1
|
Wang S, Chakraborty S, Fu Y, Lee MP, Liu J, Waldhaus J. 3D reconstruction of the mouse cochlea from scRNA-seq data suggests morphogen-based principles in apex-to-base specification. Dev Cell 2024; 59:1538-1552.e6. [PMID: 38593801 PMCID: PMC11187690 DOI: 10.1016/j.devcel.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/03/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
In the mammalian auditory system, frequency discrimination depends on numerous morphological and physiological properties of the organ of Corti, which gradually change along the apex-to-base (tonotopic) axis of the organ. For example, the basilar membrane stiffness changes tonotopically, thus affecting the tuning properties of individual hair cells. At the molecular level, those frequency-specific characteristics are mirrored by gene expression gradients; however, the molecular mechanisms controlling tonotopic gene expression in the mouse cochlea remain elusive. Through analyzing single-cell RNA sequencing (scRNA-seq) data from E12.5 and E14.5 time points, we predicted that morphogens, rather than a cell division-associated mechanism, confer spatial identity in the extending cochlea. Subsequently, we reconstructed the developing cochlea in 3D space from scRNA-seq data to investigate the molecular pathways mediating positional information. The retinoic acid (RA) and hedgehog pathways were found to form opposing apex-to-base gradients, and functional interrogation using mouse cochlear explants suggested that both pathways jointly specify the longitudinal axis.
Collapse
Affiliation(s)
- Shuze Wang
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Saikat Chakraborty
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yujuan Fu
- Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Mary P Lee
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joerg Waldhaus
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Chen M, Huang J. Retinoic acid induces differentiation of cochlear neural progenitor cells into hair cells. Braz J Otorhinolaryngol 2021; 88:962-967. [PMID: 33707121 PMCID: PMC9615533 DOI: 10.1016/j.bjorl.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 01/05/2021] [Accepted: 01/30/2021] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Inner ear progenitor cells have the potential for multi-directional differentiation. Retinoic acid is an important requirement for the development of the inner ear. Blocking the Curtyr's retinoic acid signaling pathway can significantly reduce the number of hair cells. Therefore, we believe that retinoic acid may induce the regeneration of inner ear hair cells. OBJECTIVE To investigate whether the cochlear neural progenitor cells maintain the characteristics of stem cells during recovery and subculture, whether retinoic acid can induce cochlear neural progenitor cells into hair cells in vitro, and whether retinoic acid promotes or inhibits the proliferation of cochlear neural progenitor cells during differentiation. METHODS Cochlear neural progenitor cells were cultured and induced in DMEM/F12+RA (10-6M) and then detected the expressions of hair cell markers (Math1 and MyosinVIIa) by immunofluorescence cytochemistry and realtime-polymerase chain reaction, and the proliferation of cochlear neural progenitor cells was detected by Brdu. RESULTS The nestin of cochlear neural progenitor cells was positively expressed. The ratios of Math1-positive cells in the control group and experimental group were 1.5% and 63%, respectively; the ratios of MyosinVIIa-positive cells in the control group and experimental group were 0.96% and 56%, respectively (p<0.05). The ratios of Brdu+-labeled cells in retinoic acid group, group PBS, and group FBS were 20.6%, 29.9%, and 54.3%, respectively; however, the proliferation rate in the experimental group decreased. CONCLUSION Retinoic acid can promote cochlear neural progenitor cells to differentiate into the hair cells.
Collapse
Affiliation(s)
- Minyun Chen
- The Second Affiliated Hospital of Fujian Medical University, Department of Otolaryngology, Fujian, China
| | - Jianmin Huang
- Fujian Medical University Union Hospital, Department of Otolaryngology, Fujian, China.
| |
Collapse
|
3
|
dos Santos ÍGD, de Oliveira Mendes TA, Silva GAB, Reis AMS, Monteiro-Vitorello CB, Schaker PDC, Herai RH, Fabotti ABC, Coutinho LL, Jorge EC. Didelphis albiventris: an overview of unprecedented transcriptome sequencing of the white-eared opossum. BMC Genomics 2019; 20:866. [PMID: 31730444 PMCID: PMC6858782 DOI: 10.1186/s12864-019-6240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The white-eared opossum (Didelphis albiventris) is widely distributed throughout Brazil and South America. It has been used as an animal model for studying different scientific questions ranging from the restoration of degraded green areas to medical aspects of Chagas disease, leishmaniasis and resistance against snake venom. As a marsupial, D. albiventris can also contribute to the understanding of the molecular mechanisms that govern the different stages of organogenesis. Opossum joeys are born after only 13 days, and the final stages of organogenesis occur when the neonates are inside the pouch, depending on lactation. As neither the genome of this opossum species nor its transcriptome has been completely sequenced, the use of D. albiventris as an animal model is limited. In this work, we sequenced the D. albiventris transcriptome by RNA-seq to obtain the first catalogue of differentially expressed (DE) genes and gene ontology (GO) annotations during the neonatal stages of marsupial development. RESULTS The D. albiventris transcriptome was obtained from whole neonates harvested at birth (P0), at 5 days of age (P5) and at 10 days of age (P10). The de novo assembly of these transcripts generated 85,338 transcripts. Approximately 30% of these transcripts could be mapped against the amino acid sequences of M. domestica, the evolutionarily closest relative of D. albiventris to be sequenced thus far. Among the expressed transcripts, 2077 were found to be DE between P0 and P5, 13,780 between P0 and P10, and 1453 between P5 and P10. The enriched GO terms were mainly related to the immune system, blood tissue development and differentiation, vision, hearing, digestion, the CNS and limb development. CONCLUSIONS The elucidation of opossum transcriptomes provides an out-group for better understanding the distinct characteristics associated with the evolution of mammalian species. This study provides the first transcriptome sequences and catalogue of genes for a marsupial species at different neonatal stages, allowing the study of the mechanisms involved in organogenesis.
Collapse
Affiliation(s)
- Íria Gabriela Dias dos Santos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | - Gerluza Aparecida Borges Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Amanda Maria Sena Reis
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | - Patricia Dayane Carvalho Schaker
- Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, São Paulo Brazil
| | - Roberto Hirochi Herai
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, Brazil
| | | | - Luiz Lehmann Coutinho
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, São Paulo Brazil
| | - Erika Cristina Jorge
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| |
Collapse
|
4
|
Mawson AR, Croft AM. Rubella Virus Infection, the Congenital Rubella Syndrome, and the Link to Autism. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E3543. [PMID: 31546693 PMCID: PMC6801530 DOI: 10.3390/ijerph16193543] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/09/2019] [Accepted: 09/15/2019] [Indexed: 12/16/2022]
Abstract
Rubella is a systemic virus infection that is usually mild. It can, however, cause severe birth defects known as the congenital rubella syndrome (CRS) when infection occurs early in pregnancy. As many as 8%-13% of children with CRS developed autism during the rubella epidemic of the 1960s compared to the background rate of about 1 new case per 5000 children. Rubella infection and CRS are now rare in the U.S. and in Europe due to widespread vaccination. However, autism rates have risen dramatically in recent decades to about 3% of children today, with many cases appearing after a period of normal development ('regressive autism'). Evidence is reviewed here suggesting that the signs and symptoms of rubella may be due to alterations in the hepatic metabolism of vitamin A (retinoids), precipitated by the acute phase of the infection. The infection causes mild liver dysfunction and the spillage of stored vitamin A compounds into the circulation, resulting in an endogenous form of hypervitaminosis A. Given that vitamin A is a known teratogen, it is suggested that rubella infection occurring in the early weeks of pregnancy causes CRS through maternal liver dysfunction and exposure of the developing fetus to excessive vitamin A. On this view, the multiple manifestations of CRS and associated autism represent endogenous forms of hypervitaminosis A. It is further proposed that regressive autism results primarily from post-natal influences of a liver-damaging nature and exposure to excess vitamin A, inducing CRS-like features as a function of vitamin A toxicity, but without the associated dysmorphogenesis. A number of environmental factors are discussed that may plausibly be candidates for this role, and suggestions are offered for testing the model. The model also suggests a number of measures that may be effective both in reducing the risk of fetal CRS in women who acquire rubella in their first trimester and in reversing or minimizing regressive autism among children in whom the diagnosis is suspected or confirmed.
