Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38804528

ABSTRACT

The planar polarized organization of hair cells in the vestibular maculae is unique because these sensory organs contain two groups of cells with oppositely oriented stereociliary bundles that meet at a line of polarity reversal (LPR). EMX2 is a transcription factor expressed by one hair cell group that reverses the orientation of their bundles, thereby forming the LPR. We generated Emx2-CreERt2 transgenic mice for genetic lineage tracing and demonstrate Emx2 expression before hair cell specification when the nascent utricle and saccule constitute a continuous prosensory domain. Precursors labeled by Emx2-CreERt2 at this stage give rise to hair cells located along one side of the LPR in the mature utricle or saccule, indicating that this boundary is first established in the prosensory domain. Consistent with this, Emx2-CreERt2 lineage tracing in Dreher mutants, where the utricle and saccule fail to segregate, labels a continuous field of cells along one side of a fused utriculo-saccular-cochlear organ. These observations reveal that LPR positioning is pre-determined in the developing prosensory domain, and that EMX2 expression defines lineages of hair cells with oppositely oriented stereociliary bundles.


Subject(s)
Cell Lineage , Cell Polarity , Ear, Inner , Homeodomain Proteins , Mice, Transgenic , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Cell Lineage/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Cell Polarity/genetics , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology
2.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682291

ABSTRACT

The planar polarized organization of hair cells in the vestibular maculae is unique because these sensory organs contain two groups of cells with oppositely oriented stereociliary bundles that meet at a line of polarity reversal (LPR). EMX2 is a transcription factor expressed by one hair cell group that reverses the orientation of their bundles, thereby forming the LPR. We generated Emx2-CreERt2 transgenic mice for genetic lineage tracing and demonstrate Emx2 expression before hair cell specification when the nascent utricle and saccule constitute a continuous prosensory domain. Precursors labeled by Emx2-CreERt2 at this stage give rise to hair cells located along one side of the LPR in the mature utricle or saccule, indicating that this boundary is first established in the prosensory domain. Consistent with this, Emx2-CreERt2 lineage tracing in Dreher mutants, where the utricle and saccule fail to segregate, labels a continuous field of cells along one side of a fused utriculo-saccular-cochlear organ. These observations reveal that LPR positioning is pre-determined in the developing prosensory domain, and that EMX2 expression defines lineages of hair cells with oppositely oriented stereociliary bundles.


Subject(s)
Cell Lineage , Cell Polarity , Ear, Inner , Homeodomain Proteins , Mice, Transgenic , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Cell Lineage/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Cell Polarity/genetics , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology
3.
Neural Plast ; 2021: 9950533, 2021.
Article in English | MEDLINE | ID: mdl-34122536

ABSTRACT

As part of the inner ear, the vestibular system is responsible for sense of balance, which consists of three semicircular canals, the utricle, and the saccule. Increasing evidence has indicated that the noncanonical Wnt/PCP signaling pathway plays a significant role in the development of the polarity of the inner ear. However, the role of canonical Wnt signaling in the polarity of the vestibule is still not completely clear. In this study, we found that canonical Wnt pathway-related genes are expressed in the early stage of development of the utricle and change dynamically. We conditionally knocked out ß-catenin, a canonical Wnt signaling core protein, and found that the cilia orientation of hair cells was disordered with reduced number of hair cells in the utricle. Moreover, regulating the canonical Wnt pathway (Licl and IWP2) in vitro also affected hair cell polarity and indicated that Axin2 may be important in this process. In conclusion, our results not only confirm that the regulation of canonical Wnt signaling affects the number of hair cells in the utricle but also provide evidence for its role in polarity development.


Subject(s)
Hair Cells, Auditory/physiology , Saccule and Utricle/cytology , Wnt Signaling Pathway/physiology , Animals , Axin Protein/analysis , Cell Polarity , Female , Gene Knockout Techniques , Hair Cells, Auditory/cytology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Saccule and Utricle/embryology , Saccule and Utricle/physiology , beta Catenin/deficiency , beta Catenin/physiology
4.
Sci Rep ; 11(1): 2140, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33495483

