Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 17: 1389816, 2024.
Article in English | MEDLINE | ID: mdl-38840777

ABSTRACT

Spiral ganglion neurons (SGNs) transmit auditory information from cochlear hair cells to the brain. SGNs are thus not only important for normal hearing, but also for effective functioning of cochlear implants, which stimulate SGNs when hair cells are missing. SGNs slowly degenerate following aminoglycoside-induced hair cell loss, a process thought to involve an immune response. However, the specific immune response pathways involved remain unknown. We used RNAseq to gain a deeper understanding immune-related and other transcriptomic changes that occur in the rat spiral ganglion after kanamycin-induced deafening. Among the immune and inflammatory genes that were selectively upregulated in deafened spiral ganglia, the complement cascade genes were prominent. We then assessed SGN survival, as well as immune cell numbers and activation, in the spiral ganglia of rats with a CRISPR-Cas9-mediated knockout of complement component 3 (C3). Similar to previous findings in our lab and other deafened rodent models, we observed an increase in macrophage number and increased expression of CD68, a marker of phagocytic activity and cell activation, in macrophages in the deafened ganglia. Moreover, we found an increase in MHCII expression on spiral ganglion macrophages and an increase in lymphocyte number in the deafened ganglia, suggestive of an adaptive immune response. However, C3 knockout did not affect SGN survival or increase in macrophage number/activation, implying that complement activation does not play a role in SGN death after deafening. Together, these data suggest that both innate and adaptive immune responses are activated in the deafened spiral ganglion, with the adaptive response directly contributing to cochlear neurodegeneration.

2.
Front Cell Neurosci ; 18: 1363219, 2024.
Article in English | MEDLINE | ID: mdl-38694536

ABSTRACT

Introduction: Cochlear afferent synapses connecting inner hair cells to spiral ganglion neurons are susceptible to excitotoxic trauma on exposure to loud sound, resulting in a noise-induced cochlear synaptopathy (NICS). Here we assessed the ability of cyclic AMP-dependent protein kinase (PKA) signaling to promote cochlear synapse regeneration, inferred from its ability to promote axon regeneration in axotomized CNS neurons, another system refractory to regeneration. Methods: We mimicked NICS in vitro by applying a glutamate receptor agonist, kainic acid (KA) to organotypic cochlear explant cultures and experimentally manipulated cAMP signaling to determine whether PKA could promote synapse regeneration. We then delivered the cAMP phosphodiesterase inhibitor rolipram via implanted subcutaneous minipumps in noise-exposed CBA/CaJ mice to test the hypothesis that cAMP signaling could promote cochlear synapse regeneration in vivo. Results: We showed that the application of the cell membrane-permeable cAMP agonist 8-cpt-cAMP or the cAMP phosphodiesterase inhibitor rolipram promotes significant regeneration of synapses in vitro within twelve hours after their destruction by KA. This is independent of neurotrophin-3, which also promotes synapse regeneration. Moreover, of the two independent signaling effectors activated by cAMP - the cAMP Exchange Protein Activated by cAMP and the cAMP-dependent protein kinase - it is the latter that mediates synapse regeneration. Finally, we showed that systemic delivery of rolipram promotes synapse regeneration in vivo following NICS. Discussion: In vitro experiments show that cAMP signaling promotes synapse regeneration after excitotoxic destruction of cochlear synapses and does so via PKA signaling. The cAMP phosphodiesterase inhibitor rolipram promotes synapse regeneration in vivo in noise-exposed mice. Systemic administration of rolipram or similar compounds appears to provide a minimally invasive therapeutic approach to reversing synaptopathy post-noise.

3.
Neurotherapeutics ; 20(2): 578-601, 2023 03.
Article in English | MEDLINE | ID: mdl-36697994

ABSTRACT

Destruction of cochlear hair cells by aminoglycoside antibiotics leads to gradual death of the spiral ganglion neurons (SGNs) that relay auditory information to the brain, potentially limiting the efficacy of cochlear implants. Because the reasons for this cochlear neurodegeneration are unknown, there are no neuroprotective strategies for patients. To investigate this problem, we assessed transcriptomic changes in the rat spiral ganglion following aminoglycoside antibiotic (kanamycin)-induced hair cell destruction. We observed selectively increased expression of immune and inflammatory response genes and increased abundance of activated macrophages in spiral ganglia by postnatal day 32 in kanamycin-deafened rats, preceding significant SGN degeneration. Treatment with the anti-inflammatory medications dexamethasone and ibuprofen diminished long-term SGN degeneration. Ibuprofen and dexamethasone also diminished macrophage activation. Efficacy of ibuprofen treatment was augmented by co-administration of the nicotinamide adenine dinucleotide-stabilizing agent P7C3-A20. Our results support a critical role of neuroinflammation in SGN degeneration after aminoglycoside antibiotic-mediated cochlear hair cell loss, as well as a neuroprotective strategy that could improve cochlear implant efficacy.


