffer, pH 7.4, 125 NaCl, 5 KCl, 1 MgSO4, This procedure generated an all-atoms model with a large
1.2 KH2PO4, 2 CaCl2, 10 glucose, supplemented with 0.5% number of steric clashes and internal tensions that were
BSA and loaded at room temperature with a mixture (1:2, relaxed using energy minimization (EM) and molecular
vol:vol in DMSO) of fura-2/AM (final concentration in dynamics (MD). Simulations were performed in the presence
KRH ¼ 1 mM ) and 20% Pluronic gel (Molecular Probes, of implicit solvent with the generalized Born approach (69)
Eugene, OR; final concentration in KRH ¼ 0.04%). After as implemented in the SANDER module of AMBER8.0
45 min, the loading solution was removed and the cells with a 2 ps time-step. After EM, 50 ps of constrained
washed and kept in KRH for 45 min to allow complete MD were performed, in which the initial harmonic constraints
de-esterification of the dye. Videomicroscopy and Ca2þ on the Ca carbons were linearly reduced from 1 to
measurements were carried out at room temperature. The 0 kcal/mol-Å2, followed by 450 ps of unconstrained MD.
fura-2 loaded cultures were observed on an inverted micro- Simulations were stopped when the systems reached stabiliz-
scope (Zeiss Axiovert 35 TV) with an oil immersion 40 ation of the RMSD. A dielectric constant of 78, a cutoff
objective (1.8 NA; Carl Zeiss). Cells were excited at wave- distance of 1.8 nm and a salt concentration of 0.15 M were
lengths between 340 and 380 nm with a monochromator used. The simulations used the Amber94 force field (70)
device equipped with integrated light source (Polychrome keeping the temperature at 100 K using Langevin dynamics
IV, Till Photonics). Excitation light was separated from the with a collision frequency of 2 ps21. The shake algorithm
light emitted from the sample using a 395 nm dichroic was used to constraint all chemical bonds. This procedure is
mirror. Images of emitted fluorescence .510 nm were expected to allow the entire complex to explore the energy
recorded at a frequency of one image per second by a landscape more efficiently than a simple EM, obtaining a
cooled slow-scan interline transfer camera (IMAGO CCD better relaxation of the protein complex. A similar strategy
camera, Till Photonics) and simultaneously displayed on a has already been proven to produce reliable results in other
19 inch Vivitron color monitor. The imaging system was con- protein– RNA and protein – protein complexes (71,72).
trolled by an integrating imaging software package (TILLvi- RMSDs were calculated from the last minimization step
sION, Till Photonics) using a personal computer. Video over the Ca- atoms. The distances between the extremes of
frames were then digitized, integrated and processed offline the pore-lining TM3 helices were calculated between the
to convert fluorescence data into Ca2þ maps by computing a centers of mass of the first (last) four Ca atoms of two oppo-
ratio of 340/380 nm excitation wavelength values. Mechanical site TM3 helices. The inter-helix angles were calculated
stimulation of single cells in confluent monolayer was per- between the lines defined by centers of mass of the first
formed by briefly deforming the cell surface with a micropip- and last four Ca carbons of each helix TM3. The distances
ette. A fire-polished glass micropipette with a tip diameter between OH atoms of Tyr152, corresponding to the
1 mm was positioned over a single cell by a micromanipula- maximum constriction of the pore, were calculated between
tor (Narishige). The pipette was briefly deflected downward couples of residues in opposite TM3 helices in the connexon.
manually to transiently distort the membrane. Experiments All the distances and angles were averaged over the three
were either discontinued or discarded when there was evi- possible pairs of opposite helices in the hexamer.
dence of damage to the plasma membrane, as revealed by
leak of the fluorescent probe from the cell. Results are
shown as mean + SD. Comparisons between two populations
of data were made using the Student’s unpaired t-test and
ACKNOWLEDGEMENTS
P-values of 0.05 or less were considered to be significant.
