1
|
Li P, Qian T, Sun S. Spatial architecture of the cochlear immune microenvironment in noise-induced and age-related sensorineural hearing loss. Int Immunopharmacol 2023; 114:109488. [PMID: 36470117 DOI: 10.1016/j.intimp.2022.109488] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
The cochlea encodes sound stimuli and transmits them to the central nervous system, and damage to sensory cells and synapses in the cochlea leads to hearing loss. The inner ear was previously considered to be an immune privileged organ to protect the auditory organ from reactions with the immune system. However, recent studies have revealed the presence of resident macrophages in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis. The tissue-resident macrophages are responsible for the detection, phagocytosis, and clearance of cellular debris and pathogens from the tissues, and they initiate inflammation and influence tissue repair by producing inflammatory cytokines and chemokines. Insult to the cochlea can activate the cochlear macrophages to initiate immune responses. In this review, we describe the distribution and functions of cochlear macrophages in noise-induced hearing impairment and age-related hearing disabilities. We also focus on potential therapeutic interventions concerning hearing loss by modulating local immune responses.
Collapse
Affiliation(s)
- Peifan Li
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Tingting Qian
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Shan Sun
- ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
2
|
Yamaguchi S, Yoshida M, Horie N, Satoh K, Fukuda Y, Ishizaka S, Ogawa K, Morofuji Y, Hiu T, Izumo T, Kawakami S, Nishida N, Matsuo T. Stem Cell Therapy for Acute/Subacute Ischemic Stroke with a Focus on Intraarterial Stem Cell Transplantation: From Basic Research to Clinical Trials. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010033. [PMID: 36671605 PMCID: PMC9854681 DOI: 10.3390/bioengineering10010033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Stem cell therapy for ischemic stroke holds great promise for the treatment of neurological impairment and has moved from the laboratory into early clinical trials. The mechanism of action of stem cell therapy includes the bystander effect and cell replacement. The bystander effect plays an important role in the acute to subacute phase, and cell replacement plays an important role in the subacute to chronic phase. Intraarterial (IA) transplantation is less invasive than intraparenchymal transplantation and can provide more cells in the affected brain region than intravenous transplantation. However, transplanted cell migration was reported to be insufficient, and few transplanted cells were retained in the brain for an extended period. Therefore, the bystander effect was considered the main mechanism of action of IA stem cell transplantation. In most clinical trials, IA transplantation was performed during the acute and subacute phases. Although clinical trials of IA transplantation demonstrated safety, they did not demonstrate satisfactory efficacy in improving patient outcomes. To increase efficacy, increased migration of transplanted cells and production of long surviving and effective stem cells would be crucial. Given the lack of knowledge on this subject, we review and summarize the mechanisms of action of transplanted stem cells and recent advancements in preclinical and clinical studies to provide information and guidance for further advancement of acute/subacute phase IA stem cell transplantation therapy for ischemic stroke.
Collapse
Affiliation(s)
- Susumu Yamaguchi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
- Department of Neurosurgery, Sasebo General Hospital, Nagasaki 857-8511, Japan
- Correspondence: ; Tel.: +81-095-819-7375
| | - Michiharu Yoshida
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
- Department of Neurosurgery, Sasebo General Hospital, Nagasaki 857-8511, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, Hiroshima University, Hiroshima 734-8551, Japan
| | - Katsuya Satoh
- Department of Occupational Therapy Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Yuutaka Fukuda
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Shunsuke Ishizaka
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Yoichi Morofuji
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Takeshi Hiu
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Tsuyoshi Izumo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Noriyuki Nishida
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Takayuki Matsuo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan
| |
Collapse
|
3
|
Yamaguchi S, Horie N, Satoh K, Ishikawa T, Mori T, Maeda H, Fukuda Y, Ishizaka S, Hiu T, Morofuji Y, Izumo T, Nishida N, Matsuo T. Age of donor of human mesenchymal stem cells affects structural and functional recovery after cell therapy following ischaemic stroke. J Cereb Blood Flow Metab 2018; 38:1199-1212. [PMID: 28914133 PMCID: PMC6434451 DOI: 10.1177/0271678x17731964] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cell transplantation therapy offers great potential to improve impairments after stroke. However, the importance of donor age on therapeutic efficacy is unclear. We investigated the regenerative capacity of transplanted cells focusing on donor age (young vs. old) for ischaemic stroke. The quantities of human mesenchymal stem cell (hMSC) secreted brain-derived neurotrophic factor in vitro and of monocyte chemotactic protein-1 at day 7 in vivo were both significantly higher for young hMSC compared with old hMSC. Male Sprague-Dawley rats subjected to transient middle cerebral artery occlusion that received young hMSC (trans-arterially at 24 h after stroke) showed better behavioural recovery with prevention of brain atrophy compared with rats that received old hMSC. Histological analysis of the peri-infarct cortex showed that rats treated with young hMSC had significantly fewer microglia and more vessels covered with pericytes. Interestingly, migration of neural stem/progenitor cells expressing Musashi-1 positively correlated with astrocyte process alignment, which was more pronounced for young hMSC. Aging of hMSC may be a critical factor that affects cell therapy outcomes, and transplantation of young hMSC appears to provide better functional recovery through anti-inflammatory effects, vessel maturation, and neurogenesis potentially by the dominance of trophic factor secretion.
