Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Elife ; 122024 Mar 14.
Article in English | MEDLINE | ID: mdl-38483314

ABSTRACT

Mammals harbor a limited number of sound-receptor hair cells (HCs) that cannot be regenerated after damage. Thus, investigating the underlying molecular mechanisms that maintain HC survival is crucial for preventing hearing impairment. Intriguingly, Pou4f3-/- or Gfi1-/- HCs form initially but then rapidly degenerate, whereas Rbm24-/- HCs degenerate considerably later. However, the transcriptional cascades involving Pou4f3, Gfi1, and Rbm24 remain undescribed. Here, we demonstrate that Rbm24 expression is completely repressed in Pou4f3-/- HCs but unaltered in Gfi1-/- HCs, and further that the expression of both POU4F3 and GFI1 is intact in Rbm24-/- HCs. Moreover, by using in vivo mouse transgenic reporter assays, we identify three Rbm24 enhancers to which POU4F3 binds. Lastly, through in vivo genetic testing of whether Rbm24 restoration alleviates the degeneration of Pou4f3-/- HCs, we show that ectopic Rbm24 alone cannot prevent Pou4f3-/- HCs from degenerating. Collectively, our findings provide new molecular and genetic insights into how HC survival is regulated.


Subject(s)
Genetic Therapy , Transcription Factors , Animals , Mice , Animals, Genetically Modified , Transcription Factors/genetics , Hair Cells, Auditory , Sound , Mammals , Homeodomain Proteins , Transcription Factor Brn-3C/genetics , DNA-Binding Proteins/genetics , RNA-Binding Proteins
2.
Sci Rep ; 13(1): 12584, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37537203

ABSTRACT

POU4F3, a member of the POU family of transcription factors, commonly causes autosomal dominant deafness. Exome sequencing was used to identify four novel variants in POU4F3 (NM_002700.2), including c.564dupA: p.Ala189SerfsTer26, c.743T > C:p.Leu248Pro, c.879C > A:p.Phe293Leu, and c.952G > A:p.Val318Met, and diverse aspects of the molecular consequences of their protein expression, stability, subcellular localization, and transcriptional activity were investigated. The expression of three mutant proteins, encoded by missense variants, was reduced compared to the wild-type protein, demonstrating that the mutants were unstable and vulnerable to degradation. Additionally, all the mutant proteins had distinct subcellular localization patterns. A mutant protein carrying p.Ala189SerfsTer26, in which both mono- and bi-partite nuclear localization signals were disrupted, showed abnormal subcellular localization. Resultantly, all the mutant proteins significantly reduced the transcriptional activity required to regulate the downstream target gene expression. Furthermore, we identified the altered expression of 14 downstream target genes associated with inner ear development using patient-derived lymphoblastoid cell lines. There was a significant correlation of the expression profile between patient-derived cells and the cochlear hair cells, which provided a breakthrough for cases where the collection of human cochlear samples for transcriptome studies was unfeasible. This study expanded the genotypic spectrum of POU4F3 in DFNA15, and further refined the molecular mechanisms underlying POU4F3-associated DFNA15.


Subject(s)
Hearing Loss, Sensorineural , Hearing Loss , Humans , Homeodomain Proteins/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss/genetics , Hearing Loss/metabolism , Transcription Factors/genetics , Transcription Factor Brn-3C/genetics , Pedigree
3.
Eur J Hum Genet ; 31(7): 815-823, 2023 07.
Article in English | MEDLINE | ID: mdl-37072551

ABSTRACT

Genotype-phenotype correlations add value to the management of families with hereditary hearing loss (HL), where age-related typical audiograms (ARTAs) are generated from cross-sectional regression equations and used to predict the audiogram phenotype across the lifespan. A seven-generation kindred with autosomal dominant sensorineural HL (ADSNHL) was recruited and a novel pathogenic variant in POU4F3 (c.37del) was identified by combining linkage analysis with whole exome sequencing (WES). POU4F3 is noted for large intrafamilial variation including the age of onset of HL, audiogram configuration and presence of vestibular impairment. Sequential audiograms and longitudinal analyses reveal highly variable audiogram features among POU4F3 (c.37del) carriers, limiting the utility of ARTAs for clinical prognosis and management of HL. Furthermore, a comparison of ARTAs against three previously published families (1 Israeli Jewish, 2 Dutch) reveals significant interfamilial differences, with earlier onset and slower deterioration. This is the first published report of a North American family with ADSNHL due to POU4F3, the first report of the pathogenic c.37del variant, and the first study to conduct longitudinal analysis, extending the phenotypic spectrum of DFNA15.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hearing Loss , Humans , Cross-Sectional Studies , Homeodomain Proteins/genetics , Hearing Loss/genetics , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Pedigree , Transcription Factor Brn-3C/genetics
4.
Elife ; 112022 11 29.
Article in English | MEDLINE | ID: mdl-36445327

