Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Audiological Medicine, 2009; 00: 1–6

Inner ear connexins, intercellular signalling and deafness

FABIO MAMMANO1-3 & fabio anselmi2


1Departmentof Physics ‘G.Galilei’, University of Padova, Padova, 2Venetian Institute of Molecular Medicine, Foundation for
Advanced Biomedical Research, Padova, and 3CNR Neuroscience Institute, Padova, Italy

Abstract
The genes GJB2 and GJB6, respectively encoding transmembrane proteins connexin 26 (Cx26) and connexin 30 (Cx30)
are found within 50 kb of each other in the DFNB1 complex deafness locus on chromosome 13q12. Up to 50% of all
patients with autosomal recessive non-syndromic prelingual deafness in different populations present with mutations or
deletions in this locus. Cx26 and Cx30 are abundantly expressed in the inner ear and, in recent years, have been shown to
form hemichannels that release ATP from the endolymphatic surface of supporting and epithelial cells of the organ of Corti
(OoC), as well as gap junction (GJ) channels that allow the concomitant intercellular diffusion of Ca21 mobilizing second
messengers. Released ATP in turn activates G-protein coupled P2Y2 and P2Y4 receptors, PLC-dependent generation of IP3,
release of Ca21 from intracellular stores, ensuing in the regenerative propagation of intercellular Ca21 signals (ICS) across
these coupled cells. The range of ICS propagation in the OoC is sensitive to the concentration of extracellular divalent
cations and to ectonucleotidase activity. Strictly related oscillations of the intracellular free Ca21 concentration ([Ca21]i) in
cochlear supporting cells are also evoked by nanomolar concentrations of ATP on the endolymphatic surface of the OoC.
ICS and [Ca21]i oscillations are of great interest in relation to the responses evoked by damaging stimuli delivered to hair
cells, and may play a crucial role in development of the OoC and the acquisition of hearing.

Key Words: cochlea, hearing, DFNB1, mouse models, development, organotypic cultures, CELAbs

Cochlear GJ networks
which mediate the transfer of ions, metabolites and
All cells providing mechanical support to cochlear second messengers between cells (6–8). The develop-
hair cells are designated as supporting cells (1). In ment of the cochlear GJ system precedes the func-
the mature OoC, these include inner phalangeal tional maturation of the murine inner ear (2), which
cells, inner and outer pillar cells, outer phalangeal takes place between the second and third postnatal
cells (also known as Deiters’ cells), as well as Hens- week (9).
en’s, Böttcher’s and Claudius’ cells. The inner pha- The connective tissue GJ network starts to develop
langeal cells completely surround the inner hair cells around birth and comprises interdental cells and
(IHCs). The outer phalangeal cells form cups hold- fibrocytes in the spiral limbus, fibrocytes of the spiral
ing the synaptic poles of the outer hair cells (OHCs) ligament, basal and intermediate cells of the SV. The
and send fine processes, or phalanges, to the reticular epithelial GJ network forms around embryonic day
lamina (RL). This stiff cytoplasmic plate is composed 16 (E16) and connects all supporting cells in the
of a mosaic of apposing phalangeal processes of outer OoC as well as adjacent epithelial cells (2). In the
pillar cells and outer phalangeal cells, both of which hearing cochlea, the epithelial GJ network apparently
seal the endolymphatic poles of the hair cells, extend- subdivides further in two separate, medial and lat-
ing from the innermost row of OHCs to the Hensen’s eral, buffering compartments, which are thought to
cells. Thus, only stereociliary bundles of OHCs be individually dedicated to the homeostasis of IHCs
emerge above the RL (1). and OHCs (10).