Collapse
Affiliation(s)
- Susumu Yamaguchi
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Nobutaka Horie
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuya Satoh
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Ishikawa
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tsuyoshi Mori
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hajime Maeda
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuhtaka Fukuda
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shunsuke Ishizaka
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Hiu
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yoichi Morofuji
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tsuyoshi Izumo
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Noriyuki Nishida
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takayuki Matsuo
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
4
|
Requena T, Gallego-Martinez A, Lopez-Escamez JA. Bioinformatic Integration of Molecular Networks and Major Pathways Involved in Mice Cochlear and Vestibular Supporting Cells. Front Mol Neurosci 2018; 11:108. [PMID: 29674954 PMCID: PMC5895758 DOI: 10.3389/fnmol.2018.00108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 03/19/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Cochlear and vestibular epithelial non-hair cells (ENHCs) are the supporting elements of the cellular architecture in the organ of Corti and the vestibular neuroepithelium in the inner ear. Intercellular and cell-extracellular matrix interactions are essential to prevent an abnormal ion redistribution leading to hearing and vestibular loss. The aim of this study is to define the main pathways and molecular networks in the mouse ENHCs. Methods: We retrieved microarray and RNA-seq datasets from mouse epithelial sensory and non-sensory cells from gEAR portal (http://umgear.org/index.html) and obtained gene expression fold-change between ENHCs and non-epithelial cells (NECs) against HCs for each gene. Differentially expressed genes (DEG) with a log2 fold change between 1 and -1 were discarded. The remaining genes were selected to search for interactions using Ingenuity Pathway Analysis and STRING platform. Specific molecular networks for ENHCs in the cochlea and the vestibular organs were generated and significant pathways were identified. Results: Between 1723 and 1559 DEG were found in the mouse cochlear and vestibular tissues, respectively. Six main pathways showed enrichment in the supporting cells in both tissues: (1) "Inhibition of Matrix Metalloproteases"; (2) "Calcium Transport I"; (3) "Calcium Signaling"; (4) "Leukocyte Extravasation Signaling"; (5) "Signaling by Rho Family GTPases"; and (6) "Axonal Guidance Si". In the mouse cochlea, ENHCs showed a significant enrichment in 18 pathways highlighting "axonal guidance signaling (AGS)" (p = 4.37 × 10-8) and "RhoGDI Signaling" (p = 3.31 × 10-8). In the vestibular dataset, there were 20 enriched pathways in ENHCs, the most significant being "Leukocyte Extravasation Signaling" (p = 8.71 × 10-6), "Signaling by Rho Family GTPases" (p = 1.20 × 10-5) and "Calcium Signaling" (p = 1.20 × 10-5). Among the top ranked networks, the most biologically significant network contained the "auditory and vestibular system development and function" terms. We also found 108 genes showing tonotopic gene expression in the cochlear ENHCs. Conclusions: We have predicted the main pathways and molecular networks for ENHCs in the organ of Corti and vestibular neuroepithelium. These pathways will facilitate the design of molecular maps to select novel candidate genes for hearing or vestibular loss to conduct functional studies.
Collapse
Affiliation(s)
- Teresa Requena
- Otology & Neurotology Group CTS495, Department of Genomic Medicine-Centro de Genómica e Investigación Oncológica-Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Alvaro Gallego-Martinez
- Otology & Neurotology Group CTS495, Department of Genomic Medicine-Centro de Genómica e Investigación Oncológica-Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Jose A Lopez-Escamez
- Otology & Neurotology Group CTS495, Department of Genomic Medicine-Centro de Genómica e Investigación Oncológica-Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria, ibs.GRANADA, Hospital Virgen de las Nieves, Universidad de Granada, Granada, Spain.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
5
|
Barald KF, Shen YC, Bianchi LM. Chemokines and cytokines on the neuroimmunoaxis: Inner ear neurotrophic cytokines in development and disease. Prospects for repair? Exp Neurol 2018; 301:92-99. [DOI: 10.1016/j.expneurol.2017.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/18/2017] [Accepted: 10/12/2017] [Indexed: 01/22/2023]
|
6
|
Weber LJ, Marcy HK, Shen YC, Tomkovich SE, Brooks KM, Hilk KE, Barald KF. The role of jab1, a putative downstream effector of the neurotrophic cytokine macrophage migration inhibitory factor (MIF) in zebrafish inner ear hair cell development. Exp Neurol 2017; 301:100-109. [PMID: 28928022 DOI: 10.1016/j.expneurol.2017.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/05/2017] [Accepted: 09/12/2017] [Indexed: 01/12/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a neurotrophic cytokine essential for inner ear hair cell (HC) development and statoacoustic ganglion (SAG) neurite outgrowth, and SAG survival in mouse, chick and zebrafish. Another neurotrophic cytokine, Monocyte chemoattractant protein 1 (MCP1) is known to synergize with MIF; but MCP1 alone is insufficient to support mouse/chick SAG neurite outgrowth or neuronal survival. Because of the relatively short time over which the zebrafish inner ear develops (~30hpf), the living zebrafish embryo is an ideal system to examine mif and mcp1 cytokine pathways and interactions. We used a novel technique: direct delivery of antisense oligonucleotide morpholinos (MOs) into the embryonic zebrafish otocyst to discover downstream effectors of mif as well as to clarify the relationship between mif and mcp1 in inner ear development. MOs for mif, mcp1 and the presumptive mif and mcp1 effector, c-Jun activation domain-binding protein-1 (jab1), were injected and then electroporated into the zebrafish otocyst 25-48hours post fertilization (hpf). We found that although mif is important at early stages (before 30hpf) for auditory macular HC development, jab1 is more critical for vestibular macular HC development before 30hpf. After 30hpf, mcp1 becomes important for HC development in both maculae.
