1
|
Richards LA, Schonhoff CM. Nitric oxide and sex differences in dendritic branching and arborization. J Neurosci Res 2021; 99:1390-1400. [PMID: 33538046 DOI: 10.1002/jnr.24789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is an important signaling molecule with many functions in the nervous system. Derived from the enzymatic conversion of arginine by several nitric oxide synthases (NOS), NO plays significant roles in neuronal developmental events such as the establishment of dendritic branching or arbors. A brief summary of the discovery, molecular biology, and chemistry of NO, and a description of important NO-mediated signal transduction pathways with emphasis on the role for NO in the development of dendritic branching during neurodevelopment are presented. Important sex differences in neuronal nitric oxide synthase expression during neuronal development are considered. Finally, a survey of endogenous and exogenous substances that disrupt dendritic patterning is presented with particular emphasis on how these molecules may drive NO-mediated sex differences in dendritic branching.
Collapse
Affiliation(s)
- Laura A Richards
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA
| | - Christopher M Schonhoff
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA.,Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA
| |
Collapse
|
2
|
Frick C, Fink S, Schmidbauer D, Rousset F, Eickhoff H, Tropitzsch A, Kramer B, Senn P, Glueckert R, Rask-Andersen H, Wiesmüller KH, Löwenheim H, Müller M. Age-Dependency of Neurite Outgrowth in Postnatal Mouse Cochlear Spiral Ganglion Explants. Brain Sci 2020; 10:E580. [PMID: 32839381 PMCID: PMC7564056 DOI: 10.3390/brainsci10090580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The spatial gap between cochlear implants (CIs) and the auditory nerve limits frequency selectivity as large populations of spiral ganglion neurons (SGNs) are electrically stimulated synchronously. To improve CI performance, a possible strategy is to promote neurite outgrowth toward the CI, thereby allowing a discrete stimulation of small SGN subpopulations. Brain-derived neurotrophic factor (BDNF) is effective to stimulate neurite outgrowth from SGNs. METHOD TrkB (tropomyosin receptor kinase B) agonists, BDNF, and five known small-molecule BDNF mimetics were tested for their efficacy in stimulating neurite outgrowth in postnatal SGN explants. To modulate Trk receptor-mediated effects, TrkB and TrkC ligands were scavenged by an excess of recombinant receptor proteins. The pan-Trk inhibitor K252a was used to block Trk receptor actions. RESULTS THF (7,8,3'-trihydroxyflavone) partly reproduced the BDNF effect in postnatal day 7 (P7) mouse cochlear spiral ganglion explants (SGEs), but failed to show effectiveness in P4 SGEs. During the same postnatal period, spontaneous and BDNF-stimulated neurite outgrowth increased. The increased neurite outgrowth in P7 SGEs was not caused by the TrkB/TrkC ligands, BDNF and neurotrophin-3 (NT-3). CONCLUSIONS The age-dependency of induction of neurite outgrowth in SGEs was very likely dependent on presently unidentified factors and/or molecular mechanisms which may also be decisive for the age-dependent efficacy of the small-molecule TrkB receptor agonist THF.
Collapse
Affiliation(s)
- Claudia Frick
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Stefan Fink
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Dominik Schmidbauer
- Inner Ear Laboratory Innsbruck, Medical University Innsbruck, 6020 Innsbruck, Austria; (D.S.); (R.G.)
| | - Francis Rousset
- The Inner Ear & Olfaction Lab, Department of Clinical Neurosciences, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.R.); (P.S.)
| | - Holger Eickhoff
- EMC Microcollections GmbH, 72070 Tübingen, Germany; (H.E.); (K.-H.W.)
| | - Anke Tropitzsch
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Benedikt Kramer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Mannheim, 68167 Mannheim, Germany
| | - Pascal Senn
- The Inner Ear & Olfaction Lab, Department of Clinical Neurosciences, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.R.); (P.S.)
| | - Rudolf Glueckert
- Inner Ear Laboratory Innsbruck, Medical University Innsbruck, 6020 Innsbruck, Austria; (D.S.); (R.G.)
- Tirol Kliniken Innsbruck, University Clinic of Otolaryngology, 6020 Innsbruck, Austria
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Otorhinolaryngology and Head and Neck Surgery, University of Uppsala, 751 85 Uppsala, Sweden;
| | | | - Hubert Löwenheim
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Marcus Müller
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| |
Collapse
|
3
|
Barui AK, Jhelum P, Nethi SK, Das T, Bhattacharya D, B V, Karri S, Chakravarty S, Patra CR. Potential Therapeutic Application of Zinc Oxide Nanoflowers in the Cerebral Ischemia Rat Model through Neuritogenic and Neuroprotective Properties. Bioconjug Chem 2020; 31:895-906. [PMID: 32050064 DOI: 10.1021/acs.bioconjchem.0c00030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Neuritogenesis, a complex process of the sprouting of neurites, plays a vital role in the structural and functional restoration of cerebral ischemia-injured neuronal tissue. Practically, there is no effective long-term treatment strategy for cerebral ischemia in clinical practice to date due to several limitations of conventional therapies, facilitating the urgency to develop new alternative therapeutic approaches. Herein, for the first time we report that pro-angiogenic nanomaterials, zinc oxide nanoflowers (ZONF), exhibit neuritogenic activity by elevating mRNA expression of different neurotrophins, following PI3K/Akt-MAPK/ERK signaling pathways. Further, ZONF administration to global cerebral ischemia-induced Fischer rats shows improved neurobehavior and enhanced synaptic plasticity of neurons via upregulation of Neurabin-2 and NT-3, revealing their neuroprotective activity. Altogether, this study offers the basis for exploitation of angio-neural cross talk of other pro-angiogenic nano/biomaterials for future advancement of alternative treatment strategies for cerebral ischemia, where neuritogenesis and neural repair are highly critical.
Collapse
Affiliation(s)
- Ayan Kumar Barui
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Priya Jhelum
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Susheel Kumar Nethi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Tapatee Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dwaipayan Bhattacharya
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Vinothkumar B
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Shailaja Karri
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
4
|
Van De Water TR. Historical Aspects of Gene Therapy and Stem Cell Therapy in the Treatment of Hearing and Balance Disorder. Anat Rec (Hoboken) 2020; 303:390-407. [DOI: 10.1002/ar.24332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Thomas R. Van De Water
- Cochlear Implant Research Program, Department of Otolaryngology, University of Miami Ear InstituteUniversity of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
5
|
Villarroel-Campos D, Schiavo G, Lazo OM. The many disguises of the signalling endosome. FEBS Lett 2018; 592:3615-3632. [PMID: 30176054 PMCID: PMC6282995 DOI: 10.1002/1873-3468.13235] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/29/2018] [Indexed: 01/09/2023]
Abstract
Neurons are highly complex and polarised cells that must overcome a series of logistic challenges to maintain homeostasis across their morphological domains. A very clear example is the propagation of neurotrophic signalling from distal axons, where target-released neurotrophins bind to their receptors and initiate signalling, towards the cell body, where nuclear and cytosolic responses are integrated. The mechanisms of propagation of neurotrophic signalling have been extensively studied and, eventually, the model of a 'signalling endosome', transporting activated receptors and associated complexes, has emerged. Nevertheless, the exact nature of this organelle remains elusive. In this Review, we examine the evidence for the retrograde transport of neurotrophins and their receptors in endosomes, outline some of their diverse physiological and pathological roles, and discuss the main interactors, morphological features and trafficking destinations of a highly flexible endosomal signalling organelle with multiple molecular signatures.
Collapse
Affiliation(s)
- David Villarroel-Campos
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, UK.,UK Dementia Research Institute at UCL, London, UK.,Discoveries Centre for Regenerative and Precision Medicine, University College London Campus, UK
| | - Oscar Marcelo Lazo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
6
|
Foltran RB, Diaz SL. BDNF isoforms: a round trip ticket between neurogenesis and serotonin? J Neurochem 2016; 138:204-21. [PMID: 27167299 DOI: 10.1111/jnc.13658] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/08/2016] [Accepted: 05/02/2016] [Indexed: 12/12/2022]
Abstract
The brain-derived neurotrophic factor, BDNF, was discovered more than 30 years ago and, like other members of the neurotrophin family, this neuropeptide is synthetized as a proneurotrophin, the pro-BDNF, which is further cleaved to yield mature BDNF. The myriad of actions of these two BDNF isoforms in the central nervous system is constantly increasing and requires the development of sophisticated tools and animal models to refine our understanding. This review is focused on BDNF isoforms, their participation in the process of neurogenesis taking place in the hippocampus of adult mammals, and the modulation of their expression by serotonergic agents. Interestingly, around this triumvirate of BDNF, serotonin, and neurogenesis, a series of recent research has emerged with apparently counterintuitive results. This calls for an exhaustive analysis of the data published so far and encourages thorough work in the quest for new hypotheses in the field. BDNF is synthetized as a pre-proneurotrophin. After removal of the pre-region, proBDNF can be cleaved by intracellular or extracellular proteases. Mature BDNF can bind TrkB receptors, promoting their homodimerization and intracellular phosphorylation. Phosphorylated-TrkB can activate three different signaling pathways. Whereas G-protein-coupled receptors can transactivate TrkB receptors, truncated forms can inhibit mBDNF signaling. Pro-BDNF binds p75(NTR) by its mature domain, whereas the pro-region binds co-receptors.
Collapse
Affiliation(s)
- Rocío Beatriz Foltran
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Fac. de Medicina - UBA, Buenos Aires, Argentina
| | - Silvina Laura Diaz
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Fac. de Medicina - UBA, Buenos Aires, Argentina
| |
Collapse
|
7
|
de Almeida JFA, Chen P, Henry MA, Diogenes A. Stem cells of the apical papilla regulate trigeminal neurite outgrowth and targeting through a BDNF-dependent mechanism. Tissue Eng Part A 2015; 20:3089-100. [PMID: 24837134 DOI: 10.1089/ten.tea.2013.0347] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Regenerative endodontic procedures have become a valuable alternative for the treatment of immature teeth with pulp necrosis. In addition to resolution of periradicular pathosis and promotion of continued root development, positive vitality testing has been observed in some regenerative clinical cases. Importantly, the positive response to electric stimulation of the regenerated tissue requires targeting of periradicular axons into the previously empty root canal space. However, the mechanism by which this process occurs is largely unknown. Since stem cells of the apical papilla (SCAP) have been proposed to populate the root canal following regenerative endodontic procedures, we hypothesized that SCAP regulate neurite outgrowth and axonal targeting. To test this hypothesis, we established primary co-cultures of human SCAP and rat trigeminal neurons, and performed neurite outgrowth assays using ELISA and confocal microscopy to determine the effect of increasing concentration of SCAP on the total neurite outgrowth and axonal targeting. In addition, we evaluated whether SCAP evoked axonal targeting in vivo using a matrigel subcutaneous implant assay. Data were analyzed by ANOVA with Bonferroni's post hoc test, and significance was set at p<0.05. The results demonstrated that SCAP release a soluble factor that regulates neurite outgrowth from cultured trigeminal neurons. Next, we demonstrated that this effect is completely abolished by pretreatment with a neutralizing antibody to brain-derived neurotrophic factor (BDNF), but not by antibodies to other neurotrophins. Further, SCAP release BDNF in a concentration-dependent manner as detected by ELISA, and trigger directed axonal targeting both in vitro and in vivo as demonstrated by microfluidic and matrigel implant experiments, respectively. Collectively, these results suggest that SCAP may be responsible for the chemical signal driving axons to target regenerated tissue via a BDNF-dependent mechanism.