The OoC is part of the tubular cochlear duct In the so-called potassium recycle hypothesis (3),
epithelium in which cells are connected by a network the epithelial GJ network is presumed to intervene in
of tight junctions and adherens junctions near their the cycling of K1 following activation of the mecha-
surface facing the scala media, thus providing separa- notransduction process in hair cells (11). Indeed,
tion of the endolymph from the perilymph (2). Non- several classes of supporting cells in the sensory epi-
sensory cells in the cochlear duct are also massively thelium express glial fibrillary acidic protein (GFAP)
interconnected by intercellular GJ channels (3–5), (12), a classic marker for astrocytes, and are thought

Correspondence: F. Mammano, VIMM, Via G. Orus 2, 35129 Padova, Italy. Tel/fax: +39 049 7923 231/250. E-mail: fabio.mammano@unipd.it

ISSN 1651-386X print/ISSN 1651-3835 online © 2009 Informa UK Ltd. (Informa Healthcare, Taylor & Francis AS)
DOI: 10.3109/16513860903444311
2 F. Mammano

to perform similar spatial buffering of K1 (3,5). postnatal day 12–13, and thereafter decreased sig-
Although the widespread localization of cochlear GJ nificantly in parallel with the appearance of epithe-
channels (4), K/Cl cotransporters (13), and aqua- lial breaches that compromised the integrity of the
porins (14) could provide a passive mechanism of endolymphatic compartment. Endolymphatic K1
K1 buffering and osmotic homeostasis, it is worth concentration was also significantly lower in adult
emphasizing that currently there is no direct evidence Cx26OtogCre mice compared to controls. Likewise,
for the potassium recycle hypothesis. expression of the Cx26R75W dominant negative
mutation in mice, obtained by injection of the trans-
gene into fertilized mouse eggs (28) (also called
Connexins, deafness and mouse models pronucleus injection), resulted in deafness associ-
ated with significant histological abnormalities
Most cell−cell channels in the cochlear GJ networks
within the inner ear including degeneration, by
are composed of two types of integral transmem-
postnatal week 7, of the OHCs. In a conditional (c)
brane proteins subunits, namely Cx26 and Cx30
Cx26 null mouse model (29) (obtained by crossing
(15), which share 77% amino acid identity and may
Cx26loxP/loxP mice with R26cre-ERT mice in
assemble to form heteromeric and heterotypic chan-
which Cre is activated in the presence of the syn-
nels (16). The genes encoding Cx26 (GJB2) and
thetic oestrogen 4-hydroxytamoxifen), the earliest
Cx30 (GJB6) are found within 50 kb of each other
cell death occurred around postnatal day 14 in
in the DFNB1 complex deafness locus. DFNB1
OHCs and their surrounding supporting cells in the
mutations, which are almost as frequent as those
middle cochlear turn (CT). Hence, death rapidly
causing cystic fibrosis, account for around 50% of
spread to the basal CT so that all types of cells had
genetic cases of severe to profound non-syndromic
disappeared from the OoC of cCx26 null mice a few
hearing loss in various ethnic groups (17). For both
months after birth. Unlike Cx26OtogCre mice, in
connexins, expression is evident in several sub-com-
cCx26 null mice peripheral nerve fibres and soma
partments of the cochlear duct already one week
of SGNs at corresponding cochlear locations were
before the onset of hearing (18,19), in keeping with
completely degenerated as well, whereas hair cells
the notion that intercellular connections by GJ pro-
in the apical CT were relatively preserved (29).
teins are crucial for the maturation of different tis-
In Cx30(–/–) mice (30) (obtained by deletion of
sues (20). In particular, immunolabelling with
the Cx30 coding region) the cochleae were morpho-
selective antibodies highlighted the expression of
logically indistinguishable from those of Cx30(1/–)
connexins at point of contacts between supporting
and wt mice up to postnatal day 17. However,
and epithelial cells of the OoC on both sides of the
Cx30(–/–) mice failed to develop any measurable EP,
hair cell region (HCR), as well as in basal and inter-
and adults had significantly decreased endolymphatic
mediate cells of the stria vascularis (SV). Further-
K1 levels. Auditory thresholds worsened from 84dB
more, both Cx26 and Cx30 showed marked
at postnatal day 17–18 to more than 100dB in adult
immunoreaction in the spiral limbus and the spiral
Cx30(–/–) mice; hence, hearing loss was more severe
ligament (10,18,19,21–25).
in Cx30(–/–) mice than in Cx26OtogCre mice. Corre-
Mouse models with defective expression of Cx26
spondingly, the apoptotic process in Cx30(–/–) mice,
or Cx30 confirmed that they are essential for hear-
albeit delayed by about four days relative to Cx26O-
ing. Thus, targeted ablation of Cx26 in the epithelial togCre mice, affected ultimately both IHCs and OHCs.