Subject(s)
Ibuprofen , Spiral Ganglion , Rats , Animals , Ibuprofen/metabolism , Hair Cells, Auditory/metabolism , Aminoglycosides/toxicity , Aminoglycosides/metabolism , Anti-Bacterial Agents/toxicity , Kanamycin/toxicity , Kanamycin/metabolism , Neurons , Anti-Inflammatory Agents/metabolism , Dexamethasone
4.
Proc Natl Acad Sci U S A ; 117(7): 3828-3838, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32015128

ABSTRACT

Exposure to loud sound damages the postsynaptic terminals of spiral ganglion neurons (SGNs) on cochlear inner hair cells (IHCs), resulting in loss of synapses, a process termed synaptopathy. Glutamatergic neurotransmission via α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-type receptors is required for synaptopathy, and here we identify a possible involvement of GluA2-lacking Ca2+-permeable AMPA receptors (CP-AMPARs) using IEM-1460, which has been shown to block GluA2-lacking AMPARs. In CBA/CaJ mice, a 2-h exposure to 100-dB sound pressure level octave band (8 to 16 kHz) noise results in no permanent threshold shift but does cause significant synaptopathy and a reduction in auditory brainstem response (ABR) wave-I amplitude. Chronic intracochlear perfusion of IEM-1460 in artificial perilymph (AP) into adult CBA/CaJ mice prevented the decrease in ABR wave-I amplitude and the synaptopathy relative to intracochlear perfusion of AP alone. Interestingly, IEM-1460 itself did not affect the ABR threshold, presumably because GluA2-containing AMPARs can sustain sufficient synaptic transmission to evoke low-threshold responses during blockade of GluA2-lacking AMPARs. On individual postsynaptic densities, we observed GluA2-lacking nanodomains alongside regions with robust GluA2 expression, consistent with the idea that individual synapses have both CP-AMPARs and Ca2+-impermeable AMPARs. SGNs innervating the same IHC differ in their relative vulnerability to noise. We found local heterogeneity among synapses in the relative abundance of GluA2 subunits that may underlie such differences in vulnerability. We propose a role for GluA2-lacking CP-AMPARs in noise-induced cochlear synaptopathy whereby differences among synapses account for differences in excitotoxic susceptibility. These data suggest a means of maintaining normal hearing thresholds while protecting against noise-induced synaptopathy, via selective blockade of CP-AMPARs.


Subject(s)
Calcium/metabolism , Cochlea/metabolism , Hearing Loss, Noise-Induced/metabolism , Noise/adverse effects , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Evoked Potentials, Auditory, Brain Stem , Hearing , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/physiopathology , Humans , Male , Mice , Mice, Inbred CBA , Receptors, AMPA/genetics
5.
J Neurosci ; 34(39): 13110-26, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25253857

ABSTRACT

Spiral ganglion neurons (SGNs) receive input from cochlear hair cells and project from the cochlea to the cochlear nucleus. After destruction of hair cells with aminoglycoside antibiotics or noise, SGNs gradually die. It has been assumed that SGN death is attributable to loss of neurotrophic factors (NTFs) derived from hair cells or supporting cells in the organ of Corti (OC). We used quantitative PCR (qPCR) to assay NTF expression-neurotrophin-3 (NT-3), BDNF, GDNF, neurturin, artemin, and CNTF-in the OC and cochlear nucleus at various ages from postnatal day 0 (P0) to P90 in control hearing and neonatally deafened rats. NT-3, neurturin, and CNTF were most abundant in the postnatal hearing OC; CNTF and neurturin most abundant in the cochlear nucleus. In the OC, NT-3 and CNTF showed a postnatal increase in expression approximately concomitant with hearing onset. In rats deafened by daily kanamycin injections (from P8 to P16), surviving inner hair cells were evident at P16 but absent by P19, with most postsynaptic boutons lost before P16. NT-3 and CNTF, which normally increase postnatally, had significantly reduced expression in the OC of deafened rats, although CNTF was expressed throughout the time that SGNs were dying. In contrast, neurturin expression was constant, unaffected by deafening or by age. CNTF and neurturin expression in the cochlear nucleus was unaffected by deafening or age. Thus, NTFs other than NT-3 are available to SGNs even as they are dying after deafening, apparently conflicting with the hypothesis that SGN death is attributable to lack of NTFs.