Collapse
Affiliation(s)
- Anthony R Mawson
- Department of Epidemiology and Biostatistics, School of Public Health, College of Health Sciences, Jackson State University, Jackson, MS 39213, USA.
| | - Ashley M Croft
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| |
Collapse
|
5
|
Rah YC, Park S, Koun S, Park HC, Choi J. In vivo assay of the ethanol-induced embryonic hair cell loss and the protective role of the retinoic and folic acid in zebrafish larvae (Danio rerio). Alcohol 2019; 75:113-121. [PMID: 30640074 DOI: 10.1016/j.alcohol.2018.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 06/15/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022]
Abstract
In reference to the auditory manifestation of fetal alcohol syndrome, previous work has preferentially focused on the deviant neural development of the auditory system. Changes in the sensory hair cell, the ultimate sensory organ, were not well understood. In this study, we carried out an in vivo assessment of the embryonic hair cell changes on the lateral line of zebrafish upon exposure to various ethanol concentrations (0.25%, 0.5%, 0.75%, and 1.0%). A significant decrease in the hair cell count was confirmed as the ethanol concentration increased. Long-term observation (up to 240 hours post-fertilization [hpf]) suggested an irreversible hair cell loss with little chance of a simple delayed development. For an underlying biological process, a significant increase of hair cell apoptosis and a significant decrease of cytoplasmic mitochondria were confirmed as the ethanol concentration increased. Co-treatment with retinoic (0.1 nM) or folic (0.1 mM) acid with the same concentrations of ethanol resulted in significant increases in the remaining hair cells, compared to the ethanol-only treatment group, for every ethanol concentration. The retinoic acid provided more effective protection over folic acid, resulting in no significant changes in hair cell counts for every ethanol concentration (except 1.0%), compared with that of the negative control (without chemical treatment). Hair cell counts in every ethanol concentration were significantly lower than those in negative controls without chemical treatment after folic acid co-treatment. In conclusion, gestational ethanol exposure causes developmental sensory hair cell loss. Potential underlying mechanisms include retinoic or folic acid deficiency, and mitochondrial damage with subsequent hair cell apoptosis. Hair cell loss could possibly be prevented by administering either retinoic or folic acid, with retinoic acid supplementation as the preferred treatment.
Collapse
|
6
|
Sphingosine 1-phosphate-mediated activation of ezrin-radixin-moesin proteins contributes to cytoskeletal remodeling and changes of membrane properties in epithelial otic vesicle progenitors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:554-565. [PMID: 30611767 DOI: 10.1016/j.bbamcr.2018.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/24/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022]
Abstract
Hearing loss is among the most prevalent sensory impairments in humans. Cochlear implantable devices represent the current therapies for hearing loss but have various shortcomings. ERM (ezrin- radixin -moesin) are a family of adaptor proteins that link plasma membrane with actin cytoskeleton, playing a crucial role in cell morphology and in the formation of membrane protrusions. Recently, bioactive sphingolipids have emerged as regulators of ERM proteins. Sphingosine 1-phosphate (S1P) is a pleiotropic sphingolipid which regulates fundamental cellular functions such as proliferation, survival, migration as well as processes such as development and inflammation mainly via ligation to its specific receptors S1PR (S1P1-5). Experimental findings demonstrate a key role for S1P signaling axis in the maintenance of auditory function. Preservation of cellular junctions is a fundamental function both for S1P and ERM proteins, crucial for the maintenance of cochlear integrity. In the present work, S1P was found to activate ERM in a S1P2-dependent manner in murine auditory epithelial progenitors US/VOT-E36. S1P-induced ERM activation potently contributed to actin cytoskeletal remodeling and to the appearance of ionic currents and membrane passive properties changes typical of more differentiated cells. Moreover, PKC and Akt activation was found to mediate S1P-induced ERM phosphorylation. The obtained findings contribute to demonstrate the role of S1P signaling pathway in inner ear biology and to disclose potential innovative therapeutical approaches in the field of hearing loss prevention and treatment.
Collapse
|
7
|
Сhurbanov AY, Karafet TM, Morozov IV, Mikhalskaia VY, Zytsar MV, Bondar AA, Posukh OL. Whole Exome Sequencing Reveals Homozygous Mutations in RAI1, OTOF, and SLC26A4 Genes Associated with Nonsyndromic Hearing Loss in Altaian Families (South Siberia). PLoS One 2016; 11:e0153841. [PMID: 27082237 PMCID: PMC4833413 DOI: 10.1371/journal.pone.0153841] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 04/05/2016] [Indexed: 12/15/2022] Open
Abstract
Hearing loss (HL) is one of the most common sensorineural disorders and several dozen genes contribute to its pathogenesis. Establishing a genetic diagnosis of HL is of great importance for clinical evaluation of deaf patients and for estimating recurrence risks for their families. Efforts to identify genes responsible for HL have been challenged by high genetic heterogeneity and different ethnic-specific prevalence of inherited deafness. Here we present the utility of whole exome sequencing (WES) for identifying candidate causal variants for previously unexplained nonsyndromic HL of seven patients from four unrelated Altaian families (the Altai Republic, South Siberia). The WES analysis revealed homozygous missense mutations in three genes associated with HL. Mutation c.2168A>G (SLC26A4) was found in one family, a novel mutation c.1111G>C (OTOF) was revealed in another family, and mutation c.5254G>A (RAI1) was found in two families. Sanger sequencing was applied for screening of identified variants in an ethnically diverse cohort of other patients with HL (n = 116) and in Altaian controls (n = 120). Identified variants were found only in patients of Altaian ethnicity (n = 93). Several lines of evidences support the association of homozygosity for discovered variants c.5254G>A (RAI1), c.1111C>G (OTOF), and c.2168A>G (SLC26A4) with HL in Altaian patients. Local prevalence of identified variants implies possible founder effect in significant number of HL cases in indigenous population of the Altai region. Notably, this is the first reported instance of patients with RAI1 missense mutation whose HL is not accompanied by specific traits typical for Smith-Magenis syndrome. Presumed association of RAI1 gene variant c.5254G>A with isolated HL needs to be proved by further experimental studies.
Collapse
Affiliation(s)
- Alexander Y. Сhurbanov
- Arizona Research Laboratories, Division of Biotechnology, University of Arizona, Tucson, Arizona, United States of America
| | - Tatiana M. Karafet
- Arizona Research Laboratories, Division of Biotechnology, University of Arizona, Tucson, Arizona, United States of America
| | - Igor V. Morozov
- SB RAS Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
- Novosibirsk State University, Novosibirsk, Russian Federation
| | - Valeriia Yu. Mikhalskaia
- Novosibirsk State University, Novosibirsk, Russian Federation
- Laboratory of Human Molecular Genetics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Marina V. Zytsar
- Novosibirsk State University, Novosibirsk, Russian Federation
- Laboratory of Human Molecular Genetics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Alexander A. Bondar
- SB RAS Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Olga L. Posukh
- Novosibirsk State University, Novosibirsk, Russian Federation
- Laboratory of Human Molecular Genetics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
- * E-mail:
| |
Collapse
|
8
|
Basch ML, Brown RM, Jen H, Groves AK. Where hearing starts: the development of the mammalian cochlea. J Anat 2016; 228:233-54. [PMID: 26052920 PMCID: PMC4718162 DOI: 10.1111/joa.12314] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2015] [Indexed: 12/11/2022] Open
Abstract
The mammalian cochlea is a remarkable sensory organ, capable of perceiving sound over a range of 10(12) in pressure, and discriminating both infrasonic and ultrasonic frequencies in different species. The sensory hair cells of the mammalian cochlea are exquisitely sensitive, responding to atomic-level deflections at speeds on the order of tens of microseconds. The number and placement of hair cells are precisely determined during inner ear development, and a large number of developmental processes sculpt the shape, size and morphology of these cells along the length of the cochlear duct to make them optimally responsive to different sound frequencies. In this review, we briefly discuss the evolutionary origins of the mammalian cochlea, and then describe the successive developmental processes that lead to its induction, cell cycle exit, cellular patterning and the establishment of topologically distinct frequency responses along its length.
Collapse
Affiliation(s)
- Martin L. Basch
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
| | - Rogers M. Brown
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Hsin‐I Jen
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Andrew K. Groves
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
9
|
Developmental exposure to purity-controlled polychlorinated biphenyl congeners (PCB74 and PCB95) in rats: effects on brainstem auditory evoked potentials and catalepsy. Toxicology 2014; 327:22-31. [PMID: 25449634 DOI: 10.1016/j.tox.2014.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/11/2014] [Accepted: 11/11/2014] [Indexed: 01/12/2023]
Abstract
Whereas the effects of dioxin-like polychlorinated biphenyls (DL-PCBs) are well described, less is known about non-dioxin-like PCBs (NDL-PCBs), including influences on the nervous system and related behavioral effects after developmental exposure. Following the examination of the highly purified NDL congeners PCB52 and PCB180, we report here the results of experiments with PCB74 and PCB95. Rat dams were orally exposed to equimolar doses of either congener (40μmol/kg bw - 11.68mg PCB74/kg bw or 13.06mg PCB95/kg bw) from gestational day (GD) 10 to postnatal day (PND) 7. Control dams were given the vehicle. Adult offspring were tested for cataleptic behavior after induction with haloperidol, a classical neuroleptic drug, and brainstem auditory evoked potentials (BAEPs), using clicks and tone pips of different frequencies for stimulation. Results revealed slight effects on latencies to movement onset in female offspring exposed to PCB74, whereas PCB74 males and offspring exposed to PCB95 were not affected. Pronounced changes were observed in BAEPs at low frequencies in PCB74 offspring, with elevated thresholds in both sexes. PCB95 increased thresholds in males, but not females. Small effects were detected on latency of the late wave IV in both sexes after developmental exposure to PCB74 or PCB95. Compared with the other NDL-PCB congeners tested, PCB74 caused the most pronounced effects on BAEPs.