ABSTRACT

The Hippo signaling pathway is a key regulator of tissue development and regeneration. Activation of the Hippo pathway leads to nuclear translocation of the YAP1 transcriptional coactivator, resulting in changes in gene expression and cell cycle entry. Recent studies have demonstrated the nuclear translocation of YAP1 during the development of the sensory organs of the inner ear, but the possible role of YAP1 in sensory regeneration of the inner ear is unclear. The present study characterized the cellular localization of YAP1 in the utricles of mice and chicks, both under normal conditions and after HC injury. During neonatal development, YAP1 expression was observed in the cytoplasm of supporting cells, and was transiently expressed in the cytoplasm of some differentiating hair cells. We also observed temporary nuclear translocation of YAP1 in supporting cells of the mouse utricle after short periods in organotypic culture. However, little or no nuclear translocation of YAP1 was observed in the utricles of neonatal or mature mice after ototoxic injury. In contrast, substantial YAP1 nuclear translocation was observed in the chicken utricle after streptomycin treatment in vitro and in vivo. Together, these data suggest that differences in YAP1 signaling may partially account for the differing regenerative abilities of the avian vs. mammalian inner ear.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Saccule and Utricle/embryology , Saccule and Utricle/injuries , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chickens , Diphtheria Toxin/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Protein Transport/drug effects , Saccule and Utricle/metabolism , Saccule and Utricle/pathology , Transcription Factor Brn-3C/metabolism
5.
J Cell Physiol ; 236(7): 5235-5252, 2021 07.
Article in English | MEDLINE | ID: mdl-33368220

ABSTRACT

Intratympanic injection of gentamicin has proven to be an effective therapy for intractable vestibular dysfunction. However, most studies to date have focused on the cochlea, so little is known about the distribution and uptake of gentamicin by the counterpart of the auditory system, specifically vestibular hair cells (HCs). Here, with a combination of in vivo and in vitro approaches, we used a gentamicin-Texas Red (GTTR) conjugate to investigate the mechanisms of gentamicin vestibulotoxicity in the developing mammalian utricular HCs. In vivo, GTTR fluorescence was concentrated in the apical cytoplasm and the cellular membrane of neonatal utricular HCs, but scarce in the nucleus of HCs and supporting cells. Quantitative analysis showed the GTTR uptake by striolar HCs was significantly higher than that in the extrastriola. In addition, the GTTR fluorescence intensity in the striola was increased gradually from 1 to 8 days, peaking at 8-9 days postnatally. In vitro, utricle explants were incubated with GTTR and candidate uptake conduits, including mechanotransduction (MET) channels and endocytosis in the HC, were inhibited separately. GTTR uptake by HCs could be inhibited by quinine, a blocker of MET channels, under both normal and stressed conditions. Meanwhile, endocytic inhibition only reduced GTTR uptake in the CoCl2 hypoxia model. In sum, the maturation of MET channels mediated uptake of GTTR into vestibular HCs. Under stressed conditions, MET channels play a pronounced role, manifested by channel-dependent stress enhanced GTTR permeation, while endocytosis participates in GTTR entry in a more selective manner.


Subject(s)
Biological Transport/physiology , Gentamicins/pharmacology , Gentamicins/pharmacokinetics , Hair Cells, Auditory/metabolism , Saccule and Utricle/embryology , Animals , Endocytosis/drug effects , Female , Gentamicins/chemistry , Male , Membrane Transport Modulators/pharmacology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Quinine/pharmacology , Reactive Oxygen Species/metabolism , Saccule and Utricle/metabolism , Staining and Labeling , Vestibular Diseases/drug therapy , Vestibular Diseases/pathology , Xanthenes/chemistry
6.
Development ; 147(22)2020 11 19.
Article in English | MEDLINE | ID: mdl-33046506

ABSTRACT

FGF8 signaling plays diverse roles in inner ear development, acting at multiple stages from otic placode induction to cellular differentiation in the organ of Corti. As a secreted morphogen with diverse functions, Fgf8 expression is likely to be spatially restricted and temporally dynamic throughout inner ear development. We evaluated these characteristics using genetic labeling mediated by Fgf8mcm gene-targeted mice and determined that Fgf8 expression is a specific and early marker of Type-I vestibular hair cell identity. Fgf8mcm expression initiates at E11.5 in the future striolar region of the utricle, labeling hair cells following EdU birthdating, and demonstrates that sub-type identity is determined shortly after terminal mitosis. This early fate specification is not apparent using markers or morphological criteria that are not present before birth in the mouse. Although analyses of Fgf8 conditional knockout mice did not reveal developmental phenotypes, the restricted pattern of Fgf8 expression suggests that functionally redundant FGF ligands may contribute to vestibular hair cell differentiation and supports a developmental model in which Type-I and Type-II hair cells develop in parallel rather than from an intermediate precursor.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Hair Cells, Vestibular/metabolism , Saccule and Utricle/embryology , Animals , Fibroblast Growth Factor 8/genetics , Hair Cells, Vestibular/cytology , Mice , Mice, Knockout , Saccule and Utricle/cytology
7.
Nat Commun ; 11(1): 63, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31896743