This work has been funded by grants from Regione Friuli
Venezia-Giulia (to P.D’A.), Telethon Italy (GP0043Y02 to
Generation of the all-atoms model
F.M. and P.D’A. and GGP05131 to F.M.), the Sixth Research
Our analysis is based on the Ca model of mouse Cx32 Frame Program of the European Union (FP6 Integrated Project
reported by Fleishman et al. (38; PDB entry code 1TXH). EuroHear, LSHG-CT-20054-512063 to F.M.), Fondazione
This model includes the transmembrane (TM) a-helices corre- CARIPARO (to S.P.) and National Institutes of Health
sponding to the segments Ala19 – Ala40 (TM1), Leu76 –Ala96 (RO1-NS036706 to F.F.B.).
(TM2), Leu131 – Val152 (TM3) and Val189 – Val209 (TM4).
After sequence alignment using T-COFFEE (67), these Conflict of Interest statement. None declared.
2586 Human Molecular Genetics, 2006, Vol. 15, No. 17

REFERENCES 23. Oshima, A., Doi, T., Mitsuoka, K., Maeda, S. and Fujiyoshi, Y. (2003)
Roles of Met-34, Cys-64 and Arg-75 in the assembly of human connexin
1. Bruzzone, R., White, T.W. and Paul, D.L. (1996) Connections with 26. Implication for key amino acid residues for channel formation and
connexins: the molecular basis of direct intercellular signaling. function. J. Biol. Chem., 278, 1807–1816.
Eur. J. Biochem., 238, 1 –27. 24. Skerrett, I.M., Di, W.L., Kasperek, E.M., Kelsell, D.P. and Nicholson, B.J.
2. Sosinsky, G.E. and Nicholson, B.J. (2005) Structural organization of gap (2004) Aberrant gating, but a normal expression pattern, underlies the
junction channels. Biochim. Biophys. Acta, 1711, 99 –125. recessive phenotype of the deafness mutant Connexin26M34T. FASEB J.,
3. Willecke, K., Eiberger, J., Degen, J., Eckardt, D., Romualdi, A., 18, 860–862.
Guldenagel, M., Deutsch, U. and Sohl, G. (2002) Structural and functional 25. Estivill, X., Fortina, P., Surrey, S., Rabionet, R., Melchionda, S.,
diversity of connexin genes in the mouse and human genome. Biol. D’Agruma, L., Mansfield, E., Rappaport, E., Govea, N., Mila, M. et al.
Chem., 383, 725–737. (1998) Connexin-26 mutations in sporadic and inherited sensorineural
4. Gerido, D.A. and White, T.W. (2004) Connexin disorders of the ear, skin, deafness. Lancet, 351, 394–398.
and lens. Biochim. Biophys. Acta, 1662, 159 –170. 26. Wang, H.L., Chang, W.T., Li, A.H., Yeh, T.H., Wu, C.Y., Chen, M.S. and
5. Wei, C.J., Xu, X. and Lo, C.W. (2004) Connexins and cell signaling in Huang, P.C. (2003) Functional analysis of connexin-26 mutants associated
development and disease. Annu. Rev. Cell Dev. Biol., 20, 811 –838. with hereditary recessive deafness. J. Neurochem., 84, 735– 742.
6. Kikuchi, T., Kimura, R.S., Paul, D.L., Takasaka, T. and Adams, J.C. 27. Peracchia, C. (2004) Chemical gating of gap junction channels; roles of
(2000) Gap junction systems in the mammalian cochlea. Brain Res. Brain calcium, pH and calmodulin. Biochim. Biophys. Acta, 1662, 61 –80.
Res. Rev., 32, 163–166. 28. Lautermann, J., ten Cate, W.J., Altenhoff, P., Grummer, R., Traub, O.,
7. Petit, C., Levilliers, J. and Hardelin, J.P. (2001) Molecular genetics of Frank, H., Jahnke, K. and Winterhager, E. (1998) Expression of the
hearing loss. Annu. Rev. Genet., 35, 589–646. gap-junction connexins 26 and 30 in the rat cochlea. Cell Tissue Res., 294,
8. Sabag, A.D., Dagan, O. and Avraham, K.B. (2005) Connexins in hearing 415–420.
loss: a comprehensive overview. J. Basic Clin. Physiol. Pharmacol., 16, 29. Forge, A., Marziano, N.K., Casalotti, S.O., Becker, D.L. and Jagger, D.