Collapse
Affiliation(s)
- Loren J Weber
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA.
| | - Hannah K Marcy
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Undergraduate Research Opportunity Program, 1190 Undergraduate Science Building, 204 Washtenaw Avenue, Ann Arbor, MI 48109-2215, USA.
| | - Yu-Chi Shen
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA.
| | - Sarah E Tomkovich
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Undergraduate Research Opportunity Program, 1190 Undergraduate Science Building, 204 Washtenaw Avenue, Ann Arbor, MI 48109-2215, USA.
| | - Kristina M Brooks
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA.
| | - Kelly E Hilk
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Undergraduate Research Opportunity Program, 1190 Undergraduate Science Building, 204 Washtenaw Avenue, Ann Arbor, MI 48109-2215, USA.
| | - Kate F Barald
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109-0619, USA; Department of Biomedical Engineering, College of Engineering, 2200 Bonisteel Boulevard, University of Michigan, Ann Arbor, MI 48109-2099, USA.
| |
Collapse
|
7
|
Decreased Motor Neuron Support by SMA Astrocytes due to Diminished MCP1 Secretion. J Neurosci 2017; 37:5309-5318. [PMID: 28450545 DOI: 10.1523/jneurosci.3472-16.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/07/2017] [Accepted: 04/19/2017] [Indexed: 12/22/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal-recessive disorder characterized by severe, often fatal muscle weakness due to loss of motor neurons. SMA patients have deletions and other mutations of the survival of motor neuron 1 (SMN1) gene, resulting in decreased SMN protein. Astrocytes are the primary support cells of the CNS and are responsible for glutamate clearance, metabolic support, response to injury, and regulation of signal transmission. Astrocytes have been implicated in SMA as in in other neurodegenerative disorders. Astrocyte-specific rescue of SMN protein levels has been shown to mitigate disease manifestations in mice. However, the mechanism by which SMN deficiency in astrocytes may contribute to SMA is unclear and what aspect of astrocyte activity is lacking is unknown. Therefore, it is worthwhile to identify defects in SMN-deficient astrocytes that compromise normal function. We show here that SMA astrocyte cultures derived from mouse spinal cord of both sexes are deficient in supporting both WT and SMN-deficient motor neurons derived from male, female, and mixed-sex sources and that this deficiency may be mitigated with secreted factors. In particular, SMN-deficient astrocytes have decreased levels of monocyte chemoactive protein 1 (MCP1) secretion compared with controls and MCP1 restoration stimulates outgrowth of neurites from cultured motor neurons. Correction of MCP1 deficiency may thus be a new therapeutic approach to SMA.SIGNIFICANCE STATEMENT Spinal muscular atrophy (SMA) is caused by the loss of motor neurons, but astrocyte dysfunction also contributes to the disease in mouse models. Monocyte chemoactive protein 1 (MCP1) has been shown to be neuroprotective and is released by astrocytes. Here, we report that MCP1 levels are decreased in SMA mice and that replacement of deficient MCP1 increases differentiation and neurite length of WT and SMN-deficient motor-neuron-like cells in cell culture. This study reveals a novel aspect of astrocyte dysfunction in SMA and indicates a possible approach for improving motor neuron growth and survival in this disease.
Collapse
|
8
|
Ramamurthy P, White JB, Yull Park J, Hume RI, Ebisu F, Mendez F, Takayama S, Barald KF. Concomitant differentiation of a population of mouse embryonic stem cells into neuron-like cells and schwann cell-like cells in a slow-flow microfluidic device. Dev Dyn 2017; 246:7-27. [PMID: 27761977 PMCID: PMC5159187 DOI: 10.1002/dvdy.24466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/16/2016] [Accepted: 09/30/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To send meaningful information to the brain, an inner ear cochlear implant (CI) must become closely coupled to as large and healthy a population of remaining spiral ganglion neurons (SGN) as possible. Inner ear gangliogenesis depends on macrophage migration inhibitory factor (MIF), a directionally attractant neurotrophic cytokine made by both Schwann and supporting cells (Bank et al., 2012). MIF-induced mouse embryonic stem cell (mESC)-derived "neurons" could potentially substitute for lost or damaged SGN. mESC-derived "Schwann cells" produce MIF, as do all Schwann cells (Huang et al., a; Roth et al., 2007; Roth et al., 2008) and could attract SGN to a "cell-coated" implant. RESULTS Neuron- and Schwann cell-like cells were produced from a common population of mESCs in an ultra-slow-flow microfluidic device. As the populations interacted, "neurons" grew over the "Schwann cell" lawn, and early events in myelination were documented. Blocking MIF on the Schwann cell side greatly reduced directional neurite outgrowth. MIF-expressing "Schwann cells" were used to coat a CI: Mouse SGN and MIF-induced "neurons" grew directionally to the CI and to a wild-type but not MIF-knockout organ of Corti explant. CONCLUSIONS Two novel stem cell-based approaches for treating the problem of sensorineural hearing loss are described. Developmental Dynamics 246:7-27, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Poornapriya Ramamurthy
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Joshua B White
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - Joong Yull Park