Collapse
Affiliation(s)
- Jose Flavio A de Almeida
- 1 Department of Restorative Dentistry, Endodontics Division, Piracicaba Dental School, University of Campinas , Piracicaba, São Paulo, Brazil
| | | | | | | |
Collapse
|
8
|
Kondo K, Pak K, Chavez E, Mullen L, Euteneuer S, Ryan AF. Changes in responsiveness of rat spiral ganglion neurons to neurotrophins across age: differential regulation of survival and neuritogenesis. Int J Neurosci 2013; 123:465-75. [PMID: 23301942 DOI: 10.3109/00207454.2013.764497] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Developmental changes in responsiveness of rat spiral ganglion neurons (SGNs) to neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF) were examined using an explant culture system. Spiral ganglion (SG) explants at embryonic Day 18 (E18), postnatal Day 0 (P0), P5, P10 and P20 were cultured with the addition of either NT-3 or BDNF at various concentrations (0.1-100 ng/ml) and analyzed the dose-response characteristics of three parameters: SGN survival, the number of neurites emanating from the explants and the length of neurite extension. In E18 cultures, SGN survival and neurite number were enhanced more strongly by NT-3 than by the BDNF. As the explants became more mature, the effects of NT-3 decreased, whereas those of BDNF increased, peaking at P0. Although the intrinsic capacity of SGNs to produce and extend neurites declined considerably by P20, they still retained the capacity to respond to both NT-3 and BDNF. These temporal patterns in responsiveness of SGNs to neurotrophins correspond well to the expression pattern of the two neurotrophins in cochlear sensory epithelium in vivo and also correlate with the time course of developmental events in SGNs such as cell death and the establishment of mature hair cell innervation patterns.
Collapse
Affiliation(s)
- Kenji Kondo
- Division of Otolaryngology and Neurosciences, Department of Surgery and Veterans Administration Medical Center, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
9
|
Palmgren B, Jiao Y, Novozhilova E, Stupp SI, Olivius P. Survival, migration and differentiation of mouse tau-GFP embryonic stem cells transplanted into the rat auditory nerve. Exp Neurol 2012; 235:599-609. [DOI: 10.1016/j.expneurol.2012.03.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 02/18/2012] [Accepted: 03/25/2012] [Indexed: 01/13/2023]
|
10
|
Aburto MR, Magariños M, Leon Y, Varela-Nieto I, Sanchez-Calderon H. AKT signaling mediates IGF-I survival actions on otic neural progenitors. PLoS One 2012; 7:e30790. [PMID: 22292041 PMCID: PMC3264639 DOI: 10.1371/journal.pone.0030790] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 12/29/2011] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Otic neurons and sensory cells derive from common progenitors whose transition into mature cells requires the coordination of cell survival, proliferation and differentiation programmes. Neurotrophic support and survival of post-mitotic otic neurons have been intensively studied, but the bases underlying the regulation of programmed cell death in immature proliferative otic neuroblasts remains poorly understood. The protein kinase AKT acts as a node, playing a critical role in controlling cell survival and cell cycle progression. AKT is activated by trophic factors, including insulin-like growth factor I (IGF-I), through the generation of the lipidic second messenger phosphatidylinositol 3-phosphate by phosphatidylinositol 3-kinase (PI3K). Here we have investigated the role of IGF-dependent activation of the PI3K-AKT pathway in maintenance of otic neuroblasts. METHODOLOGY/PRINCIPAL FINDINGS By using a combination of organotypic cultures of chicken (Gallus gallus) otic vesicles and acoustic-vestibular ganglia, Western blotting, immunohistochemistry and in situ hybridization, we show that IGF-I-activation of AKT protects neural progenitors from programmed cell death. IGF-I maintains otic neuroblasts in an undifferentiated and proliferative state, which is characterised by the upregulation of the forkhead box M1 (FoxM1) transcription factor. By contrast, our results indicate that post-mitotic p27(Kip)-positive neurons become IGF-I independent as they extend their neuronal processes. Neurons gradually reduce their expression of the Igf1r, while they increase that of the neurotrophin receptor, TrkC. CONCLUSIONS/SIGNIFICANCE Proliferative otic neuroblasts are dependent on the activation of the PI3K-AKT pathway by IGF-I for survival during the otic neuronal progenitor phase of early inner ear development.
Collapse
Affiliation(s)
- Maria R. Aburto
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
- Departamento de Biologia, Universidad Autonoma de Madrid, Madrid, Spain
| | - Yolanda Leon
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- Departamento de Biologia, Universidad Autonoma de Madrid, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
| | - Hortensia Sanchez-Calderon
- Instituto de Investigaciones Biomedicas “Alberto Sols”, CSIC-UAM, Madrid, Spain
- CIBERER, Unit 761, ISCIII, Madrid, Spain
| |
Collapse
|
11
|
Jang Y, Jung J, Kim H, Oh J, Jeon JH, Jung S, Kim KT, Cho H, Yang DJ, Kim SM, Kim IB, Song MR, Oh U. Axonal neuropathy-associated TRPV4 regulates neurotrophic factor-derived axonal growth. J Biol Chem 2011; 287:6014-24. [PMID: 22187434 DOI: 10.1074/jbc.m111.316315] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Spinal muscular atrophy and hereditary motor and sensory neuropathies are characterized by muscle weakness and atrophy caused by the degenerations of peripheral motor and sensory nerves. Recent advances in genetics have resulted in the identification of missense mutations in TRPV4 in patients with these hereditary neuropathies. Neurodegeneration caused by Ca(2+) overload due to the gain-of-function mutation of TRPV4 was suggested as the molecular mechanism for the neuropathies. Despite the importance of TRPV4 mutations in causing neuropathies, the precise role of TRPV4 in the sensory/motor neurons is unknown. Here, we report that TRPV4 mediates neurotrophic factor-derived neuritogenesis in developing peripheral neurons. TRPV4 was found to be highly expressed in sensory and spinal motor neurons in early development as well as in the adult, and the overexpression or chemical activation of TRPV4 was found to promote neuritogenesis in sensory neurons as well as PC12 cells, whereas its knockdown and pharmacologic inhibition had the opposite effect. More importantly, nerve growth factor or cAMP treatment up-regulated the expression of phospholipase A(2) and TRPV4. Neurotrophic factor-derived neuritogenesis appears to be regulated by the phospholipase A(2)-mediated TRPV4 pathway. These findings show that TRPV4 mediates neurotrophic factor-induced neuritogenesis in developing peripheral nerves. Because neurotrophic factors are essential for the maintenance of peripheral nerves, these findings suggest that aberrant TRPV4 activity may lead to some types of pathology of sensory and motor nerves.
Collapse
Affiliation(s)
- Yongwoo Jang
- Sensory Research Center, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fantetti KN, Fekete DM. Dissection and culture of chick statoacoustic ganglion and spinal cord explants in collagen gels for neurite outgrowth assays. J Vis Exp 2011:3600. [PMID: 22215150 PMCID: PMC3369645 DOI: 10.3791/3600] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The sensory organs of the chicken inner ear are innervated by the peripheral processes of statoacoustic ganglion (SAG) neurons. Sensory organ innervation depends on a combination of axon guidance cues and survival factors located along the trajectory of growing axons and/or within their sensory organ targets. For example, functional interference with a classic axon guidance signaling pathway, semaphorin-neuropilin, generated misrouting of otic axons. Also, several growth factors expressed in the sensory targets of the inner ear, including Neurotrophin-3 (NT-3) and Brain Derived Neurotrophic Factor (BDNF), have been manipulated in transgenic animals, again leading to misrouting of SAG axons. These same molecules promote both survival and neurite outgrowth of chick SAG neurons in vitro. Here, we describe and demonstrate the in vitro method we are currently using to test the responsiveness of chick SAG neurites to soluble proteins, including known morphogens such as the Wnts, as well as growth factors that are important for promoting SAG neurite outgrowth and neuron survival. Using this model system, we hope to draw conclusions about the effects that secreted ligands can exert on SAG neuron survival and neurite outgrowth. SAG explants are dissected on embryonic day 4 (E4) and cultured in three-dimensional collagen gels under serum-free conditions for 24 hours. First, neurite responsiveness is tested by culturing explants with protein-supplemented medium. Then, to ask whether point sources of secreted ligands can have directional effects on neurite outgrowth, explants are co-cultured with protein-coated beads and assayed for the ability of the bead to locally promote or inhibit outgrowth. We also include a demonstration of the dissection (modified protocol) and culture of E6 spinal cord explants. We routinely use spinal cord explants to confirm bioactivity of the proteins and protein-soaked beads, and to verify species cross-reactivity with chick tissue, under the same culture conditions as SAG explants. These in vitro assays are convenient for quickly screening for molecules that exert trophic (survival) or tropic (directional) effects on SAG neurons, especially before performing studies in vivo. Moreover, this method permits the testing of individual molecules under serum-free conditions, with high neuron survival.
Collapse
|
13
|
Shibata SB, Budenz CL, Bowling SA, Pfingst BE, Raphael Y. Nerve maintenance and regeneration in the damaged cochlea. Hear Res 2011; 281:56-64. [PMID: 21596129 PMCID: PMC3196294 DOI: 10.1016/j.heares.2011.04.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 04/22/2011] [Accepted: 04/23/2011] [Indexed: 12/22/2022]
Abstract
Following the onset of sensorineural hearing loss, degeneration of mechanosensitive hair cells and spiral ganglion cells (SGCs) in humans and animals occurs to variable degrees, with a trend for greater neural degeneration with greater duration of deafness. Emergence of the cochlear implant prosthesis has provided much needed aid to many hearing impaired patients and has become a well-recognized therapy worldwide. However, ongoing peripheral nerve fiber regression and subsequent degeneration of SGC bodies can reduce the neural targets of cochlear implant stimulation and diminish its function. There is increasing interest in bio-engineering approaches that aim to enhance cochlear implant efficacy by preventing SGC body degeneration and/or regenerating peripheral nerve fibers into the deaf sensory epithelium. We review the advancements in maintaining and regenerating nerves in damaged animal cochleae, with an emphasis on the therapeutic capacity of neurotrophic factors delivered to the inner ear after an insult. Additionally, we summarize the histological process of neuronal degeneration in the inner ear and describe different animal models that have been employed to study this mechanism. Research on enhancing the biological infrastructure of the deafened cochlea in order to improve cochlear implant efficacy is of immediate clinical importance.