Subject(s)
Cochlear Nucleus/metabolism , Deafness/metabolism , Hair Cells, Auditory/metabolism , Nerve Growth Factors/metabolism , Spiral Ganglion/metabolism , Animals , Cochlear Nucleus/cytology , Cochlear Nucleus/growth & development , Deafness/chemically induced , Gene Expression Regulation, Developmental , Kanamycin/toxicity , Nerve Growth Factors/genetics , Organ Specificity , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Spiral Ganglion/cytology , Spiral Ganglion/growth & development
6.
Otolaryngol Head Neck Surg ; 149(5): 745-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23907267

ABSTRACT

OBJECTIVE: To establish the intracellular consequences of electrical stimulation to spiral ganglion neurons after deafferentation. Here we use a rat model to determine the effect of both low and high pulse rate acute electrical stimulation on activation of the proapoptotic transcription factor Jun in deafferented spiral ganglion neurons in vivo. STUDY DESIGN: Experimental animal study. SETTING: Hearing research laboratories of the University of Iowa Departments of Biology and Otolaryngology. METHODS: A single electrode was implanted through the round window of kanamycin-deafened rats at either postnatal day 32 (P32, n = 24) or P60 (n = 22) for 4 hours of stimulation (monopolar, biphasic pulses, amplitude twice electrically evoked auditory brainstem response [eABR] threshold) at either 100 or 5000 Hz. Jun phosphorylation was assayed by immunofluorescence to quantitatively assess the effect of electrical stimulation on proapoptotic signaling. RESULTS: Jun phosphorylation was reliably suppressed by 100 Hz stimuli in deafened cochleae of P32 but not P60 rats. This effect was not significant in the basal cochlear turns. Stimulation frequency may be consequential: 100 Hz was significantly more effective than was 5 kHz stimulation in suppressing phospho-Jun. CONCLUSIONS: Suppression of Jun phosphorylation occurs in deafferented spiral ganglion neurons after only 4 hours of electrical stimulation. This finding is consistent with the hypothesis that electrical stimulation can decrease spiral ganglion neuron death after deafferentation.


Subject(s)
Apoptosis/physiology , Cochlear Implants , Deafness/therapy , Electric Stimulation/methods , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing/physiology , Spiral Ganglion/pathology , Animals , Deafness/pathology , Deafness/physiopathology , Disease Models, Animal , Rats , Spiral Ganglion/physiopathology
7.
Anat Rec (Hoboken) ; 295(11): 1877-95, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23044862

ABSTRACT

The spiral ganglion neurons (SGNs) are the afferent neurons of the cochlea, connecting the auditory sensory cells-hair cells-to the brainstem cochlear nuclei. The neurotrophins neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF) are expressed in the cochlea and both support SGN survival during development. These neurotrophins remain expressed in the postnatal cochlea and continue to play additional roles for SGNs, contributing to maintenance of hair cell-SGN synapses and regulating expression of ion channels, presynaptic and postsynaptic proteins, and SGN membrane electrical properties in a physiologically important spatial pattern. Remarkably, NT-3 and BDNF have different, even opposing, effects on SGN physiology despite the close similarity of their receptors TrkB and TrkC. Recent studies have also raised the possibility that precursor proneurotrophin forms of the neurotrophins play a role in responses to trauma in the cochlea, signaling through the proneurotrophin receptor p75(NTR) . Here, we review expression and function of neurotrophins and their p75(NTR) and Trk-family receptors in the cochlea. We focus, in particular, on neurotrophin functions other than support of SGN survival, including regulation of SGN neurite growth, synaptic and membrane physiology. These functions, unlike survival, are ones for which BDNF and NT-3 substantially differ in their effects. Signal transduction mechanisms of p75(NTR) and of Trk-family receptors are discussed, indicating how these lead to different responses, and we speculate on how BDNF and NT-3 can cause different phenotypic changes in SGNs. Because these complex signaling interactions remain incompletely understood, use of neurotrophins as therapeutic agents in the cochlea should be approached with caution.


Subject(s)
Cochlea/metabolism , Nerve Growth Factors/metabolism , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Animals , Cochlea/cytology , Humans , Signal Transduction
8.
J Comp Neurol ; 519(16): 3327-45, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21800311

ABSTRACT

The rat auditory cortex is organized as a tonotopic map of sound frequency. This map is broadly tuned at birth and is refined during the first 3 weeks postnatal. The structural correlates underlying tonotopic map maturation and reorganization during development are poorly understood. We employed fluorescent dye ballistic labeling ("DiOlistics") alone, or in conjunction with immunohistochemistry, to quantify synaptogenesis in the auditory cortex of normal hearing rats. We show that the developmental appearance of dendritic protrusions, which include both immature filopodia and mature spines, on layers 2/3, 4, and 5 pyramidal and layer 4 spiny nonpyramidal neurons occurs in three phases: slow addition of dendritic protrusions from postnatal day 4 (P4) to P9, rapid addition of dendritic protrusions from P9 to P19, and a final phase where mature protrusion density is achieved (>P21). Next, we combined DiOlistics with immunohistochemical labeling of bassoon, a presynaptic scaffolding protein, as a novel method to categorize dendritic protrusions as either filopodia or mature spines in cortex fixed in vivo. Using this method we observed an increase in the spine-to-filopodium ratio from P9-P16, indicating a period of rapid spine maturation. Previous studies report mature spines as being shorter in length compared to filopodia. We similarly observed a reduction in protrusion length between P9 and P16, corroborating our immunohistochemical spine maturation data. These studies show that dendritic protrusion formation and spine maturation occur rapidly at a time previously shown to correspond to auditory cortical tonotopic map refinement (P11-P14), providing a structural correlate of physiological maturation.