ABSTRACT

Reprogramming of the cochlea with hair-cell-specific transcription factors such as ATOH1 has been proposed as a potential therapeutic strategy for hearing loss. ATOH1 expression in the developing cochlea can efficiently induce hair cell regeneration but the efficiency of hair cell reprogramming declines rapidly as the cochlea matures. We developed Cre-inducible mice to compare hair cell reprogramming with ATOH1 alone or in combination with two other hair cell transcription factors, GFI1 and POU4F3. In newborn mice, all transcription factor combinations tested produced large numbers of cells with the morphology of hair cells and rudimentary mechanotransduction properties. However, 1 week later, only a combination of ATOH1, GFI1 and POU4F3 could reprogram non-sensory cells of the cochlea to a hair cell fate, and these new cells were less mature than cells generated by reprogramming 1 week earlier. We used scRNA-seq and combined scRNA-seq and ATAC-seq to suggest at least two impediments to hair cell reprogramming in older animals. First, hair cell gene loci become less epigenetically accessible in non-sensory cells of the cochlea with increasing age. Second, signaling from hair cells to supporting cells, including Notch signaling, can prevent reprogramming of many supporting cells to hair cells, even with three hair cell transcription factors. Our results shed light on the molecular barriers that must be overcome to promote hair cell regeneration in the adult cochlea.


Subject(s)
Cellular Reprogramming , Hair Cells, Auditory, Inner , Mechanotransduction, Cellular , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Homeodomain Proteins , Signal Transduction , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics , Hair Cells, Auditory, Inner/cytology
5.
Proc Natl Acad Sci U S A ; 119(15): e2116973119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35380897

ABSTRACT

Sensory hair cells (HCs) in the utricle are mechanoreceptors required to detect linear acceleration. After damage, the mammalian utricle partially restores the HC population and organ function, although regenerated HCs are primarily type II and immature. Whether native, surviving HCs can repair and contribute to this recovery is unclear. Here, we generated the Pou4f3DTR/+; Atoh1CreERTM/+; Rosa26RtdTomato/+ mouse to fate map HCs prior to ablation. After HC ablation, vestibular evoked potentials were abolished in all animals, with ∼57% later recovering responses. Relative to nonrecovery mice, recovery animals harbored more Atoh1-tdTomato+ surviving HCs. In both groups, surviving HCs displayed markers of both type I and type II subtypes and afferent synapses, despite distorted lamination and morphology. Surviving type II HCs remained innervated in both groups, whereas surviving type I HCs first lacked and later regained calyces in the recovery, but not the nonrecovery, group. Finally, surviving HCs initially displayed immature and subsequently mature-appearing bundles in the recovery group. These results demonstrate that surviving HCs are capable of self-repair and may contribute to the recovery of vestibular function.


Subject(s)
Hair Cells, Vestibular , Regeneration , Saccule and Utricle , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Survival/genetics , Hair Cells, Vestibular/physiology , Homeodomain Proteins/genetics , Mice , Mice, Mutant Strains , RNA, Untranslated/genetics , Regeneration/genetics , Saccule and Utricle/cytology , Saccule and Utricle/injuries , Saccule and Utricle/physiology , Transcription Factor Brn-3C/genetics
6.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34544869

ABSTRACT

Mutations in the gene for Norrie disease protein (Ndp) cause syndromic deafness and blindness. We show here that cochlear function in an Ndp knockout mouse deteriorated with age: At P3-P4, hair cells (HCs) showed progressive loss of Pou4f3 and Gfi1, key transcription factors for HC maturation, and Myo7a, a specialized myosin required for normal function of HC stereocilia. Loss of expression of these genes correlated to increasing HC loss and profound hearing loss by 2 mo. We show that overexpression of the Ndp gene in neonatal supporting cells or, remarkably, up-regulation of canonical Wnt signaling in HCs rescued HCs and cochlear function. We conclude that Ndp secreted from supporting cells orchestrates a transcriptional network for the maintenance and survival of HCs and that increasing the level of ß-catenin, the intracellular effector of Wnt signaling, is sufficient to replace the functional requirement for Ndp in the cochlea.