Collapse
|
10
|
Tornari C, Towers ER, Gale JE, Dawson SJ. Regulation of the orphan nuclear receptor Nr2f2 by the DFNA15 deafness gene Pou4f3. PLoS One 2014; 9:e112247. [PMID: 25372459 PMCID: PMC4221282 DOI: 10.1371/journal.pone.0112247] [Citation(s) in RCA: 11] [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: 09/09/2014] [Accepted: 10/08/2014] [Indexed: 12/23/2022] Open
Abstract
Hair cells are the mechanotransducing cells of the inner ear that are essential for hearing and balance. POU4F3--a POU-domain transcription factor selectively expressed by these cells--has been shown to be essential for hair cell differentiation and survival in mice and its mutation in humans underlies late-onset progressive hearing loss (DFNA15). The downstream targets of POU4F3 are required for hair cell differentiation and survival. We aimed to identify such targets in order to elucidate the molecular pathways involved in hair cell production and maintenance. The orphan thyroid nuclear receptor Nr2f2 was identified as a POU4F3 target using a subtractive hybridization strategy and EMSA analysis showed that POU4F3 binds to two sites in the Nr2f2 5' flanking region. These sites were shown to be required for POU4F3 activation as their mutation leads to a reduction in the response of an Nr2f2 5' flanking region reporter construct to POU4F3. Immunocytochemistry was carried out in the developing and adult inner ear in order to investigate the relevance of this interaction in hearing. NR2F2 expression in the postnatal mouse organ of Corti was shown to be detectable in all sensory epithelia examined and characterised. These data demonstrate that Nr2f2 is a direct target of POU4F3 in vitro and that this regulatory relationship may be relevant to hair cell development and survival.
Collapse
Affiliation(s)
| | - Emily R. Towers
- UCL Ear Institute, University College London, London, United Kingdom
| | - Jonathan E. Gale
- UCL Ear Institute, University College London, London, United Kingdom
| | - Sally J. Dawson
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
11
|
Fares S, Sethom MM, Khouaja-Mokrani C, Jabnoun S, Feki M, Kaabachi N. VitaminA, E, and D deficiencies in tunisian very low birth weight neonates: prevalence and risk factors. Pediatr Neonatol 2014; 55:196-201. [PMID: 24289974 DOI: 10.1016/j.pedneo.2013.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 07/09/2013] [Accepted: 09/26/2013] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Preterm neonates are at high risk of vitamin deficiencies, which may expose them to increased morbidity and mortality. This study aimed to determine the prevalence and risk factors for vitamin A, E, and D deficiencies in Tunisian very low birth weight (VLBW) neonates. METHODS A total of 607 VLBW and 300 term neonates were included in the study. Plasma vitamins A and E were assessed by high performance liquid chromatography and vitamin D was assessed by radioimmunoassay. RESULTS Prevalence of vitamin A, E, and D deficiencies were dramatically elevated in VLBW neonates and were significantly higher than term neonates (75.9% vs. 63.3%; 71.3% vs. 55.5%; and 65.2% vs. 40.4%, respectively). In VLBW neonates, the prevalence of vitamin deficiencies was significantly higher in lower classes of gestational age and birth weight. Vitamin E deficiency was associated with pre-eclampsia [odds ratio (OR) (95% confidence interval, 95% CI), 1.56 (1.01-2.44); p < 0.01] and gestational diabetes [4.01 (1.05-17.0); p < 0.01]. Vitamin D deficiency was associated with twin pregnancy [OR (95% CI), 2.66 (1.33-5.35); p < 0.01] and pre-eclampsia [2.89 (1.36-6.40); p < 0.01]. CONCLUSION Vitamin A, E, and D deficiencies are very common in Tunisian VLBW neonates and are associated with pre-eclampsia. Improved nutritional and health support for pregnant women and high dose vitamins A, E, and D supplementation in VLBW neonates are strongly required in Tunisia.
Collapse
Affiliation(s)
- Samira Fares
- UR05/08-08, Department of Biochemistry, Rabta Hospital and Faculty of Medicine of Tunis, El Manar University, 1007 Tunis, Tunisia
| | - Mohamed Marouane Sethom
- UR05/08-08, Department of Biochemistry, Rabta Hospital and Faculty of Medicine of Tunis, El Manar University, 1007 Tunis, Tunisia
| | - Chahnez Khouaja-Mokrani
- Service of Neonatology, Center of Maternity and Neonatology and Faculty of Medicine of Tunis, El Manar University, 1007 Tunis, Tunisia
| | - Sami Jabnoun
- Service of Neonatology, Center of Maternity and Neonatology and Faculty of Medicine of Tunis, El Manar University, 1007 Tunis, Tunisia
| | - Moncef Feki
- UR05/08-08, Department of Biochemistry, Rabta Hospital and Faculty of Medicine of Tunis, El Manar University, 1007 Tunis, Tunisia.
| | - Naziha Kaabachi
- UR05/08-08, Department of Biochemistry, Rabta Hospital and Faculty of Medicine of Tunis, El Manar University, 1007 Tunis, Tunisia
| |
Collapse
|
12
|
Thiede BR, Mann ZF, Chang W, Ku YC, Son YK, Lovett M, Kelley MW, Corwin JT. Retinoic acid signalling regulates the development of tonotopically patterned hair cells in the chicken cochlea. Nat Commun 2014; 5:3840. [PMID: 24845860 DOI: 10.1038/ncomms4840] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 04/08/2014] [Indexed: 01/07/2023] Open
Abstract
Precise frequency discrimination is a hallmark of auditory function in birds and mammals and is required for distinguishing similar sounding words, like 'bat,' 'cat' and 'hat.' In the cochlea, tuning and spectral separation result from longitudinal differences in basilar membrane stiffness and numerous individual gradations in sensory hair cell phenotypes, but it is unknown what patterns the phenotypes. Here we used RNA-seq to compare transcriptomes from proximal, middle and distal regions of the embryonic chicken cochlea, and found opposing longitudinal gradients of expression for retinoic acid (RA)-synthesizing and degrading enzymes. In vitro experiments showed that RA is necessary and sufficient to induce the development of distal-like hair cell phenotypes and promotes expression of the actin-crosslinking proteins, Espin and Fscn2. These and other findings highlight a role for RA signalling in patterning the development of a longitudinal gradient of frequency-tuned hair cell phenotypes in the cochlea.
Collapse
Affiliation(s)
- Benjamin R Thiede
- Department of Neuroscience, University of Virginia School of Medicine, 409 Lane Road, Charlottesville, Virginia 22908, USA
| | - Zoë F Mann
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 35A Convent Drive, Bethesda, Maryland 20892-3729, USA
| | - Weise Chang
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 35A Convent Drive, Bethesda, Maryland 20892-3729, USA
| | - Yuan-Chieh Ku
- Division of Human Genetics, Department of Genetics, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Yena K Son
- Department of Neuroscience, University of Virginia School of Medicine, 409 Lane Road, Charlottesville, Virginia 22908, USA
| | - Michael Lovett
- Division of Human Genetics, Department of Genetics, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 35A Convent Drive, Bethesda, Maryland 20892-3729, USA
| | - Jeffrey T Corwin
- 1] Department of Neuroscience, University of Virginia School of Medicine, 409 Lane Road, Charlottesville, Virginia 22908, USA [2] Department of Cell Biology, University of Virginia School of Medicine, 409 Lane Road, Charlottesville, Virginia 22908, USA
| |
Collapse
|
13
|
Emmett SD, West KP. Gestational vitamin A deficiency: a novel cause of sensorineural hearing loss in the developing world? Med Hypotheses 2014; 82:6-10. [PMID: 24120698 PMCID: PMC4391953 DOI: 10.1016/j.mehy.2013.09.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/11/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022]
Abstract
Hearing loss is a substantial public health problem with profound social and economic consequences in the developing world. The World Health Organization (WHO) estimates that there are 360 million people living with disabling hearing loss globally, and 80% of these individuals are from low- and middle-income countries. The epidemiology of hearing impairment remains poorly defined in most impoverished societies. Middle ear infections in childhood are a key determinant; however, congenital anomalies may also comprise an important etiology and may arise from gestational malnutrition. While evidence exists that preventable vitamin A deficiency exacerbates the severity of ear infections and, consequently, hearing loss, antenatal vitamin A deficiency during sensitive periods of fetal development may represent an etiologically distinct and virtually unexplored causal pathway. Evidence from multiple animal systems clearly shows that fetal inner ear development requires adequate vitamin A nutriture to proceed normally. Inner ear malformations occur in experimentally imposed maternal vitamin A deficiency in multiple species in a dose-response manner. These anomalies are likely due to the loss of retinoic acid-dependent regulation of both hindbrain development and otic morphogenic processes. Based on in vivo evidence in experimental animals, we hypothesize that preventable gestational vitamin A deficiency, especially during early stages of fetal development, may predispose offspring to inner ear malformations and sensorineural hearing loss. As vitamin A deficiency affects an estimated 20 million pregnant women globally, we hypothesize that, in undernourished settings, routine provision of supplemental vitamin A at the recommended allowance throughout pregnancy may promote normal inner ear development and reduce risk of an as yet unknown fraction of sensorineural hearing loss. If our hypothesis proves correct, gestational vitamin A deficiency would represent a potentially preventable etiology of sensorineural hearing loss of substantial public health significance.