Subject(s)
Auditory Cortex/growth & development , Dendritic Spines/ultrastructure , Neurogenesis , Animals , Female , Fluorescent Antibody Technique , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley
9.
J Biol Chem ; 286(30): 26496-506, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21652711

ABSTRACT

The A kinase anchor protein AKAP150 recruits the cAMP-dependent protein kinase (PKA) to dendritic spines. Here we show that in AKAP150 (AKAP5) knock-out (KO) mice frequency of miniature excitatory post-synaptic currents (mEPSC) and inhibitory post-synaptic currents (mIPSC) are elevated at 2 weeks and, more modestly, 4 weeks of age in the hippocampal CA1 area versus litter mate WT mice. Linear spine density and ratio of AMPAR to NMDAR EPSC amplitudes were also increased. Amplitude and decay time of mEPSCs, decay time of mIPSCs, and spine size were unaltered. Mice in which the PKA anchoring C-terminal 36 residues of AKAP150 are deleted (D36) showed similar changes. Furthermore, whereas acute stimulation of PKA (2-4 h) increases spine density, prolonged PKA stimulation (48 h) reduces spine density in apical dendrites of CA1 pyramidal neurons in organotypic slice cultures. The data from the AKAP150 mutant mice show that AKAP150-anchored PKA chronically limits the number of spines with functional AMPARs at 2-4 weeks of age. However, synaptic transmission and spine density was normal at 8 weeks in KO and D36 mice. Thus AKAP150-independent mechanisms correct the aberrantly high number of active spines in juvenile AKAP150 KO and D36 mice during development.


Subject(s)
A Kinase Anchor Proteins/metabolism , Aging/physiology , Dendrites/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Knockout , Pyramidal Cells/cytology , Pyramidal Cells/metabolism
10.
Mol Cell Neurosci ; 47(4): 306-15, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21658451

ABSTRACT

Spiral ganglion Schwann cells (SGSCs) myelinate spiral ganglion neurons (SGNs) and represent a potential source of neurotrophic support for SGNs. Deafening due to loss of hair cells results in gradual degeneration and death of SGNs. Successful efforts to maintain or regenerate a functional auditory nerve may depend on a healthy population of SGSCs, yet the responses of SGSCs to neural injury remain largely unknown. Here we investigate the role of p75(NTR) in SGSC responses to gradual denervation. Following deafening, SGSCs in the osseous spiral lamina (OSL) and, subsequently, in Rosenthal's canal (RC) expressed elevated p75(NTR) compared to hearing controls. p75(NTR)-positive cells co-labeled with S100 and RIP antibodies (Schwann cell markers), but not with anti-neurofilament. The pattern of p75(NTR) expression mirrored the pattern of neural degeneration, beginning in the OSL of the cochlea base and later extending into the apex. SGSCs expressed sortilin, a p75(NTR) co-receptor for pro-neurotrophins. Both pro-nerve growth factor (pro-NGF) and pro-brain derived neurotrophic factor (proBDNF) induced apoptosis in cultured SGSCs. Deafened animals exhibited significantly higher levels of SGSC proliferation (as measured by BrdU uptake) compared to hearing animals while total Schwann cell density remained stable, suggesting a tight regulation of SGSC proliferation and cell death. SGSCs undergoing cell division lose p75(NTR) expression from the cell surface and demonstrate nuclear localization of the intracellular domain (ICD), raising the possibility that p75(NTR) cleavage and ICD nuclear localization regulate SGSC proliferation. These results suggest that p75(NTR) contributes to SGSC responses to deafening and neural degeneration.


