Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
2.
Pharmacol Ther ; 255: 108589, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38295906

ABSTRACT

The original paradigm of classical - also referred to as canonical - cellular signal transduction of heterotrimeric G proteins (G protein) is defined by a hierarchical, orthograde interaction of three players: the agonist-activated G protein-coupled receptor (GPCR), which activates the transducing G protein, that in turn regulates its intracellular effectors. This receptor-transducer-effector concept was extended by the identification of regulators and adapters such as the regulators of G protein signaling (RGS), receptor kinases like ßARK, or GPCR-interacting arrestin adapters that are integrated into this canonical signaling process at different levels to enable fine-tuning. Finally, the identification of atypical signaling mechanisms of classical regulators, together with the discovery of novel modulators, added a new and fascinating dimension to the cellular G protein signal transduction. This heterogeneous group of accessory G protein modulators was coined "activators of G protein signaling" (AGS) proteins and plays distinct roles in canonical and non-canonical G protein signaling pathways. AGS proteins contribute to the control of essential cellular functions such as cell development and division, intracellular transport processes, secretion, autophagy or cell movements. As such, they are involved in numerous biological processes that are crucial for diseases, like diabetes mellitus, cancer, and stroke, which represent major health burdens. Although the identification of a large number of non-canonical G protein signaling pathways has broadened the spectrum of this cellular communication system, their underlying mechanisms, functions, and biological effects are poorly understood. In this review, we highlight and discuss atypical G protein-dependent signaling mechanisms with a focus on inhibitory G proteins (Gi) involved in canonical and non-canonical signal transduction, review recent developments and open questions, address the potential of new approaches for targeted pharmacological interventions.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Signal Transduction , Humans , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism
3.
Mol Cell Proteomics ; 23(2): 100704, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38128648

ABSTRACT

In the ear, inner hair cells (IHCs) employ sophisticated glutamatergic ribbon synapses with afferent neurons to transmit auditory information to the brain. The presynaptic machinery responsible for neurotransmitter release in IHC synapses includes proteins such as the multi-C2-domain protein otoferlin and the vesicular glutamate transporter 3 (VGluT3). Yet, much of this likely unique molecular machinery remains to be deciphered. The scarcity of material has so far hampered biochemical studies which require large amounts of purified samples. We developed a subcellular fractionation workflow combined with immunoisolation of VGluT3-containing membrane vesicles, allowing for the enrichment of glutamatergic organelles that are likely dominated by synaptic vesicles (SVs) of IHCs. We have characterized their protein composition in mice before and after hearing onset using mass spectrometry and confocal imaging and provide a fully annotated proteome with hitherto unidentified proteins. Despite the prevalence of IHC marker proteins across IHC maturation, the profiles of trafficking proteins differed markedly before and after hearing onset. Among the proteins enriched after hearing onset were VAMP-7, syntaxin-7, syntaxin-8, syntaxin-12/13, SCAMP1, V-ATPase, SV2, and PKCα. Our study provides an inventory of the machinery associated with synaptic vesicle-mediated trafficking and presynaptic activity at IHC ribbon synapses and serves as a foundation for future functional studies.


Subject(s)
Hair Cells, Auditory, Inner , Proteomics , Mice , Animals , Hair Cells, Auditory, Inner/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Qa-SNARE Proteins/metabolism , Membrane Proteins/metabolism
4.
Front Cell Neurosci ; 15: 677543, 2021.
Article in English | MEDLINE | ID: mdl-34335185

ABSTRACT

Deficiency of otoferlin causes profound prelingual deafness in humans and animal models. Here, we closely analyzed developmental deficits and degenerative mechanisms in Otof knock-out (Otof -/-) mice over the course of 48 weeks. We found otoferlin to be required for proper synapse development in the immature rodent cochlea: In absence of otoferlin, synaptic pruning was delayed, and postsynaptic boutons appeared enlarged at 2 weeks of age. At postnatal day 14 (P14), we found on average ∼15 synapses per inner hair cell (IHC) in Otof -/- cochleae as well as in wild-type controls. Further on, the number of synapses in Otof -/- IHCs was reduced to ∼7 at 8 weeks of age and to ∼6 at 48 weeks of age. In the same period, the number of spiral ganglion neurons (SGNs) declined in Otof -/- animals. Importantly, we found an age-progressive loss of IHCs to an overall number of 75% of wildtype IHCs. The IHC loss more prominently but not exclusively affected the basal aspects of the cochlea. For outer hair cells (OHCs), we observed slightly accelerated age-dependent degeneration from base to apex. This was associated with a progressive decay in DPOAE amplitudes for high frequency stimuli, which could first be observed at the age of 24 weeks in Otof -/- mice. Our data will help to plan and predict the outcome of a gene therapy applied at various ages of DFNB9 patients.