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Richard I Hume
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| | - Fumi Ebisu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Flor Mendez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shuichi Takayama
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - Kate F Barald
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Cochlear afferent innervation development. Hear Res 2015; 330:157-69. [DOI: 10.1016/j.heares.2015.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/02/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023]
|
10
|
Niemi JP, DeFrancesco-Lisowitz A, Cregg JM, Howarth M, Zigmond RE. Overexpression of the monocyte chemokine CCL2 in dorsal root ganglion neurons causes a conditioning-like increase in neurite outgrowth and does so via a STAT3 dependent mechanism. Exp Neurol 2015; 275 Pt 1:25-37. [PMID: 26431741 DOI: 10.1016/j.expneurol.2015.09.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/08/2015] [Accepted: 09/27/2015] [Indexed: 12/24/2022]
Abstract
Neuroinflammation plays a critical role in the regeneration of peripheral nerves following axotomy. An injury to the sciatic nerve leads to significant macrophage accumulation in the L5 DRG, an effect not seen when the dorsal root is injured. We recently demonstrated that this accumulation around axotomized cell bodies is necessary for a peripheral conditioning lesion response to occur. Here we asked whether overexpression of the monocyte chemokine CCL2 specifically in DRG neurons of uninjured mice is sufficient to cause macrophage accumulation and to enhance regeneration or whether other injury-derived signals are required. AAV5-EF1α-CCL2 was injected intrathecally, and this injection led to a time-dependent increase in CCL2 mRNA expression and macrophage accumulation in L5 DRG, with a maximal response at 3 weeks post-injection. These changes led to a conditioning-like increase in neurite outgrowth in DRG explant and dissociated cell cultures. This increase in regeneration was dependent upon CCL2 acting through its primary receptor CCR2. When CCL2 was overexpressed in CCR2-/- mice, macrophage accumulation and enhanced regeneration were not observed. To address the mechanism by which CCL2 overexpression enhances regeneration, we tested for elevated expression of regeneration-associated genes in these animals. Surprisingly, we found that CCL2 overexpression led to a selective increase in LIF mRNA and neuronal phosphorylated STAT3 (pSTAT3) in L5 DRGs, with no change in expression seen in other RAGs such as GAP-43. Blockade of STAT3 phosphorylation by each of two different inhibitors prevented the increase in neurite outgrowth. Thus, CCL2 overexpression is sufficient to induce macrophage accumulation in uninjured L5 DRGs and increase the regenerative capacity of DRG neurons via a STAT3-dependent mechanism.
Collapse
Affiliation(s)
- Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | | | - Jared M Cregg
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Madeline Howarth
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA.
| |
Collapse
|
11
|
Shen YC, Upadhyayula R, Cevallos S, Messick RJ, Hsia T, Leese MP, Jewett DM, Ferrer-Torres D, Roth TM, Dohle W, Potter BVL, Barald KF. Targeted NF1 cancer therapeutics with multiple modes of action: small molecule hormone-like agents resembling the natural anticancer metabolite, 2-methoxyoestradiol. Br J Cancer 2015; 113:1158-67. [PMID: 26461061 PMCID: PMC4647869 DOI: 10.1038/bjc.2015.345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/12/2022] Open
Abstract
Background: Both the number and size of tumours in NF1 patients increase in response to the rise in steroid hormones seen at puberty and during pregnancy. The size of tumours decreases after delivery, suggesting that hormone-targeting therapy might provide a viable new NF1 treatment approach. Our earlier studies demonstrated that human NF1 tumour cell lines either went through apoptosis or ceased growth in the presence of 2-methoxyoestradiol (2ME2), a naturally occurring anticancer metabolite of 17-β estradiol. Previous reports of treatment with sulfamoylated steroidal and non-steroidal derivatives of 2ME2 showed promising reductions in tumour burden in hormone-responsive cancers other than NF1. Here we present the first studies indicating that 2ME2 derivatives could also provide an avenue for treating NF1, for which few treatment options are available. Methods: STX3451, (2-(3-Bromo-4,5-dimethoxybenzyl)-7-methoxy-6-sulfamoyloxy-1,2,3,4-tetrahydroisoquinoline), a non-steroidal sulphamate analogue of 2ME2, was tested in dose-dependent studies of malignant and benign NF1 human tumour cell lines and cell lines with variable controlled neurofibromin expression. The mechanisms of action of STX3451 were also analysed. Results: We found that STX3451-induced apoptosis in human malignant peripheral nerve sheath tumour (MPNST) cell lines, even in the presence of elevated oestrogen and progesterone. It inhibits both PI3 kinase and mTOR signalling pathways. It disrupts actin- and microtubule-based cytoskeletal structures in cell lines derived from human MPNSTs and in cells derived from benign plexiform neurofibromas. STX3451 selectively kills MPNST-derived cells, but also halts growth of other tumour-derived NF1 cell lines. Conclusion: STX3451 provides a new approach for inducing cell death and lowering tumour burden in NF1 and other hormone-responsive cancers with limited treatment options.