GJ network of Cx26OtogCre mice (26) (obtained by
Transgenic expression of extra copies of the Cx26
crossing Cx26loxP/loxP mice with Otog-Cre mice line
gene from a modified bacterial artificial chromosome
expressing the Cre gene under the control of the
(BAC) in a Cx30(–/–) background restored cochlea
murine Otog promoter) ensued in cell death accom-
development and hearing (31).
panied by epithelial breaches shortly after the onset
of hearing (which, in mice, occurs at postnatal day
12 (27)), along with progressive and significant
Permselectivity of GJ channels
hearing loss ranging from 30dB to 70dB. The apop-
totic process affected first the two supporting cells Although mouse models confirmed that Cx26 and
that surround the IHCs and later extended to OHCs Cx30 are essential for auditory function, as well as
and supporting cells around them. IHCs were pre- survival and development of the OoC (26,28–30),
served in adult Cx26OtogCre mice, and cell death was the physiopathological mechanisms leading to deaf-
not detected at any stage either in spiral ganglion ness when connexins are absent or mutated remain
neurons (SGNs), in the fibrocytes of the spiral lim- unclear (15). Thus, deafness and lack of EP in
bus and spiral ligament or in the SV. Endochlear Cx30(–/–) mice correlate with: 1) disruption of the
potential (EP) values developed normally up to endothelial barrier of the capillaries supplying the SV
Inner ear connexins 3
before EP onset; 2) significant down-regulation of cells. Nanomolar levels of ATP on the endolym-
Bhmt; and 3) local increase in homocysteine, a known phatic surface of the OoC, which have been linked
factor of endothelial dysfunction (32), with no obvious to sound exposure (44), in turn activate G-protein
link to GJ channel function. Likewise, the K1 recycle coupled P2Y2 and P2Y4 receptors, PLC-dependent
hypothesis is challenged by the identification of Cx26 generation of IP3 and release of Ca2 from intracel-
recessive deafness mutants, e.g.V84L (33), which retain lular stores, ensuing in the regenerative propagation
partial channel function (34) and are as permeable to of ICS across the networks of cochlear supporting
K1 as the wt channels. However, the transfer of the and epithelial cells (43,45–47). The range of ICS
Ca21-mobilizing second messenger IP3 (and possibly propagation is sensitive to the concentration of
other signalling molecules) is impaired between the divalent cations in the extracellular medium and is
cells expressing these mutant proteins (35). controlled by ATP degradation (43) due to ecto-
The concept that each GJ channel is unique in nucleotidases expressed at the endolymphatic sur-
terms of permselectivity is supported by a large num- face of the OoC (48,49). By manipulating the
ber of studies (36) and suggests that permeability to bathing medium one can either reduce the rate of
larger metabolites, rather then small inorganic ions, ATP degradation, by inhibiting ectonucleotidases
plays an important role in the development, physiol- with ARL67156, or else increase this rate, e.g. by
ogy and aetiology of connexin related diseases supplementing soluble apyrase. ICS propagation is
(7,8,37). This is a crucial issue, for it has been dem- blocked altogether by membrane permeable block-
onstrated that many disease leading mutations cause ers of connexin channels, such as CBX, whereas
changes in the permeability of the channel to signal- inhibitors of the P2Y receptors, such as suramin, or
ling molecules (7). Furthermore, several lines of blockers of connexin hemichannels, such as La3,
experiments showed that the effect of single point confine ICS propagation to cells directly coupled
mutations can be subtle, discriminating between to the stimulated cells through GJ channels
molecules having the same net charge but different (35,43,45,46).
charge distribution; however, the molecular mecha- The mechanisms underlying the spreading of ICS
nisms remain unclear (7). The recent publication have been analysed in detail and shown to entail
of the structure of the Cx26 at 3.5 Å resolution release of ATP from connexin hemichannels at the
(38) will undoubtedly pave the way to the study, endolymphatic surface of the epithelium, as well as
at atomic level, of connexin channel function and diffusion of Ca21-mobilizing second messengers
dysfunction. across GJ channels (43) (Figure 1). IP3 is a key inter-
mediate that mediates ATP-dependent Ca21
responses of cochlear sensory (50) and non-sensory
Connexin hemichannels, ATP release and (35,46,51,52) cells.
cell–cell signalling
While the exact function of inner ear connexins
What could be the significance of ICS
remains unclear (15), we note that Cx26 and Cx30
exchange, and of the strictly related [Ca21]i
also form unpaired connexons (39), i.e. non-junc-
oscillations?