Collapse
Affiliation(s)
- Susan D Emmett
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, 601 N Caroline Street, Baltimore, MD 21287, USA; Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, W2041, Baltimore, MD 21205, USA.
| | - Keith P West
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, W2041, Baltimore, MD 21205, USA
| |
Collapse
|
14
|
Uribe PM, Asuncion JD, Matsui JI. Ethanol affects the development of sensory hair cells in larval zebrafish (Danio rerio). PLoS One 2013; 8:e83039. [PMID: 24324841 PMCID: PMC3855788 DOI: 10.1371/journal.pone.0083039] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/30/2013] [Indexed: 12/20/2022] Open
Abstract
Children born to mothers with substantial alcohol consumption during pregnancy can present a number of morphological, cognitive, and sensory abnormalities, including hearing deficits, collectively known as fetal alcohol syndrome (FAS). The goal of this study was to determine if the zebrafish lateral line could be used to study sensory hair cell abnormalities caused by exposure to ethanol during embryogenesis. Some lateral line sensory hair cells are present at 2 days post-fertilization (dpf) and are functional by 5 dpf. Zebrafish embryos were raised in fish water supplemented with varying concentrations of ethanol (0.75%-1.75% by volume) from 2 dpf through 5 dpf. Ethanol treatment during development resulted in many physical abnormalities characteristic of FAS in humans. Also, the number of sensory hair cells decreased as the concentration of ethanol increased in a dose-dependent manner. The dye FM 1-43FX was used to detect the presence of functional mechanotransduction channels. The percentage of FM 1-43-labeled hair cells decreased as the concentration of ethanol increased. Methanol treatment did not affect the development of hair cells. The cell cycle markers proliferating cell nuclear antigen (PCNA) and bromodeoxyuridine (BrdU) demonstrated that ethanol reduced the number of sensory hair cells, as a consequence of decreased cellular proliferation. There was also a significant increase in the rate of apoptosis, as determined by TUNEL-labeling, in neuromasts following ethanol treatment during larval development. Therefore, zebrafish are a useful animal model to study the effects of hair cell developmental disorders associated with FAS.
Collapse
Affiliation(s)
- Phillip M. Uribe
- Department of Neuroscience, Pomona College, Claremont, California, United States of America
| | - James D. Asuncion
- Department of Neuroscience, Pomona College, Claremont, California, United States of America
| | - Jonathan I. Matsui
- Department of Neuroscience, Pomona College, Claremont, California, United States of America
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Otolaryngology and Communication Enhancement, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
15
|
Postnatal development, maturation and aging in the mouse cochlea and their effects on hair cell regeneration. Hear Res 2012; 297:68-83. [PMID: 23164734 DOI: 10.1016/j.heares.2012.11.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/22/2012] [Accepted: 11/07/2012] [Indexed: 12/23/2022]
Abstract
The organ of Corti in the mammalian inner ear is comprised of mechanosensory hair cells (HCs) and nonsensory supporting cells (SCs), both of which are believed to be terminally post-mitotic beyond late embryonic ages. Consequently, regeneration of HCs and SCs does not occur naturally in the adult mammalian cochlea, though recent evidence suggests that these cells may not be completely or irreversibly quiescent at earlier postnatal ages. Furthermore, regenerative processes can be induced by genetic and pharmacological manipulations, but, more and more reports suggest that regenerative potential declines as the organ of Corti continues to age. In numerous mammalian systems, such effects of aging on regenerative potential are well established. However, in the cochlea, the problem of regeneration has not been traditionally viewed as one of aging. This is an important consideration as current models are unable to elicit widespread regeneration or full recovery of function at adult ages yet regenerative therapies will need to be developed specifically for adult populations. Still, the advent of gene targeting and other genetic manipulations has established mice as critically important models for the study of cochlear development and HC regeneration and suggests that auditory HC regeneration in adult mammals may indeed be possible. Thus, this review will focus on the pursuit of regeneration in the postnatal and adult mouse cochlea and highlight processes that occur during postnatal development, maturation, and aging that could contribute to an age-related decline in regenerative potential. Second, we will draw upon the wealth of knowledge pertaining to age related senescence in tissues outside of the ear to synthesize new insights and potentially guide future research aimed at promoting HC regeneration in the adult cochlea.
Collapse
|
16
|
Rudnicki A, Avraham KB. microRNAs: the art of silencing in the ear. EMBO Mol Med 2012; 4:849-59. [PMID: 22745034 PMCID: PMC3491818 DOI: 10.1002/emmm.201100922] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 05/15/2012] [Accepted: 05/24/2012] [Indexed: 11/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression through the RNA interference (RNAi) pathway and by inhibition of mRNA translation. miRNAs first made their appearance in the auditory and vestibular systems in 2005, with the discovery of a triad of hair cell-specific miRNAs later found to be involved in both human and mouse deafness. Since then, miRNAs have been implicated in other medical conditions related to these systems, such as cholesteatomas, vestibular schwannomas and otitis media. Due to the limitations in studying miRNAs and their targets derived from human inner ears, animal models are vital in this field of research. Therefore their role in inner ear development and function has been demonstrated by studies in zebrafish and mice. Transcriptomic and proteomic approaches have been undertaken to identify miRNAs and their targets. Finally, it has been suggested that miRNAs may be used in the future in regeneration of inner ear hair cells and ultimately play a role in therapeutics.
Collapse
Affiliation(s)
- Anya Rudnicki
- Sackler Faculty of Medicine, Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, Israel
| | | |
Collapse
|
17
|
Gross J, Stute K, Fuchs J, Angerstein M, Amarjargal N, Mazurek B. Effects of retinoic acid and butyric acid on the expression of prestin and Gata-3 in organotypic cultures of the organ of corti of newborn rats. Dev Neurobiol 2011; 71:650-61. [PMID: 21344672 DOI: 10.1002/dneu.20881] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prestin is the motor protein of the outer hair cells of the organ of Corti and a key factor in ensuring a high level of sensitivity of mammalian hearing. The factors that influence prestin expression are still largely unknown. We studied the effects of the application of retinoic acid, a ligand of a nuclear receptor, and of butyric acid, an inhibitor of histone deacetylase activity, on the expression of mRNA of prestin and Gata-3 in the organotypic culture of the organ of Corti of newborn rats using RT-PCR. Application of retinoic acid at concentrations of 1-50 μM results in a dose-dependent expression decrease after two days in culture. Treatment with sodium butyrate (0.5-2 mM) elevated the expression of prestin and Gata-3. Statistically significant correlations between Gata-3 and prestin mRNA levels were observed under all conditions. The data indicate that retinoid nuclear transcription factors, GATA-3 and histone acetylation/deacetylation processes may have a regulatory role to play in prestin expression.
Collapse
Affiliation(s)
- Johann Gross
- Department of Otorhinolaryngology, Molecular Biology Research Laboratory, Charité-Universitätsmedizin Berlin, 10117 Berlin, Charitéplatz 1, Germany.
| | | | | | | | | | | |
Collapse
|
18
|
Lilienthal H, Heikkinen P, Andersson PL, van der Ven LTM, Viluksela M. Auditory Effects of Developmental Exposure to Purity-Controlled Polychlorinated Biphenyls (PCB52 and PCB180) in Rats. Toxicol Sci 2011; 122:100-11. [DOI: 10.1093/toxsci/kfr077] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
19
|
Huang M, Sage C, Tang Y, Lee SG, Petrillo M, Hinds PW, Chen ZY. Overlapping and distinct pRb pathways in the mammalian auditory and vestibular organs. Cell Cycle 2011; 10:337-51. [PMID: 21239885 DOI: 10.4161/cc.10.2.14640] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Retinoblastoma gene (Rb1) is required for proper cell cycle exit in the developing mouse inner ear and its deletion in the embryo leads to proliferation of sensory progenitor cells that differentiate into hair cells and supporting cells. In a conditional hair cell Rb1 knockout mouse, Pou4f3-Cre-pRb(-/-), pRb(-/-) utricular hair cells differentiate and survive into adulthood whereas differentiation and survival of pRb(-/-) cochlear hair cells are impaired. To comprehensively survey the pRb pathway in the mammalian inner ear, we performed microarray analysis of (pRb(-/-) cochlea and utricle. The comparative analysis shows that the core pathway shared between pRb(-/-) cochlea and utricle is centered on E2F, the key pathway that mediates pRb function. A majority of differentially expressed genes and enriched pathways are not shared but uniquely associated with pRb(-/-) cochlea or utricle. In pRb(-/-) cochlea, pathways involved in early inner ear development such as Wnt/β-catenin and Notch were enriched, whereas pathways involving in proliferation and survival are enriched in pRb(-/-) utricle. Clustering analysis showed that the pRb(-/-) inner ear has characteristics of a younger control inner ear, an indication of delayed differentiation. We created a transgenic mouse model (ER-Cre-pRb(flox/flox)) in which Rb1 can be acutely deleted postnatally. Acute Rb1 deletion in the adult mouse fails to induce proliferation or cell death in inner ear, strongly indicating that Rb1 loss in these postmitotic tissues can be effectively compensated for, or that pRb-mediated changes in the postmitotic compartment result in events that are functionally irreversible once enacted. This study thus supports the concept that pRb-regulated pathways relevant to hair cell development, encompassing proliferation, differentiation and survival, act predominantly during early development.