Subject(s)
Cell Proliferation , Receptor, Nerve Growth Factor/metabolism , Schwann Cells/metabolism , Spiral Ganglion/cytology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Deafness/pathology , Deafness/physiopathology , Nerve Growth Factor/metabolism , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/genetics , Schwann Cells/cytology , Spiral Ganglion/metabolism
11.
J Neurosci ; 31(21): 7938-49, 2011 May 25.
Article in English | MEDLINE | ID: mdl-21613508

ABSTRACT

Spiral ganglion neurons (SGNs) are postsynaptic to hair cells and project to the brainstem. The inner hair cell (IHC) to SGN synapse is susceptible to glutamate excitotoxicity and to acoustic trauma, with potentially adverse consequences to long-term SGN survival. We used a cochlear explant culture from P6 rat pups consisting of a portion of organ of Corti maintained intact with the corresponding portion of spiral ganglion to investigate excitotoxic damage to IHC-SGN synapses in vitro. The normal innervation pattern is preserved in vitro. Brief treatment with NMDA and kainate results in loss of IHC-SGN synapses and degeneration of the distal type 1 SGN peripheral axons, mimicking damage to SGN peripheral axons caused by excitotoxicity or noise in vivo. The number of IHC presynaptic ribbons is not significantly altered. Reinnervation of IHCs occurs and regenerating axons remain restricted to the IHC row. However, the number of postsynaptic densities (PSDs) does not fully recover and not all axons regrow to the IHCs. Addition of either neurotrophin-3 (NT-3) or BDNF increases axon growth and synaptogenesis. Selective blockade of endogenous NT-3 signaling with TrkC-IgG reduced regeneration of axons and PSDs, but TrkB-IgG, which blocks BDNF, has no such effect, indicating that endogenous NT-3 is necessary for SGN axon growth and synaptogenesis. Remarkably, TrkC-IgG reduced axon growth and synaptogenesis even in the presence of BDNF, indicating that endogenous NT-3 has a distinctive role, not mimicked by BDNF, in promoting SGN axon growth in the organ of Corti and synaptogenesis on IHCs.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Hair Cells, Auditory, Inner/physiology , Neurotrophin 3/physiology , Regeneration/physiology , Spiral Ganglion/physiology , Synapses/physiology , Animals , Axons/physiology , Female , Hair Cells, Auditory, Inner/drug effects , Male , Neurotrophin 3/antagonists & inhibitors , Neurotrophin 3/biosynthesis , Organ Culture Techniques , Rats , Regeneration/drug effects , Spiral Ganglion/drug effects , Synapses/drug effects
12.
Hear Res ; 278(1-2): 77-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21554942

ABSTRACT

Jun N-terminal kinase (JNK) is a multifunctional protein kinase crucial for neuronal apoptosis as well as neurite growth. We have previously shown that JNK activity is correlated with spiral ganglion neuron (SGN) apoptosis following hair cell loss in rats (Alam et al., 2007) implying that JNK inhibition may have therapeutic potential to protect SGNs in deaf individuals. Here we investigated the role of JNK in neurite outgrowth from cultured neonatal rat and mouse SGNs. We show that JNK is required for initial growth of neurites and for continued extension of already established neurites. The effect of JNK inhibition on neurite growth is rapid and is also rapidly reversible after washout of the inhibitor. Using phosphoJNK immunoreactivity as an indicator, we show that JNK is activated in growth cones within 30 min after transfer to medium lacking neurotrophic stimuli (5 K medium) but activation in the nucleus and soma requires hours. By transfecting epitope-tagged JNK1, JNK2, or JNK3 isoforms into SGNs, we found that all are present in the nucleus and cytoplasm and that there is no preferential redistribution to the nucleus after transfer to 5 K medium. Cotransfection of dominant-negative (dn) JNK1 and JNK2 into SGNs reduced neurite growth, although transfection of dnJNK1 or dnJNK2 alone had no significant effect. SGNs cultured from JNK3(-/-) mice showed reduced neurite growth that was further reduced by transfection of dnJNK1 and dnJNK2. This indicates that all three JNK isoforms promote SGN neurite growth although there may be functional redundancy between JNK1 and JNK2.


Subject(s)
MAP Kinase Signaling System , Neurites/enzymology , Neurites/ultrastructure , Spiral Ganglion/enzymology , Spiral Ganglion/innervation , Animals , Cells, Cultured , Enzyme Activation , Kinetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Neurons/enzymology , Neurons/ultrastructure , Phosphorylation , Rats , Spiral Ganglion/ultrastructure , Subcellular Fractions/enzymology , Transfection
13.
PLoS Biol ; 9(4): e1000612, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21526220