5.
Sci Rep ; 11(1): 14271, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253772

ABSTRACT

Inhibitory G proteins (Gi proteins) are highly homologous but play distinct biological roles. However, their isoform-specific detection remains challenging. To facilitate the analysis of Gαi3 expression, we generated a Gnai3- iresGFP reporter mouse line. An internal ribosomal entry site (IRES) was inserted behind the stop-codon of the Gnai3 gene to initiate simultaneous translation of the GFP cDNA together with Gαi3. The expression of GFP was confirmed in spleen and thymus tissue by immunoblot analysis. Importantly, the GFP knock-in (ki) did not alter Gαi3 expression levels in all organs tested including spleen and thymus compared to wild-type littermates. Flow cytometry of thymocytes, splenic and blood cell suspensions revealed significantly higher GFP fluorescence intensities in homozygous ki/ki animals compared to heterozygous mice (+/ki). Using cell-type specific surface markers GFP fluorescence was assigned to B cells, T cells, macrophages and granulocytes from both splenic and blood cells and additionally blood-derived platelets. Moreover, immunofluorescent staining of the inner ear from knock-in mice unraveled GFP expression in sensory and non-sensory cell types, with highest levels in Deiter's cells and in the first row of Hensen's cells in the organ of Corti, indicating a novel site for Gαi3 expression. In summary, the Gnai3- iresGFP reporter mouse represents an ideal tool for precise analyses of Gαi3 expression patterns and sites.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/biosynthesis , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genes, Reporter , Green Fluorescent Proteins/metabolism , Animals , Flow Cytometry , Gene Expression Profiling , Genotype , Heterozygote , Mice , Mice, Inbred C57BL , Spleen/metabolism , Thymus Gland/metabolism
6.
Genes (Basel) ; 11(12)2020 11 26.
Article in English | MEDLINE | ID: mdl-33256196

ABSTRACT

The OTOF gene encodes otoferlin, a critical protein at the synapse of auditory sensory cells, the inner hair cells (IHCs). In the absence of otoferlin, signal transmission of IHCs fails due to impaired release of synaptic vesicles at the IHC synapse. Biallelic pathogenic and likely pathogenic variants in OTOF predominantly cause autosomal recessive profound prelingual deafness, DFNB9. Due to the isolated defect of synaptic transmission and initially preserved otoacoustic emissions (OAEs), the clinical characteristics have been termed "auditory synaptopathy". We review the broad phenotypic spectrum reported in patients with variants in OTOF that includes milder hearing loss, as well as progressive and temperature-sensitive hearing loss. We highlight several challenges that must be addressed for rapid clinical and genetic diagnosis. Importantly, we call for changes in newborn hearing screening protocols, since OAE tests fail to diagnose deafness in this case. Continued research appears to be needed to complete otoferlin isoform expression characterization to enhance genetic diagnostics. This timely review is meant to sensitize the field to clinical characteristics of DFNB9 and current limitations in preparation for clinical trials for OTOF gene therapies that are projected to start in 2021.


Subject(s)
Genetic Variation/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss/genetics , Membrane Proteins/genetics , Animals , Humans
7.
Hear Res ; 394: 107857, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31810595

ABSTRACT

Adeno-associated viruses (AAVs) are preferred vectors for gene replacement therapy, as they are non-pathogenic, non-inflammatory, induce stable transgene expression in terminally differentiated cells, and a series of natural and engineered capsid proteins can be employed to target the vectors to specific cells. Only one feature of AAVs is limiting: the low cargo capacity for foreign DNA, restricting their application to coding sequences of <4 kb. In the last decade, splitting larger cDNAs into two AAVs and co-transducing tissue with such dual-AAV vectors has shown to result in the expression of the full-length protein in different tissues like retina, muscle and liver. This is due to the intrinsic capability of the AAV genomes to undergo homologous recombination and/or head-to-tail multimerization in nuclei of target cells. Recently, two groups independently found that a dual-AAV approach successfully delivered the 6 kb full-length otoferlin cDNA into inner hair cells of otoferlin knock-out mice and restored hearing. These pioneering studies pave the way for gene therapeutics that use dual-AAV vectors to restore hearing in forms of deafness caused by mutations in large genes.