Collapse
Affiliation(s)
- Mingqian Huang
- Department of Otology and Laryngology, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Estephane D, Anctil M. Retinoic acid and nitric oxide promote cell proliferation and differentially induce neuronal differentiation in vitro in the cnidarian Renilla koellikeri. Dev Neurobiol 2010; 70:842-52. [DOI: 10.1002/dneu.20824] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Taura A, Taura K, Choung YH, Masuda M, Pak K, Chavez E, Ryan AF. Histone deacetylase inhibition enhances adenoviral vector transduction in inner ear tissue. Neuroscience 2010; 166:1185-93. [PMID: 20060033 DOI: 10.1016/j.neuroscience.2009.12.064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 12/16/2009] [Accepted: 12/23/2009] [Indexed: 10/20/2022]
Abstract
Adenovirus vectors (AdVs) are efficient tools for gene therapy in many tissues. Several studies have demonstrated successful transgene transduction with AdVs in the inner ear of rodents [Kawamoto K, Ishimoto SI, Minoda R, Brough DE, Raphael Y (2003) J Neurosci 23:4395-4400]. However, toxicity of AdVs [Morral N, O'Neal WK, Rice K, Leland MM, Piedra PA, Aguilar-Cordova E, Carey KD, Beaudet AL, Langston C (2002) Hum Gene Ther 13:143-154.] or lack of tropism to important cell types such as hair cells [Shou J, Zheng JL, Gao WQ (2003) Mol Cell Neurosci 23:169-179] appears to limit their experimental and potential clinical utility. Histone deacetylase inhibitors (HDIs) are known to enhance AdV-mediated transgene expression in various organs [Dion LD, Goldsmith KT, Tang DC, Engler JA, Yoshida M, Garver RI Jr (1997) Virology 231:201-209], but their effects in the inner ear have not been documented. We investigated the ability of one HDI, trichostatin A (TSA), to enhance AdV-mediated transgene expression in inner ear tissue. We cultured neonatal rat macular and cochlear explants, and transduced them with an AdV encoding green fluorescent protein (Ad-GFP) under the control of a constitutive promoter for 24 h. In the absence of TSA, GFP expression was limited, and very few hair cells were transduced. TSA did not enhance transduction when applied at the onset of Ad-GFP transduction. However, administration of TSA during or just after Ad-GFP application increased GFP expression in supporting cells approximately fourfold. Moreover, vestibular hair cell transduction was enhanced approximately sixfold, and that of inner hair cells by more than 17-fold. These results suggest that TSA increases AdV-mediated transgene expression in the inner ear, including the successful transduction of hair cells. HDIs, some of which are currently under clinical trials (Sandor et al., 2002), could be useful tools in overcoming current limitations of gene therapy in the inner ear using Ad-GFP.
Collapse
Affiliation(s)
- A Taura
- Division of Otolaryngology, Departments of Surgery, UCSD School of Medicine, San Diego, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Dollé P. Developmental expression of retinoic acid receptors (RARs). NUCLEAR RECEPTOR SIGNALING 2009; 7:e006. [PMID: 19471585 PMCID: PMC2686085 DOI: 10.1621/nrs.07006] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Accepted: 04/21/2009] [Indexed: 12/11/2022]
Abstract
Here, I review the developmental expression features of genes encoding the retinoic acid receptors (RARs) and the 'retinoid X' or rexinoid receptors (RXRs). The first detailed expression studies were performed in the mouse over two decades ago, following the cloning of the murine Rar genes. These studies revealed complex expression features at all stages of post-implantation development, one receptor gene (Rara) showing widespread expression, the two others (Rarb and Rarg) with highly regionalized and/or cell type-specific expression in both neural and non-neural tissues. Rxr genes also have either widespread (Rxra, Rxrb), or highly-restricted (Rxrg) expression patterns. Studies performed in zebrafish and Xenopus demonstrated expression of Rar and Rxr genes (both maternal and zygotic), at early pre-gastrulation stages. The eventual characterization of specific enzymes involved in the synthesis of retinoic acid (retinol/retinaldehyde dehydrogenases), or the triggering of its catabolism (CYP26 cytochrome P450s), all of them showing differential expression patterns, led to a clearer understanding of the phenomenons regulated by retinoic acid signaling during development. Functional studies involving targeted gene disruptions in the mouse, and additional approaches such as dominant negative receptor expression in other models, have pinpointed the specific, versus partly redundant, roles of the RARs and RXRs in many developing organ systems. These pleiotropic roles are summarized hereafter in relationship to the receptors' expression patterns.
Collapse
Affiliation(s)
- Pascal Dollé
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), France.
| |
Collapse
|
23
|
Effects of the brominated flame retardant hexabromocyclododecane (HBCD) on dopamine-dependent behavior and brainstem auditory evoked potentials in a one-generation reproduction study in Wistar rats. Toxicol Lett 2009; 185:63-72. [DOI: 10.1016/j.toxlet.2008.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 11/28/2008] [Accepted: 12/01/2008] [Indexed: 01/23/2023]
|
24
|
Löwenheim H, Waldhaus J, Hirt B, Sandke S, Müller M. [Regenerative medicine in the treatment of sensorineural hearing loss]. HNO 2008; 56:288-300. [PMID: 18288464 DOI: 10.1007/s00106-008-1689-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Regenerative medicine offers the prospect of causal treatment of sensorineural hearing loss. In humans, the loss of sensory hair cells is irreversible and results in chronic hearing loss. Other vertebrates, particularly birds, have the capability to spontaneously regenerate lost sensory hair cells and restore hearing. In the bird model, regeneration of hair cells is based on the proliferation of supporting cells. In mammals, supporting cells have lost their proliferative capacity and are terminally differentiated. To gain an understanding about regeneration of hair cells in mammals, cell division of supporting cells has to be controlled. Gene disruption of the cell cycle inhibitor p27(Kip1) allows supporting cell proliferation in the organ of Corti in vivo. Furthermore, in vitro studies indicate that newly generated cells may differentiate into hair cells after p27(Kip1) disruption. Other current methods to induce hair cell regeneration include the gene transfer of Math1 and transplantation of stem cells to the inner ear.
Collapse
Affiliation(s)
- H Löwenheim
- Klinik für Hals-Nasen-Ohren-Heilkunde, Universitätsklinikum Tübingen, Elfriede-Aulhorn-Strasse 5, 72076 Tübingen, Deutschland.
| | | | | | | | | |
Collapse
|
25
|
Helyer R, Cacciabue-Rivolta D, Davies D, Rivolta MN, Kros CJ, Holley MC. A model for mammalian cochlear hair cell differentiation in vitro: effects of retinoic acid on cytoskeletal proteins and potassium conductances. Eur J Neurosci 2007; 25:957-73. [PMID: 17331193 DOI: 10.1111/j.1460-9568.2007.05338.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We have established a model for the in-vitro differentiation of mouse cochlear hair cells and have used it to explore the influence of retinoic acid on proliferation, cytoskeletal proteins and voltage-gated potassium conductances. The model is based on the conditionally immortal cell line University of Sheffield/ventral otocyst-epithelial cell line clone 36 (US/VOT-E36), derived from ventral otic epithelial cells of the mouse at embryonic day 10.5 and transfected with a reporter for myosin VIIa. Retinoic acid did not increase cell proliferation but led to up-regulation of myosin VIIa and formation of prominent actin rings that gave rise to numerous large, linear actin bundles. Cells expressing myosin VIIa had larger potassium conductances and did not express the cyclin-dependent kinase inhibitor p27(kip1). US/VOT-E36 endogenously expressed the voltage-gated potassium channel alpha-subunits Kv1.3 and Kv2.1, which we subsequently identified in embryonic and neonatal hair cells in both auditory and vestibular sensory epithelia in vivo. These subunits could underlie the embryonic and neonatal delayed-rectifiers recorded in nascent hair cells in vivo. Kv2.1 was particularly prominent on the basolateral membrane of cochlear inner hair cells. Kv1.3 was distributed throughout all hair cells but tended to be localized to the cuticular plates. US/VOT-E36 recapitulates a coherent pattern of cell differentiation under the influence of retinoic acid and will provide a convenient model for screening the effects of other extrinsic factors on the differentiation of cochlear epithelial cell types in vitro.