101 –116. (2003) The inner ear contains heteromeric channels composed of cx26 and
9. Jentsch, T.J. (2000) Neuronal KCNQ potassium channels: physiology and cx30 and deafness-related mutations in cx26 have a dominant negative
role in disease. Nat. Rev. Neurosci., 1, 21–30. effect on cx30. Cell Commun. Adhes., 10, 341–246.
10. Boettger, T., Hubner, C.A., Maier, H., Rust, M.B., Beck, F.X. and 30. Valiunas, V., Manthey, D., Vogel, R., Willecke, K. and Weingart, R.
Jentsch, T.J. (2002) Deafness and renal tubular acidosis in mice lacking (1999) Biophysical properties of mouse connexin30 gap junction channels
the K–Cl co-transporter Kcc4. Nature, 416, 874–878. studied in transfected human HeLa cells. J. Physiol., 519, 631 –644.
11. Nuttall, A.L. (1999) Sound-induced cochlear ischemia/hypoxia as a 31. Dahl, E., Manthey, D., Chen, Y., Schwarz, H.J., Chang, Y.S., Lalley, P.A.,
mechanism of hearing loss. Noise Health, 2, 17 –32. Nicholson, B.J. and Willecke, K. (1996) Molecular cloning and functional
12. Rio, C., Dikkes, P., Liberman, M.C. and Corfas, G. (2002) Glial fibrillary expression of mouse connexin-30, a gap junction gene highly expressed in
acidic protein expression and promoter activity in the inner ear of adult brain and skin. J. Biol. Chem., 271, 17903–17910.
developing and adult mice. J. Comp. Neurol., 442, 156– 162. 32. Bruzzone, R., Veronesi, V., Gomes, D., Bicego, M., Duval, N., Marlin, S.,
13. Kelsell, D.P., Dunlop, J., Stevens, H.P., Lench, N.J., Liang, J.N., Petit, C., D’Andrea, P. and White, T.W. (2003) Loss-of-function and
Parry, G., Mueller, R.F. and Leigh, I.M. (1997) Connexin 26 mutations in residual channel activity of connexin26 mutations associated with
hereditary non-syndromic sensorineural deafness. Nature, 387, 80–83. non-syndromic deafness. FEBS Lett., 533, 79– 88.
14. White, T.W., Deans, M.R., Kelsell, D.P. and Paul, D.L. (1998) Connexin 33. Beltramello, M., Piazza, V., Bukauskas, F.F., Pozzan, T. and Mammano, F.
mutations in deafness. Nature, 394, 630–631. (2005) Impaired permeability to Ins(1,4,5)P3 in a mutant connexin
15. Houseman, M.J., Ellis, L.A., Pagnamenta, A., Di, W.L., Rickard, S., underlies recessive hereditary deafness. Nat. Cell Biol., 7, 63–69.
Osborn, A.H., Dahl, H.H., Taylor, G.R., Bitner-Glindzicz, M., Reardon, W. 34. Beltramello, M., Bicego, M., Piazza, V., Ciubotaru, C.D., Mammano, F.
et al. (2001) Genetic analysis of the connexin-26 M34T variant: and D’Andrea, P. (2003) Permeability and gating properties of human
identification of genotype M34T/M34T segregating with mild-moderate connexins 26 and 30 expressed in HeLa cells. Biochem. Biophys. Res.
non-syndromic sensorineural hearing loss. J. Med. Genet., 38, 20 –25. Commun., 305, 1024– 1033.
16. Denoyelle, F., Weil, D., Maw, M.A., Wilcox, S.A., Lench, N.J., 35. Allbritton, N.L. and Meyer, T. (1993) Localized calcium spikes and
Allen-Powell, D.R., Osborn, A.H., Dahl, H.H., Middleton, A., propagating calcium waves. Cell Calcium, 14, 691 –697.