Collapse
Affiliation(s)
- Yu-chi Shen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.,Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ravi Upadhyayula
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.,Neuroscience Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Stephanie Cevallos
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.,NIH PREP program, Ann Arbor, Michigan 48109, USA
| | - Ryan J Messick
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | - Tammy Hsia
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.,Cancer Biology Summer Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Mathew P Leese
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Douglas M Jewett
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | - Daysha Ferrer-Torres
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.,Cancer Biology Summer Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Therese M Roth
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | - Wolfgang Dohle
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Barry V L Potter
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK.,Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Kate F Barald
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA.,Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA.,Neuroscience Program, University of Michigan, Ann Arbor, Michigan 48109, USA.,NIH PREP program, Ann Arbor, Michigan 48109, USA.,Cancer Biology Summer Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
12
|
Battisti AC, Fantetti KN, Moyers BA, Fekete DM. A subset of chicken statoacoustic ganglion neurites are repelled by Slit1 and Slit2. Hear Res 2014; 310:1-12. [PMID: 24456709 PMCID: PMC3979322 DOI: 10.1016/j.heares.2014.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 12/20/2013] [Accepted: 01/09/2014] [Indexed: 01/23/2023]
Abstract
Mechanosensory hair cells in the chicken inner ear are innervated by bipolar afferent neurons of the statoacoustic ganglion (SAG). During development, individual SAG neurons project their peripheral process to only one of eight distinct sensory organs. These neuronal subtypes may respond differently to guidance cues as they explore the periphery in search of their target. Previous gene expression data suggested that Slit repellants might channel SAG neurites into the sensory primordia, based on the presence of robo transcripts in the neurons and the confinement of slit transcripts to the flanks of the prosensory domains. This led to the prediction that excess Slit proteins would impede the outgrowth of SAG neurites. As predicted, axonal projections to the primordium of the anterior crista were reduced 2-3 days after electroporation of either slit1 or slit2 expression plasmids into the anterior pole of the otocyst on embryonic day 3 (E3). The posterior crista afferents, which normally grow through and adjacent to slit expression domains as they are navigating towards the posterior pole of the otocyst, did not show Slit responsiveness when similarly challenged by ectopic delivery of slit to their targets. The sensitivity to ectopic Slits shown by the anterior crista afferents was more the exception than the rule: responsiveness to Slits was not observed when the entire E4 SAG was challenged with Slits for 40 h in vitro. The corona of neurites emanating from SAG explants was unaffected by the presence of purified human Slit1 and Slit2 in the culture medium. Reduced axon outgrowth from E8 olfactory bulbs cultured under similar conditions for 24 h confirmed bioactivity of purified human Slits on chicken neurons. In summary, differential sensitivity to Slit repellents may influence the directional outgrowth of otic axons toward either the anterior or posterior otocyst.
Collapse
Affiliation(s)
- Andrea C Battisti
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| | - Kristen N Fantetti
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| | - Belle A Moyers
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| | - Donna M Fekete
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| |
Collapse
|
13
|
A critical role for macrophages near axotomized neuronal cell bodies in stimulating nerve regeneration. J Neurosci 2013; 33:16236-48. [PMID: 24107955 DOI: 10.1523/jneurosci.3319-12.2013] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Macrophages have been implicated in peripheral nerve regeneration for some time, supposedly through their involvement in Wallerian degeneration, the process by which the distal nerve degenerates after axotomy and is cleared by phagocytosis. Thus, in several studies in which macrophage accumulation in the distal nerve was reduced and Wallerian degeneration inhibited, regeneration was delayed. However, this interpretation ignores the more recent findings that macrophages also accumulate around axotomized cell bodies. The function of macrophage action at this second site has not been clear. In two mutant strains of mice, the slow Wallerian degeneration (Wld(s)) mouse and the chemokine receptor CCR2 knock-out mouse, we report that macrophage accumulation after axotomy was abolished in both the dorsal root ganglion (DRG) and the distal sciatic nerve. To measure neurite outgrowth, DRG neurons were given a conditioning lesion, and outgrowth was measured in vitro 7 d later in the absence of the distal nerve segment. The increased growth normally seen after a conditioning lesion did not occur or was reduced in Wld(s) or CCR2(-/-) mice. In the superior cervical ganglion (SCG), particularly in Wld(s) mice, macrophage accumulation was reduced but not abolished after axotomy. In SCG neurons from Wld(s) mice, the conditioning lesion response was unchanged; however, in CCR2(-/-) mice in which the effect on macrophage accumulation was greater, SCG neurite outgrowth was significantly reduced. These results indicate that macrophages affect neurite outgrowth by acting at the level of peripheral ganglia in addition to any effects they might produce by facilitation of Wallerian degeneration.
Collapse
|
14
|
Bank LM, Bianchi LM, Ebisu F, Lerman-Sinkoff D, Smiley EC, Shen YC, Ramamurthy P, Thompson DL, Roth TM, Beck CR, Flynn M, Teller RS, Feng L, Llewellyn GN, Holmes B, Sharples C, Coutinho-Budd J, Linn SA, Chervenak AP, Dolan DF, Benson J, Kanicki A, Martin CA, Altschuler R, Koch AE, Koch AE, Jewett EM, Germiller JA, Barald KF. Macrophage migration inhibitory factor acts as a neurotrophin in the developing inner ear. Development 2013; 139:4666-74. [PMID: 23172918 DOI: 10.1242/dev.066647] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study is the first to demonstrate that macrophage migration inhibitory factor (MIF), an immune system 'inflammatory' cytokine that is released by the developing otocyst, plays a role in regulating early innervation of the mouse and chick inner ear. We demonstrate that MIF is a major bioactive component of the previously uncharacterized otocyst-derived factor, which directs initial neurite outgrowth from the statoacoustic ganglion (SAG) to the developing inner ear. Recombinant MIF acts as a neurotrophin in promoting both SAG directional neurite outgrowth and neuronal survival and is expressed in both the developing and mature inner ear of chick and mouse. A MIF receptor, CD74, is found on both embryonic SAG neurons and adult mouse spiral ganglion neurons. Mif knockout mice are hearing impaired and demonstrate altered innervation to the organ of Corti, as well as fewer sensory hair cells. Furthermore, mouse embryonic stem cells become neuron-like when exposed to picomolar levels of MIF, suggesting the general importance of this cytokine in neural development.