tional connexin hemichannels (40,41). Using anti-
bodies against Cx26 extracellular loop peptides Although precise answers are not yet available, it is
(CELAbs) (42), immunofluorescence signals cor- tempting to propose a developmental role. Thus, in
responding to unpaired connexons (i.e. connexin non-excitable cells, the dynamics of gene expression
hemichannels) were detected in the plasma mem- have been shown to depend on [Ca2]i changes
brane of cells facing the endolymphatic surface of through NFκB signalling (53–55). Ortolano et al.
the OoC, except the hair cells, from inner sulcus (56) showed that impaired propagation of ICS,
(IS) cells across the RL to outer sulcus (OS) cells evoked by focal UV photorelease of IP3 in this imma-
(43). Using organotypic cultures of the cochlea ture OoC is associated with Cx26 down-regulation
from mice with defective expression of pannexin 1 at both protein and mRNA levels in Cx30(–/–) cul-
(Px1), P2X7 receptors, Cx30 or Cx26, Anselmi et tures. Conversely, ablation of Cx26 caused concur-
al. (43) further demonstrated that, in response to rent down-regulation of Cx30 and impaired ICS
activation of a P2Y/PLC/IP3/Ca2 signalling cas- propagation. Molecular characterization of the
cade, hemichannels formed by these connexins coregulation mechanisms implicates IP3, intracellular
release ATP from the endolymphatic surface of Ca2 and NFκB, a Ca2-dependent transcription
cochlear supporting and epithelial cells, whereas GJ factor, as important players. Interestingly, a binding
channels allow the concomitant diffusion of Ca2 site for NFκB has been identified in the promoter
mobilizing second messengers across these coupled region of GJB2 (57,58), and NFκB is reported to
4 F. Mammano
mono for print colour online

Figure 1. Schematic representation of ICS propagation across the network of GJ coupled cochlear supporting and epithelial cells. CBX,
carbenoxolone, is a non-specific inhibitor connexin hemichannels and GJ channels (74); ARL67156, 6-N,N-diethyl-D-β-y-dibromomethylene
adenosine triphosphate, originally named FPL 67156, is a commercially available inhibitor of ecto-ATPases (75). Apyrase is an enzyme
that catalyses the hydrolysis of ATP to yield AMP and inorganic phosphate (76,77). La31 is a blocker of connexin hemichannels (40) that
does not affect GJ channels when applied extracellularly to cochlear cultures (43). P2YR, G-protein coupled P2Y receptor; IP3R, receptor
for inositol 1,4,5-trisphosphate (also commonly known as triphosphoinositol, abbreviated InsP3 or IP3).

regulate the expression of at least one other connexin of OHC afferent signalling remains obscure (72),
(connexin 43, reference (59)). These data (56) also the endolymphatic surface of the OHC is a primary
offer crucial insight into the observations that some region for purinergic influence, where ATP activates
deafness associated DFNB1 alleles are characterized Ca2 signalling through the combined action of P2X
by hereditable significant reduction of both GJB2 and P2Y receptors (50,66).
and GJB6 (60) and that GJB6 deletions in trans with ICS and [Ca21]i oscillations of are of great inter-
recessive mutations of GJB2 cause deafness in est also in relation to the responses evoked by damag-
humans (61–63) (see also reference (64)). ing stimuli delivered to hair cells (45,73). Although
Furthermore, it is known that the acquisition the picture is far from complete, what is beginning
and maturation of mechanoelectrical transduction to emerge is an intriguing set of interactions between
in hair cells occurs in a gradient from the base to cochlear sensory hair cells and their supporting cells,
the apex of the cochlea, and that IHCs differentiate with ATP playing a key signalling role.
prior to OHCs (65). P2X2 and transiently expressed
P2X3 ionotropic channels on the endolymphatic
surface of IHCs activate in response to extracellular Acknowledgements
ATP (66,67), which is released from supporting
This work has been funded by grants to FM from
cells into the endolymph through hemichannels
Fondazione Cariparo (Progetti di Eccellenza 2006),
formed by Cx26 and Cx30 (43), causing nearby
Telethon Italy, Italian Ministry of Research and the
IHCs to fire bursts of Ca2 action potentials in the
European commission FP6 Integrated Project Euro-
developing OoC (68). This firing activity promotes
Hear under the Sixth Research Frame Program of
the release of the afferent neurotransmitter gluta-
The European Union.
mate at the synaptic pole of the IHC (69,70), which
in turn triggers discrete bursts of Ca2 action poten-
tials in primary auditory neurons (71). Further- Declaration of interest: The authors report no
more, although immature OHCs are not known to conflicts of interest. The authors alone are respon-
fire spontaneous action potentials (66) and the role sible for the content and writing of the paper.