Subject(s)
DNA-Binding Proteins/metabolism , Eye Proteins/physiology , Hair Cells, Auditory/pathology , Hearing Loss/pathology , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/physiology , Transcription Factor Brn-3C/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , DNA-Binding Proteins/genetics , Female , Hair Cells, Auditory/metabolism , Hearing Loss/etiology , Hearing Loss/metabolism , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics , Wnt Signaling Pathway
7.
Elife ; 102021 09 21.
Article in English | MEDLINE | ID: mdl-34545809

ABSTRACT

Nearly 50 different mouse retinal ganglion cell (RGC) types sample the visual scene for distinct features. RGC feature selectivity arises from their synapses with a specific subset of amacrine (AC) and bipolar cell (BC) types, but how RGC dendrites arborize and collect input from these specific subsets remains poorly understood. Here we examine the hypothesis that RGCs employ molecular recognition systems to meet this challenge. By combining calcium imaging and type-specific histological stains, we define a family of circuits that express the recognition molecule Sidekick-1 (Sdk1), which include a novel RGC type (S1-RGC) that responds to local edges. Genetic and physiological studies revealed that Sdk1 loss selectively disrupts S1-RGC visual responses, which result from a loss of excitatory and inhibitory inputs and selective dendritic deficits on this neuron. We conclude that Sdk1 shapes dendrite growth and wiring to help S1-RGCs become feature selective.


Subject(s)
Calcium Signaling , Dendrites/metabolism , Immunoglobulin G/metabolism , Membrane Proteins/metabolism , Neuronal Plasticity , Retinal Ganglion Cells/metabolism , Synapses/metabolism , Vision, Ocular , Visual Perception , Animals , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Excitatory Postsynaptic Potentials , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin G/genetics , Inhibitory Postsynaptic Potentials , Male , Membrane Proteins/genetics , Mice, Knockout , Neural Inhibition , Photic Stimulation , Synapses/genetics , Time Factors , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Visual Pathways/metabolism
8.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34266958

ABSTRACT

During embryonic development, hierarchical cascades of transcription factors interact with lineage-specific chromatin structures to control the sequential steps in the differentiation of specialized cell types. While examples of transcription factor cascades have been well documented, the mechanisms underlying developmental changes in accessibility of cell type-specific enhancers remain poorly understood. Here, we show that the transcriptional "master regulator" ATOH1-which is necessary for the differentiation of two distinct mechanoreceptor cell types, hair cells in the inner ear and Merkel cells of the epidermis-is unable to access much of its target enhancer network in the progenitor populations of either cell type when it first appears, imposing a block to further differentiation. This block is overcome by a feed-forward mechanism in which ATOH1 first stimulates expression of POU4F3, which subsequently acts as a pioneer factor to provide access to closed ATOH1 enhancers, allowing hair cell and Merkel cell differentiation to proceed. Our analysis also indicates the presence of both shared and divergent ATOH1/POU4F3-dependent enhancer networks in hair cells and Merkel cells. These cells share a deep developmental lineage relationship, deriving from their common epidermal origin, and suggesting that this feed-forward mechanism preceded the evolutionary divergence of these very different mechanoreceptive cell types.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Mechanoreceptors/metabolism , Transcription Factor Brn-3C/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cochlea/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Hair Cells, Auditory/cytology , Homeodomain Proteins/genetics , Humans , Merkel Cells/metabolism , Mice , Transcription Factor Brn-3C/genetics
9.
Biomed Res Int ; 2021: 5574136, 2021.
Article in English | MEDLINE | ID: mdl-34250087