Subject(s)
Ear, Inner , Animals , Dependovirus/genetics , Genetic Therapy , Genetic Vectors , Membrane Proteins , Transgenes
8.
EMBO Mol Med ; 11(1)2019 01.
Article in English | MEDLINE | ID: mdl-30509897

ABSTRACT

Normal hearing and synaptic transmission at afferent auditory inner hair cell (IHC) synapses require otoferlin. Deafness DFNB9, caused by mutations in the OTOF gene encoding otoferlin, might be treated by transferring wild-type otoferlin cDNA into IHCs, which is difficult due to the large size of this transgene. In this study, we generated two adeno-associated viruses (AAVs), each containing half of the otoferlin cDNA Co-injecting these dual-AAV2/6 half-vectors into the cochleae of 6- to 7-day-old otoferlin knock-out (Otof-/-) mice led to the expression of full-length otoferlin in up to 50% of IHCs. In the cochlea, otoferlin was selectively expressed in auditory hair cells. Dual-AAV transduction of Otof-/- IHCs fully restored fast exocytosis, while otoferlin-dependent vesicle replenishment reached 35-50% of wild-type levels. The loss of 40% of synaptic ribbons in these IHCs could not be prevented, indicating a role of otoferlin in early synapse maturation. Acoustic clicks evoked auditory brainstem responses with thresholds of 40-60 dB. Therefore, we propose that gene delivery mediated by dual-AAV vectors might be suitable to treat deafness forms caused by mutations in large genes such as OTOF.


Subject(s)
Deafness/pathology , Deafness/therapy , Exocytosis , Genetic Therapy/methods , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Membrane Proteins/deficiency , Animals , Dependovirus/genetics , Genetic Vectors , Mice, Knockout , Transduction, Genetic , Treatment Outcome
9.
Pediatrics ; 142(1)2018 07.
Article in English | MEDLINE | ID: mdl-29875180

ABSTRACT

BACKGROUND AND OBJECTIVES: Timely provision of developmental services can improve outcomes for children 0 to 3 years old with developmental delays. Early Intervention (EI) provides free developmental services to children under age 3 years; however, data suggests that many children referred to EI never connect to the program. We sought to ensure that 70% of patients referred to EI from an academic primary care clinic serving a low-income population were evaluated within 120 days of referral. METHODS: Recognizing that our baseline system of EI referrals had multiple routes to referral without an ability to track referral outcome, we implemented a multifaceted referral process with (1) a centralized electronic referral system used by providers, (2) patient navigators responsible for processing all EI referrals submitted by providers, and (3) a tracking system postreferral to facilitate identification of patients failing to connect with EI. RESULTS: The percentage of patients evaluated by EI within 120 days increased from a baseline median of 50% to a median of 72% after implementation of the systems (N = 309). After implementation, the centralized referral system was used a median of 90% of the time. Tracking of referral outcomes revealed decreases in families refusing evaluations and improvements in exchange of information with EI. CONCLUSIONS: Rates of connection to EI improved substantially when referrals were centralized in the clinic and patient navigators were responsible for tracking referral outcomes. Knowledge of EI intake processes and relationships between the practice and the EI site are essential to ensure successful connections.