Houseman, M.J. et al. (1997) Prelingual deafness: high prevalence of a 36. Osipchuk, Y. and Cahalan, M. (1992) Cell-to-cell spread of calcium
30delG mutation in the connexin 26 gene. Hum. Mol. Genet., 6, signals mediated by ATP receptors in mast cells. Nature, 359, 241–244.
2173–2177. 37. Scemes, E., Dermietzel, R. and Spray, D.C. (1998) Calcium waves
17. Kelley, P.M., Harris, D.J., Comer, B.C., Askew, J.W., Fowler, T., between astrocytes from Cx43 knockout mice. Glia, 24, 65 –73.
Smith, S.D. and Kimberling, W.J. (1998) Novel mutations in the connexin 38. Fleishman, S.J., Unger, V.M., Yeager, M. and Ben-Tal, N. (2004) A
26 gene (GJB2) that cause autosomal recessive (DFNB1) hearing loss. Calpha model for the transmembrane alpha helices of gap junction
Am. J. Hum. Genet., 62, 792 –799. intercellular channels. Mol. Cell., 15, 879–888.
18. Scott, D.A., Kraft, M.L., Carmi, R., Ramesh, A., Elbedour, K., Yairi, Y., 39. Unger, V.M., Kumar, N.M., Gilula, N.B. and Yeager, M. (1999)
Srisailapathy, C.R., Rosengren, S.S., Markham, A.F., Mueller, R.F. et al. Three-dimensional structure of a recombinant gap junction membrane
(1998) Identification of mutations in the connexin 26 gene that cause channel. Science, 283, 1176–1180.
autosomal recessive nonsyndromic hearing loss. Hum. Mutat., 11, 40. Wilcox, S.A., Saunders, K., Osborn, A.H., Arnold, A., Wunderlich, J.,
387 –394. Kelly, T., Collins, V., Wilcox, L.J., McKinlay Gardner, R.J., Kamarinos, M.
19. Feldmann, D., Denoyelle, F., Loundon, N., Weil, D., Garabedian, E.N., et al. (2000) High frequency hearing loss correlated with mutations in the
Couderc, R., Joannard, A., Schmerber, S., Delobel, B., Leman, J. et al. GJB2 gene. Hum. Genet., 106, 399–405.
(2004) Clinical evidence of the nonpathogenic nature of the M34T variant 41. Kenneson, A., Van Naarden Braun, K. and Boyle, C. (2002) GJB2
in the connexin 26 gene. Eur. J. Hum. Genet., 12, 279– 284. (connexin 26) variants and nonsyndromic sensorineural hearing loss: a
20. D’Andrea, P., Veronesi, V., Bicego, M., Melchionda, S., Zelante, L., HuGE review. Genet. Med., 4, 258– 274.
Di Iorio, E., Bruzzone, R. and Gasparini, P. (2002) Hearing loss: 42. Wu, B.L., Lindeman, N., Lip, V., Adams, A., Amato, R.S., Cox, G.,
frequency and functional studies of the most common connexin26 alleles. Irons, M., Kenna, M., Korf, B., Raisen, J. et al. (2002) Effectiveness of
Biochem. Biophys. Res. Commun., 296, 685– 691. sequencing connexin 26 (GJB2) in cases of familial or sporadic childhood
21. Martin, P.E., Coleman, S.L., Casalotti, S.O., Forge, A. and Evans, W.H. deafness referred for molecular diagnostic testing. Genet. Med., 4,
(1999) Properties of connexin26 gap junctional proteins derived from 279–288.
mutations associated with non-syndromal heriditary deafness. Hum. Mol. 43. Cucci, R.A., Prasad, S., Kelley, P.M., Green, G.E., Storm, K., Willcox, S.,
Genet., 8, 2369–2376. Cohn, E.S., Van Camp, G. and Smith, R.J. (2000) The M34T allele variant
22. Thonnissen, E., Rabionet, R., Arbones, M.L., Estivill, X., Willecke, K. of connexin 26. Genet. Test, 4, 335–344.
and Ott, T. (2002) Human connexin26 (GJB2) deafness mutations affect 44. Kenna, M.A., Wu, B.L., Cotanche, D.A., Korf, B.R. and Rehm, H.L.
the function of gap junction channels at different levels of protein (2001) Connexin 26 studies in patients with sensorineural hearing loss.
expression. Hum. Genet., 111, 190–197. Arch. Otolaryngol. Head Neck Surg., 127, 1037–1042.