ABSTRACT

Mitochondrial shape is determined by fission and fusion reactions catalyzed by large GTPases of the dynamin family, mutation of which can cause neurological dysfunction. While fission-inducing protein phosphatases have been identified, the identity of opposing kinase signaling complexes has remained elusive. We report here that in both neurons and non-neuronal cells, cAMP elevation and expression of an outer-mitochondrial membrane (OMM) targeted form of the protein kinase A (PKA) catalytic subunit reshapes mitochondria into an interconnected network. Conversely, OMM-targeting of the PKA inhibitor PKI promotes mitochondrial fragmentation upstream of neuronal death. RNAi and overexpression approaches identify mitochondria-localized A kinase anchoring protein 1 (AKAP1) as a neuroprotective and mitochondria-stabilizing factor in vitro and in vivo. According to epistasis studies with phosphorylation site-mutant dynamin-related protein 1 (Drp1), inhibition of the mitochondrial fission enzyme through a conserved PKA site is the principal mechanism by which cAMP and PKA/AKAP1 promote both mitochondrial elongation and neuronal survival. Phenocopied by a mutation that slows GTP hydrolysis, Drp1 phosphorylation inhibits the disassembly step of its catalytic cycle, accumulating large, slowly recycling Drp1 oligomers at the OMM. Unopposed fusion then promotes formation of a mitochondrial reticulum, which protects neurons from diverse insults.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mitochondria/physiology , Neurons/physiology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dynamins/metabolism , Hippocampus/cytology , Hippocampus/enzymology , Homeostasis , Humans , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Neurons/drug effects , Neurons/enzymology , Organelle Shape/drug effects , Phosphorylation , Protein Multimerization , Protein Transport , Rats
14.
J Neurosci Res ; 87(9): 1969-79, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19235894

ABSTRACT

Epileptiform activity (EA) in vivo and in vitro induces a loss of dendritic spines and synapses. Because CaMKII has been implicated in synaptogenesis and synaptic plasticity, we investigated the role of CaMKII in the effects of EA on spines, using rat hippocampal slice cultures. To visualize dendrites and postsynaptic densities (PSDs) in pyramidal neurons in the slices, we used biolistic transfection to express either free GFP or a PSD95-YFP construct that specifically labels PSDs. This allowed us to distinguish two classes of dendritic protrusions: spines that contain PSDs, and filopodia that lack PSDs and that are, on average, longer than spines. By these criteria, 48 hr of EA caused a decrease specifically in the number of spines. Immunoblots showed that EA increased CaMKII activity in the slices. Inhibition of CaMKII by expression of AIP, a specific peptide inhibitor of CaMKII, reduced spine number under basal conditions and failed to prevent EA-induced spine loss. However, under EA conditions, AIP increased the number of filopodia and the number of PSDs on the dendritic shaft. These data show at least two roles for CaMKII activity in maintenance and remodeling of dendritic spines under basal or EA conditions. First, CaMKII activity promotes the maintenance of spines and spine PSDs. Second, CaMKII activity suppresses EA-induced formation of filopodia and suppresses an increase in shaft PSDs, apparently by promoting translocation of PSDs from dendritic shafts to spines and/or selectively stabilizing spine rather than shaft PSDs.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/enzymology , Epilepsy/enzymology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , Cerebral Cortex/enzymology , Cerebral Cortex/ultrastructure , Dendritic Spines/ultrastructure , Disks Large Homolog 4 Protein , Epilepsy/physiopathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organ Culture Techniques , Protein Transport/physiology , Pseudopodia/enzymology , Pseudopodia/ultrastructure , Pyramidal Cells/enzymology , Pyramidal Cells/ultrastructure , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synaptic Membranes/enzymology , Synaptic Membranes/ultrastructure
15.
Mol Cell Neurosci ; 37(2): 376-87, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18055215

ABSTRACT

The effect of membrane electrical activity on spiral ganglion neuron (SGN) neurite growth remains unknown despite its relevance to cochlear implant technology. We demonstrate that membrane depolarization delays the initial formation and inhibits the subsequent extension of cultured SGN neurites. This inhibition depends directly on the level of depolarization with higher levels of depolarization causing retraction of existing neurites. Cultured SGNs express subunits for L-type, N-type, and P/Q type voltage-gated calcium channels (VGCCs) and removal of extracellular Ca(2+) or treatment with a combination of L-type, N-type, and P/Q-type VGCC antagonists rescues SGN neurite growth under depolarizing conditions. By measuring the fluorescence intensity of SGNs loaded with the fluorogenic calpain substrate t-butoxy carbonyl-Leu-Met-chloromethylaminocoumarin (20 microM), we demonstrate that depolarization activates calpains. Calpeptin (15 microM), a calpain inhibitor, prevents calpain activation by depolarization and rescues neurite growth in depolarized SGNs suggesting that calpain activation contributes to the inhibition of neurite growth by depolarization.