Collapse
Affiliation(s)
- Lisa M Bank
- Department of Cell and Developmental Biology, University of Michigan Medical School, 3728 BSRB 109, Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Shen YC, Thompson DL, Kuah MK, Wong KL, Wu KL, Linn SA, Jewett EM, Shu-Chien AC, Barald KF. The cytokine macrophage migration inhibitory factor (MIF) acts as a neurotrophin in the developing inner ear of the zebrafish, Danio rerio. Dev Biol 2011; 363:84-94. [PMID: 22210003 DOI: 10.1016/j.ydbio.2011.12.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/06/2011] [Accepted: 12/21/2011] [Indexed: 10/14/2022]
Abstract
Macrophage migration inhibitory factor (MIF) plays versatile roles in the immune system. MIF is also widely expressed during embryonic development, particularly in the nervous system, although its roles in neural development are only beginning to be understood. Evidence from frogs, mice and zebrafish suggests that MIF has a major role as a neurotrophin in the early development of sensory systems, including the auditory system. Here we show that the zebrafish mif pathway is required for both sensory hair cell (HC) and sensory neuronal cell survival in the ear, for HC differentiation, semicircular canal formation, statoacoustic ganglion (SAG) development, and lateral line HC differentiation. This is consistent with our findings that MIF is expressed in the developing mammalian and avian auditory systems and promotes mouse and chick SAG neurite outgrowth and neuronal survival, demonstrating key instructional roles for MIF in vertebrate otic development.
Collapse
Affiliation(s)
- Yu-chi Shen
- Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Garred MM, Wang MM, Guo X, Harrington CA, Lein PJ. Transcriptional responses of cultured rat sympathetic neurons during BMP-7-induced dendritic growth. PLoS One 2011; 6:e21754. [PMID: 21765909 PMCID: PMC3135585 DOI: 10.1371/journal.pone.0021754] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 06/06/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dendrites are the primary site of synapse formation in the vertebrate nervous system; however, relatively little is known about the molecular mechanisms that regulate the initial formation of primary dendrites. Embryonic rat sympathetic neurons cultured under defined conditions extend a single functional axon, but fail to form dendrites. Addition of bone morphogenetic proteins (BMPs) triggers these neurons to extend multiple dendrites without altering axonal growth or cell survival. We used this culture system to examine differential gene expression patterns in naïve vs. BMP-treated sympathetic neurons in order to identify candidate genes involved in regulation of primary dendritogenesis. METHODOLOGY/PRINCIPAL FINDINGS To determine the critical transcriptional window during BMP-induced dendritic growth, morphometric analysis of microtubule-associated protein (MAP-2)-immunopositive processes was used to quantify dendritic growth in cultures exposed to the transcription inhibitor actinomycin-D added at varying times after addition of BMP-7. BMP-7-induced dendritic growth was blocked when transcription was inhibited within the first 24 hr after adding exogenous BMP-7. Thus, total RNA was isolated from sympathetic neurons exposed to three different experimental conditions: (1) no BMP-7 treatment; (2) treatment with BMP-7 for 6 hr; and (3) treatment with BMP-7 for 24 hr. Affymetrix oligonucleotide microarrays were used to identify differential gene expression under these three culture conditions. BMP-7 significantly regulated 56 unique genes at 6 hr and 185 unique genes at 24 hr. Bioinformatic analyses implicate both established and novel genes and signaling pathways in primary dendritogenesis. CONCLUSIONS/SIGNIFICANCE This study provides a unique dataset that will be useful in generating testable hypotheses regarding transcriptional control of the initial stages of dendritic growth. Since BMPs selectively promote dendritic growth in central neurons as well, these findings may be generally applicable to dendritic growth in other neuronal cell types.
Collapse
Affiliation(s)
- Michelle M. Garred
- Gene Microarray Shared Resource, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Michael M. Wang
- Departments of Neurology and Molecular & Integrative Physiology, University of Michigan, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
| | - Xin Guo
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Christina A. Harrington
- Gene Microarray Shared Resource, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
17
|
Yang T, Kersigo J, Jahan I, Pan N, Fritzsch B. The molecular basis of making spiral ganglion neurons and connecting them to hair cells of the organ of Corti. Hear Res 2011; 278:21-33. [PMID: 21414397 DOI: 10.1016/j.heares.2011.03.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 03/01/2011] [Accepted: 03/07/2011] [Indexed: 11/28/2022]
Abstract
The bipolar spiral ganglion neurons apparently delaminate from the growing cochlear duct and migrate to Rosenthal's canal. They project radial fibers to innervate the organ of Corti (type I neurons to inner hair cells, type II neurons to outer hair cells) and also project tonotopically to the cochlear nuclei. The early differentiation of these neurons requires transcription factors to regulate migration, pathfinding and survival. Neurog1 null mice lack formation of neurons. Neurod1 null mice show massive neuronal death combined with aberrant central and peripheral projections. Prox1 protein is necessary for proper type II neuron process navigation, which is also affected by the neurotrophins Bdnf and Ntf3. Neurotrophin null mutants show specific patterns of neuronal loss along the cochlea but remaining neurons compensate by expanding their target area. All neurotrophin mutants have reduced radial fiber growth proportional to the degree of loss of neurotrophin alleles. This suggests a simple dose response effect of neurotrophin concentration. Keeping overall concentration constant, but misexpressing one neurotrophin under regulatory control of another one results in exuberant fiber growth not only of vestibular fibers to the cochlea but also of spiral ganglion neurons to outer hair cells suggesting different effectiveness of neurotrophins for spiral ganglion neurite growth. Finally, we report here for the first time that losing all neurons in double null mutants affects extension of the cochlear duct and leads to formation of extra rows of outer hair cells in the apex, possibly by disrupting the interaction of the spiral ganglion with the elongating cochlea.