Inner ear connexins 5
References 22. Lautermann J, ten Cate WJ, Altenhoff P, Grummer R, Traub
O, Frank H, et al. Expression of the gap junction connexins 26
1. Krstic RV. Human Microscopic Anatomy. Berlin:Springer- and 30 in the rat cochlea. Cell Tissue Res. 1998;294:415–20.
Verlag; 1997. 23. Marziano NK, Casalotti SO, Portelli AE, Becker DL, Forge
2. Cohen-Salmon M, del Castillo FJ, Petit C. Connexins A. Mutations in the gene for connexin 26 (GJB2) that cause
responsible for hereditary deafness: the tale unfolds. In: Win- hearing loss have a dominant negative effect on connexin 30.
terhager E, editor. Gap Junctions in Development and Dis- Hum Mol Genet. 2003;12;805–12.
ease. Berlin: Springer-Verlag; 2005. p. 111–34. 24. Ahmad S, Chen S, Sun J, Lin X. Connexins 26 and 30 are
3. Kikuchi T, Adams JC, Miyabe Y, So E, Kobayashi T. Potas- coassembled to form gap junctions in the cochlea of mice.
sium ion recycling pathway via gap junction systems in the Biochem Biophys Res Commun. 2003;307: 362–8.
mammalian cochlea and its interruption in hereditary non- 25. Zhao HB, Yu N. Distinct and gradient distributions of con-
syndromic deafness. Med Electron Microsc. 2000;33:51–6. nexin 26 and connexin 30 in the cochlear sensory epithelium
4. Forge A, Becker D, Casalotti S, Edwards J, Marziano N, Nevill of guinea pigs. J Comp Neurol. 2006;499:506–18.
G. Gap junctions in the inner ear: comparison of distribution 26. Cohen-Salmon M, Ott T, Michel V, Hardelin JP, Perfettini I,
patterns in different vertebrates and assessement of connexin Eybalin M, et al. Targeted ablation of connexin 26 in the inner
composition in mammals. J Comp Neurol. 2003;467:207–31. ear epithelial gap junction network causes hearing impair-
5. Zhao HB, Kikuchi T, Ngezahayo A, White TW. Gap junctions ment and cell death. Curr Biol. 2002;12:1106–11.
and cochlear homeostasis. J Membr Biol. 2006;209:177–86. 27. Mikaelian D, Alford BR, Ruben RJ. Cochlear potentials and
6. Kumar NM, Gilula NB. The gap junction communication VIIIth nerve action potentials in normal and genetically deaf
channel. Cell. 1996;84: 381–8. mice. Ann Otol Rhinol Laryngol. 1965;74:146–57.
7. Harris AL. Connexin channel permeability to cytoplasmic 28. Kudo T, Kure S, Ikeda K, Xia AP, Katori Y, Suzuki M, et al.
molecules. Prog Biophys Mol Biol. 2007;94:120–43. Transgenic expression of a dominant-negative connexin 26
8. Hernandez VH, Bortolozzi M, Pertegato V, Beltramello M, causes degeneration of the organ of Corti and non-syndromic
Giarin M, Zaccolo M, et al. Unitary permeability of gap deafness. Hum Mol Genet. 2003;12:995–1004.
junction channels to second messengers measured by FRET 29. Sun Y, Tang W, Chang Q, Wang Y, Kong W, Lin X. Connexin
microscopy. Nat Methods. 2007;4:353–8. 30 null and conditional connexin 26 null mice display distinct
9. Eatock RA, Hurley KM. Functional development of hair pattern and time-course of cellular degeneration in the coch-
cells. Curr Top Dev Biol. 2003;57:389–448. lea. J Comp Neurol. 2009;516:569–79.