ABSTRACT

BACKGROUND: The pathogenic variant, POU class 4 transcription factor 3 (POU4F3), is reported to cause autosomal dominant nonsyndromic hearing loss (ADNSHL). Previously, we have examined a four-generation midfrequency sensorineural hearing loss (MFSNHL) family (no. 6126) and established POU4F3 c.602T>C (p.Leu201Pro) as a potential disease-causing variant. OBJECTIVES: We explored the structural and functional alterations that the c.602T>C (p.Leu201Pro) variant enforces on the POU4F3 protein. METHODS: We utilized wild-type (WT) and mutant (MUT) POU4F3 c.602T>C plasmid incorporation into HeLa cells to assess functional changes, by immunofluorescence and luciferase assays. To predict protein structural alterations in the MUT versus WT POU4F3, we also generated 3D structures to compare both types of POU4F3 proteins. RESULTS: The WT POU4F3 is ubiquitously present in the nucleus, whereas the MUT form of POU4F3 exhibits a more restricted nuclear presence. This finding is different from other publications, which report a cytoplasmic localization of the MUT POU4F3. We also demonstrated that, as opposed to WT POU4F3, the MUT POU4F3 had 40% reduced luciferase activity. CONCLUSIONS: The reduced nuclear presence, combined with reduced transcriptional activity, suggests that the POU4F3 c.602T>C variant alters cellular activity and may contribute to the pathogenicity of POU4F3-related hearing loss. It, also, provides more evidence of the pathophysiological characteristics of MFSNHL.


Subject(s)
Cell Nucleus/metabolism , Genes, Dominant , Hearing Loss, Sensorineural/genetics , Homeodomain Proteins/genetics , Mutation, Missense/genetics , Transcription Factor Brn-3C/genetics , Transcription, Genetic , Base Sequence , HeLa Cells , Homeodomain Proteins/chemistry , Humans , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Transport , Transcription Factor Brn-3C/chemistry
10.
Cell Rep ; 35(3): 109016, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882317

ABSTRACT

The mammalian cochlea cannot regenerate functional hair cells (HCs) spontaneously. Atoh1 overexpression as well as other strategies are unable to generate functional HCs. Here, we simultaneously upregulated the expression of Gfi1, Pou4f3, and Atoh1 in postnatal cochlear supporting cells (SCs) in vivo, which efficiently converted SCs into HCs. The newly regenerated HCs expressed HC markers Myo7a, Calbindin, Parvalbumin, and Ctbp2 and were innervated by neurites. Importantly, many new HCs expressed the mature and terminal marker Prestin or vesicular glutamate transporter 3 (vGlut3), depending on the subtypes of the source SCs. Finally, our patch-clamp analysis showed that the new HCs in the medial region acquired a large K+ current, fired spikes transiently, and exhibited signature refinement of ribbon synapse functions, in close resemblance to native wild-type inner HCs. We demonstrated that co-upregulating Gfi1, Pou4f3, and Atoh1 enhances the efficiency of HC generation and promotes the functional maturation of new HCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , Hair Cells, Auditory/metabolism , Homeodomain Proteins/genetics , Labyrinth Supporting Cells/metabolism , Organogenesis/genetics , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics , Action Potentials/physiology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calbindins/genetics , Calbindins/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Homeodomain Proteins/metabolism , Ion Transport , Labyrinth Supporting Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Myosin VIIa/genetics , Myosin VIIa/metabolism , Neurites/metabolism , Neurites/ultrastructure , Parvalbumins/genetics , Parvalbumins/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Signal Transduction , Transcription Factor Brn-3C/metabolism , Transcription Factors/metabolism
11.
FEBS J ; 288(1): 325-353, 2021 01.
Article in English | MEDLINE | ID: mdl-32323465

ABSTRACT

Cochlear development is a complex process with precise spatiotemporal patterns. A detailed understanding of this process is important for studies of congenital hearing loss and regenerative medicine. However, much of our understanding of cochlear development is based on rodent models. Animal models that bridge the gap between humans and rodents are needed. In this study, we investigated the development of hearing organs in a small New World monkey species, the common marmoset (Callithrix jacchus). We describe the general stages of cochlear development in comparison with those of humans and mice. Moreover, we examined more than 25 proteins involved in cochlear development and found that expression patterns were generally conserved between rodents and primates. However, several proteins involved in supporting cell processes and neuronal development exhibited interspecific expression differences. Human fetal samples for studies of primate-specific cochlear development are extremely rare, especially for late developmental stages. Our results support the use of the common marmoset as an effective alternative for analyses of primate cochlear development.