ABSTRACT

Each vestibular sensory epithelium in the inner ear is divided morphologically and physiologically into two zones, called the striola and extrastriola in otolith organ maculae, and the central and peripheral zones in semicircular canal cristae. We found that formation of striolar/central zones during embryogenesis requires Cytochrome P450 26b1 (Cyp26b1)-mediated degradation of retinoic acid (RA). In Cyp26b1 conditional knockout mice, formation of striolar/central zones is compromised, such that they resemble extrastriolar/peripheral zones in multiple features. Mutants have deficient vestibular evoked potential (VsEP) responses to jerk stimuli, head tremor and deficits in balance beam tests that are consistent with abnormal vestibular input, but normal vestibulo-ocular reflexes and apparently normal motor performance during swimming. Thus, degradation of RA during embryogenesis is required for formation of highly specialized regions of the vestibular sensory epithelia with specific functions in detecting head motions.


Subject(s)
Otolithic Membrane/embryology , Retinoic Acid 4-Hydroxylase/metabolism , Tretinoin/metabolism , Animals , Evoked Potentials/genetics , Evoked Potentials/physiology , Female , Gene Expression Regulation, Developmental , Head/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/metabolism , Otolithic Membrane/cytology , Otolithic Membrane/metabolism , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Saccule and Utricle/cytology , Saccule and Utricle/embryology , Tremor/genetics , Tremor/physiopathology , Vestibular Function Tests , Vestibule, Labyrinth/embryology , Vestibule, Labyrinth/metabolism
8.
Exp Neurol ; 301(Pt B): 100-109, 2018 03.
Article in English | MEDLINE | ID: mdl-28928022

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.


Subject(s)
COP9 Signalosome Complex/physiology , Hair Cells, Auditory, Inner/physiology , Macrophage Migration-Inhibitory Factors/physiology , Acoustic Maculae/embryology , Acoustic Maculae/growth & development , Actins/metabolism , Animals , Axons/drug effects , COP9 Signalosome Complex/genetics , Chemokine CCL2/metabolism , Cytokines/biosynthesis , Embryo, Nonmammalian , Macrophage Migration-Inhibitory Factors/genetics , Oligonucleotides, Antisense/pharmacology , Oocysts/growth & development , Saccule and Utricle/embryology , Saccule and Utricle/growth & development , Tubulin/metabolism , Zebrafish
9.
Elife ; 62017 07 25.
Article in English | MEDLINE | ID: mdl-28742024

ABSTRACT

Dysfunctions of hearing and balance are often irreversible in mammals owing to the inability of cells in the inner ear to proliferate and replace lost sensory receptors. To determine the molecular basis of this deficiency we have investigated the dynamics of growth and cellular proliferation in a murine vestibular organ, the utricle. Based on this analysis, we have created a theoretical model that captures the key features of the organ's morphogenesis. Our experimental data and model demonstrate that an elastic force opposes growth of the utricular sensory epithelium during development, confines cellular proliferation to the organ's periphery, and eventually arrests its growth. We find that an increase in cellular density and the subsequent degradation of the transcriptional cofactor Yap underlie this process. A reduction in mechanical constraints results in accumulation and nuclear translocation of Yap, which triggers proliferation and restores the utricle's growth; interfering with Yap's activity reverses this effect.


Subject(s)
Elasticity , Epithelium/embryology , Epithelium/growth & development , Morphogenesis , Saccule and Utricle/embryology , Saccule and Utricle/growth & development , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Mice , Models, Theoretical , Phosphoproteins/metabolism , YAP-Signaling Proteins
10.
Sci Rep ; 6: 38253, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27917898

ABSTRACT

The role of Sox2 in neurosensory development is not yet fully understood. Using mice with conditional Islet1-cre mediated deletion of Sox2, we explored the function of Sox2 in neurosensory development in a model with limited cell type diversification, the inner ear. In Sox2 conditional mutants, neurons initially appear to form normally, whereas late- differentiating neurons of the cochlear apex never form. Variable numbers of hair cells differentiate in the utricle, saccule, and cochlear base but sensory epithelium formation is completely absent in the apex and all three cristae of the semicircular canal ampullae. Hair cells differentiate only in sensory epithelia known or proposed to have a lineage relationship of neurons and hair cells. All initially formed neurons lacking hair cell targets die by apoptosis days after they project toward non-existing epithelia. Therefore, late neuronal development depends directly on Sox2 for differentiation and on the survival of hair cells, possibly derived from common neurosensory precursors.