Collapse
Affiliation(s)
- Tian Yang
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, 143 BB, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
18
|
Appler JM, Goodrich LV. Connecting the ear to the brain: Molecular mechanisms of auditory circuit assembly. Prog Neurobiol 2011; 93:488-508. [PMID: 21232575 DOI: 10.1016/j.pneurobio.2011.01.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 12/09/2010] [Accepted: 01/03/2011] [Indexed: 12/21/2022]
Abstract
Our sense of hearing depends on precisely organized circuits that allow us to sense, perceive, and respond to complex sounds in our environment, from music and language to simple warning signals. Auditory processing begins in the cochlea of the inner ear, where sounds are detected by sensory hair cells and then transmitted to the central nervous system by spiral ganglion neurons, which faithfully preserve the frequency, intensity, and timing of each stimulus. During the assembly of auditory circuits, spiral ganglion neurons establish precise connections that link hair cells in the cochlea to target neurons in the auditory brainstem, develop specific firing properties, and elaborate unusual synapses both in the periphery and in the CNS. Understanding how spiral ganglion neurons acquire these unique properties is a key goal in auditory neuroscience, as these neurons represent the sole input of auditory information to the brain. In addition, the best currently available treatment for many forms of deafness is the cochlear implant, which compensates for lost hair cell function by directly stimulating the auditory nerve. Historically, studies of the auditory system have lagged behind other sensory systems due to the small size and inaccessibility of the inner ear. With the advent of new molecular genetic tools, this gap is narrowing. Here, we summarize recent insights into the cellular and molecular cues that guide the development of spiral ganglion neurons, from their origin in the proneurosensory domain of the otic vesicle to the formation of specialized synapses that ensure rapid and reliable transmission of sound information from the ear to the brain.
Collapse
Affiliation(s)
- Jessica M Appler
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
19
|
Moore NH, Costa LG, Shaffer SA, Goodlett DR, Guizzetti M. Shotgun proteomics implicates extracellular matrix proteins and protease systems in neuronal development induced by astrocyte cholinergic stimulation. J Neurochem 2010. [DOI: 10.1111/j.0022-3042.2008.05836.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Alteration of cytokine profiles in mice exposed to chronic low-dose ionizing radiation. Biochem Biophys Res Commun 2010; 397:644-9. [DOI: 10.1016/j.bbrc.2010.05.121] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 05/22/2010] [Indexed: 01/20/2023]
|
21
|
Katayama KI, Zine A, Ota M, Matsumoto Y, Inoue T, Fritzsch B, Aruga J. Disorganized innervation and neuronal loss in the inner ear of Slitrk6-deficient mice. PLoS One 2009; 4:e7786. [PMID: 19936227 PMCID: PMC2777407 DOI: 10.1371/journal.pone.0007786] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 10/14/2009] [Indexed: 12/24/2022] Open
Abstract
Slitrks are type I transmembrane proteins that share conserved leucine-rich repeat domains similar to those in the secreted axonal guidance molecule Slit. They also show similarities to Ntrk neurotrophin receptors in their carboxy-termini, sharing a conserved tyrosine residue. Among 6 Slitrk family genes in mammals, Slitrk6 has a unique expression pattern, with strong expression in the sensory epithelia of the inner ear. We generated Slitrk6-knockout mice and investigated the development of their auditory and vestibular sensory organs. Slitrk6-deficient mice showed pronounced reduction in the cochlear innervation. In the vestibule, the innervation to the posterior crista was often lost, reduced, or sometimes misguided. These defects were accompanied by the loss of neurons in the spiral and vestibular ganglia. Cochlear sensory epithelia from Slitrk6-knockout mice have reduced ability in promoting neurite outgrowth of spiral ganglion neurons. Indeed the Slitrk6-deficient inner ear showed a mild but significant decrease in the expression of Bdnf and Ntf3, both of which are essential for the innervation and survival of sensory neurons. In addition, the expression of Ntrk receptors, including their phosphorylated forms was decreased in Slitrk6-knockout cochlea. These results suggest that Slitrk6 promotes innervation and survival of inner ear sensory neurons by regulating the expression of trophic and/or tropic factors including neurotrophins from sensory epithelia.