Collapse
Affiliation(s)
- Seiji B. Shibata
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Cameron L. Budenz
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Sara A. Bowling
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Bryan E. Pfingst
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| |
Collapse
|
14
|
Meen E, Blakley B, Quddusi T. Brain-derived nerve growth factor in the treatment of sensorineural hearing loss. Laryngoscope 2009; 119:1590-3. [PMID: 19479743 DOI: 10.1002/lary.20515] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVES/HYPOTHESIS A possible medical treatment for sensorineural hearing loss using brain-derived nerve growth factor (BDNF) was explored. The hypothesis is that direct intracochlear application of BDNF will result in improved hearing. STUDY DESIGN Animal research study. METHODS Significant hearing loss was created using cisplatin in 11 guinea pigs. One month later, bilateral cochleostomies were performed placing 0.05 microg of BDNF in one cochlea of each animal prior to plugging with connective tissue. The other cochlea served as a control. Auditory brain-stem response (ABR) testing was then carried out for three months at 6,000, 8,000, 12,000, and 24,000 Hz. RESULTS ABR thresholds were better in the treated ear for all frequencies. Threshold differences were statistically significantly better two months after treatment (general linear model, repeated measures P = .045). CONCLUSIONS Intracochlear application of BDNF may prevent hearing loss.
Collapse
Affiliation(s)
- Eric Meen
- Department of Otolaryngology, University of Manitoba, Winnipeg, Canada.
| | | | | |
Collapse
|
15
|
El Seady R, Huisman MA, Löwik CWGM, Frijns JHM. Uncomplicated differentiation of stem cells into bipolar neurons and myelinating glia. Biochem Biophys Res Commun 2008; 376:358-62. [PMID: 18789899 DOI: 10.1016/j.bbrc.2008.08.166] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 08/28/2008] [Indexed: 02/07/2023]
Abstract
Epidermal neural crest stem cells (EPI-NCSCs), derived from the bulge of hair follicles, appear to be promising donor stem cell candidates. In the current work, EPI-NCSCs were harvested from rodents and humans. Isolation procedures revealed high levels of nestin-positive neural stem cells and the percentage of human neural stem cells (95+/-0.6%) is even higher than the percentage found in cultures of hair follicles from rodents (90+/-0.9%). Furthermore, differentiation of EPI-NCSCs into bipolar neurons, myelinating Schwann cells and oligodendrocytes occurred by applying a simple and straightforward method. Bipolar neurons could be obtained by culturing on a collagen matrix and are of great interest for auditory neuron regeneration since auditory neurons are bipolar. We propose that this type of stem cells, would make an excellent model for autologous transplantation and offers great potential for neural regeneration in diseases, such as Parkinson's and Alzheimer's disease.
Collapse
Affiliation(s)
- Ragad El Seady
- Department of Otorhinolaryngology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | | | | |
Collapse
|
16
|
Van De Water TR, Staecker H, Ernfors P, Moonen G, Lefebvre PP. Neurotrophic factors as pharmacological agents for the treatment of injured auditory neurons. CIBA FOUNDATION SYMPOSIUM 2007; 196:149-62; discussion 162-6. [PMID: 8866133 DOI: 10.1002/9780470514863.ch11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mature auditory neurons degenerate in response to a loss of target-derived trophic factors. Neurotrophic factors influence the health and viability of auditory neurons. This suggests that neurotrophic factors can be used as therapeutic agents to prevent neuronal cell death and to initiate repair of damaged neuronal processes in the injured auditory system. Both in vitro and in vivo experiments have been performed to determine which of the vast array of neurotrophic factors affect mature auditory neurons and how they can be delivered to the sites of injury within the auditory system. Neurotrophin 3 was found to be a major survival factor for auditory neurons. Brain-derived neurotrophic factor is a minor survival factor for auditory neurons and nerve growth factor, although not promoting survival, does elicit the repair of neuronal processes. Fibroblast growth factor 2, transforming growth factor beta 1 and ciliary neurotrophic factor function as injury-response factors in the auditory system. Combination of different classes of growth factors has an additive effect on neuronal survival. Growth factors may be able to be delivered to sites of injury within the cochlea by either direct perfusion or gene therapy (e.g. a defective virus expressing a growth factor gene).
Collapse
Affiliation(s)
- T R Van De Water
- Department of Otolaryngology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
17
|
Zhou Z, Liu Q, Davis RL. Complex regulation of spiral ganglion neuron firing patterns by neurotrophin-3. J Neurosci 2006; 25:7558-66. [PMID: 16107643 PMCID: PMC6725401 DOI: 10.1523/jneurosci.1735-05.2005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Auditory information is conveyed into the CNS via the spiral ganglion neurons, cells that possess distinctive electrophysiological properties that vary according to their cochlear innervation. Neurons from the base of the cochlea fire action potentials with shorter latencies and durations with more rapid accommodation than apical neurons (Adamson et al., 2002b). Interestingly, these features are altered by exposure to brain-derived neurotrophic factor and neurotrophin-3 (NT-3), suggesting that the electrophysiological diversity is not preprogrammed into the neurons but instead results from extrinsic regulation. In support of this, gradients of neurotrophins exist in the cochlea that could account for the apex- base differences in firing. To understand the determinants of spiral ganglion function, we characterized the NT-3 concentration dependence and mode of action on spiral ganglion neurons. Whole-cell current-clamp recordings were made from mouse basal spiral ganglion neurons (postnatal day 5) exposed to different concentrations of NT-3 for 3 d in vitro. Measurements of accommodation, latency, onset time course, and action potential latency revealed a nonmonotonic dependence on NT-3 concentration, with a peak effect occurring at 10 ng/ml. Addition of NT-3 at different time points showed that neurotrophin exposure altered the firing features of existing neurons rather than supporting differential survival. These experiments establish that the electrophysiological phenotype of spiral ganglion neurons depends critically on the precise concentration of NT-3 and that the functional organization of this component of the peripheral auditory system results from a complex interplay between multiple kinds of neurotrophins and their cognate receptors.
Collapse
Affiliation(s)
- Zhiping Zhou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Degeneration of spiral ganglion neurons following hair cell loss carries critical implications for efforts to rehabilitate severe cases of hearing loss with cochlear implants or hair cell regeneration. This review considers recently identified neurotrophic factors and therapeutic strategies which promote spiral ganglion neuron survival and neurite growth. Replacement of these factors may help preserve or regenerate the auditory nerve in patients with extensive hair cell loss. RECENT FINDINGS Spiral ganglion neurons depend on neurotrophic factors supplied by hair cells and other targets for their development and continued survival. Loss of this trophic support leads to spiral ganglion neuron death via apoptosis. Hair cells support spiral ganglion neuron survival by producing several peptide neurotrophic factors such as neurotrophin-3 and glial derived neurotrophic factor. In addition, neurotransmitter release from the hair cells drives membrane electrical activity in spiral ganglion neurons which also supports their survival. In animal models, replacement of peptide neurotrophic factors or electrical stimulation with an implanted electrode attenuates spiral ganglion neuron degeneration following deafferentation. Cell death inhibitors can also preserve spiral ganglion neuron populations. Preliminary studies show that transfer of stem cells or neurons from other ganglia are two potential strategies to replace lost spiral ganglion neurons. Inducing the regrowth of spiral ganglion neuron peripheral processes to approximate or contact cochlear implant electrodes may help optimize signaling from a diminished population of neurons. SUMMARY Recent studies of spiral ganglion neuron development and survival have identified several trophic and neuritogenic factors which protect these specialized cells from degeneration following hair cell loss. While still preliminary, such strategies show promise for future clinical applications.
Collapse
Affiliation(s)
- Pamela C Roehm
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
19
|
Sliwinska-Kowalska M, Rzadzinska A, Rajkowska E, Malczyk M. Expression of bFGF and NGF and their receptors in chick’s auditory organ following overexposure to noise. Hear Res 2005; 210:93-103. [PMID: 16243462 DOI: 10.1016/j.heares.2005.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2005] [Revised: 04/17/2005] [Accepted: 08/13/2005] [Indexed: 11/30/2022]
Abstract
Growth factors are known to activate signaling cascades for DNA replication; they participate in the regulation of cell differentiation and are required as positive signals for cell survival. Thus, many of them may be regarded as potential candidates stimulating regeneration processes in the inner ear. We analyzed the expression of basic fibroblast growth factor (bFGF) and nerve growth factor (NGF) and their receptor (bFGFR and NGFR)-like immunoreactivity in chick basilar papillae, along with bFGF and NGF mRNA expression. The evaluation was made 1 and 5 days after exposure to wide-band noise with two increasing levels of acoustic energy. For both factors, the immunoreactivity was shown predominantly in the middle part of basilar papilla, in noise-exposed, but not control birds. It was localized in the cytoplasm of hair cells, nuclei of supporting cells and cytoplasm of ganglion cells. Strong immunoreactivity of bFGFR and NGFR was found both in control and noise-exposed animals, with the cell localization similar to that of growth factors. The increase in mRNA expression for bFGF and NGF was found in noise-exposed animals only after lower exposure to noise, on day 5 after exposure (p<0.01). A lack of increased expression after higher exposure could be excused by larger damage of hair cells followed by the increase of mRNA for beta-actin to which the results were referred. The results suggest bFGF and NGF involvement in postinjury regeneration of the basilar papilla.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Base Sequence
- Chickens
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/metabolism
- Gene Expression
- Hearing Loss, Noise-Induced/genetics
- Hearing Loss, Noise-Induced/metabolism
- Hearing Loss, Noise-Induced/pathology
- Hearing Loss, Noise-Induced/physiopathology
- Immunohistochemistry
- Nerve Growth Factor/genetics
- Nerve Growth Factor/metabolism
- Organ of Corti/injuries
- Organ of Corti/metabolism
- Organ of Corti/pathology
- Organ of Corti/physiopathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Nerve Growth Factor/genetics
- Receptor, Nerve Growth Factor/metabolism
- Regeneration/genetics
- Regeneration/physiology
Collapse
Affiliation(s)
- Mariola Sliwinska-Kowalska
- Department of Physical Hazards, The Nofer Institute of Occupational Medicine, Teresy St. 8, 91-348 Lodz, Poland.
| | | | | | | |
Collapse
|
20
|
Nicholl AJ, Kneebone A, Davies D, Cacciabue-Rivolta DI, Rivolta MN, Coffey P, Holley MC. Differentiation of an auditory neuronal cell line suitable for cell transplantation. Eur J Neurosci 2005; 22:343-53. [PMID: 16045487 DOI: 10.1111/j.1460-9568.2005.04213.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The auditory neuroblast cell line US/VOT-N33 (N33), which is conditionally immortal, was studied as an in vitro model for the differentiation of spiral ganglion neurons (SGNs) and as a candidate for cell transplantation in rodents. It expresses numerous molecular markers characteristic of auditory neuroblasts, including the transcription factors GATA3, NeuroD, Brn3a and Islet1, as well as the neuronal cytoskeletal protein beta3-tubulin. It displays active migratory behaviour in vitro and in vivo. In the presence of the fibroblast growth factors FGF1 or FGF2 it differentiates bipolar morphologies similar to those of native SGNs. In coculture with neonatal cochlear tissue it is repelled from epithelial surfaces but not from native SGNs, alongside which it extends parallel neuronal processes. When injected into the retina in vivo, EGFP-labelled N33 cells were traced for 1-2 weeks and migrated rapidly within the subretinal space. Cells that found their way into the retinal ganglion cell layer extended multiple processes but did not express beta3-tubulin. The ability of N33 to migrate, to differentiate, to localize with native SGNs in vitro and to survive in vivo suggests that they provide an effective model for SGN differentiation and for cell transplantation into the ear.