10. Jagger DJ, Forge A. Compartmentalized and signal-selective 30. Teubner B, Michel V, Pesch J, Lautermann J, Cohen-Salmon
gap junctional coupling in the hearing cochlea. J Neurosci. M, Sohl G, et al. Connexin 30 (GJB6) deficiency causes
2006;26:1260–8. severe hearing impairment and lack of endocochlear poten-
11. Fettiplace R, Ricci AJ. Mechanoelectrical transduction in tial. Hum Mol Genet. 2003;12:13–21.
auditory hair cells. In: Eatock RA, Fay RR, Popper AN, edi- 31. Ahmad S, Tang W, Chang Q, Qu Y, Hibshman J, Li Y, et al. Res-
tors. Vertebrate Hair Cells. New York: Springer Science Inc.; toration of connexin 26 protein level in the cochlea completely
2006. Vol 27: p. 154–203. rescues hearing in a mouse model of human connexin 30-linked
12. Rio C, Dikkes P, Liberman MC, Corfas G. Glial fibrillary deafness. Proc Natl Acad Sci U S A.2007;104:1337–41.
acidic protein expression and promoter activity in the inner 32. Cohen-Salmon M, Regnault B, Cayet N, Caille D, Demuth
ear of developing and adult mice. J Comp Neurol. K, Hardelin JP, et al. Connexin 30 deficiency causes instras-
2002;442:156–62. trial fluid-blood barrier disruption within the cochlear stria
13. Boettger T, Hubner CA, Maier H, Rust MB, Beck FX, Jent- vascularis. Proc Natl Acad Sci U S A. 2007;104:6229–34.
sch TJ. Deafness and renal tubular acidosis in mice lacking 33. Kelley PM, Harris DJ, Comer BC, Askew JW, Fowler T, Smith
the K-Cl cotransporter Kcc4. Nature. 2002; 416:874–8. SD, et al. Novel mutations in the connexin 26 gene (GJB2)
14. Huang D, Chen P, Chen S, Nagura M, Lim DJ, Lin X. that cause autosomal recessive (DFNB1) hearing loss. Am J
Expression patterns of aquaporins in the inner ear: evidence Hum Genet. 1998;62:792–9.
for concerted actions of multiple types of aquaporins to facil- 34. Bruzzone R, Veronesi V, Gomes D, Bicego M, Duval N, Mar-
itate water transport in the cochlea. Hear Res. lin S, et al. Loss of function and residual channel activity of
2002;165:85–95. connexin 26 mutations associated with non-syndromic deaf-
15. Nickel R, Forge A. Gap junctions and connexins in the inner ness. FEBS Lett. 2003;533:79–88.
ear: their roles in homeostasis and deafness. Curr Opin 35. Beltramello M, Piazza V, Bukauskas FF, Pozzan T, Mammano
Otolaryngol Head Neck Surg. 2008;16:452–7. F. Impaired permeability to Ins(1,4,5)P3 in a mutant con-
16. Yum SW, Zhang J, Valiunas V, Kanaporis G, Brink PR, White nexin underlies recessive hereditary deafness. Nat Cell Biol.
TW, et al. Human connexin 26 and connexin 30 form func- 2005;7:63–9.
tional heteromeric and heterotypic channels. Am J Physiol 36. Mese G, Richard G, White TW. Gap junctions: basic structure
Cell Physiol. 2007;293:1032–48. and function. J Invest Dermatol. 2007;127:2516–24.
17. Sabag AD, Dagan O, Avraham KB. Connexins in hearing loss: 37. Nicholson SM, Bruzzone R. Gap junctions: getting the mes-
a comprehensive overview. J Basic Clin Physiol Pharmacol. sage through. Curr Biol. 1997;7:R340–4.
2005;16:101–16. 38. Maeda S, Nakagawa S, Suga M, Yamashita E, Oshima A,
18. Lautermann J, Frank HG, Jahnke K, Traub O, Winterhager Fujiyoshi Y, et al. Structure of the connexin 26 gap junction
E. Developmental expression patterns of connexin 26 and 30 channel at 3.5 A resolution. Nature. 2009;458:597–602.
in the rat cochlea. Dev Genet. 1999; 25:306–11. 39. Zhao HB, Yu N, Fleming CR. Gap junctional hemichannel-
19. Xia A, Katori Y, Oshima T, Watanabe K, Kikuchi T, Ikeda K. mediated ATP release and hearing controls in the inner ear.
Expression of connexin 30 in the developing mouse cochlea. Proc Natl Acad Sci U S A. 2005;102:18724–9.
Brain Res. 2001;898:364–7. 40. Bennett MV, Contreras JE, Bukauskas FF, Saez JC. New
20. Levin M. Gap junctional communication in morphogenesis. roles for astrocytes: gap junction hemichannels have some-
Prog Biophys Mol Biol. 2007;94:186–206. thing to communicate. Trends Neurosci. 2003;26:610–7.