Subject(s)
Callithrix/genetics , Cochlea/metabolism , Gene Expression Regulation, Developmental , Models, Animal , Organogenesis/genetics , Animals , Aquaporin 4/genetics , Aquaporin 4/metabolism , Calbindin 1/genetics , Calbindin 1/metabolism , Callithrix/embryology , Callithrix/growth & development , Callithrix/metabolism , Cochlea/anatomy & histology , Cochlea/cytology , Cochlea/growth & development , Conserved Sequence , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Embryo, Mammalian , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Myosin VIIa/genetics , Myosin VIIa/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Peripherins/genetics , Peripherins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Species Specificity , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Tubulin/genetics , Tubulin/metabolism
12.
J Comp Neurol ; 529(8): 1926-1953, 2021 06.
Article in English | MEDLINE | ID: mdl-33135183

ABSTRACT

Members of the POU4F/Brn3 transcription factor family have an established role in the development of retinal ganglion cell (RGCs) types, the main transducers of visual information from the mammalian eye to the brain. Our previous work using sparse random recombination of a conditional knock-in reporter allele expressing alkaline phosphatase (AP) and intersectional genetics had identified three types of Brn3c positive (Brn3c+ ) RGCs. Here, we describe a novel Brn3cCre mouse allele generated by serial Dre to Cre recombination and use it to explore the expression overlap of Brn3c with Brn3a and Brn3b and the dendritic arbor morphologies and visual stimulus response properties of Brn3c+ RGC types. Furthermore, we explore brain nuclei that express Brn3c or receive input from Brn3c+ neurons. Our analysis reveals a much larger number of Brn3c+ RGCs and more diverse set of RGC types than previously reported. Most RGCs expressing Brn3c during development are still Brn3c positive in the adult, and all express Brn3a while only about half express Brn3b. Genetic Brn3c-Brn3b intersection reveals an area of increased RGC density, extending from dorsotemporal to ventrolateral across the retina and overlapping with the mouse binocular field of view. In addition, we report a Brn3c+ RGC projection to the thalamic reticular nucleus, a visual nucleus that was not previously shown to receive retinal input. Furthermore, Brn3c+ neurons highlight a previously unknown subdivision of the deep mesencephalic nucleus. Thus, our newly generated allele provides novel biological insights into RGC type classification, brain connectivity, and cytoarchitectonic.


Subject(s)
Brain/cytology , Brain/metabolism , Homeodomain Proteins/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Transcription Factor Brn-3C/metabolism , Alleles , Animals , Gene Knock-In Techniques/methods , Homeodomain Proteins/genetics , Integrases , Mice , Transcription Factor Brn-3C/genetics , Visual Pathways/cytology , Visual Pathways/metabolism
13.
PLoS Genet ; 16(9): e1009040, 2020 09.
Article in English | MEDLINE | ID: mdl-32970669

ABSTRACT

Genetic hearing loss is a common health problem with no effective therapy currently available. DFNA15, caused by mutations of the transcription factor POU4F3, is one of the most common forms of autosomal dominant non-syndromic deafness. In this study, we established a novel mouse model of the human DFNA15 deafness, with a Pou4f3 gene mutation (Pou4f3Δ) identical to that found in a familial case of DFNA15. The Pou4f3(Δ/+) mice suffered progressive deafness in a similar manner to the DFNA15 patients. Hair cells in the Pou4f3(Δ/+) cochlea displayed significant stereociliary and mitochondrial pathologies, with apparent loss of outer hair cells. Progression of hearing and outer hair cell loss of the Pou4f3(Δ/+) mice was significantly modified by other genetic and environmental factors. Using Pou4f3(-/+) heterozygous knockout mice, we also showed that DFNA15 is likely caused by haploinsufficiency of the Pou4f3 gene. Importantly, inhibition of retinoic acid signaling by the aldehyde dehydrogenase (Aldh) and retinoic acid receptor inhibitors promoted Pou4f3 expression in the cochlear tissue and suppressed the progression of hearing loss in the mutant mice. These data demonstrate Pou4f3 haploinsufficiency as the main underlying cause of human DFNA15 deafness and highlight the therapeutic potential of Aldh inhibitors for treatment of progressive hearing loss.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/pathology , Hearing Loss/drug therapy , Hearing Loss/etiology , Homeodomain Proteins/genetics , Transcription Factor Brn-3C/genetics , Animals , Benzaldehydes/pharmacology , Disease Models, Animal , Haploinsufficiency/genetics , Hearing Loss/genetics , Hearing Loss/pathology , Homeodomain Proteins/metabolism , Humans , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Noise/adverse effects , Quinolines/pharmacology , Transcription Factor Brn-3C/metabolism , Tretinoin/pharmacology , para-Aminobenzoates/pharmacology
14.
Neural Plast ; 2020: 6137083, 2020.
Article in English | MEDLINE | ID: mdl-32684921