Collapse
Affiliation(s)
- A J Nicholl
- Department of Biomedical Sciences, Addison Building, Western Bank, Sheffield, S10 2TN, UK
| | | | | | | | | | | | | |
Collapse
|
21
|
Xu B, Michalski B, Racine RJ, Fahnestock M. Continuous infusion of neurotrophin-3 triggers sprouting, decreases the levels of TrkA and TrkC, and inhibits epileptogenesis and activity-dependent axonal growth in adult rats. Neuroscience 2003; 115:1295-308. [PMID: 12453498 DOI: 10.1016/s0306-4522(02)00384-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurotrophin-3 (NT-3), a member of the neurotrophin family of neurotrophic factors, is important for cell survival, axonal growth and neuronal plasticity. Epileptiform activation can regulate the expression of neurotrophins, and increases or decreases in neurotrophins can affect both epileptogenesis and seizure-related axonal growth. Interestingly, the expression of nerve growth factor and brain-derived neurotrophic factor is rapidly up-regulated following seizures, while NT-3 mRNA remains unchanged or undergoes a delayed down-regulation, suggesting that NT-3 might have a different function in epileptogenesis. In the present study, we demonstrate that continuous intraventricular infusion of NT-3 in the absence of kindling triggers mossy fiber sprouting in the inner molecular layer of the dentate gyrus and the stratum oriens of the CA3 region. Furthermore, despite this NT-3-related sprouting effect, continuous infusion of NT-3 retards the development of behavioral seizures and inhibits kindling-induced mossy fiber sprouting in the inner molecular layer of the dentate gyrus. We also show that prolonged infusion of NT-3 leads to a decrease in kindling-induced Trk phosphorylation and a down-regulation of the high-affinity Trk receptors, TrkA and TrkC, suggesting an involvement of both cholinergic nerve growth factor receptors and hippocampal NT-3 receptors in these effects. Our results demonstrate an important inhibitory role for NT-3 in seizure development and seizure-related synaptic reorganization.
Collapse
MESH Headings
- Animals
- Cell Count
- Cytochrome c Group/pharmacology
- Drug Administration Schedule
- Epilepsy/drug therapy
- Epilepsy/metabolism
- Epilepsy/physiopathology
- Growth Cones/drug effects
- Growth Cones/metabolism
- Kindling, Neurologic/drug effects
- Kindling, Neurologic/metabolism
- Male
- Molecular Weight
- Mossy Fibers, Hippocampal/drug effects
- Mossy Fibers, Hippocampal/growth & development
- Mossy Fibers, Hippocampal/metabolism
- Neuronal Plasticity/drug effects
- Neuronal Plasticity/physiology
- Neuropil/cytology
- Neuropil/drug effects
- Neurotrophin 3/metabolism
- Neurotrophin 3/pharmacology
- Phosphorylation/drug effects
- Rats
- Rats, Long-Evans
- Receptor Protein-Tyrosine Kinases/drug effects
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, trkA/drug effects
- Receptor, trkA/metabolism
- Receptor, trkB/drug effects
- Receptor, trkB/metabolism
- Receptor, trkC/drug effects
- Receptor, trkC/metabolism
- Seizures/drug therapy
- Seizures/metabolism
- Seizures/physiopathology
Collapse
Affiliation(s)
- B Xu
- Department of Psychology, McMaster University, L8S 4K1, Hamilton, ON, Canada
| | | | | | | |
Collapse
|
22
|
Alsina B, Giraldez F, Varela-Nieto I. Growth Factors and Early Development of Otic Neurons: Interactions between Intrinsic and Extrinsic Signals. Curr Top Dev Biol 2003; 57:177-206. [PMID: 14674481 DOI: 10.1016/s0070-2153(03)57006-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Berta Alsina
- DCEXS-Universitat Pomepu Fabra, Dr Aiguader 80, 08003 Barcelona, Spain
| | | | | |
Collapse
|
23
|
Lalwani AK, Han JJ, Castelein CM, Carvalho GJ, Mhatre AN. In vitro and in vivo assessment of the ability of adeno-associated virus-brain-derived neurotrophic factor to enhance spiral ganglion cell survival following ototoxic insult. Laryngoscope 2002; 112:1325-34. [PMID: 12172239 DOI: 10.1097/00005537-200208000-00001] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES/HYPOTHESIS Auditory dysfunction following ototoxic insult results from loss of cochlear hair cells. Secondary degeneration of auditory neurons ensues from withdrawal of neurotrophic support from hair cells and can be prevented with administration of neurotrophins. Administration of adeno-associated virus containing the gene for brain-derived neurotrophic factor will promote spiral ganglion neuron survival after the destruction of hair cells. METHODS Prevention of aminoglycoside-induced spiral ganglion neuron loss through the expression of brain-derived neurotrophic factor mediated by means of the adeno-associated virus was tested in vitro in cochlear explants and in vivo in mammalian cochlea. RESULTS Neuronal survival was significantly enhanced in adeno-associated virus-brain-derived neurotrophic factor transfected rat cochlear explants compared with control samples (30% vs. 19%, P <.05) following exposure to aminoglycoside. Following deafening with aminoglycoside and loop diuretic and introduction of adeno-associated virus-brain-derived neurotrophic factor through osmotic minipump, the experimental group of animals infused with adeno-associated virus-brain-derived neurotrophic factor displayed enhanced spiral ganglion neuron survival in the basal turn of the cochlea when compared with the control group infused with adeno-associated virus containing green fluorescent protein reporter gene. CONCLUSIONS Administration of adeno-associated virus-brain-derived neurotrophic factor enhances spiral ganglion neuron survival following ototoxic exposure in vitro and in vivo. These studies lay the groundwork for further exploration of its application as an adjunct therapy for patients undergoing cochlear implantation because the success of implantation depends directly on the population of neurons available for electrical stimulation.
Collapse
Affiliation(s)
- Anil K Lalwani
- Laboratory of Molecular Otology, Epstein Laboratories, Department of Otolaryngology-Head and Neck Surgery, University of California at San Francisco, 94143-0342, USA.
| | | | | | | | | |
Collapse
|
24
|
Cristobal R, Popper P, Lopez I, Micevych P, De Vellis J, Honrubia V. In vivo and in vitro localization of brain-derived neurotrophic factor, fibroblast growth factor-2 and their receptors in the bullfrog vestibular end organs. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 102:83-99. [PMID: 12191497 DOI: 10.1016/s0169-328x(02)00202-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The inner ear sensory epithelia of vertebrates are composed mainly of supporting cells and hair cells (HCs). Brain-derived neurotrophic factor (BDNF) and fibroblast growth factor-2 (FGF-2) are trophins that are believed to play an essential role in the development and innervation of inner ear epithelia. Both trophins also may play a crucial role in the maintenance and regeneration of hair cells in the adult vertebrate ear. In the bullfrog vestibular system, hair cells are produced throughout life, and the epithelia regenerates following ototoxicity. The expression of BDNF and FGF-2 in the vestibular organs of the adult bullfrog was investigated at a cellular level both in histological sections and in vitro in dissociated cell cultures. In histological sections of the crista ampullaris, in situ hybridization and immunocytochemical techniques demonstrated that HCs express both BDNF and its receptor trkB, while the supporting cells express the receptor trkB alone. Following dissociation and in vitro cell culture no changes in the pattern of BDNF and trkB receptor were observed. Immunocytochemical studies demonstrated that in vivo hair cells express FGF-2 and the receptors FGFR-1 and FGFR-2 while supporting cells do not express either molecule. Following dissociation, HCs continue to express FGF-2 and its two receptors, while supporting cells upregulate the expression of FGF-2 and its receptor FGFR-2. These data confirm the potential role of BDNF and FGF-2 trophic regulation of the sensory epithelia of the adult inner ear. The findings suggest that BDNF has a role in the maintenance of the vestibular epithelia while FGF-2 may regulate the proliferation of supporting cells.
Collapse
MESH Headings
- Animals
- Autocrine Communication/physiology
- Brain-Derived Neurotrophic Factor/metabolism
- Cell Communication/physiology
- Cells, Cultured
- Fibroblast Growth Factor 2/metabolism
- Hair Cells, Vestibular/cytology
- Hair Cells, Vestibular/metabolism
- Immunohistochemistry
- Labyrinth Supporting Cells/cytology
- Labyrinth Supporting Cells/metabolism
- Rana catesbeiana/anatomy & histology
- Rana catesbeiana/metabolism
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, Fibroblast Growth Factor, Type 2
- Receptor, trkB/metabolism
- Receptors, Fibroblast Growth Factor/metabolism
- Vestibule, Labyrinth/cytology
- Vestibule, Labyrinth/metabolism
Collapse
Affiliation(s)
- Ricardo Cristobal
- Victor Goodhill Ear Center, Division of Head and Neck Surgery, University of California, Los Angeles School of Medicine, CHS, Room 62-129, 10833 Le Conte Ave., Los Angeles, CA 90095-1624, USA
| | | | | | | | | | | |
Collapse
|
25
|
Carnicero E, Knipper M, Tan J, Alonso MT, Schimmang T. Herpes simplex virus type 1-mediated transfer of neurotrophin-3 stimulates survival of chicken auditory sensory neurons. Neurosci Lett 2002; 321:149-52. [PMID: 11880194 DOI: 10.1016/s0304-3940(01)02501-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Neurotrophin-3 (NT-3) is one of the most potent stimulators for survival of auditory sensory neurons. Viral transfer of neurotrophins into auditory neurons may offer a route to provide a permanent supply of the growth factor and guarantee their long-term survival. Herpes simplex virus type 1 (HSV-1)-based vectors have demonstrated their effectiveness to transfer genes into peripheral sensory neurons. In the present report, we have produced a HSV-1-based amplicon vector expressing NT-3. This vector efficiently infects isolated auditory neurons and stimulates their survival during distinct developmental stages of the inner ear. Therefore, this vector may present a unique entry point to develop therapies preventing or treating hearing impairment caused by the degeneration of auditory neurons.