21. Kikuchi T, Kimura RS, Paul DL, Adams JC. Gap junctions 41. Goodenough DA, Paul DL. Beyond the gap: functions of
in the rat cochlea: immunohistochemical and ultrastructural unpaired connexon channels. Nat Rev Mol Cell Biol.
analysis. Anat Embryol (Berl). 1995; 191:101–18. 2003;4:285–94.
6 F. Mammano
42. Clair C, Combettes L, Pierre F, Sansonetti P, Tran Van Nhieu G. and function in astrocytes by a mechanism involving the NF-
Extracellular-loop peptide antibodies reveal a predominant kappaB and PI3 kinase pathways. Glia. 2006;54:775–85.
hemichannel organization of connexins in polarized intestinal 60. Wilch E, Zhu M, Burkhart KB, Regier M, Elfenbein JL,
cells. Exp Cell Res. 2008;314:1250–65. Fisher RA, et al. Expression of GJB2 and GJB6 is reduced in
43. Anselmi F, Hernandez VH, Crispino G, Seydel A, Ortolano S, a novel DFNB1 allele. Am J Hum Genet. 2006;79:174–9.
Roper SD, et al. ATP release through connexin hemichannels 61. del Castillo FJ, Rodriguez-Ballesteros M, Alvarez A, Hutchin T,
and gap junction transfer of second messengers propagate Leonardi E, de Oliveira CA, et al. A novel deletion involving
Ca21 signals across the inner ear. Proc Natl Acad Sci U S A. the connexin 30 gene, del(GJB6-d13s1854), found in trans
2008;105:18770–5. with mutations in the GJB2 gene (connexin 26) in subjects
44. Munoz DJ, Kendrick IS, Rassam M, Thorne PR. Vesicular with DFNB1 non-syndromic hearing impairment. J Med
storage of adenosine triphosphate in the guinea pig cochlear Genet. 2005;42:588–94.
lateral wall and concentrations of ATP in the endolymph dur- 62. del Castillo I, Moreno-Pelayo MA, del Castillo FJ, Brown-
ing sound exposure and hypoxia. Acta Otolaryngol. stein Z, Marlin S, Adina Q, et al. Prevalence and evolutionary
2001;121:10–5. origins of the del(GJB6-D13S1830) mutation in the DFNB1
45. Gale JE, Piazza V, Ciubotaru CD, Mammano F. A mechanism locus in hearing impaired subjects: a multicentre study. Am J
for sensing noise damage in the inner ear. Curr Biol. Hum Genet. 2003;73:1452–8.
2004;14:526–9. 63. del Castillo I, Villamar M, Moreno-Pelayo MA, del Castillo
46. Piazza V, Ciubotaru CD, Gale JE, Mammano F. Purinergic FJ, Alvarez A, Telleria D, et al. A deletion involving the con-
signalling and intercellular Ca21 wave propagation in the nexin 30 gene in non-syndromic hearing impairment. N Engl
organ of Corti. Cell Calcium. 2007;41:77–86. J Med. 2002;346:243–9.
47. Mann ZF, Duchen MR, Gale JE. Mitochondria modulate the 64. Rodriguez-Paris J, Schrijver I. The digenic hypothesis unrav-
spatio-temporal properties of intra- and inter-cellular Ca21 elled: the GJB6 del(GJB6-D13S1830) mutation causes allele-
signals in cochlear supporting cells. Cell Calcium. specific loss of GJB2 expression in cis. Biochem Biophys Res
2009;46:136–46. Commun. 2009;389:354–9.
48. Housley GD, Jagger DJ, Greenwood D, Raybould NP, Salih 65. Kelly MC, Chen P. Development of form and function in
SG, Jarlebark LE, et al. Purinergic regulation of sound trans- the mammalian cochlea. Curr Opin Neurobiol. 2009;19:
duction and auditory neurotransmission. Audiol Neurootol. 395–401.
2002;7:55–61. 66. Housley GD, Marcotti W, Navaratnam D, Yamoah EN.
49. Vlajkovic SM, Housley GD, Munoz DJ, Robson SC, Sevigny J, Hair cells: beyond the transducer. J Membr Biol. 2006;209:
Wang CJ, et al. Noise exposure induces up-regulation of ecto- 89–118.
nucleoside triphosphate diphosphohydrolases 1 and 2 in rat 67. Mammano F, Bortolozzi M, Ortolano S, Anselmi F. Ca21
cochlea. Neuroscience. 2004;126:763–73. signalling in the inner ear. Physiology (Bethesda).