Subject(s)
Developmental Disabilities/rehabilitation , Early Intervention, Educational/statistics & numerical data , Quality Improvement/statistics & numerical data , Referral and Consultation/statistics & numerical data , Child, Preschool , Early Intervention, Educational/methods , Female , Humans , Infant , Male , Time Factors
10.
Article in English | MEDLINE | ID: mdl-29046633

ABSTRACT

Otoferlin is essential for fast Ca2+-triggered transmitter release from auditory inner hair cells (IHCs), playing key roles in synaptic vesicle release, replenishment and retrieval. Dysfunction of otoferlin results in profound prelingual deafness. Despite its crucial role in cochlear synaptic processes, mechanisms regulating otoferlin activity have not been studied to date. Here, we identified Ca2+/calmodulin-dependent serine/threonine kinase II delta (CaMKIIδ) as an otoferlin binding partner by pull-downs from chicken utricles and reassured interaction by a co-immunoprecipitation with heterologously expressed proteins in HEK cells. We confirmed the expression of CaMKIIδ in rodent IHCs by immunohistochemistry and real-time PCR. A proximity ligation assay indicates close proximity of the two proteins in rat IHCs, suggesting that otoferlin and CaMKIIδ also interact in mammalian IHCs. In vitro phosphorylation of otoferlin by CaMKIIδ revealed ten phosphorylation sites, five of which are located within C2-domains. Exchange of serines/threonines at phosphorylated sites into phosphomimetic aspartates reduces the Ca2+ affinity of the recombinant C2F domain 10-fold, and increases the Ca2+ affinity of the C2C domain. Concordantly, we show that phosphorylation of otoferlin and/or its interaction partners are enhanced upon hair cell depolarization and blocked by pharmacological CaMKII inhibition. We therefore propose that otoferlin activity is regulated by CaMKIIδ in IHCs.

11.
EMBO J ; 35(23): 2519-2535, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27729456

ABSTRACT

The multi-C2 domain protein otoferlin is required for hearing and mutated in human deafness. Some OTOF mutations cause a mild elevation of auditory thresholds but strong impairment of speech perception. At elevated body temperature, hearing is lost. Mice homozygous for one of these mutations, OtofI515T/I515T, exhibit a moderate hearing impairment involving enhanced adaptation to continuous or repetitive sound stimulation. In OtofI515T/I515T inner hair cells (IHCs), otoferlin levels are diminished by 65%, and synaptic vesicles are enlarged. Exocytosis during prolonged stimulation is strongly reduced. This indicates that otoferlin is critical for the reformation of properly sized and fusion-competent synaptic vesicles. Moreover, we found sustained exocytosis and sound encoding to scale with the amount of otoferlin at the plasma membrane. We identified a 20 amino acid motif including an RXR motif, presumably present in human but not in mouse otoferlin, which reduces the plasma membrane abundance of Ile515Thr-otoferlin. Together, this likely explains the auditory synaptopathy at normal temperature and the temperature-sensitive deafness in humans carrying the Ile515Thr mutation.


Subject(s)
Auditory Fatigue , Hair Cells, Auditory/physiology , Membrane Proteins/metabolism , Mutant Proteins/genetics , Mutation, Missense , Protein Stability/radiation effects , Synapses/metabolism , Animals , Exocytosis , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mutant Proteins/chemistry , Temperature
12.
EMBO J ; 34(21): 2686-702, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26446278

ABSTRACT

Active zones (AZs) of inner hair cells (IHCs) indefatigably release hundreds of vesicles per second, requiring each release site to reload vesicles at tens per second. Here, we report that the endocytic adaptor protein 2µ (AP-2µ) is required for release site replenishment and hearing. We show that hair cell-specific disruption of AP-2µ slows IHC exocytosis immediately after fusion of the readily releasable pool of vesicles, despite normal abundance of membrane-proximal vesicles and intact endocytic membrane retrieval. Sound-driven postsynaptic spiking was reduced in a use-dependent manner, and the altered interspike interval statistics suggested a slowed reloading of release sites. Sustained strong stimulation led to accumulation of endosome-like vacuoles, fewer clathrin-coated endocytic intermediates, and vesicle depletion of the membrane-distal synaptic ribbon in AP-2µ-deficient IHCs, indicating a further role of AP-2µ in clathrin-dependent vesicle reformation on a timescale of many seconds. Finally, we show that AP-2 sorts its IHC-cargo otoferlin. We propose that binding of AP-2 to otoferlin facilitates replenishment of release sites, for example, via speeding AZ clearance of exocytosed material, in addition to a role of AP-2 in synaptic vesicle reformation.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Hair Cells, Auditory/physiology , Synaptic Vesicles/metabolism , Action Potentials , Animals , Evoked Potentials, Auditory, Brain Stem , Hearing , Mice, Inbred C57BL , Mice, Transgenic , Synapses/physiology , Synaptic Transmission
13.
J Cell Sci ; 128(4): 638-44, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25609709