Collapse
Affiliation(s)
- Estela Carnicero
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Cientificas, Departamento de Bioquímica, Biología Molecular y Fisiología, Facultad de Medicina, 47005 Valladolid, Spain
| | | | | | | | | |
Collapse
|
26
|
Hansen MR, Vijapurkar U, Koland JG, Green SH. Reciprocal signaling between spiral ganglion neurons and Schwann cells involves neuregulin and neurotrophins. Hear Res 2001; 161:87-98. [PMID: 11744285 DOI: 10.1016/s0378-5955(01)00360-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
To investigate the role of neuron-glial cell interactions in the auditory nerve, we asked whether spiral ganglion neurons (SGNs) express neuregulin and whether neuregulin regulates proliferation and/or neurotrophin expression in spiral ganglion Schwann cells (SGSCs). Using immunocytochemistry, we found that type I and type II SGNs express neuregulin in vivo and in vitro. Cultured SGSCs express the neuregulin receptors ErbB2 and ErbB3, but not ErbB4. Neuregulin activates ErbB2 and ErbB3 in cultured SGSCs, evidenced by increased tyrosine phosphorylation of the receptors following neuregulin treatment. Neuregulin treatment increased the proliferation rate of cultured SGSCs by 2.5-fold. Fibroblast growth factor-2 (FGF-2) and transforming growth factor beta (TGF-beta) also increased SGSC proliferation. The mitogenic effect of neuregulin and FGF-2 was blocked by inhibition of mitogen-activated protein kinase signaling but not by inhibition of phosphatidylinositol-3'-OH kinase. Using RT-PCR, we found that cultured SGSCs express neurotrophins, including brain-derived neurotrophic factor and neurotrophin-3 (NT-3), raising the possibility that SGSCs contribute to the trophic support of SGNs. Treatment with neither neuregulin nor TGF-beta increased neurotrophin expression in cultured SGSCs, as had been observed in developing sympathetic ganglia, but appeared to negatively regulate NT-3 expression. Thus, neuregulin and neurotrophins may mediate reciprocal neuron-glial interactions in the auditory nerve.
Collapse
Affiliation(s)
- M R Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
27
|
Keith CH, Wilson MT. Factors controlling axonal and dendritic arbors. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 205:77-147. [PMID: 11336394 DOI: 10.1016/s0074-7696(01)05003-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The sculpting and maintenance of axonal and dendritic arbors is largely under the control of molecules external to the cell. These factors include both substratum-associated and soluble factors that can enhance or inhibit the outgrowth of axons and dendrites. A large number of factors that modulate axonal outgrowth have been identified, and the first stages of the intracellular signaling pathways by which they modify process outgrowth have been characterized. Relatively fewer factors and pathways that affect dendritic outgrowth have been described. The factors that affect axonal arbors form an incompletely overlapping set with those that affect dendritic arbors, allowing selective control of the development and maintenance of these critical aspects of neuronal morphology.
Collapse
Affiliation(s)
- C H Keith
- Department of Cellular Biology. University of Georgia, Athens, 30605, USA
| | | |
Collapse
|
28
|
Zha XM, Bishop JF, Hansen MR, Victoria L, Abbas PJ, Mouradian MM, Green SH. BDNF synthesis in spiral ganglion neurons is constitutive and CREB-dependent. Hear Res 2001; 156:53-68. [PMID: 11377882 DOI: 10.1016/s0378-5955(01)00267-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), which supports spiral ganglion neuron (SGN) survival in vivo and in vitro, is synthesized by SGNs. The BDNF gene generates multiple different transcripts, each from its own promoter region. Using reverse transcriptase-polymerase chain reaction (RT-PCR), we find that SGNs express only the downstream transcripts III and IV in vivo and in vitro. Using RT-PCR assays of BDNF transcripts and transfection of BDNF promoter-reporter constructs, we tested the hypothesis, originally derived from studies of cortical neurons, that depolarization induces BDNF expression via a signaling pathway that includes Ca2+/calmodulin-dependent kinases (CaMKs) and the transcription factor, Ca2+/cyclic AMP response element binding protein (CREB). In contrast, we found that in SGNs in vivo BDNF expression is constitutive and is not increased by electrical activation. Similarly, BDNF expression in vitro is not increased by stimuli that activate CREB, including depolarization, cAMP, or transfection of activated CaMK mutants. However, transfection of dominant-negative CREB mutants did abrogate gene expression driven by BDNF promoters III and IV, indicating that CREB is necessary for constitutive BDNF expression. Thus, BDNF synthesis within SGNs makes possible an autocrine or paracrine mechanism that can contribute to support SGN survival but SGNs are distinctive in that this mechanism is constitutive and not activity-regulated.
Collapse
Affiliation(s)
- X M Zha
- Department of Biological Sciences, University of Iowa, Iowa City 52242-1324, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Multiple distinct signal pathways, including an autocrine neurotrophic mechanism, contribute to the survival-promoting effect of depolarization on spiral ganglion neurons in vitro. J Neurosci 2001. [PMID: 11264301 DOI: 10.1523/jneurosci.21-07-02256.2001] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have shown previously that BDNF, neurotrophin-3 (NT-3), chlorphenylthio-cAMP (cpt-cAMP) (a permeant cAMP analog), and membrane depolarization promote spiral ganglion neuron (SGN) survival in vitro in an additive manner, depolarization having the greatest efficacy. Expression of both BDNF and of NT-3 is detectable in cultured SGNs after plating in either depolarizing or nondepolarizing medium. These neurotrophins promote survival by an autocrine mechanism; TrkB-IgG or TrkC-IgG, which block neurotrophin binding to, respectively, TrkB and TrkC, partially inhibit the trophic effect of depolarization. The mitogen-activated protein kinase kinase inhibitor PD98059 and the phosphatidylinositol-3-OH kinase inhibitor LY294002 both abolish trophic support by neurotrophins but only partially inhibit support by depolarization. Inhibition by these compounds is not additive with inhibition by Trk-IgGs. The cAMP antagonist Rp-adenosine-3',5'-cyclic-phosphorothioate (Rp-cAMPS) abolishes survival attributable to cpt-cAMP but has no effect on that attributable to neurotrophins, nor do inhibitors of neurotrophin-dependent survival affect survival attributable to cpt-cAMP. However, Rp-cAMPS does partially inhibit depolarization-dependent survival, an inhibition that is additive with that by Trk-IgGs, PD98059, or LY294002. Moreover, Rp-cAMPS prevents depolarization-dependent survival of PC12 cells maintained in subthreshold levels of NGF. Inhibition of Ca(2+)/calmodulin-dependent protein kinases (CaMKs) with KN-62 reduces SGN survival independently of Rp-cAMPS, Trk-IgGs, and LY294002 and additively with them. Combined inhibition of Trk, cAMP, and CaMK signaling prevents depolarization-dependent survival. Thus, survival of SGNs under depolarizing conditions involves additivity among a depolarization-independent autocrine pathway, a cAMP-dependent pathway, and a CaMK-dependent pathway.
Collapse
|
30
|
Morishita H, Makishima T, Kaneko C, Lee YS, Segil N, Takahashi K, Kuraoka A, Nakagawa T, Nabekura J, Nakayama K, Nakayama KI. Deafness Due to Degeneration of Cochlear Neurons in Caspase-3-Deficient Mice. Biochem Biophys Res Commun 2001; 284:142-9. [PMID: 11374883 DOI: 10.1006/bbrc.2001.4939] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mice that lack caspase-3, which functions in apoptosis, were generated by gene targeting and shown to undergo hearing loss. The ABR threshold of the caspase-3(-/-) mice was significantly elevated compared to that of caspase-3(+/+) mice at 15 days of age and was progressively elevated further by 30 days. Distortion product otoacoustic emissions were not detectable in caspase-3(-/-) mice at 15 days of age. Caspase-3(-/-) mice exhibited marked degeneration of spiral ganglion neurons and a loss of inner and outer hair cells in the cochlea at 30 days of age, although no such changes were apparent at 15 days. The degenerating neurons manifested features, including cytoplasmic vacuolization, distinct from those characteristic of apoptosis. Spiral ganglion neurons and cochlear hair cells thus appear to require caspase-3 for survival but not for initial development. The mapping of both the human caspase-3 gene and the locus responsible for an autosomal dominant, nonsyndromic form of hearing loss (DFNA24) to chromosome 4q35 suggests that the caspase-3(-/-) mice may represent a model of this human condition.
Collapse
MESH Headings
- Aging/pathology
- Animals
- Auditory Threshold
- Caspase 3
- Caspases/biosynthesis
- Caspases/deficiency
- Caspases/genetics
- Cell Count
- Cell Death/genetics
- Cochlea/innervation
- Cochlea/metabolism
- Cochlea/pathology
- Deafness/congenital
- Deafness/genetics
- Deafness/pathology
- Disease Models, Animal
- Evoked Potentials, Auditory, Brain Stem/genetics
- Hair Cells, Auditory, Inner/pathology
- Hair Cells, Auditory, Outer/pathology
- Immunohistochemistry
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons/metabolism
- Neurons/pathology
- Otoacoustic Emissions, Spontaneous/genetics
- Spiral Ganglion/metabolism
- Spiral Ganglion/pathology
- Vacuoles/pathology
Collapse
Affiliation(s)
- H Morishita
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Okamura H, Spicer SS, Schulte BA. Immunohistochemical localization of phospholipase C isozymes in mature and developing gerbil cochlea. Neuroscience 2001; 102:451-9. [PMID: 11166131 DOI: 10.1016/s0306-4522(00)00456-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The possibility that phospholipase C contributes to intracellular signaling in the cochlea was investigated by immunostaining for eight different isoforms of the enzyme. In the mature gerbil cochlea, expression of the isozymes varied widely among different cell types. The phospholipase C-beta1 isoform was detected in inner and outer hair cells, and spiral ganglion neurons where it may participate in regulating Ca(2+) flux. The beta3 isozyme was expressed in epithelial cells thought to mediate lateral and medial circulation of potassium. The beta2 isozyme was present in border, inner phalangeal and Hensen cells, the stria vascularis, and suprastrial and supralimbal fibrocytes where it also may be involved in regulating ion transport activities. The phospholipase C-gamma isozymes were expressed in supporting cells, the stria vascularis, and certain fibrocytes where they possibly participate in activating tyrosine kinase and modulating ion conductances. The delta2 isoform was found in pillar, outer sulcus and strial marginal cells as well as spiral ganglion neurons and their radial processes. Documentation of changes in the expression pattern of phospholipase C isoforms during postnatal development and knowledge of their distribution in several positive control tissues provided further data for speculation about the biologic significance of the cochlear reactivity. The results demonstrate a wide diversity of isozyme distribution in the cochlea and suggest that the enzymes affect activities of various cochlear cell types in different ways.