Subject(s)
Hair Cells, Auditory/metabolism , Neurogenesis/physiology , SOXB1 Transcription Factors/metabolism , Saccule and Utricle/embryology , Animals , Gene Deletion , Hair Cells, Auditory/cytology , Mice , Mice, Transgenic , SOXB1 Transcription Factors/genetics , Saccule and Utricle/cytology
11.
Ciênc. Saúde Colet. (Impr.) ; 20(2): 433-440, fev. 2015.
Article in Portuguese | LILACS, BDS | ID: lil-742216

ABSTRACT

A medicina regenerativa implica em uma mudança de paradigma, a regeneração do organismo ao nível celular ou tecidual – um assunto contemporâneo controverso e de difícil estandardização. O artigo apresenta um resumo das tendências científicas, econômicas, sociais e de regulamentação global nessa área, analisadas em relação a dilemas teóricos relevantes em antropologia médica e sociologia da ciência e da saúde. Em especial, aqueles que tratam da construção de um ‘aparato coletivo de sentido’ para as novas entidades biológicas e ontológicas, a formação da cidadania biológica e a governança pela incerteza. Apresentam-se, também, evidências empíricas sobre um fenômeno chave para a governança e a regulamentação, qual seja a instalação de uma nova demanda transnacional em pesquisa e saúde através de mercados paralelos de óvulos e de terapias celulares em experimentação. Utilizam-se dados qualitativos coletados para uma pesquisa mais abrangente, resenhas jornalísticas e entrevistas com lideranças internacionais. Conclui-se com uma reflexão sobre a importância da governança internacional em ensaios clínicos e dos caminhos a serem explorados, visando uma harmonização da diversidade de práticas normativas.


Regenerative medicine involves a paradigm change due to organism regeneration at cellular and tissue level – a controversial contemporary issue and difficult to regulate. This article presents a summary of the main scientific, economic, social and regulatory global trends, analyzed according to relevant theoretical dilemmas in medical anthropology and in the sociology of science and health. This is especially true of the construction of a ‘collective frame of reference’ on the new biological and ontological entities, the shaping of biological citizenship, and governance through uncertainty. Empirical evidence is also presented on a key aspect in regulation and governance, namely the emergence of a new transnational demand in health research through the establishment of parallel markets for ova and experimental cellular therapies. Qualitative data collected for a broader research paper is analyzed, as well as journal reviews and information gathered during interviews with international leaders. The paper concludes with a discussion on the importance on international governance of clinical trials and on further exploration, towards a multilevel harmonization of a diversity of normative practices.


Subject(s)
Humans , Animals , Male , Female , Adult , Mice , Adherens Junctions/metabolism , Cadherins/metabolism , Hair Cells, Auditory/metabolism , Postural Balance/physiology , Saccule and Utricle/metabolism , Adherens Junctions/ultrastructure , Animals, Newborn , Cell Count , Cells, Cultured , Hair Cells, Auditory/cytology , Hair Cells, Auditory/ultrastructure , Hair Cells, Vestibular/cytology , Hair Cells, Vestibular/metabolism , Hair Cells, Vestibular/ultrastructure , Mice, Transgenic , Saccule and Utricle/embryology , Saccule and Utricle/ultrastructure
12.
Dev Dyn ; 243(10): 1328-37, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24888499