ABSTRACT

Ribbon synapses of cochlear inner hair cells (IHCs) employ efficient vesicle replenishment to indefatigably encode sound. In neurons, neuroendocrine and immune cells, vesicle replenishment depends on proteins of the mammalian uncoordinated 13 (Munc13, also known as Unc13) and Ca(2+)-dependent activator proteins for secretion (CAPS) families, which prime vesicles for exocytosis. Here, we tested whether Munc13 and CAPS proteins also regulate exocytosis in mouse IHCs by combining immunohistochemistry with auditory systems physiology and IHC patch-clamp recordings of exocytosis in mice lacking Munc13 and CAPS isoforms. Surprisingly, we did not detect Munc13 or CAPS proteins at IHC presynaptic active zones and found normal IHC exocytosis as well as auditory brainstem responses (ABRs) in Munc13 and CAPS deletion mutants. Instead, we show that otoferlin, a C2-domain protein that is crucial for vesicular fusion and replenishment in IHCs, clusters at the plasma membrane of the presynaptic active zone. Electron tomography of otoferlin-deficient IHC synapses revealed a reduction of short tethers holding vesicles at the active zone, which might be a structural correlate of impaired vesicle priming in otoferlin-deficient IHCs. We conclude that IHCs use an unconventional priming machinery that involves otoferlin.


Subject(s)
Evoked Potentials, Auditory, Brain Stem/physiology , Hair Cells, Auditory, Inner/metabolism , Membrane Proteins/genetics , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Animals , Calcium-Binding Proteins/genetics , Electron Microscope Tomography , Exocytosis/physiology , Female , Hair Cells, Auditory, Inner/cytology , Hearing/genetics , Hearing/physiology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Patch-Clamp Techniques
14.
J Cell Biol ; 205(4): 591-606, 2014 May 26.
Article in English | MEDLINE | ID: mdl-24862576

ABSTRACT

The molecular composition of the organelles involved in membrane recycling is difficult to establish as a result of the absence of suitable labeling tools. We introduce in this paper a novel probe, named membrane-binding fluorophore-cysteine-lysine-palmitoyl group (mCLING), which labels the plasma membrane and is taken up during endocytosis. It remains attached to membranes after fixation and permeabilization and can therefore be used in combination with immunostaining and super-resolution microscopy. We applied mCLING to mammalian-cultured cells, yeast, bacteria, primary cultured neurons, Drosophila melanogaster larval neuromuscular junctions, and mammalian tissue. mCLING enabled us to study the molecular composition of different trafficking organelles. We used it to address several questions related to synaptic vesicle recycling in the auditory inner hair cells from the organ of Corti and to investigate molecular differences between synaptic vesicles that recycle actively or spontaneously in cultured neurons. We conclude that mCLING enables the investigation of trafficking membranes in a broad range of preparations.


Subject(s)
Cell Membrane/physiology , Microscopy, Fluorescence/methods , Organelles/physiology , Protein Transport/physiology , Animals , Bacteria , COS Cells , Chlorocebus aethiops , Drosophila melanogaster , Endocytosis/physiology , Exocytosis/physiology , Fluorescent Dyes , Hair Cells, Auditory, Inner/physiology , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Organ of Corti/physiology , Primary Cell Culture , Rats , Saccharomyces cerevisiae , Synaptic Vesicles/physiology
15.
Trends Neurosci ; 35(11): 671-80, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22959777

ABSTRACT

Sound is encoded at synapses between cochlear inner hair cells and the auditory nerve. These synapses are anatomically and functionally specialized to transmit acoustic information with high fidelity over a lifetime. The molecular mechanisms of hair-cell transmitter release have recently attracted substantial interest. Here we review progress toward understanding otoferlin, a multi-C2 domain protein identified a decade ago by genetic analysis of human deafness. Otoferlin functions in hair-cell exocytosis. Several otoferlin C2 domains bind to Ca2+, phospholipids, and proteins. Current research reveals requirements for otoferlin in priming and fusion of synaptic vesicles during sound encoding. Understanding the molecular mechanisms through which otoferlin functions also has important implications for understanding the disease mechanisms that lead to deafness.