Collapse
Affiliation(s)
- R Helyer
- Department of Biomedical Science, Addison Building, Western Bank, Sheffield, UK
| | | | | | | | | | | |
Collapse
|
26
|
Winter H, Braig C, Zimmermann U, Geisler HS, Fränzer JT, Weber T, Ley M, Engel J, Knirsch M, Bauer K, Christ S, Walsh EJ, McGee J, Köpschall I, Rohbock K, Knipper M. Thyroid hormone receptors TRalpha1 and TRbeta differentially regulate gene expression of Kcnq4 and prestin during final differentiation of outer hair cells. J Cell Sci 2006; 119:2975-84. [PMID: 16803873 DOI: 10.1242/jcs.03013] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thyroid hormone (TH or T3) and TH-receptor beta (TRbeta) have been reported to be relevant for cochlear development and hearing function. Mutations in the TRbeta gene result in deafness associated with resistance to TH syndrome. The effect of TRalpha1 on neither hearing function nor cochlear T3 target genes has been described to date. It is also uncertain whether TRalpha1 and TRbeta can act simultaneously on different target genes within a single cell. We focused on two concomitantly expressed outer hair cell genes, the potassium channel Kcnq4 and the motor protein prestin Slc26a5. In outer hair cells, TH enhanced the expression of the prestin gene through TRbeta. Simultaneously Kcnq4 expression was activated in the same cells by derepression of TRalpha1 aporeceptors mediated by an identified THresponse element, which modulates KCNQ4 promoter activity. We show that T3 target genes can differ in their sensitivity to TH receptors having the ligand either bound (holoreceptors) or not bound (aporeceptors) within single cells, and suggest a role for TRalpha1 in final cell differentiation.
Collapse
Affiliation(s)
- Harald Winter
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Laboratory of Molecular Neurobiology, University of Tübingen, Elfriede-Aulhorn-Strasse 5, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sage C, Huang M, Vollrath MA, Brown MC, Hinds PW, Corey DP, Vetter DE, Chen ZY. Essential role of retinoblastoma protein in mammalian hair cell development and hearing. Proc Natl Acad Sci U S A 2006; 103:7345-50. [PMID: 16648263 PMCID: PMC1450112 DOI: 10.1073/pnas.0510631103] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The retinoblastoma protein pRb is required for cell-cycle exit of embryonic mammalian hair cells but not for their early differentiation. However, its role in postnatal hair cells is unknown. To study the function of pRb in mature animals, we created a new conditional mouse model, with the Rb gene deleted primarily in the inner ear. Progeny survive up to 6 months. During early postnatal development, pRb(-/-) hair cells continue to divide and can transduce mechanical stimuli. However, adult pRb(-/-) mice exhibit profound hearing loss due to progressive degeneration of the organ of Corti. We show that pRb is required for the full maturation of cochlear outer hair cells, likely in a gene-specific manner, and is also essential for their survival. In addition, lack of pRb results in cell division in postnatal auditory supporting cells. In contrast, many pRb(-/-) vestibular hair cells survive and continue to divide in adult mice. Significantly, adult pRb(-/-) vestibular hair cells are functional, and pRb(-/-) mice maintain partial vestibular function. Therefore, the functional adult vestibular pRb(-/-) hair cells, derived from proliferation of postnatal hair cells, are largely integrated into vestibular pathways. This study reveals essential yet distinct roles of pRb in cochlear and vestibular hair cell maturation, function, and survival and suggests that transient block of pRb function in mature hair cells may lead to propagation of functional hair cells.
Collapse
Affiliation(s)
- Cyrille Sage
- *Neurology Service, Center for Nervous System Repair, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Mingqian Huang
- *Neurology Service, Center for Nervous System Repair, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Melissa A. Vollrath
- Howard Hughes Medical Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - M. Christian Brown
- Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary and Harvard Medical School, Boston, MA 02114
| | - Philip W. Hinds
- Department of Radiation Oncology, Molecular Oncology Research Institute, Tufts–New England Medical Center, Boston, MA 02111; and
| | - David P. Corey
- Howard Hughes Medical Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Douglas E. Vetter
- Departments of Neuroscience and Biomedical Engineering, Tufts University School of Medicine, Boston, MA 02111
| | - Zheng-Yi Chen
- *Neurology Service, Center for Nervous System Repair, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
28
|
Abstract
The inner ear originates from an embryonic ectodermal placode and rapidly develops into a three-dimensional structure (the otocyst) through complex molecular and cellular interactions. Many genes and their products are involved in inner ear induction, organogenesis, and cell differentiation. Retinoic acid (RA) is an endogenous signaling molecule that may play a role during different phases of inner ear development, as shown from pathological observations. To gain insight into the function of RA during inner ear development, we have investigated the spatio-temporal expression patterns of major components of RA signaling pathway, including cellular retinoic acid binding proteins (CRABPs), cellular retinoid binding proteins (CRBPs), retinaldehyde dehydrogenases (RALDHs), catabolic enzymes (CYP26s), and nuclear receptors (RARs). Although the CrbpI, CrabpI, and -II genes are specifically expressed in the inner ear throughout development, loss-of-function studies have revealed that these proteins are dispensable for inner development and function. Several Raldh and Cyp26 gene transcripts are expressed at embryological day (E) 9.0-9.5 in the otocyst and show mainly complementary distributions in the otic epithelium and mesenchyme during following stages. From Western blot, RT-PCR, and in situ hybridization analysis, there is a low expression of Raldhs in the early otocyst at E9, while Cyp26s are strongly expressed. During the following days, there is an up-regulation of Raldhs and a down-regulation for Cyp26s. Specific RA receptor (Rar and Rxr) genes are expressed in the otocyst and during further development of the inner ear. At the otocyst stage, most of the components of the retinoid pathway are present, suggesting that the embryonic inner ear might act as an autocrine system, which is able to synthesize and metabolize RA necessary for its development. We propose a model in which two RA-dependent pathways may control inner ear ontogenesis: one indirect with RA from somitic mesoderm acting to regulate gene expression within the hindbrain neuroepithelium, and another with RA acting directly on the otocyst. Current evidence suggests that RA may regulate several genes involved in mesenchyme-epithelial interactions, thereby controlling inner ear morphogenesis. Our investigations suggest that RA signaling is a critical component not only of embryonic development, but also of postnatal maintenance of the inner ear.
Collapse
Affiliation(s)
- Raymond Romand
- Institut Clinique de la Souris and Institut de Génétique et de Biologie Moléculaire et cellulaire, B.P. 10142, 67404 Illkirch Cedex, France.
| | | | | |
Collapse
|
29
|
Stone IM, Lurie DI, Kelley MW, Poulsen DJ. Adeno-associated virus-mediated gene transfer to hair cells and support cells of the murine cochlea. Mol Ther 2005; 11:843-8. [PMID: 15922954 DOI: 10.1016/j.ymthe.2005.02.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 02/03/2005] [Accepted: 02/03/2005] [Indexed: 11/19/2022] Open
Abstract
More than 28 million Americans suffer from various forms of hearing loss. The lack of effective treatments for many forms of hearing disorders has prompted interest in the potential application of gene delivery techniques to treat both inherited and pathological hearing disorders. However, to develop a gene therapy strategy that will successfully treat hearing disorders, appropriate vectors that are capable of transducing cochlear hair cells and support cells must be identified. In the present study, we examined the efficiency with which AAV vectors (serotypes 1, 2, and 5) transduce hair cells and support cells in cochlear explants from P0 and E13 mice. We further examined the ability of the CBA and GFAP promoters to drive expression of a GFP marker gene in hair cells and support cells. Robust GFP expression was observed in hair cells and support cells following transduction of primary murine cochlear explants with AAV serotypes 1 and 2, but not serotype 5. The CBA promoter predominantly drove GFP expression in hair cells. In contrast, strong expression from the GFAP promoter was observed primarily in support cells. Thus, using AAV vectors and specific promoters, cell-type-specific expression of transgenes can be established within the cochlea.
Collapse
Affiliation(s)
- Ida M Stone
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, No. 1552, Missoula, MT 59812, USA
| | | | | | | |
Collapse
|
30
|
Abstract
The rate of identification of genes for hearing has clearly outpaced the rate of determination of the functions of these genes' products. The use of transgenic and knock-out mouse models is a powerful approach to the elucidation of gene function in the ear. A large number of gene-targeted mice with auditory defects have recently been created and characterized, and nine independent mouse lines in which Cre recombinase activity begins to be expressed during early embryonic development of the ear or is specifically expressed in hair cells during postnatal development will be useful for ear-specific gene manipulation when combined with mouse lines that have loxP sites flanking the genes of interest. Existing gene-trapped embryonic stem (ES) cells and existing targeting constructs are readily available; new targeting constructs can easily be created by modifying bacterial artificial chromosomes and using them to directly transfect and screen ES cells; and N-ethyl-N-nitrosourea mutagenesis of ES cells can create point mutations in specific genes. To minimize variation in hearing phenotypes and avoid undesired hearing defects, mutant mice in the common gene-targeting background strains (129 and C57BL/6) should be transferred into congenic CBA/CaJ, a strain with "gold standard" normal hearing. Valuable mutant strains can be maintained, distributed, and cryopreserved in one of four NIH-sponsored Mutant Mouse Regional Resource Centers. Targeting hearing genes in mice will provide unprecedented opportunities for collaboration and new directions in the hearing research community.