Human Molecular Genetics, 2006, Vol. 15, No. 17 2587

45. Marlin, S., Garabedian, E.N., Roger, G., Moatti, L., Matha, N., Lewin, P., 59. Boitano, S., Dirksen, E.R. and Sanderson, M.J. (1992) Intercellular
Petit, C. and Denoyelle, F. (2001) Connexin 26 gene mutations in propagation of calcium waves mediated by inositol trisphosphate. Science,
congenitally deaf children: pitfalls for genetic counseling. Arch. 258, 292–295.
Otolaryngol. Head Neck Surg., 127, 927– 933. 60. Newman, E.A. and Zahs, K.R. (1997) Calcium waves in retinal glial cells.
46. Snoeckx, R.L., Huygen, P.L., Feldmann, D., Marlin, S., Denoyelle, F., Science, 275, 844 –847.
Waligora, J., Mueller-Malesinska, M., Pollak, A., Ploski, R., Murgia, A. 61. Giaume, C. and Venance, L. (1998) Intercellular calcium signaling and
et al. (2005) GJB2 mutations and degree of hearing loss: a multicenter gap junctional communication in astrocytes. Glia, 24, 50–64.
study. Am. J. Hum. Genet., 77, 945–957. 62. Bruzzone, R. and Cohen-Salmon, M. (2005) Hearing the messenger:
47. Roux, A.F., Pallares-Ruiz, N., Vielle, A., Faugere, V., Templin, C., Ins(1,4,5)P3 and deafness. Nat. Cell Biol., 7, 14 –16.
Leprevost, D., Artieres, F., Lina, G., Molinari, N., Blanchet, P. et al. 63. del Castillo, I., Villamar, M., Moreno-Pelayo, M.A., del Castillo, F.J.,
(2004) Molecular epidemiology of DFNB1 deafness in France. BMC Med. Alvarez, A., Telleria, D., Menendez, I. and Moreno, F. (2002) A deletion
Genet., 5, 5. involving the connexin 30 gene in nonsyndromic hearing impairment.
48. Levi, H., Tell, L. and Cohen, T. (2004) Sensorineural hearing loss in N. Engl. J. Med., 346, 243– 249.
Jewish children born in Jerusalem. Int. J. Pediatr. Otorhinolaryngol., 68, 64. GenDeaf Study GroupMazzoli, M., Van Camp, G., Newton, V.,
1245–1250. Giarbini, M., Declau, F. and Parving, A. (2003) Recommendations for the
49. Liu, X.Z., Pandya, A., Angeli, S., Telischi, F.F., Arnos, K.S., Nance, W.E. description of genetic and audiological data for families with
and Balkany, T. (2005) Audiological features of GJB2 (connexin 26) nonsyndromic hereditary hearing impairment. Audiological Medicine, 1,
deafness. Ear Hear., 26, 361 –369. 148–150.
50. Cryns, K., Orzan, E., Murgia, A., Huygen, P.L., Moreno, F., 65. Miyawaki, A., Llopis, J., Heim, R., McCaffery, J.M., Adams, J.A.,
del Castillo, I., Chamberlin, G.P., Azaiez, H., Prasad, S., Cucci, R.A. et al. Ikura, M. and Tsien, R.Y. (1997) Fluorescent indicators for Ca2þ
(2004) A genotype–phenotype correlation for GJB2 (connexin 26) based on green fluorescent proteins and calmodulin. Nature, 388,
deafness. J. Med. Genet., 41, 147–154.
882–887.