Subject(s)
Exocytosis/physiology , Hair Cells, Auditory, Inner/physiology , Hair Cells, Vestibular/physiology , Hearing Loss, Sensorineural/genetics , Membrane Proteins/physiology , Animals , Calcium Signaling/physiology , Humans , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Neurologic Mutants , Models, Neurological , Mutation , Patch-Clamp Techniques , Phospholipids/metabolism , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Structure-Activity Relationship , Synaptic Vesicles/metabolism
16.
BMC Neurosci ; 13: 40, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22533376

ABSTRACT

BACKGROUND: Transgenic mice have become an important tool to elucidate the genetic foundation of the human language faculty. While learning is an essential prerequisite for the acquisition of human speech, it is still a matter of debate whether auditory learning plays any role in the development of species-specific vocalizations in mice. To study the influence of auditory input on call development, we compared the occurrence and structure of ultrasonic vocalizations from deaf otoferlin-knockout mice, a model for human deafness DFNB9, to those of hearing wild-type and heterozygous littermates. RESULTS: We found that the occurrence and structure of ultrasonic vocalizations recorded from deaf otoferlin-knockout mice and hearing wild-type and heterozygous littermates do not differ. Isolation calls from 16 deaf and 15 hearing pups show the same ontogenetic development in terms of the usage and structure of their vocalizations as their hearing conspecifics. Similarly, adult courtship 'songs' produced by 12 deaf and 16 hearing males did not differ in the latency to call, rhythm of calling or acoustic structure. CONCLUSION: The results indicate that auditory experience is not a prerequisite for the development of species-specific vocalizations in mice. Thus, mouse models are of only limited suitability to study the evolution of vocal learning, a crucial component in the development of human speech. Nevertheless, ultrasonic vocalizations of mice constitute a valuable readout in studies of the genetic foundations of social and communicative behavior.


Subject(s)
Deafness/physiopathology , Hearing/physiology , Sexual Behavior, Animal/physiology , Vocalization, Animal/physiology , Acoustic Stimulation , Animals , Deafness/genetics , Female , Hearing/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Knockout
17.
J Neurosci ; 31(13): 4886-95, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21451027

ABSTRACT

Cochlear inner hair cells (IHCs) use Ca(2+)-dependent exocytosis of glutamate to signal sound information. Otoferlin (Otof), a C(2) domain protein essential for IHC exocytosis and hearing, may serve as a Ca(2+) sensor in vesicle fusion in IHCs that seem to lack the classical neuronal Ca(2+) sensors synaptotagmin 1 (Syt1) and Syt2. Support for the Ca(2+) sensor of fusion hypothesis for otoferlin function comes from biochemical experiments, but additional roles in late exocytosis upstream of fusion have been indicated by physiological studies. Here, we tested the functional equivalence of otoferlin and Syt1 in three neurosecretory model systems: auditory IHCs, adrenal chromaffin cells, and hippocampal neurons. Long-term and short-term ectopic expression of Syt1 in IHCs of Otof (-/-) mice by viral gene transfer in the embryonic inner ear and organotypic culture failed to rescue their Ca(2+) influx-triggered exocytosis. Conversely, virally mediated overexpression of otoferlin did not restore phasic exocytosis in Syt1-deficient chromaffin cells or neurons but enhanced asynchronous release in the latter. We further tested exocytosis in Otof (-/-) hippocampal neurons and in Syt1(-/-) IHCs but found no deficits in vesicle fusion. Expression analysis of different synaptotagmin isoforms indicated that Syt1 and Syt2 are absent from mature IHCs. Our data argue against a simple functional equivalence of the two C(2) domain proteins in exocytosis of IHC ribbon synapses, chromaffin cells, and hippocampal synapses.