50. Mammano F, Frolenkov GI, Lagostena L, Belyantseva IA, 2007;22:131–44.
Kurc M, Dodane V, et al. ATP-induced Ca21 release in coch- 68. Tritsch NX, Yi E, Gale JE, Glowatzki E, Bergles DE. The
lear outer hair cells: localization of an inositol triphosphate- origin of spontaneous activity in the developing auditory sys-
gated Ca21 store to the base of the sensory hair bundle. J tem. Nature. 2007;450:50–5.
Neurosci. 1999;19:6918–29. 69. Glowatzki E, Grant L, Fuchs P. Hair cell afferent synapses.
51. Lagostena L, Mammano F. Intracellular calcium dynamics Curr Opin Neurobiol. 2008;18:389–95.
and membrane conductance changes evoked by Deiters’ cell 70. Meyer AC, Frank T, Khimich D, Hoch G, Riedel D, Chapoch-
purinoceptor activation in the organ of Corti. Cell Calcium. nikov NM, et al. Tuning of synapse number, structure and
2001;29:191–8. function in the cochlea. Nat Neurosci. 2009;12:444–53.
52. Lagostena L, Ashmore JF, Kachar B, Mammano F. Purinergic 71. Goutman JD, Glowatzki E. Time-course and calcium depend-
control of intercellular communication between Hensen’s cells ence of transmitter release at a single ribbon synapse. Proc
of the guinea pig cochlea. J Physiol. 2001;531:693–706. Natl Acad Sci U S A. 2007;104:16341–6.
53. Dolmetsch RE, Xu K, Lewis RS. Calcium oscillations increase 72. Knirsch M, Brandt N, Braig C, Kuhn S, Hirt B, Munkner S,
the efficiency and specificity of gene expression. Nature. et al. Persistence of Ca(v)1.3 Ca21 channels in mature outer
1998;392:933–6. hair cells support outer hair cell afferent signalling. J Neuro-
54. Hu Q, Deshpande S, Irani K, Ziegelstein RC. [Ca21](i) oscil- sci. 2007;27:6442–51.
lation frequency regulates agonist-stimulated NF-kappaB 73. Lahne M, Gale JE. Damage induced activation of ERK1/2 in
transcriptional activity. J Biol Chem. 1999;274:33995–8. cochlear supporting cells is a hair cell death-promoting signal
55. Nelson DE, Ihekwaba AE, Elliott M, Johnson JR, Gibney CA, that depends on extracellular ATP and calcium. J Neurosci.
Foreman BE, et al. Oscillations in NF-kappaB signalling con- 2008;28:4918–28.
trol the dynamics of gene expression. Science. 2004;306:704– 74. Rozental R, Miduturu S, Spray DC. How to close a gap junc-
8. tion channel. In: Bruzzone R, Giaume C, editors. Connexin
56. Ortolano S, di Pasquale G, Crispino G, Anselmi F, Mam- Methods and Protocols. Totowa, NJ: Humana Press; 2001.
mano F, Chiorini JA. Coordinated control of connexin 26 p. 447–76.
and connexin 30 at the regulatory and functional level in the 75. Levesque SA, Lavoie EG, Lecka J, Bigonnesse F, Sevigny J.
inner ear. Proc Natl Acad Sci U S A. 2008;105:18776–81. Specificity of the ecto-ATPase inhibitor ARL 67156 on
57. Hennemann H, Kozjek G, Dahl E, Nicholson B, Willecke K. human and mouse ectonucleotidases. Br J Pharmacol.
Molecular cloning of mouse connexins 26 and 32: similar 2007;152:141–50.
genomic organization but distinct promoter sequences of two 76. Komoszynski MA. Comparative studies on animal and plant
gap junction genes. Eur J Cell Biol. 1992;58:81–9. apyrases (ATP diphosphohydrolase EC 3.6.1.5) with applica-
58. Tu ZJ, Kiang DT. Mapping and characterization of the basal tion of immunological techniques and various ATPase inhib-
promoter of the human connexin 26 gene. Biochim Biophys itors. Comp Biochem Physiol B Biochem Mol Biol.
Acta. 1998;1443:169–81. 1996;113:581–91.
59. Zhao Y, Rivieccio MA, Lutz S, Scemes E, Brosnan CF. The 77. Molnar J, Lorand L. Studies on apyrases. Arch Biochem
TLR3 ligand polyI: C down-regulates connexin 43 expression Biophys. 1961;93:353–63.

You might also like