ABSTRACT

BACKGROUND: Cisplatin is a widely used chemotherapeutic agent that can also cause ototoxic injury. One potential treatment for cisplatin-induced hearing loss involves the activation of endogenous inner ear stem cells, which may then produce replacement hair cells. In this series of experiments, we examined the effects of cisplatin exposure on both hair cells and resident stem cells of the mouse inner ear. RESULTS: Treatment for 24 hr with 10 µM cisplatin caused significant loss of hair cells in the mouse utricle, but such damage was not evident until 4 days after the cisplatin exposure. In addition to killing hair cells, cisplatin treatment also disrupted the actin cytoskeleton in remaining supporting cells, and led to increased histone H2AX phosphorylation within the sensory epithelia. Finally, treatment with 10 µM cisplatin appeared to have direct toxic effects on resident stem cells in the mouse utricle. Exposure to cisplatin blocked the proliferation of isolated stem cells and prevented sphere formation when those cells were maintained in suspension culture. CONCLUSION: The results suggest that inner ear stem cells may be injured during cisplatin ototoxicity, thus limiting their ability to mediate sensory repair.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Ear, Inner/drug effects , Ear, Inner/embryology , Embryonic Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Ear, Inner/cytology , Embryonic Stem Cells/physiology , Female , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/physiology , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Saccule and Utricle/cytology , Saccule and Utricle/drug effects , Saccule and Utricle/embryology
13.
Dev Biol ; 381(2): 401-10, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23796904

ABSTRACT

Atoh1 function is required for the earliest stages of inner ear hair cell development, which begins during the second week of gestation. Atoh1 expression in developing hair cells continues until early postnatal ages, but the function of this late expression is unknown. To test the role of continued Atoh1 expression in hair cell maturation we conditionally deleted the gene in the inner ear at various embryonic and postnatal ages. In the organ of Corti, deletion of Atoh1 at E15.5 led to the death of all hair cells. In contrast, deletion at E16.5 caused death only in apical regions, but abnormalities of stereocilia formation were present throughout the cochlea. In the utricle, deletion at E14.5 or E16.5 did not cause cell death but led to decreased expression of myosin VIIa and failure of stereocilia formation. Furthermore, we show that maintained expression of Barhl1 and Gfi1, two transcription factors implicated in cochlear hair cell survival, depends upon continued Atoh1 expression. However, maintained expression of Pou4f3 and several hair cell-specific markers is independent of Atoh1 expression. These data reveal novel late roles for Atoh1 that are separable from its initial role in hair cell development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Gene Expression Regulation, Developmental , Saccule and Utricle/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Cell Death , Cell Survival , Cochlea/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Female , Gene Deletion , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pregnancy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccule and Utricle/embryology , Saccule and Utricle/metabolism , Stereocilia/metabolism , Tamoxifen , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
PLoS One ; 7(10): e46863, 2012.
Article in English | MEDLINE | ID: mdl-23056493

ABSTRACT

BACKGROUND: Intrauterine exposure to amniotic fluid (AF) cytokines is thought to predispose to bronchopulmonary dysplasia (BPD). We evaluated the effects of GBS exposure on RNA expression in fetal lung tissue to determine early molecular pathways associated with fetal lung injury that may progress to BPD. METHODS: Ten chronically catheterized pregnant monkeys (Macaca nemestrina) at 118-125 days gestation (term = 172 days) received choriodecidual inoculation of either: 1) Group B Streptococcus (n = 5) or 2) saline (n = 5). Cesarean section and fetal necropsy was performed in the first week after GBS or saline inoculation regardless of labor. RNA was extracted from fetal lungs and profiled by microarray. Results were analyzed using single gene, Gene Set, and Ingenuity Pathway Analysis. Validation was by RT-PCR and immunohistochemistry. RESULTS: Despite uterine quiescence in most cases, fetal lung injury occurred in four GBS cases (intra-alveolar neutrophils, interstitial thickening) and one control (peri-mortem hemorrhage). Significant elevations of AF cytokines (TNF-α, IL-8, IL-1ß, IL-6) were detected in GBS versus controls (p<0.05). Lung injury was not directly caused by GBS, because GBS was undetectable by culture and PCR in the AF and fetal lungs. A total of 335 genes were differentially expressed greater than 1.5 fold (p<0.05) with GBS exposure associated with a striking upregulation of genes in innate and adaptive immunity and downregulation of pathways for angiogenesis, morphogenesis, and cellular growth and development. CONCLUSIONS: A transient choriodecidual infection may induce fetal lung injury with profound alterations in the genetic program of the fetal lung before signs of preterm labor. Our results provide a window for the first time into early molecular pathways disrupting fetal lung angiogenesis and morphogenesis before preterm labor occurs, which may set the stage for BPD. A strategy to prevent BPD should target the fetus in utero to attenuate alterations in the fetal lung genetic program.