Collapse
Affiliation(s)
- H Okamura
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
32
|
Carnicero E, Garrido JJ, Alonso MT, Schimmang T. Roles of fibroblast growth factor 2 during innervation of the avian inner ear. J Neurochem 2001; 77:786-95. [PMID: 11331407 DOI: 10.1046/j.1471-4159.2001.00283.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The importance of individual members of the fibroblast growth factor gene family during innervation of the vertebrate inner ear is not clearly defined. Here we address the role of fibroblast growth factor 2 (FGF-2 or basic FGF) during development of the chicken inner ear. We found that FGF-2 stimulated survival of isolated cochlear and vestibular neurons during distinct phases of inner ear innervation. The potential neurotrophic role of FGF-2 was confirmed by its expression in the corresponding sensory epithelia and the detection of one of its high-affinity receptors in inner ear neurons. Finally, we have analysed the potential of the amplicon system based on defective herpes simplex virus type 1 (HSV-1) vectors to express FGF-2 in cochlear neurons. Overexpression of FGF-2 in cochlear neurons resulted in neuronal differentiation demonstrating the presence of biologically active growth factor. This study underlines the potential of FGF-2 to control innervation and development of sensory epithelia in the avian inner ear. Furthermore, amplicon vectors may provide a useful tool to analyse gene function in isolated neurons of the vertebrate inner ear.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cells, Cultured
- Chick Embryo
- Chickens
- Cochlea/embryology
- Cochlea/innervation
- Ear, Inner/embryology
- Ear, Inner/innervation
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/pharmacology
- Fibroblast Growth Factor 2/physiology
- Gene Expression
- Genetic Vectors
- Herpesvirus 1, Human/genetics
- Immunohistochemistry
- Neurons, Afferent/chemistry
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- Receptor Protein-Tyrosine Kinases/analysis
- Receptor, Fibroblast Growth Factor, Type 1
- Receptors, Fibroblast Growth Factor/analysis
- Transfection
- Vestibule, Labyrinth/embryology
- Vestibule, Labyrinth/innervation
Collapse
Affiliation(s)
- E Carnicero
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Cientificas, Departamento de Bioquímica, Biología Molecular y Fisiología, Facultad de Medicina, Valladolid, Spain
| | | | | | | |
Collapse
|
33
|
Tashiro Y, Miyahara M, Shirasaki R, Okabe M, Heizmann CW, Murakami F. Local nonpermissive and oriented permissive cues guide vestibular axons to the cerebellum. Development 2001; 128:973-81. [PMID: 11222151 DOI: 10.1242/dev.128.6.973] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Information that originates from peripheral sensory organs is conveyed by axons of cephalic sensory cranial ganglia connecting the sensory organs to appropriate central targets in the brain. Thus, the establishment of correct axonal projections by sensory afferents is one of the most important issues in neural development. Previously, we examined the development of the vestibular nerve that originates from the VIIIth ganglion using a flat whole-mount preparation of the rat hindbrain and developed an in vitro, culture preparation that can recapitulate vestibular nerve development (Tashiro, Y., Endo, T., Shirasaki, R., Miyahara, M., Heizmann, C. W. and Murakami, F. (2000) J. Comp. Neurol. 417, 491–500). Both in vivo and in vitro, the ascending branch of the VIIIth ganglion projecting to the cerebellum reaches the base of the cerebellar primordium and starts to splay out towards the rhombic lip, apparently avoiding the ventral metencephalon. We now examine the nature of cues that guide vestibulocerebellar axons by applying various manipulations to the flat whole-mount in vitro preparation. Our observations suggest that local nonpermissive cues and oriented cues play a pivotal role in the guidance of vestibular axons to their central target.
Collapse
Affiliation(s)
- Y Tashiro
- Laboratory of Neuroscience, Division of Biophysical Engineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Gillespie LN, Clark GM, Bartlett PF, Marzella PL. LIF is more potent than BDNF in promoting neurite outgrowth of mammalian auditory neurons in vitro. Neuroreport 2001; 12:275-9. [PMID: 11209934 DOI: 10.1097/00001756-200102120-00019] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neurotrophic factors are known to play a crucial role in the elongation and guidance of auditory nerve fibres to their targets within the organ of Corti. Maintenance of these neural connections following deafness would clearly influence the efficacy of therapies for hearing recovery. The growth factors leukaemia inhibitory factor (LIF), brain-derived neurotrophic factor (BDNF) and transforming growth factor-beta 5 (TGF-beta5) were tested for their efficacy in promoting neurite outgrowth from dissociated cultures of early postnatal rat auditory neurons. Our results indicate that while BDNF enhances neurite outgrowth in a strong fashion, LIF is more potent; moreover, the combined administration of both factors has even greater neuritogenic capacities. TGF-beta5, although neurotrophic, has no neuritogenic activity on cultured auditory neurons. LIF and BDNF may therefore be potential candidates when developing pharmacological therapies for hearing recovery.
Collapse
Affiliation(s)
- L N Gillespie
- Department of Otolaryngology, The University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | | | | | | |
Collapse
|
35
|
Sekiya T, Shimamura N, Hatayama T, Suzuki S. Cerebellopontine angle cisternal infusion of NGF, BDNF and NT-3: effects on cochlear neurons disconnected from central target, cochlear nucleus. An in vivo quantitative study. Acta Otolaryngol 2000; 120:473-9. [PMID: 10958397 DOI: 10.1080/000164800750045956] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Cochlear neurons need their synaptic contacts with both their peripheral (organ of Corti) and central (cochlear nucleus) targets for survival. We examined the in vivo effectiveness of the neurotrophins (NGF, BDNF and NT-3) on cochlear neuronal survival using our in vivo model, in which the central connection alone was selectively and quantitatively interrupted. The particular neurotrophins evaluated in the present study did not appear to have cochlear nerve rescue potential. However, the experimental model reported here can serve as a useful tool to investigate cochlear neuronal degeneration from the central side, which may lead to identification of effective mediators in the future.
Collapse
Affiliation(s)
- T Sekiya
- Department of Neurosurgery, Hirosaki University School of Medicine, Hirosaki, Japan.
| | | | | | | |
Collapse
|
36
|
Shoji F, Yamasoba T, Magal E, Dolan DF, Altschuler RA, Miller JM. Glial cell line-derived neurotrophic factor has a dose dependent influence on noise-induced hearing loss in the guinea pig cochlea. Hear Res 2000; 142:41-55. [PMID: 10748327 DOI: 10.1016/s0378-5955(00)00007-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We examined the effectiveness of glial cell line-derived neurotrophic factor (GDNF) to attenuate cochlear damage from intense noise stress. Subjects were exposed to 115 dB SPL one octave band noise centered at 4 kHz for 5 h. They received artificial perilymph with or without GDNF into the left scala tympani at 0.5 microliter/h from 4 days before noise exposure through 8 days following noise exposure. Different concentrations of GDNF (1 ng/ml, 10 ng/ml, 100 ng/ml, and 1 microgram/ml) were applied chronically directly into the guinea pig cochlea via a microcannula and osmotic pump. Noise-induced hearing loss was assessed with pure tone auditory brainstem responses (at 2, 4, 8 and 20 kHz), measured prior to surgery, 1 day before noise exposure, and 7 days following noise exposure. Subjects were killed on day 8 following exposure for histological preparation and quantitative assessment of hair cell (HC) damage. A dose-dependent protective effect of GDNF on both sensory cell preservation and hearing function was found in the treated ears. At 1 ng/ml, GDNF showed no significant protection; at 10 ng/ml, GDNF showed significant HC protection; and at 100ng/ml, it was greater and bilateral. At 1 microgram/ml, GDNF appeared to have a toxic effect under noise stress in some cochleae. These findings indicate that GDNF at certain concentrations can effectively protect the inner ear from noise-induced hearing loss.
Collapse
Affiliation(s)
- F Shoji
- Kresge Hearing Research Institute, The University of Michigan, 1301 East Ann Street, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
37
|
Brumwell CL, Hossain WA, Morest DK, Bernd P. Role for basic fibroblast growth factor (FGF-2) in tyrosine kinase (TrkB) expression in the early development and innervation of the auditory receptor: in vitro and in situ studies. Exp Neurol 2000; 162:121-45. [PMID: 10716894 DOI: 10.1006/exnr.2000.7317] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A previous study showed that basic fibroblast growth factor (FGF-2) promotes the effects of brain-derived neurotrophic factor (BDNF) on migration and neurite outgrowth from the cochleovestibular ganglion (CVG). This suggests that FGF-2 may up-regulate the receptor for BDNF. Thus we have examined TrkB expression during CVG formation and otic innervation in vitro and in the chicken embryo using immunohistochemistry. Following anatomical staging according to Hamburger-Hamilton, results were compared with mRNA expression in vitro using in situ hybridization. In the embryo at stage 16 (E2+) clusters of either lightly stained or immunonegative cells occurred within the otocyst and among those migrating to the CVG. By stage 22 (E3.5), immunostaining was concentrated in the CVG perikarya and invaded the processes growing into the otic epithelium but not into the rhombencephalon. Subsequently TrkB expression decreased in the perikarya and became localized in the leading processes of the fibers invading the epithelium and in the structures participating in synapse formation with the hair cells. In vitro there was moderate immunostaining and modest in situ hybridization for trkB in the neuroblasts migrating from the otocyst under control conditions. In contrast, neuroblasts previously exposed to FGF-2 exhibited accelerated migration and differentiation, with increased trkB mRNA expression. Morphological differentiation was associated with more intense immunostaining of processes than cell bodies. Evidently TrkB shifts its expression sequentially from sites engaged in migration, ganglion cell differentiation, axonal outgrowth, epithelial innervation, and synapse formation. FGF-2 may promote the role of BDNF in these developmental events by upregulating the TrkB receptor.
Collapse
Affiliation(s)
- C L Brumwell
- Department of Anatomy, University of Connecticut Health Center, Farmington, Connecticut, 06030-3405, USA
| | | | | | | |
Collapse
|
38
|
Tashiro Y, Endo T, Shirasaki R, Miyahara M, Heizmann CW, Murakami F. Afferents of cranial sensory ganglia pathfind to their target independent of the site of entry into the hindbrain. J Comp Neurol 2000; 417:491-500. [PMID: 10701868 DOI: 10.1002/(sici)1096-9861(20000221)417:4<491::aid-cne7>3.0.co;2-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In vertebrates, sensory neurons interconnect a variety of peripheral tissues and central targets, conveying sensory information from different types of sensory receptors to appropriate second-order neurons in the central nervous system (CNS). To explore the possibility that the different rhombomere environments where sensory neurons enter into the hindbrain affect the pathfinding capability of growth cones, we studied the development of the VIIIth ganglion afferent both in vivo and in vitro. We focused on the vestibular nerve because it is the only cranial nerve projecting to the cerebellum, allowing for ready identification from its pattern of projection. Embryonic rat brain was cut along the dorsal midline and, with the VIIIth and Vth ganglia still attached, flat mounted and visualized with antibodies specific for sensory ganglia. Axons reached the cerebellar primordium at embryonic day (E) 13, then splayed out towards the edges of the rhombic lip of rostral hindbrain. In vitro, the VIIIth ganglion showed development similar to that in vivo and innervated the cerebellum, an appropriate target, indicating that mechanisms for axon guidance and target recognition are preserved in vitro. When the VIIIth ganglion was transplanted to the position of the Vth ganglion, axons from the transplanted ganglion entered the cerebellar primordium with a trajectory characteristic of the VIIIth nerve. These results indicate that the central projection pattern of the VIIIth nerve is not affected by the environment of nerve entry into the brainstem, suggesting that axons of sensory cranial ganglion intrinsically possess the capacity to find their target correctly.