Subject(s)
Exocytosis/physiology , Membrane Proteins/physiology , Synaptotagmin I/physiology , Acoustic Stimulation/methods , Animals , Animals, Newborn , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Exocytosis/genetics , Hippocampus/cytology , Hippocampus/physiology , Membrane Fusion/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Knockout , Neural Inhibition/genetics , Neurons/metabolism , Organ Culture Techniques , Synapses/genetics , Synapses/physiology , Synaptotagmin I/deficiency , Synaptotagmin I/genetics
18.
Nat Neurosci ; 14(4): 411-3, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21378973

ABSTRACT

SNARE proteins mediate membrane fusion. Neurosecretion depends on neuronal soluble NSF attachment protein receptors (SNAREs; SNAP-25, syntaxin-1, and synaptobrevin-1 or synaptobrevin-2) and is blocked by neurotoxin-mediated cleavage or genetic ablation. We found that exocytosis in mouse inner hair cells (IHCs) was insensitive to neurotoxins and genetic ablation of neuronal SNAREs. mRNA, but no synaptically localized protein, of neuronal SNAREs was present in IHCs. Thus, IHC exocytosis is unconventional and may operate independently of neuronal SNAREs.


Subject(s)
Exocytosis/physiology , Hair Cells, Auditory, Inner/metabolism , SNARE Proteins/genetics , Synapses/metabolism , Animals , Cattle , Cells, Cultured , Exocytosis/drug effects , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/ultrastructure , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Organ Culture Techniques , SNARE Proteins/deficiency , Synapses/drug effects , Synapses/ultrastructure
19.
J Mol Biol ; 406(3): 479-90, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21216247

ABSTRACT

Otoferlin (Otof), whose genetic mutations cause profound deafness in humans, is a protein composed of at least six C(2) domains, which are known as Ca(2)(+)-binding and phospholipid-binding regions. Mammalian ferlin proteins are proposed to act in membrane fusion events, with Otof being specifically required for exocytosis in auditory hair cells. Ferlin C(2) domains exhibit a rather low level of sequence similarity to those of synaptotagmins, protein kinase C isoforms, or phospholipases. Here, we report the crystal structure of the N-terminal C(2) domain of Otof (C2A) at 1.95-Å resolution. In contrast to previous predictions, we found that this C(2) domain is complete with eight ß-strands. Comparing the structure of Otof C2A to those of other C(2) domains revealed one top loop in Otof to be significantly shorter. This results in a depression of the surface, which is positively charged for the Otof C2A domain, and contrasts with the head-like protrusion surrounded by a negatively charged "neck" typically found in other C(2) domains. Isothermal titration calorimetry and circular dichroism spectroscopy studies confirmed that Otof C2A is unable to bind Ca(2+), while the synaptotagmin-1 C2A domain exhibited Ca(2+) binding under the same conditions. Furthermore, floatation assays revealed a failure of Otof C(2)A to bind to phospholipid membranes. Accordingly, no positively charged ß-groove-like surface structure, which is known to bind phosphatidylinositol-4,5-bisphosphate in other C(2) domains, was found at the respective position in Otof C2A. Taken together, these data demonstrate that the Otof C2A domain differs structurally and functionally from other C(2) domains.


Subject(s)
Membrane Proteins/chemistry , Models, Molecular , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Exocytosis , Hair Cells, Auditory/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Mutagenesis , Mutation , Phospholipids/chemistry , Protein Binding , Rats
20.
Eur J Hum Genet ; 18(12): 1327-32, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20648058

ABSTRACT

The exquisite performance of the mammalian hearing organ results from a finely orchestrated array of cell types, and their highly specialized functions are determined by their gene expression profile. In rodents, this profile is established mainly during the first 2 weeks of postnatal maturation. In this paper, we used the differential display technique on the rat organ of Corti to uncover transcripts upregulated in expression between postnatal stages P0 and P14. A total of 176 different genes were identified, the mRNA amount of which increased during early postnatal development. The transcripts code for proteins serving a broad spectrum of cellular functions including intracellular signaling, control of growth/differentiation, regulation of protein synthesis/degradation/modification, metabolism and synaptic function. In addition, the set of upregulated transcripts contained several proteins of yet unknown function, as well as hypothetical proteins and so far unknown mRNA sequences. Thus, this study unravels the broad and specific transcription program that operates the maturation of the mammalian hearing organ. Further, as 49 of the genes found here map to at least one unspecified deafness locus, our study provides candidate genes for these and novel deafness loci.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Hearing/genetics , Organ of Corti/metabolism , Animals , Animals, Newborn , DNA, Complementary/genetics , Gene Frequency/genetics , Genetic Predisposition to Disease , Hearing Disorders/genetics , Organ of Corti/growth & development , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...