Subject(s)
Down-Regulation , Fetus/microbiology , Lung/embryology , Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Pregnancy Complications/genetics , Streptococcal Infections/genetics , Animals , Cytokines/metabolism , Female , Fetus/embryology , Fetus/physiology , Gene Expression Profiling , Lung/microbiology , Macaca nemestrina , Oligonucleotide Array Sequence Analysis , Placenta/microbiology , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Pregnancy Complications/physiopathology , Saccule and Utricle/embryology , Streptococcal Infections/metabolism , Streptococcal Infections/pathology , Streptococcal Infections/physiopathology , Streptococcus agalactiae/physiology
15.
J Neurosci ; 31(33): 11855-66, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849546

ABSTRACT

Mammals experience permanent impairments from hair cell (HC) losses, but birds and other non-mammals quickly recover hearing and balance senses after supporting cells (SCs) give rise to replacement HCs. Avian HC epithelia express little or no E-cadherin, and differences in the thickness of F-actin belts at SC junctions strongly correlate with different species' capacities for HC replacement, so we investigated junctional cadherins in human and murine ears. We found strong E-cadherin expression at SC-SC junctions that increases more than sixfold postnatally in mice. When we cultured utricles from young mice with γ-secretase inhibitors (GSIs), striolar SCs completely internalized their E-cadherin, without affecting N-cadherin. Hes and Hey expression also decreased and the SCs began to express Atoh1. After 48 h, those SCs expressed myosins VI and VIIA, and by 72 h, they developed hair bundles. However, some scattered striolar SCs retained E-cadherin and the SC phenotype. In extrastriolar regions, the vast majority of SCs also retained E-cadherin and failed to convert into HCs even after long GSI treatments. Microscopic measurements revealed that the junctions between extrastriolar SCs were more developed than those between striolar SCs. In GSI-treated utricles as old as P12, differentiated striolar SCs converted into HCs, but such responses declined with age and ceased by P16. Thus, temporal and spatial differences in postnatal SC-to-HC phenotype conversion capacity are linked to the structural attributes of E-cadherin containing SC junctions in mammals, which differ substantially from their counterparts in non-mammalian vertebrates that readily recover from hearing and balance deficits through hair cell regeneration.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Hair Cells, Auditory/metabolism , Postural Balance/physiology , Saccule and Utricle/metabolism , Adherens Junctions/ultrastructure , Adult , Animals , Animals, Newborn , Cell Count , Cells, Cultured , Female , Hair Cells, Auditory/cytology , Hair Cells, Auditory/ultrastructure , Hair Cells, Vestibular/cytology , Hair Cells, Vestibular/metabolism , Hair Cells, Vestibular/ultrastructure , Humans , Male , Mice , Mice, Transgenic , Saccule and Utricle/embryology , Saccule and Utricle/ultrastructure
16.
PLoS One ; 6(3): e17949, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21423764

ABSTRACT

Loss-of-function mutations of SLC26A4/pendrin are among the most prevalent causes of deafness. Deafness and vestibular dysfunction in the corresponding mouse model, Slc26a4(-/-), are associated with an enlargement and acidification of the membranous labyrinth. Here we relate the onset of expression of the HCO(3) (-) transporter pendrin to the luminal pH and to enlargement-associated epithelial cell stretching. We determined expression with immunocytochemistry, cell stretching by digital morphometry and pH with double-barreled ion-selective electrodes. Pendrin was first expressed in the endolymphatic sac at embryonic day (E) 11.5, in the cochlear hook-region at E13.5, in the utricle and saccule at E14.5, in ampullae at E16.5, and in the upper turn of the cochlea at E17.5. Epithelial cell stretching in Slc26a4(-/-) mice began at E14.5. pH changes occurred first in the cochlea at E15.5 and in the endolymphatic sac at E17.5. At postnatal day 2, stria vascularis, outer sulcus and Reissner's membrane epithelial cells, and utricular and saccular transitional cells were stretched, whereas sensory cells in the cochlea, utricle and saccule did not differ between Slc26a4(+/-) and Slc26a4(-/-) mice. Structural development of stria vascularis, including vascularization, was retarded in Slc26a4(-/-) mice. In conclusion, the data demonstrate that the enlargement and stretching of non-sensory epithelial cells precedes luminal acidification in the cochlea and the endolymphatic sac. Stretching and luminal acidification may alter cell-to-cell communication and lead to the observed retarded development of stria vascularis, which may be an important step on the path to deafness in Slc26a4(-/-) mice, and possibly in humans, lacking functional pendrin expression.