Collapse
Affiliation(s)
- Y Tashiro
- Division of Biophysical Engineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Cheng AG, Huang T, Stracher A, Kim A, Liu W, Malgrange B, Lefebvre PP, Schulman A, Van de Water TR. Calpain inhibitors protect auditory sensory cells from hypoxia and neurotrophin-withdrawal induced apoptosis. Brain Res 1999; 850:234-43. [PMID: 10629769 DOI: 10.1016/s0006-8993(99)01983-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inhibitors of calpain have been shown to protect nerve growth factor (NGF)-deprived ciliary ganglion neurons and hypoxic cortical neurons. Calpains have been identified in the cochlea and are active during ischemic injury. Since apoptosis can be initiated by loss of neurotrophic support, hypoxia, and ototoxins (e.g., cisplatin, CDDP), the role of calpain inhibitors under these conditions was examined in auditory hair cells and neurons. Dissociated spiral ganglion neuron (SGN) cell cultures and organ of Corti explants from P3 rats were used to test the efficacy of calpain inhibitors as otoprotective molecules. Our results indicate that calpain inhibitor I, calpain inhibitor II, and leupeptin all provided significant protection of SGNs against neurotrophin-withdrawal and hypoxia-induced apoptosis. The increase in neuronal survival ranged from 2.16 to 2.31 times greater than in untreated neurotrophin-withdrawn SGN cell cultures. BOC-Asp(Ome)-Fluoromethyl Ketone (B-D-FMK), a general caspase inhibitor, increased neuronal survival 2.16 times more. Neuronal survival rates were from 1.88 to 2.27 times greater than in untreated, hypoxic neurons and hair cell survival rates were from 1.98 to 2.03 times greater than untreated, hypoxic organ of Corti explants. However, protection of auditory hair cells and neurons from CDDP-induced damage (10 and 6 micrograms/ml, respectively) was limited with any of these calpain inhibitors. Apoptotic pathways initiated by neurotrophin-deprivation and ototoxic stress (e.g., CDDP) have been shown to be different. Our results agree with this finding, with neurotrophin-withdrawal and hypoxia, but not CDDP damage-induced apoptosis being calpain-dependent.
Collapse
Affiliation(s)
- A G Cheng
- Department of Otolaryngology, Albert Einstein College of Medicine, Rose F. Kennedy Center, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cochran SL, Stone JS, Bermingham-McDonogh O, Akers SR, Lefcort F, Rubel EW. Ontogenetic expression of trk neurotrophin receptors in the chick auditory system. J Comp Neurol 1999; 413:271-88. [PMID: 10524339 DOI: 10.1002/(sici)1096-9861(19991018)413:2<271::aid-cne8>3.0.co;2-l] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neurotrophins and their cognate receptors are critical to normal nervous system development. Trk receptors are high-affinity receptors for nerve-growth factor (trkA), brain-derived neurotrophic factor and neurotrophin-4/5 (trkB), and neurotrophin-3 (trkC). We examine the expression of these three neurotrophin tyrosine kinase receptors in the chick auditory system throughout most of development. Trks were localized in the auditory brainstem, the cochlear ganglion, and the basilar papilla of chicks from embryonic (E) day 5 to E21, by using antibodies and standard immunocytochemical methods. TrkB mRNA was localized in brainstem nuclei by in situ hybridization. TrkB and trkC are highly expressed in the embryonic auditory brainstem, and their patterns of expression are both spatially and temporally dynamic. During early brainstem development, trkB and trkC are localized in the neuronal cell bodies and in the surrounding neuropil of nucleus magnocellularis (NM) and nucleus laminaris (NL). During later development, trkC is expressed in the cell bodies of NM and NL, whereas trkB is expressed in the nerve calyces surrounding NM neurons and in the ventral, but not the dorsal, dendrites of NL. In the periphery, trkB and trkC are located in the cochlear ganglion neurons and in peripheral fibers innervating the basilar papilla and synapsing at the base of hair cells. The protracted expression of trks seen in our materials is consistent with the hypothesis that the neurotrophins/tyrosine kinase receptors play one or several roles in the development of auditory circuitry. In particular, the polarized expression of trkB in NL is coincident with refinement of NM terminal arborizations on NL.
Collapse
Affiliation(s)
- S L Cochran
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology/Head and Neck Surgery, University of Washington, Seattle 98195-7923, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Neurons of the cochlear ganglion are endowed with a set of voltage-gated ion channels that enable them to encode and transmit sound information from the cochlear receptors to the brain. The temporal expression pattern of the K+ currents in chick cochlear ganglion neurons during embryonic development was analyzed using whole-cell voltage clamp techniques. In acutely isolated neurons, slowly activating delayed rectifier K+ currents appear at embryonic day 7 (E7) and increase in amplitude during development. A fast activating, fast inactivating K+ current of the A type is first expressed at E10, increasing in amplitude thereafter. To investigate the possible role of neurotrophins in the induction of these K+ channels, neurons were grown in culture in the presence or absence of brain-derived neurotrophic factor (BDNF) or neurotrophin-3 (NT-3). Neurons isolated at E8 and grown in culture for 1 day exhibit a high expression of A-current, together with the outgrowth of neurites. A-currents are not seen in acutely dissociated neurons from age-matched embryos (E9) which lack neurites, cut off by the isolation procedure. This suggests a preferential neuritic location of the channels carrying the A-current. However, the level of expression of the K+ currents was independent of BDNF or NT-3 application. Similarly, neurons isolated at E10 and grown in culture for up to 4 days maintain the amplitude of the K+ currents independently of the presence of the neurotrophins. These results indicate that BDNF and NT-3 may not directly regulate the expression of K+ channels in chick cochlear ganglion neurons. The notable expression of the fast inactivating A-current suggests that it plays a significant role in the modulation of synaptic efficacy and the encoding of auditory stimuli.
Collapse
Affiliation(s)
- J F García-Díaz
- Department of Physiology, Boston University School of Medicine, MA 02118, USA.
| |
Collapse
|
42
|
Gao XB, van den Pol AN. Neurotrophin-3 potentiates excitatory GABAergic synaptic transmission in cultured developing hypothalamic neurones of the rat. J Physiol 1999; 518:81-95. [PMID: 10373691 PMCID: PMC2269394 DOI: 10.1111/j.1469-7793.1999.0081r.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
1. Neurotrophin-3 (NT-3) supports the survival and differentiation of neurones in the central and peripheral nervous systems through a number of mechanisms that occur in a matter of hours or days. NT-3 may also have a more rapid mode of action that influences synaptic activity in mature neurones. In the present study, the effect of NT-3 on developing GABAergic synapses was investigated in 3- to 7-day-old cultures of rat hypothalamic neurones with whole-cell patch-clamp recording. 2. NT-3 induced a substantial dose-dependent potentiation of the frequency of spontaneous postsynaptic currents (sPSCs; 160 %) in developing neurones during a period when GABA evoked inward (depolarizing) current, as determined with gramicidin-perforated patch recordings. The NT-3 effect was long lasting; continued enhancement was found > 30 min after NT-3 wash-out. NT-3 evoked a substantial 202 % increase in total GABA-mediated inward current, measured as the time-current integral. Action potential frequency was also increased by NT-3 (to 220 %). 3. The frequency of GABA-mediated miniature postsynaptic currents in developing neurones in the presence of tetrodotoxin was potentiated (to 140%) by NT-3 with no change in the mean amplitude, suggesting a presynaptic locus of the effect. 4. In striking contrast to immature neurones, when more mature neurones were studied, NT-3 did not enhance the frequency of GABA-mediated spontaneous postsynaptic currents (sPSCs), but instead evoked a slight (16%) decrease. The frequency of miniature post-synaptic currents was also slightly decreased (16%) by the NT-3, with no change in amplitude. These results were recorded during a later period of neuronal maturity when GABA would evoke outward (hyperpolarizing) currents. NT-3 had no effect on the mean amplitude of GABA-evoked postsynaptic currents in either developing or mature neurones. 5. Intracellular application of K252a, a non-selective tyrosine kinase inhibitor, did not block the NT-3 effect postsynaptically. In contrast, bath application of K252a prevented the enhancement of sPSCs by NT-3, consistent with NT-3 acting through presynaptic induction of tyrosine kinase. Decreasing extracellular calcium with BAPTA or inhibiting calcium channels with Cd2+ blocked the augmentation of sPSC frequency by NT-3, suggesting that an increase of calcium entry may be required for the facilitation of NT-3. 6. Together, our results suggest NT-3 enhances GABA release during the developmental period when GABA is depolarizing and calcium elevating, but not later when GABA is inhibitory, suggesting that one mechanism through which NT-3 may influence neuronal development is via presynaptic potentiation of GABA excitation.
Collapse
Affiliation(s)
- X B Gao
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
43
|
Hashino E, Dolnick RY, Cohan CS. Developing vestibular ganglion neurons switch trophic sensitivity from BDNF to GDNF after target innervation. JOURNAL OF NEUROBIOLOGY 1999; 38:414-27. [PMID: 10022582 DOI: 10.1002/(sici)1097-4695(19990215)38:3<414::aid-neu9>3.0.co;2-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence showing a distinctive cell loss in vestibular and cochlear ganglia of brain-derived neurotrophic factor (BDNF) versus neurotrophin-3 (NT-3) null mutant mice demonstrates that these neurotrophins play a critical role in inner ear development. In this study, biological functions of BDNF and NT-3 in the chick vestibular and cochlear ganglion development was assessed in vitro and compared to those of other neurotrophic factors. The embryonic day (E)8-12 vestibular ganglion neurons showed an extensive outgrowth in response to BDNF with less outgrowth to NT-3. In contrast, NT-3 had stronger neurotrophic effects on the E12 cochlear ganglion neurons compared to BDNF. These results support previous evidence that neurotrophins play important roles in the vestibular and cochlear ganglion neuron development. However, the responsiveness to the neurotrophins declined and became undetectable by E16. Unexpectedly, glial cell line-derived neurotrophic factor (GDNF) promoted neurite outgrowth from vestibular ganglia at E12-16, later than the stages at which BDNF had neurotrophic effects. The time of switching sensitivity of the vestibular ganglion neurons from BDNF to GDNF correlated with the time of completion of synaptogenesis on their peripheral and central targets. Furthermore, a factor released from E12 inner ears exerted neurotrophic effects on late-stage vestibular ganglion neurons that were not responsive to the E4 otocyst-derived factor. These results raise the possibility that the vestibular ganglion neurons become responsive to GDNF upon target innervation and that the changes in sensitivity are regulated by changes in available factors released from their peripheral targets, the inner ear epithelia.