51. Wilch, E., Zhu, M., Burkhart, K.B., Regier, M., Elfenbein, J.L.,
66. Elfgang, C., Eckert, R., Lichtenberg-Frate, H., Butterweck, A., Traub, O.,
Fisher, R.A. and Friderici, K.H. (2006) Expression of GJB2 and GJB6 is
Klein, R.A., Hulser, D.F. and Willecke, K. (1995) Specific permeability
reduced in a novel DFNB1 allele. Am. J. Hum. Genet., 79, 174–179.
and selective formation of gap junction channels in connexin-transfected
52. Palmada, M., Schmalisch, K., Bohmer, C., Schug, N., Pfister, M., Lang, F.
HeLa cells. J. Cell Biol., 129, 805– 817.
and Blin, N. (2006) Loss of function mutations of the GJB2 gene detected
in patients with DFNB1-associated hearing impairment. Neurobiol. Dis., 67. Notredame, C., Higgins, D.G. and Heringa, J. (2000) T-Coffee: a novel
22, 112 –118. method for fast and accurate multiple sequence alignment. J. Mol. Biol.,
53. White, T.W. and Bruzzone, R. (2000) Gap junctions: fates worse than 302, 205–217.
death? Curr. Biol., 10, R685– R688. 68. Case, D.A., Darden, T.A., Cheatham, T.E., III, Simmerling, C.L.,
54. Cohen-Salmon, M., Ott, T., Michel, V., Hardelin, J.P., Perfettini, I., Wang, J., Duke, R.E., Lou, R., Merz, K.M., Wang, B., Pearlman, D.A.
Eybalin, M., Wu, T., Marcus, D.C., Wangemann, P., Willecke, K. et al. et al. (2004) AMBER 8.0. Amber 8.0.
(2002) Targeted ablation of connexin26 in the inner ear epithelial gap 69. Tsui, V. and Case, D.A. (2000) Theory and applications of the generalized
junction network causes hearing impairment and cell death. Curr. Biol., Born solvation model in macromolecular simulations. Biopolymers, 56,
12, 1106–1111. 275–291.
55. Zhou, X.W., Pfahnl, A., Werner, R., Hudder, A., Llanes, A., Luebke, A. 70. Cornell, W.D., Cieplak, P., Bayly, C.I., Gould, I.R., Merz, K.M.,
and Dahl, G. (1997) Identification of a pore lining segment in gap junction Ferguson, D.M., Spellmayer, D.C., Fox, T., Caldwell, J.W. and
hemichannels. Biophys. J., 72, 1946–1953. Kollman, P.A. (1995) A second generation force field for the simulation of
56. Kronengold, J., Trexler, E.B., Bukauskas, F.F., Bargiello, T.A. and proteins, nucleic acids, and organic molecules. J. Am. Chem. Soc., 117,
Verselis, V.K. (2003) Single-channel SCAM identifies pore-lining 5179–5197.
residues in the first extracellular loop and first transmembrane domains of 71. Ayala, Y.M., Pantano, S., D’Ambrogio, A., Buratti, E., Brindisi, A.,
Cx46 hemichannels. J. Gen. Physiol., 122, 389–405. Marchetti, C., Romano, M. and Baralle, F.E. (2005) Human, Drosophila,
57. Gale, J.E., Piazza, V., Ciubotaru, C.D. and Mammano, F. (2004) A and C. elegans TDP43: nucleic acid binding properties and splicing
mechanism for sensing noise damage in the inner ear. Curr. Biol., 14, regulatory function. J. Mol. Biol., 348, 575 –588.
526 –529. 72. Pantano, S., Marcello, A., Ferrari, A., Gaudiosi, D., Sabo, A., Pellegrini, V.,
58. Sanderson, M.J., Charles, A.C., Boitano, S. and Dirksen, E.R. (1994) Beltram, F., Giacca, M. and Carloni, P. (2005) Insights on HIV-1 Tat:
Mechanisms and function of intercellular calcium signaling. Mol. Cell. P/CAF bromodomain molecular recognition from in vivo experiments and
Endocrinol., 98, 173–187. molecular dynamics simulations. Proteins, 62, 1062–1073.

You might also like