Subject(s)
Acids/metabolism , Anion Transport Proteins/deficiency , Deafness/embryology , Deafness/pathology , Epithelial Cells/pathology , Stria Vascularis/metabolism , Stria Vascularis/pathology , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Connexin 26 , Connexins/metabolism , Deafness/metabolism , Epithelial Cells/metabolism , Hydrogen-Ion Concentration , Mice , Saccule and Utricle/embryology , Saccule and Utricle/metabolism , Saccule and Utricle/pathology , Sodium-Potassium-Exchanging ATPase/metabolism , Stria Vascularis/enzymology , Sulfate Transporters
17.
J Chem Neuroanat ; 41(2): 73-85, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21126574

ABSTRACT

PINK1 is a 581 amino acid protein with a serine/threonine kinase domain and an N-terminal mitochondrial targeting motif. The enzyme is expressed in the brain as well as in several tissues such as heart, skeletal muscle, liver, kidney, pancreas and testis. In the present study, we have investigated by Western blot analysis and immunohistochemistry the presence and distribution of PINK1 in the brain, eye and inner ear of mouse during embryonic development. In the brain we detected two PINK1 molecular isoforms of 55 kDa and 66 kDa. Immunoreactive perikarya first appeared at stage E15 in the diencephalon within the thalamus, the hypothalamus, the periventricular layers of the third ventricle and in the rhombencephalon at level of the pons. Subsequently, new PINK1-positive neurons were found in the midbrain within the floor and the periventricular layers of the ventral wall of the mesencephalic vesicle (stage E17) as well as in the neopallial cortex, the tegmentum of the midbrain and the periventricular region of the caudal part of the rhombencephalon (stage E19). At P0, PINK1-immunoreactive cells appeared in the striatum, the mantle layer and caudal part of the medulla oblongata and the cerebellum. The spatio-temporal expression of PINK1 and its heterogeneous distribution suggest that the enzyme might be involved in neuroregulatory processes during embryogenesis. In the eye, PINK1-immunoreactivity was found in the lens and in the cornea, whereas in the inner ear the enzyme was expressed in the ependymal and subependymal cells of the saccule and in the semicircular canals indicating that PINK1 plays a role in the development of these sensory organs.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/metabolism , Protein Kinases/analysis , Animals , Cornea/embryology , Cornea/growth & development , Cornea/metabolism , Diencephalon/embryology , Diencephalon/growth & development , Diencephalon/metabolism , Embryonic Development , Female , Immunohistochemistry , Lens, Crystalline/embryology , Lens, Crystalline/growth & development , Lens, Crystalline/metabolism , Male , Mesencephalon/embryology , Mesencephalon/growth & development , Mesencephalon/metabolism , Mice , Neurons/cytology , Organ Specificity , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Kinases/genetics , Rhombencephalon/embryology , Rhombencephalon/growth & development , Rhombencephalon/metabolism , Saccule and Utricle/embryology , Saccule and Utricle/growth & development , Saccule and Utricle/metabolism , Semicircular Canals/embryology , Semicircular Canals/growth & development , Semicircular Canals/metabolism
18.
Acta Otolaryngol ; 130(7): 773-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20082566

ABSTRACT

UNLABELLED: Abstract Conclusion: There is no change in the total cell population and hair cell:supporting cell ratio in the human utricular macula from gestational week 16 and onwards, whereas the lower hair cell:supporting cell ratio and lower total number of cells in the youngest specimens indicate that the utricle is still differentiating and adding new cells at the 10th to 12th gestational week. OBJECTIVES: Archival temporal bones were investigated to quantify cell numbers in the utricular macula in fetuses and children. METHODS: The age of the subjects ranged from gestational week 10 to 15 years. The optical fractionator was used to estimate the total number of cells in the utricular macula. RESULTS: The total cell number was found to be 143 000 in subjects older than gestational week 16. The number of hair cells and supporting cells did not change between the 16th gestational week and 15 years and was 36 000 and 107 000, respectively. In the youngest specimen (10th and 12th gestational week) there was a statistically significant lower total number of cells (62 000) and a lower hair cell:supporting cell ratio, resulting in a mean number of 13 000 hair cells and 49 000 supporting cells.