Collapse
Affiliation(s)
- Jiangang Gao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105-2794, USA
| | | | | |
Collapse
|
31
|
Abstract
The sensory epithelium of the mammalian cochlea is composed of a regular mosaic of sensory hair cells and nonsensory supporting cells. During development, differentiation occurs in a gradient that progresses along the axis of the cochlea from base to apex. To begin to identify some of the factors that regulate this developmental process, the potential roles of planar and vertical signals were examined during early stages of cochlear development. We demonstrate roles for both underlying mesenchymal cells and adjacent epithelial cells in the differentiation and patterning of the sensory epithelium, and in particular in the development of mechanosensory hair cells. As development proceeds, the requirements for both planar and vertical signals decrease, and development of the sensory epithelium becomes essentially independent from these cues. Finally, we demonstrate that the temporal gradient of cellular differentiation is not dependent on planar signals within the developing sensory epithelium.
Collapse
|
32
|
Löwenheim H. Regenerative Medicine for Diseases of the Head and Neck: Principles ofIn vivoRegeneration. DNA Cell Biol 2003; 22:571-92. [PMID: 14577910 DOI: 10.1089/104454903322405464] [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: 11/13/2022] Open
Abstract
The application of endogenous regeneration in regenerative medicine is based on the concept of inducing regeneration of damaged or lost tissues from residual tissues in situ. Therefore, endogenous regeneration is also termed in vivo regeneration as opposed to mechanisms of ex vivo regeneration which are applied, for example, in the field of tissue engineering. The basic science foundation for mechanisms of endogenous regeneration is provided by the field of regenerative biology. The ambitious vision for the application of endogenous regeneration in regenerative medicine is stimulated by investigations in the model organisms of regenerative biology, most notably hydra, planarians and urodeles. These model organisms demonstrate remarkable regenerative capabilities, which appear to be conserved over large phylogenetical stretches with convincing evidence for a homologue origin of an endogenous regenerative capability. Although the elucidation of the molecular and cellular mechanisms of these endogenous regenerative phenomena is still in its beginning, there are indications that these processes have potential to become useful for human benefit. Such indications also exist for particular applications in diseases of the head and neck region. As such epimorphic regeneration without blastema formation may be relevant to regeneration of sensorineural epithelia of the inner ear or the olphactory epithelium. Complex tissue lesions of the head and neck as they occur after trauma or tumor resections may be approached on the basis of relevant mechanisms in epimorphic regeneration with blastema formation.
Collapse
Affiliation(s)
- H Löwenheim
- Department of Otolaryngology-Head & Neck Surgery, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
33
|
Affiliation(s)
- Raymond Romand
- Institut Clinique de la Souris, 67404 Illkirch Cedex, France
| |
Collapse
|
34
|
Romand R, Hashino E, Dollé P, Vonesch JL, Chambon P, Ghyselinck NB. The retinoic acid receptors RARalpha and RARgamma are required for inner ear development. Mech Dev 2002; 119:213-23. [PMID: 12464434 DOI: 10.1016/s0925-4773(02)00385-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To define the signal transduction pathway of retinoic acid during inner ear development, we analyzed the expression patterns of transcripts encoding the three retinoic acid receptors (RARalpha, beta, and gamma) and related them to phenotypes resulting from single or compound inactivation of these nuclear receptors. The expression of all three RARs was observed in the developing mouse otocyst as early as embryonic day 10.5 (E10.5)-E12.5 and continued into adulthood. Expression domains of the three RAR receptors, however, were largely non-overlapping: RARalpha was predominantly expressed in the developing sensory epithelium, RARbeta in inner ear mesenchymal tissues and RARgamma in the differentiating otic capsule. In the adult, RARalpha and RARgamma transcripts were found in the organ of Corti and the spiral ganglion, whereas RARbeta transcripts were localized in mesenchyme-derived tissues. RARalpha, beta, and gamma null mutant mice, as well as RARalpha/RARbeta and RARbeta/RARgamma combined null fetuses, did not present any noticeable morphological abnormalities in the inner ear. In contrast, RARalpha/RARgamma null mutants displayed a severe hypoplasia of the otocyst that was already visible at E10.5 without any visible endolymphatic duct. The hypoplastic otocyst in RARalpha/RARgamma null mutants was characterized by impaired chondrocyte differentiation and neural development. After the second week of gestation, these mutant fetuses lacked all of the semi-circular canals and the endolymphatic duct and displayed strong anomalies in the inner ear structures. The morphological deficits were generally more severe in the cochlear portion than in the vestibular portion of the inner ear. Altogether, these results demonstrate that RARalpha and RARgamma play an essential role in the initial differentiation of otic placode derivatives, whereas RARbeta plays a minimal role in this process.
Collapse
Affiliation(s)
- Raymond Romand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université Louis Pasteur, Collège de France, B.P. 10142, 67404 Illkirch Cedex, France.
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
One of the most striking aspects of the cellular pattern within the sensory epithelium of the mammalian cochlea is the presence of two rows of pillar cells in the region between the single row of inner hair cells and the first row of outer hair cells. The factors that regulate pillar cell development have not been determined; however, previous results suggested a key role for fibroblast growth factor receptor 3 (FGFR3). To examine the specific effects of FGFR3 on pillar cell development, we inhibited receptor activation in embryonic cochlear explant cultures. Results indicated that differentiation of pillar cells is dependent on continuous activation of FGFR3. Moreover, transient inhibition of FGFR3 did not inhibit the pillar cell fate permanently, because reactivation of FGFR3 resulted in the resumption of pillar cell differentiation. The effects of increased FGFR3 activation were determined by exposing cochlear explants to FGF2, a strong ligand for several FGF receptors. Treatment with FGF2 led to a significant increase in the number of pillar cells and to a small increase in the number of inner hair cells. These effects were not dependent on cellular proliferation, suggesting that additional pillar cells and inner hair cells were a result of increased recruitment into the prosensory domain. These results indicate that FGF signaling plays a critical role in the commitment and differentiation of pillar cells. Moreover, the position of the pillar cells appears to be determined by the activation of FGFR3 in a subset of the progenitor cells that initially express this receptor.
Collapse
|
36
|
Maden M. Role and distribution of retinoic acid during CNS development. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 209:1-77. [PMID: 11580199 DOI: 10.1016/s0074-7696(01)09010-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Retinoic acid (RA), the biologically active derivative of vitamin A, induces a variety of embryonal carcinoma and neuroblastoma cell lines to differentiate into neurons. The molecular events underlying this process are reviewed with a view to determining whether these data can lead to a better understanding of the normal process of neuronal differentiation during development. Several transcription factors, intracellular signaling molecules, cytoplasmic proteins, and extracellular molecules are shown to be necessary and sufficient for RA-induced differentiation. The evidence that RA is an endogenous component of the developing central nervous system (CNS) is then reviewed, data which include high-pressure liquid chromotography (HPLC) measurements, reporter systems and the distribution of the enzymes that synthesize RA. The latter is particularly relevant to whether RA signals in a paracrine fashion on adjacent tissues or whether it acts in an autocrine manner on cells that synthesize it. It seems that a paracrine system may operate to begin early patterning events within the developing CNS from adjacent somites and later within the CNS itself to induce subsets of neurons. The distribution of retinoid-binding proteins, retinoid receptors, and RA-synthesizing enzymes is described as well as the effects of knockouts of these genes. Finally, the effects of a deficiency and an excess of RA on the developing CNS are described from the point of view of patterning the CNS, where it seems that the hindbrain is the most susceptible part of the CNS to altered levels of RA or RA receptors and also from the point of view of neuronal differentiation where, as in the case of embryonal carcinoma (EC) cells, RA promotes neuronal differentiation. The crucial roles played by certain genes, particularly the Hox genes in RA-induced patterning processes, are also emphasized.
Collapse
Affiliation(s)
- M Maden
- MRC Centre for Developmental Neurobiology, King's College London, United Kingdom
| |
Collapse
|
37
|
Bryant J, Goodyear RJ, Richardson GP. Sensory organ development in the inner ear: molecular and cellular mechanisms. Br Med Bull 2002; 63:39-57. [PMID: 12324383 DOI: 10.1093/bmb/63.1.39] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The molecular mechanisms underlying the specification of sensory organs in the inner ear and the development of hair and supporting cells within these organs are described. The different organs are all derived from a common pro-sensory region, and may be specified by their proximity to the boundaries between compartments - broad domains within the otocyst defined by the asymmetric expression patterns of transcription factors. Activation of Notch may specify the pro-sensory region, and lateral inhibition mediated by Notch signalling influences whether cells of common lineage in a sensory patch differentiate as either hair cells or supporting cells. The transcription factors Math1 and Brn3.1 are required for hair cell differentiation, and supporting cells express negative regulators of neurogenesis, Hes1 and Hes5. Retinoic acid and thyroid hormone influence early aspects and timing of hair cell differentiation, respectively. Development of the hair cell's mechanosensory hair bundle involves interactions between the cytoskeleton, cell-surface adhesion molecules, receptors and associated extracellular matrix.