Collapse
Affiliation(s)
- Kei-ichi Katayama
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Wako-shi, Saitama, Japan
| | - Azel Zine
- University of Montpellier I, Institute of Neurosciences, INSERM U583, Montpellier, France
| | - Maya Ota
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Wako-shi, Saitama, Japan
| | - Yoshifumi Matsumoto
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Wako-shi, Saitama, Japan
| | - Takashi Inoue
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Wako-shi, Saitama, Japan
| | - Bernd Fritzsch
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Jun Aruga
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute (BSI), Wako-shi, Saitama, Japan
| |
Collapse
|
22
|
Shen YC, Li D, Al-Shoaibi A, Bersano-Begey T, Chen H, Ali S, Flak B, Perrin C, Winslow M, Shah H, Ramamurthy P, Schmedlen RH, Takayama S, Barald KF. A student team in a University of Michigan biomedical engineering design course constructs a microfluidic bioreactor for studies of zebrafish development. Zebrafish 2009; 6:201-13. [PMID: 19292670 DOI: 10.1089/zeb.2008.0572] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The zebrafish is a valuable model for teaching developmental, molecular, and cell biology; aquatic sciences; comparative anatomy; physiology; and genetics. Here we demonstrate that zebrafish provide an excellent model system to teach engineering principles. A seven-member undergraduate team in a biomedical engineering class designed, built, and tested a zebrafish microfluidic bioreactor applying microfluidics, an emerging engineering technology, to study zebrafish development. During the semester, students learned engineering and biology experimental design, chip microfabrication, mathematical modeling, zebrafish husbandry, principles of developmental biology, fluid dynamics, microscopy, and basic molecular biology theory and techniques. The team worked to maximize each person's contribution and presented weekly written and oral reports. Two postdoctoral fellows, a graduate student, and three faculty instructors coordinated and directed the team in an optimal blending of engineering, molecular, and developmental biology skill sets. The students presented two posters, including one at the Zebrafish meetings in Madison, Wisconsin (June 2008).
Collapse
Affiliation(s)
- Yu-chi Shen
- Department of Cell and Developmental Biology, Medical School, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Moore NH, Costa LG, Shaffer SA, Goodlett DR, Guizzetti M. Shotgun proteomics implicates extracellular matrix proteins and protease systems in neuronal development induced by astrocyte cholinergic stimulation. J Neurochem 2008; 108:891-908. [PMID: 19077055 DOI: 10.1111/j.1471-4159.2008.05836.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astrocytes play an important role in neuronal development through the release of soluble factors that affect neuronal maturation. Shotgun proteomics followed by gene ontology analysis was used in this study to identify proteins present in the conditioned medium of primary rat astrocytes. One hundred and thirty three secreted proteins were identified, the majority of which were never before reported to be produced by astrocytes. Extracellular proteins were classified based on their biological and molecular functions; most of the identified proteins were involved in neuronal development. Semi-quantitative proteomic analysis was carried out to identify changes in the levels of proteins released by astrocytes after stimulation with the cholinergic agonist carbachol, as we have previously reported that carbachol-treated astrocytes elicit neuritogenesis in hippocampal neurons through the release of soluble factors. Carbachol up-regulated secretion of 15 proteins and down-regulated the release of 17 proteins. Changes in the levels of four proteins involved in neuronal differentiation (thrombospondin-1, fibronectin, plasminogen activator inhibitor-1, and plasminogen activator urokinase) were verified by western blot or ELISA. In conclusion, this study identified a large number of proteins involved in neuronal development in the astrocyte secretome and implicated extracellular matrix proteins and protease systems in neuronal development induced by astrocyte cholinergic stimulation.
Collapse
Affiliation(s)
- Nadia H Moore
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, USA
| | | | | | | | | |
Collapse
|
24
|
Roth TM, Ramamurthy P, Ebisu F, Lisak RP, Bealmear BM, Barald KF. A mouse embryonic stem cell model of Schwann cell differentiation for studies of the role of neurofibromatosis type 1 in Schwann cell development and tumor formation. Glia 2007; 55:1123-33. [PMID: 17597122 DOI: 10.1002/glia.20534] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The neurofibromatosis Type 1 (NF1) gene functions as a tumor suppressor gene. One known function of neurofibromin, the NF1 protein product, is to accelerate the slow intrinsic GTPase activity of Ras to increase the production of inactive rasGDP, with wide-ranging effects on p21ras pathways. Loss of neurofibromin in the autosomal dominant disorder NF1 is associated with tumors of the peripheral nervous system, particularly neurofibromas, benign lesions in which the major affected cell type is the Schwann cell (SC). NF1 is the most common cancer predisposition syndrome affecting the nervous system. We have developed an in vitro system for differentiating mouse embryonic stem cells (mESC) that are NF1 wild type (+/+), heterozygous (+/-), or null (-/-) into SC-like cells to study the role of NF1 in SC development and tumor formation. These mES-generated SC-like cells, regardless of their NF1 status, express SC markers correlated with their stage of maturation, including myelin proteins. They also support and preferentially direct neurite outgrowth from primary neurons. NF1 null and heterozygous SC-like cells proliferate at an accelerated rate compared to NF1 wild type; this growth advantage can be reverted to wild type levels using an inhibitor of MAP kinase kinase (Mek). The mESC of all NF1 types can also be differentiated into neuron-like cells. This novel model system provides an ideal paradigm for studies of the role of NF1 in cell growth and differentiation of the different cell types affected by NF1 in cells with differing levels of neurofibromin that are neither transformed nor malignant.
Collapse
Affiliation(s)
- Therese M Roth
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | | | | |
Collapse
|