Subject(s)
Hair Cells, Vestibular/cytology , Saccule and Utricle/cytology , Adolescent , Cell Count/instrumentation , Cell Count/methods , Child , Child, Preschool , Female , Fetus , Gestational Age , Humans , Infant , Infant, Newborn , Male , Saccule and Utricle/embryology
19.
Cell Tissue Res ; 334(3): 339-58, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18985389

ABSTRACT

At embryonic day 8.5, the LIM-homeodomain factor Lmx1a is expressed throughout the otic placode but becomes developmentally restricted to non-sensory epithelia of the ear (endolymphatic duct, ductus reuniens, cochlea lateral wall). We confirm here that the ears of newborn dreher (Lmx1a (dr)) mutants are dysmorphic. Hair cell markers such as Atoh1 and Myo7 reveal, for the first time, that newborn Lmx1a mutants have only three sensory epithelia: two enlarged canal cristae and one fused epithelium comprising an amalgamation of the cochlea, saccule, and utricle (a "cochlear-gravistatic" endorgan). The enlarged anterior canal crista develops by fusion of horizontal and anterior crista, whereas the posterior crista fuses with an enlarged papilla neglecta that may extend into the cochlear lateral wall. In the fused endorgan, the cochlear region is distinguished from the vestibular region by markers such as Gata3, the presence of a tectorial membrane, and cochlea-specific innervation. The cochlea-like apex displays minor disorganization of the hair and supporting cells. This contrasts with the basal half of the cochlear region, which shows a vestibular epithelium-like organization of hair cells and supporting cells. The dismorphic features of the cochlea are also reflected in altered gene expression patterns. Fgf8 expression expands from inner hair cells in the apex to most hair cells in the base. Two supporting cell marker proteins, Sox2 and Prox1, also differ in their cellular distribution between the base and the apex. Sox2 expression expands in mutant canal cristae prior to their enlargement and fusion and displays a more diffuse and widespread expression in the base of the cochlear region, whereas Prox1 is not detected in the base. These changes in Sox2 and Prox1 expression suggest that Lmx1a expression restricts and sharpens Sox2 expression, thereby defining non-sensory and sensory epithelium. The adult Lmx1a mutant organ of Corti shows a loss of cochlear hair cells, suggesting that the long-term maintenance of hair cells is also disrupted in these mutants.


Subject(s)
Ear/embryology , Epithelium/embryology , Homeodomain Proteins/metabolism , Morphogenesis , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Ear/pathology , Epithelium/innervation , Epithelium/pathology , Epithelium/ultrastructure , Gene Expression Regulation , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Inner/ultrastructure , LIM-Homeodomain Proteins , Mice , Mutation/genetics , Organ of Corti/embryology , Organ of Corti/pathology , Organ of Corti/ultrastructure , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Saccule and Utricle/embryology , Saccule and Utricle/pathology , Saccule and Utricle/ultrastructure , Transcription Factors
20.
Dev Biol ; 322(1): 33-45, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18652815

ABSTRACT

Inner ear hair cells and supporting cells arise from common precursors and, in mammals, do not show phenotypic conversion. Here, we studied the role of the homeodomain transcription factor Prox1 in the inner ear sensory epithelia. Adenoviral-mediated Prox1 transduction into hair cells in explant cultures led to strong repression of Atoh1 and Gfi1, two transcription factors critical for hair cell differentiation and survival. Luciferase assays showed that Prox1 can repress transcriptional activity of Gfi1 independently of Atoh1. Prox1 transduction into cochlear outer hair cells resulted in degeneration of these cells, consistent with the known phenotype of Gfi1-deficient mice. These results together with the widespread expression of endogenous Prox1 within the population of inner ear supporting cells point to the role for Prox1 in antagonizing the hair cell phenotype in these non-sensory cells. Further, in vivo analyses of hair cells from Gfi1-deficient mice suggest that the cyclin-dependent kinase inhibitor p57(Kip2) mediates the differentiation- and survival-promoting functions of Gfi1. These data reveal novel gene interactions and show that these interactions regulate cellular differentiation within the inner ear sensory epithelia. The data point to the tight regulation of phenotypic characteristics of hair cells and supporting cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Ear, Inner/embryology , Epithelial Cells/physiology , Homeodomain Proteins/physiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/physiology , Adenoviridae/genetics , Animals , Apoptosis/physiology , Cell Differentiation/genetics , Cells, Cultured , Cochlea/cytology , Cochlea/embryology , Cochlea/metabolism , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Ear, Inner/cytology , Epithelial Cells/cytology , Gene Expression Regulation, Developmental , Gene Transfer Techniques , Genes, Reporter , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/virology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Knockout , NIH 3T3 Cells , Organ Culture Techniques , Saccule and Utricle/cytology , Saccule and Utricle/embryology , Saccule and Utricle/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...