Subject(s)
Calcium Channels/metabolism , Calpain/metabolism , Cell Membrane/metabolism , Neurites/metabolism , Spiral Ganglion/growth & development , Spiral Ganglion/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/metabolism , Calcium Channels, Q-Type/drug effects , Calcium Channels, Q-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calpain/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Membrane/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurites/drug effects , Neurites/ultrastructure , Rats , Spiral Ganglion/drug effects , Transfection/methods
16.
Mol Cell Neurosci ; 36(1): 13-26, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17651987

ABSTRACT

By fusing the CaMKII-inhibitory peptide AIP to GFP, we constructed a specific and effective CaMKII inhibitor, GFP-AIP. Expression of GFP-AIP and/or dominant-inhibitory CaMKIV in cultured neonatal rat spiral ganglion neurons (SGNs) shows that CaMKII and CaMKIV act additively and in parallel to mediate the prosurvival effect of depolarization. Depolarization or expression of constitutively active CaMKII functionally inactivates Bad, indicating that this is one means by which CaMKII promotes neuronal survival. CaMKIV, but not CaMKII, requires CREB to promote SGN survival, consistent with the exclusively nuclear localization of CaMKIV and indicating that the principal prosurvival function of CaMKIV is activation of CREB. Consistent with this, a constitutively active CREB construct that provides a high level of CREB activity promotes SGN survival, although low levels of CREB activity did not do so. Also, in apoptotic SGNs, activation of CREB by depolarization is disabled, presumably as part of a cellular commitment to apoptosis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 4/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , CREB-Binding Protein/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/genetics , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Activation/genetics , Enzyme Activation/physiology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , Neurofilament Proteins/metabolism , Neurons/drug effects , Neurons/ultrastructure , Peptides/metabolism , Potassium Chloride/pharmacology , Rats , Spiral Ganglion/cytology , Transfection , bcl-Associated Death Protein/metabolism
17.
J Comp Neurol ; 503(6): 832-52, 2007 Aug 20.
Article in English | MEDLINE | ID: mdl-17570507

ABSTRACT

Neurons depend on afferent input for survival. Rats were given daily kanamycin injections from P8 to P16 to destroy hair cells, the sole afferent input to spiral ganglion neurons (SGNs). Most SGNs die over an approximately 14-week period after deafferentation. During this period, the SGN population is heterogeneous. At any given time, some SGNs exhibit apoptotic markers--TUNEL and cytochrome c loss--whereas others appear nonapoptotic. We asked whether differences among SGNs in intracellular signaling relevant to apoptotic regulation could account for this heterogeneity. cAMP response element binding protein (CREB) phosphorylation, which reflects neurotrophic signaling, is reduced in many SGNs at P16, P23, and P32, when SGNs begin to die. In particular, nearly all apoptotic SGNs exhibit reduced phospho-CREB, implying that apoptosis is due to insufficient neurotrophic support. However, >32% of SGNs maintain high phospho-CREB levels, implying access to neurotrophic support. By P60, when approximately 50% of the SGNs have died, phospho-CREB levels in surviving neurons are not reduced, and SGN death is no longer correlated with reduced phospho-CREB. Activity in the proapoptotic Jun N-terminal kinase (JNK)-Jun signaling pathway is elevated in SGNs during the cell death period. This too is heterogeneous: <42% of the SGNs exhibited high phospho-Jun levels, but nearly all SGNs undergoing apoptosis exhibited elevated phospho-Jun. Thus, heterogeneity among SGNs in prosurvival and proapoptotic signaling is correlated with apoptosis. SGN death following deafferentation has an early phase in which apoptosis is correlated with reduced phospho-CREB and a later phase in which it is not. Proapoptotic JNK-Jun signaling is tightly correlated with SGN apoptosis.


Subject(s)
Apoptosis/physiology , Deafness/physiopathology , Hair Cells, Auditory/pathology , Signal Transduction/physiology , Spiral Ganglion/metabolism , Animals , Anti-Bacterial Agents/toxicity , Blotting, Western , Cell Survival/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Deafness/chemically induced , Fluorescent Antibody Technique , Hair Cells, Auditory/drug effects , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases/metabolism , Kanamycin/toxicity , Neurons/pathology , Neurons/physiology , Phosphorylation , Rats , Rats, Sprague-Dawley , Spiral Ganglion/pathology
18.
Dev Neurobiol ; 67(3): 316-25, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17443790

ABSTRACT

Spiral ganglion neurons (SGNs) provide afferent innervation to the cochlea and rely on contact with hair cells (HCs) for their survival. Following deafferentation due to hair cell loss, SGNs gradually die. In a rat culture model, we explored the ability of prosurvival members of the Bcl-2 family of proteins to support the survival and neurite outgrowth of SGNs. We found that overexpression of either Bcl-2 or Bcl-xL significantly increases SGN survival in the absence of neurotrophic factors, establishing that the Bcl-2 pathway is sufficient for SGN cell survival and that SGN deprived of trophic support die by an apoptotic mechanism. However, in contrast to observations in central neurons and PC12 cells where Bcl-2 appears to promote neurite growth, both Bcl-2 and Bcl-xL overexpression dramatically inhibit neurite outgrowth in SGNs. This inhibition of neurite growth by Bcl-2 occurs in nearly all SGNs even in the presence of multiple neurotrophic factors implying that Bcl-2 directly inhibits neurite growth rather than simply rescuing a subpopulation of neurons incapable of extending neurites without additional stimuli. Thus, although overexpression of prosurvival members of the Bcl-2 family prevents SGN loss following trophic factor deprivation, the inhibition of neurite growth by these molecules may limit their efficacy for support of auditory nerve maintenance or regeneration following hair cell loss.