Collapse
Affiliation(s)
- Jane Bryant
- School of Biological Sciences, University of Sussex, Brighton, UK
| | | | | |
Collapse
|
38
|
Romand R, Albuisson E, Niederreither K, Fraulob V, Chambon P, Dollé P. Specific expression of the retinoic acid-synthesizing enzyme RALDH2 during mouse inner ear development. Mech Dev 2001; 106:185-9. [PMID: 11472854 DOI: 10.1016/s0925-4773(01)00447-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Retinoid binding proteins and nuclear receptors are expressed in the developing mouse inner ear. Here, we report that the retinaldehyde dehydrogenase 2 (Raldh2) gene, whose product is involved in the enzymatic generation of retinoic acid (RA), exhibits a restricted expression pattern during mouse inner ear ontogenesis. The Raldh2 gene is first expressed at embryonic day (E) 10.5 in a V-shaped medio-dorsal region of the otocyst outer epithelium, which evolves as two separate domains upon otocyst morphogenesis. At E14.5, Raldh2 is expressed in two areas of the utricle epithelium and specific regions of the saccule and cochlear mesenchyme. Later, Raldh2 transcripts are restricted to two cochlear areas, the stria vascularis and Reissner membrane. Raldh2 mesenchymal expression did not correlate with migrating neural crest-derived melanoblasts. These restricted expression domains may correspond to specific sites of RA synthesis during inner ear morphogenesis.
Collapse
Affiliation(s)
- R Romand
- Laboratoire de Neurobiologie, Université Blaise Pascal, 63177 Cedex, Aubière, France.
| | | | | | | | | | | |
Collapse
|
39
|
Mothe AJ, Brown IR. Expression of mRNA encoding extracellular matrix glycoproteins SPARC and SC1 is temporally and spatially regulated in the developing cochlea of the rat inner ear. Hear Res 2001; 155:161-74. [PMID: 11335086 DOI: 10.1016/s0378-5955(01)00246-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
SPARC is a multifunctional extracellular matrix (ECM) glycoprotein that shares partial sequence homology with SC1/hevin. These ECM molecules exhibit calcium-binding properties and modulate cellular interactions. This study examines the expression of SC1 and SPARC mRNA in the developing cochlea of the rat inner ear prior to and after the onset of hearing. At all ages examined, SC1 mRNA is highly expressed in neurons of the spiral ganglion. In contrast, SPARC transcripts are not detected in the spiral ganglion but are enriched in the temporal bone and cartilaginous otic capsule surrounding the cochlea. Both SC1 and SPARC mRNA are expressed in connective tissue elements involved in maintaining ionic homeostasis of cochlear fluids. SC1 mRNA is localized to type III fibrocytes of the spiral ligament (slg) and marginal cells of the stria vascularis, while SPARC mRNA is apparent in the spiral limbus and type I fibrocytes of the slg. At postnatal day 10, SPARC mRNA shows a dramatic change in expression. High levels of SPARC transcripts are induced in Deiters cells (dc) of the organ of Corti. Interestingly, this induction of SPARC mRNA correlates with the onset of hearing. This suggests that SPARC may play a role in calcium regulation in dc when functional maturation of the cochlea is attained and rapid changes in calcium levels are required.
Collapse
Affiliation(s)
- A J Mothe
- Department of Zoology, University of Toronto at Scarborough, M1C 1A4, Toronto, ON, Canada
| | | |
Collapse
|
40
|
Abstract
A decade ago it was discovered that mature birds are able to regenerate hair cells, the receptors for auditory perception. This surprising finding generated hope in the field of auditory neuroscience that new hair cells someday may be coaxed to form in another class of warm-blooded vertebrates, mammals. We have made considerable progress toward understanding some cellular and molecular events that lead to hair cell regeneration in birds. This review discusses our current understanding of avian hair cell regeneration, with some comparisons to other vertebrate classes and other regenerative systems.
Collapse
Affiliation(s)
- J S Stone
- Virginia Merrill Bloedel Hearing Research Center, University of Washington School of Medicine, Seattle, WA 98195-7923, USA
| | | |
Collapse
|
41
|
Romand R, Sapin V, Ghyselinck NB, Avan P, Le Calvez S, Dollé P, Chambon P, Mark M. Spatio-temporal distribution of cellular retinoid binding protein gene transcripts in the developing and the adult cochlea. Morphological and functional consequences in CRABP- and CRBPI-null mutant mice. Eur J Neurosci 2000; 12:2793-804. [PMID: 10971621 DOI: 10.1046/j.1460-9568.2000.00165.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The expression patterns of the mouse cellular retinoid binding protein genes were investigated by in situ hybridization analysis in the inner ear from 10.5 days post coïtum (dpc) up to the adult stage. The cellular retinoic acid binding protein II (CRABPII) and cellular retinol binding protein I (CRBPI) were present in a widespread and abundant pattern in cochlear structures during embryogenesis. Expression of the cellular retinoic acid binding protein I (CRABPI) is restricted during development in Kölliker's organ whilst cellular retinol binding protein II (CRBPII) is only visible after birth with a ubiquitous distribution in most regions of the cochlea including nervous components. No CRABP or CRBP transcripts were observed in the auditory receptors. Morphological observations of CRBPI- and CRABPI/CRABPII-null mutant fetus at 18.5 dpc do not show any structural modification at the level of the organ of Corti. Furthermore, electrophysiological tests performed by measuring distorsion-product otoacoustic emissions and auditory brainstem evoked responses did not present significant alteration of the auditory function for the different types of mutants. The expression of retinoid binding proteins in cochlear structures during embryogenesis could suggest important roles for these proteins during ontogenesis and morphogenesis of the inner ear. Despite these observations, morphological and functional data from mutant mice did not present obvious modifications of the cochlear structures and auditory thresholds. It is therefore unlikely that CRABPs and CRBPI are directly involved in development of the cochlea and hair cell differentiation.
Collapse
MESH Headings
- Age Factors
- Animals
- Audiometry, Pure-Tone
- Auditory Threshold/physiology
- Cochlea/cytology
- Cochlea/growth & development
- Cochlea/physiology
- Evoked Potentials, Auditory, Brain Stem/physiology
- Gene Expression Regulation, Developmental
- Hair Cells, Auditory, Inner/chemistry
- Hair Cells, Auditory, Inner/physiology
- Hair Cells, Auditory, Outer/chemistry
- Hair Cells, Auditory, Outer/physiology
- In Situ Hybridization
- Membrane Potentials/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Knockout
- RNA, Messenger/analysis
- Receptors, Retinoic Acid/genetics
- Retinol-Binding Proteins/genetics
- Retinol-Binding Proteins, Cellular
- Transcription, Genetic/physiology
Collapse
Affiliation(s)
- R Romand
- Laboratoire de Neurobiologie, Université Blaise Pascal, 63177 Aubière Cedex, France.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Pickles JO, van Heumen WR. Lateral interactions account for the pattern of the hair cell array in the chick basilar papilla. Hear Res 2000; 145:65-74. [PMID: 10867278 DOI: 10.1016/s0378-5955(00)00073-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
It has been suggested that lateral interactions set up the array of hair cells and supporting cells in the chick basilar papilla. The presence of a hair cell would inhibit adjacent cells from becoming hair cells, and promote the formation of supporting cells. Models of cell specification were tested, starting with a closely packed array of multipotent progenitor cells. Lateral interactions, in which emerging hair cells promoted a supporting cell phenotype in adjacent cells, and in which emerging supporting cells promoted a hair cell phenotype in adjacent cells, produced an array of cells similar to that observed experimentally in the distal and central parts of the basilar papilla. In these areas, the ratio of supporting cells to hair cells is very close to 2:1, each hair cell on average being surrounded by six supporting cells, and each supporting cell being surrounded by three hair cells and three supporting cells. Identical patterns of hair and supporting cells could be produced by models in which either of the lateral inhibitory factors was replaced by a diffusive factor, i.e. a factor which acts on all cells in the model irrespective of position. The agreement of the model with observed cell ratios supports the view that the fate of both hair cells and supporting cells in the chick basilar papilla is a product of cell interactions within the papilla. It is possible that one factor, that provides contact lateral inhibition and promotes the formation of supporting cells, is the Notch/Delta system. It is possible that the other factor is retinoic acid, a diffusive factor that promotes the formation of hair cells.
Collapse
Affiliation(s)
- J O Pickles
- Vision, Touch and Hearing Research Centre, Department of Physiology and Pharmacology, University of Queensland, 4072, Brisbane, Qld., Australia.
| | | |
Collapse
|