Collapse
Affiliation(s)
- E Hashino
- Department of Anatomy and Cell Biology, State University of New York at Buffalo, 14214, USA
| | | | | |
Collapse
|
44
|
Mufson EJ, Kroin JS, Sendera TJ, Sobreviela T. Distribution and retrograde transport of trophic factors in the central nervous system: functional implications for the treatment of neurodegenerative diseases. Prog Neurobiol 1999; 57:451-84. [PMID: 10080385 DOI: 10.1016/s0301-0082(98)00059-8] [Citation(s) in RCA: 232] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotrophins play a crucial role in the maintenance, survival and selective vulnerability of various neuronal populations within the normal and diseased brain. Several families of growth promoting substances have been identified within the central nervous system (CNS) including the superfamily of nerve growth factor related neurotrophin factors, glial derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF). In addition, other non-neuronal growth factors such as fibroblast growth factor (FGF) have also been identified. This article reviews the trophic anatomy of these factors within the CNS. Intraventricular and intraparenchymal injections of exogenous nerve growth factor result in retrograde labeling mainly within the cholinergic basal forebrain. Distribution of brain derived neurotrophic factor (BDNF) following intraventricular injection is minimal due to the binding to the trkB receptor along the ventricular wall. In contrast, intraparenchymal injections of BDNF results in widespread retrograde transport throughout the CNS. BDNF has also been shown to be transported anterogradely within the CNS. Infusion of GDNF into the CNS results in retrograde transport limited to the nigrostriatal pathway. Hippocampal injections of NT-3 retrogradely label mainly basal forebrain neurons. Retrograde transport of radiolabeled CNTF has only been observed in sensory neurons of the sciatic nerve. Following intraventricular and intraparenchymal infusion of radiolabeled bFGF, retrograde neuronal labeling was found in the telecephalon, diencephalon, mesencephalon and pons. In contrast retrograde labeling for aFGF was found only in the hypothalamus and midbrain. Since select neurotrophins traffic anterogradely and retrogradely within the nervous system, these proteins could be used to treat neurological diseases such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- E J Mufson
- Research Center for Brain Repair, Department of Neurological Sciences, Rush Presbyterian-Luke's Medical Center, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
45
|
Abstract
OBJECTIVES Describe quantitatively the number of ganglion cells in the geniculate (G) and meatal (M) segments of the human facial nerve. STUDY DESIGN One hundred human temporal bone specimens that were sectioned horizontally and stained with hematoxylin and eosin were selected from a temporal bone collection on the basis of minimal artifact and absence of pathology involving the facial nerve. METHODS Cells with a nucleolus in all sections through the facial nerve were projected on tracing paper with a camera lucida and counted manually. A modified Abercrombie technique was employed to compute total cells in the G and M segments. RESULTS Ages of patients ranged from 1 month to 92 years; the male-to-female ratio was 56:44. The total number of cells in individual temporal bones ranged from 589 to 4183 (mean, 2162 cells). The range of cells in the G ganglion was from 66 to 4017 (mean, 1713 cells); in the M ganglion the number ranged from 0 to 2764 (mean, 448 cells). There was no correlation of total ganglion cell number to age or sex. The majority of cells were found in the G ganglion in 88% of temporal bones. In 8% temporal bones the majority of cells were in the M ganglion and in 4% the M and G ganglions contained an equal number of cells. CONCLUSIONS The facial nerve sensory ganglion consists of two components: G and M. The G ganglion outnumbers the M component in the majority of temporal bones (88%). The M ganglion was equal to or greater in number than the G ganglion in 12% of temporal bones.
Collapse
Affiliation(s)
- R R Gacek
- State University of New York Health Science Center, Department of Otolaryngology and Communication Sciences, Syracuse, USA
| |
Collapse
|
46
|
Bianchi LM, Dolnick R, Medd A, Cohan CS. Developmental changes in growth factors released by the embryonic inner ear. Exp Neurol 1998; 150:98-106. [PMID: 9514837 DOI: 10.1006/exnr.1997.6774] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies have demonstrated a role for neurotrophins in regulating the survival of developing auditory and vestibular neurons. However, the developmental time-course for neurotrophin production and release by inner ear tissues has not been defined. In the present study, neurotrophin-like activity was evaluated from culture medium conditioned by early- or midembryonic stage inner ears. Examination of the proteinaceous properties of conditioned medium revealed a developmental change in growth factor release by the inner ear. Neurotrophin-like molecules were not detected in medium conditioned by early stage otocysts. In contrast, neurotrophin-like bioactivity was detected in medium conditioned by middevelopmental stage inner ears. Western blot analysis revealed that NT-3 was released by the rat inner ear at midstages of inner ear development. ELISA measurements revealed that both NT-3 and BDNF are produced by the middevelopmental stage inner ear, and that NT-3 protein levels are higher than BDNF levels. These results suggest that there are developmental changes in the release of growth factors by the inner ear.
Collapse
Affiliation(s)
- L M Bianchi
- Department of Otolaryngology and Communicative Sciences, Medical University of South Carolina, Charleston 29425, USA
| | | | | | | |
Collapse
|
47
|
Montcouquiol M, Valat J, Travo C, Sans A. A role for BDNF in early postnatal rat vestibular epithelia maturation: implication of supporting cells. Eur J Neurosci 1998; 10:598-606. [PMID: 9749722 DOI: 10.1046/j.1460-9568.1998.00070.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The early development of the inner ear is largely determined by two members of the neurotrophic family: brain-derived neurotrophic factor (BDNF) and neurotrophin 3 (NT-3). Little information is available on the role of these neurotrophins during the late stages of vestibular development in the rat which take place during the first postnatal weeks. At this period where terminal synaptogenesis and maturation occur, we have investigated the expression and the activity of BDNF, the most important neurotrophin in the vestibular system. Using different experimental approaches, we show that BDNF is released by vestibular epithelia on postnatal day 3 (P3) and continues to have a trophic effect on vestibular neurones in vitro. Immunocytochemistry coupled to confocal microscopy revealed a remarkable evolution in BDNF localization during later stages of development. Whereas BDNF is present in both supporting cells and hair cells at P3, its distribution gradually changed and is highly compartmentalized within the upper part of supporting cells at P8 and P15. In parallel, we observed the presence of a truncated form of the BDNF receptor in sensory hair cells. These results suggest an original role for supporting cells, which could be involved in the release of BDNF during the late stages of synaptogenesis in mammalian vestibular epithelia. In particular, BDNF could participate to the set up of the calyx, a specific nerve structure surrounding type I vestibular hair cells.
Collapse
Affiliation(s)
- M Montcouquiol
- INSERM U432, Neurobiologie et développement du système vestibulaire, Université de Montpellier II, France.
| | | | | | | |
Collapse
|
48
|
Abstract
The inner ear is a complex sensory organ responsible for balance and sound detection in vertebrates. It originates from a transient embryonic structure, the otic vesicle, that contains all of the information to develop autonomously into the mature inner ear. We review here the development of the otic vesicle, bringing together classical embryological experiments and recent genetic and molecular data. The specification of the prospective ectoderm and its commitment to the otic fate are very early events and can be related to the expression of genes with restricted expression domains. A combinatorial gene expression model for placode specification and diversification, based on classical embryological evidence and gene expression patterns, is discussed. The formation of the otic vesicle is dependent on inducing signals from endoderm, mesoderm and neuroectoderm. Ear induction consists of a sequence of discrete instructions from those tissues that confer its final identity on the otic field, rather than a single all-or-none process. The important role of the neural tube in otic development is highlighted by the abnormalities observed in mouse mutants for the Hoxa1, kreisler and fgf3 genes and those reported in retinoic acid-deficient quails. Still, the nature of the relation between the neural tube and otic development remains unclear. Gene targeting experiments in the mouse have provided evidence for genes potentially involved in regional and cell-fate specification in the inner ear. The disruption of the mouse Brn3.1 gene identifies the first mutation affecting sensory hair-cell specification, and mutants for Pax2 and Nkx5.1 genes show their requirement for the development of specific regions of the otic vesicle. Several growth-factors contribute to the patterned cell proliferation of the otic vesicle. Among these, IGF-I and FGF-2 are expressed in the otic vesicle and may act in an autocrine manner. Finally, little is known about early mechanisms involved in guiding ear innervation. However, targeted disruption of genes coding for neurotrophins and Trk receptors have shown that once synaptic contacts are established, they depend on specific trophic interactions that involve these two gene families. The accessibility of new cellular and molecular approaches are opening new perspectives in vertebrate development and are also starting to be applied to ear development. This will allow this classical and attractive model system to see a rapid progress in the near future.
Collapse
Affiliation(s)
- M Torres
- Departamento de Inmunologiá y Oncologiá, Centro Nacional de Biotecnología CSIC, Cantoblanco, E-28049, Madrid, Spain
| | | |
Collapse
|
49
|
Abstract
Dissociated primary cell cultures were derived from the cochlear nuclei (CN) of postnatal rats using standard techniques. Cultured cells differentiated morphologically, but their dendritic profiles were generally less specialized than those of CN cells in vivo. Physiologically, cultured cells could be divided into three classes: tonic, phasic and non-spiking cells, which differed in many of their fundamental biophysical properties. The percentage of cultured cells that spiked repetitively increased over time to a maximum of 85% at 6 days. However, the percentage of cells that produced action potentials decreased with time in culture, from 91% during the first 8 days to less than 40% after 9 days. CN cells were successfully cultured in both serum-supplemented and serum-free (Neurobasal) media. More neurons survived at low plating densities in Neurobasal than in medium containing serum, although neuronal survival was similar at higher densities. Few neurons raised in the serum-free medium were spontaneously active; other response properties were similar to those of cells grown in the presence of serum. Although differentiation of CN cells in culture did not completely mirror the in vivo developmental pattern, these experiments demonstrate that primary culture represents a viable method for the in vitro study of CN neurons.
Collapse
Affiliation(s)
- J L Fitzakerley
- Center for Hearing Sciences and Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
50
|
Don DM, Newman AN, Micevych PE, Popper P. Expression of brain-derived neurotrophic factor and its receptor mRNA in the vestibuloauditory system of the bullfrog. Hear Res 1997; 114:10-20. [PMID: 9447914 DOI: 10.1016/s0378-5955(97)00113-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin which has been suggested to play a crucial role in the development and maintenance of the inner ear. In the present study, we investigated the expression of mRNAs of BDNF and its high-affinity receptor trkB in the vestibuloauditory system of the adult bullfrog. In situ hybridization was performed using riboprobes transcribed from Xenopus BDNF and trkB cDNA clones. BDNF mRNA was expressed in the sensory epithelia of the ampullary cristae, utricular and saccular maculae, lagena, and amphibian and basilar papillae. Strong hybridization for BDNF mRNA was also found in neuron somata of the vestibuloauditory nuclear complex. trkB mRNA was detected in the sensory epithelia of all vestibular and auditory endorgans. High levels of both BDNF and trkB mRNAs were found in vestibuloauditory ganglion cells. These results support the hypothesis that BDNF participates in the maintenance of vestibuloauditory neurons and may be important for the trophic regulation of vestibular and auditory sensory epithelia in this animal model.
Collapse
Affiliation(s)
- D M Don
- Department of Surgery, University of California, Los Angeles School of Medicine, 90095-1624, USA
| | | | | | | |
Collapse
|