Subject(s)
Neural Inhibition/physiology , Neurites/physiology , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , Spiral Ganglion/cytology , bcl-X Protein/metabolism , Analysis of Variance , Animals , Animals, Newborn , Cell Count/methods , Cell Death/physiology , Cells, Cultured , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/metabolism , Rats , Transfection
19.
Proc Natl Acad Sci U S A ; 103(44): 16556-61, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17060608

ABSTRACT

Extracellular proton concentrations in the brain may be an important signal for neuron function. Proton concentrations change both acutely when synaptic vesicles release their acidic contents into the synaptic cleft and chronically during ischemia and seizures. However, the brain receptors that detect protons and their physiologic importance remain uncertain. Using organotypic hippocampal slices and biolistic transfection, we found the acid-sensing ion channel 1a (ASIC1a), localized in dendritic spines where it functioned as a proton receptor. ASIC1a also affected the density of spines, the postsynaptic site of most excitatory synapses. Decreasing ASIC1a reduced the number of spines, whereas overexpressing ASIC1a had the opposite effect. Ca(2+)-mediated Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) signaling was probably responsible, because acid evoked an ASIC1a-dependent elevation of spine intracellular Ca(2+) concentration, and reducing or increasing ASIC1a levels caused parallel changes in CaMKII phosphorylation in vivo. Moreover, inhibiting CaMKII prevented ASIC1a from increasing spine density. These data indicate that ASIC1a functions as a postsynaptic proton receptor that influences intracellular Ca(2+) concentration and CaMKII phosphorylation and thereby the density of dendritic spines. The results provide insight into how protons influence brain function and how they may contribute to pathophysiology.


Subject(s)
Dendritic Spines/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protons , Sodium Channels/metabolism , Synapses/metabolism , Acid Sensing Ion Channels , Acids/metabolism , Animals , Binding, Competitive , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Dendritic Spines/drug effects , Hippocampus/metabolism , Mice , Peptides/pharmacology , Phosphorylation , Tissue Culture Techniques
20.
Glia ; 53(6): 593-600, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16432850

ABSTRACT

Vestibular schwannomas (VSs) are benign tumors that arise from the Schwann cells (SCs) lining the vestibular nerve. VS cells survive and proliferate far from neurons and axonally derived growth factors. We have previously shown that VSs produce the glial growth factor, neuregulin-1 (NRG1), and its receptors, ErbB2 and ErbB3. In the present work, we explore the contribution of constitutive NRG1:ErbB signaling to human VS cell proliferation. We confirm that human VSs, which express markers of immature and denervated SCs, also express endogenous NRG1 and activated ErbB2. We find that a blocking anti-NRG1 antibody and trastuzumab (Herceptin, HCN), a humanized anti-ErbB2 inhibitory monoclonal antibody, effectively inhibit NRG1 induced SC proliferation. Treatment of primary VS cultures with anti-NRG1 or HCN reduces cell proliferation in the absence of exogenous NRG1. Furthermore, conditioned medium from VS cell cultures contains NRG1 and stimulates SC proliferation in SC cultures, an effect that is inhibited by anti-NRG1 and HCN. These data suggest an autocrine pathway of VS growth stimulation involving NRG and ErbB receptors. Inhibition of constitutive NRG:ErbB signaling reduces VS cell proliferation in vitro and may have therapeutic potential for patients with VSs.


Subject(s)
Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Neuregulin-1/metabolism , Neuroma, Acoustic/metabolism , Oncogene Proteins v-erbB/metabolism , Vestibular Nerve/metabolism , Animals , Animals, Newborn , Antibodies/pharmacology , Autocrine Communication/drug effects , Autocrine Communication/physiology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Culture Media, Conditioned/pharmacology , Down-Regulation/drug effects , Down-Regulation/physiology , Gene Expression Regulation, Neoplastic/physiology , Glycoproteins/metabolism , Humans , Neuregulin-1/antagonists & inhibitors , Neuregulin-1/genetics , Neuroma, Acoustic/genetics , Neuroma, Acoustic/pathology , Oncogene Proteins v-erbB/genetics , Rats , Receptor, ErbB-2 , Schwann Cells/drug effects , Schwann Cells/metabolism , Signal Transduction/physiology , Stem Cells/drug effects , Stem Cells/metabolism , Vestibular Nerve/pathology , Vestibular Nerve/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...