ABSTRACT

Hereditary hearing loss is one of the most common sensory disabilities worldwide. Mutation of POU domain class 4 transcription factor 3 (POU4F3) is considered the pathogenic cause of autosomal dominant nonsyndromic hearing loss (ADNSHL), designated as autosomal dominant nonsyndromic deafness 15. In this study, four novel variants in POU4F3, c.696G>T (p.Glu232Asp), c.325C>T (p.His109Tyr), c.635T>C (p.Leu212Pro), and c.183delG (p.Ala62Argfs∗22), were identified in four different Chinese families with ADNSHL by targeted next-generation sequencing and Sanger sequencing. Based on the American College of Medical Genetics and Genomics guidelines, c.183delG (p.Ala62Argfs∗22) is classified as a pathogenic variant, c.696G>T (p.Glu232Asp) and c.635T>C (p.Leu212Pro) are classified as likely pathogenic variants, and c.325C>T (p.His109Tyr) is classified as a variant of uncertain significance. Based on previous reports and the results of this study, we speculated that POU4F3 pathogenic variants are significant contributors to ADNSHL in the East Asian population. Therefore, screening of POU4F3 should be a routine examination for the diagnosis of hereditary hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Homeodomain Proteins/genetics , Mutation, Missense , Pedigree , Transcription Factor Brn-3C/genetics , Adolescent , Child , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Young Adult
15.
J Cell Mol Med ; 24(12): 6978-6987, 2020 06.
Article in English | MEDLINE | ID: mdl-32390314

ABSTRACT

Autosomal dominant non-syndromic hearing loss is genetically heterogeneous with 47 genes identified to date, including POU4F3. In this study, by using a next-generation sequencing panel targeting 127 deafness genes, we identified a pathogenic frameshift mutation c.704_705del and a missense mutation c.593G>A in two three-generation Chinese families with late-onset progressive ADNSHL, respectively. The novel mutations of POU4F3 co-segregated with the deafness phenotype in these two families. c.704_705del caused a frameshift p.T235fs and c.593G>A caused an amino acid substitution of p.R198H. Both mutations led to an abnormal and incomplete protein structure. POU4F3 with either of the two mutations was transiently transfected into HEI-OC1 and HEK 293 cell lines and immunofluorescence assay was performed to investigate the subcellular localization of mutated protein. The results indicated that both c.704_705del (p.T235fs) and c.593G>A (p.R198H) could impair the nuclear localization function of POU4F3. The p.R198H POU4F3 protein was detected as a weak band of the correct molecular weight, indicating that the stability of p.R198H POU4F3 differed from that of the wild-type protein. While, the p.T235fs POU4F3 protein was expressed with a smaller molecular weight, implying this mutation result in a frameshift and premature termination of the POU4F3 protein. In summary, we report two novel mutations of POU4F3 associated with progressive ADNSHL and explored their effects on POU4F3 nuclear localization. These findings expanded the mutation spectrum of POU4F3 and provided new knowledge for the pathogenesis of POU4F3 in hearing loss.


Subject(s)
Asian People/genetics , Genes, Dominant , Genetic Association Studies , Genetic Predisposition to Disease , Hearing Loss/genetics , Homeodomain Proteins/genetics , Mutation/genetics , Transcription Factor Brn-3C/genetics , Adult , Amino Acid Sequence , Base Sequence , Family , Female , Genome, Human , Homeodomain Proteins/chemistry , Humans , Male , Middle Aged , Models, Molecular , Pedigree , Subcellular Fractions/metabolism , Transcription Factor Brn-3C/chemistry
16.
Wiley Interdiscip Rev Dev Biol ; 9(4): e374, 2020 07.
Article in English | MEDLINE | ID: mdl-32012462

ABSTRACT

One approach to understand the construction of complex systems is to investigate whether there are simple design principles that are commonly used in building such a system. In the context of nervous system development, one may ask whether the generation of its highly diverse sets of constituents, that is, distinct neuronal cell types, relies on genetic mechanisms that share specific common features. Specifically, are there common patterns in the function of regulatory genes across different neuron types and are those regulatory mechanisms not only used in different parts of one nervous system, but are they conserved across animal phylogeny? We address these questions here by focusing on one specific, highly conserved and well-studied regulatory factor, the POU homeodomain transcription factor UNC-86. Work over the last 30 years has revealed a common and paradigmatic theme of unc-86 function throughout most of the neuron types in which Caenorhabditis elegans unc-86 is expressed. Apart from its role in preventing lineage reiterations during development, UNC-86 operates in combination with distinct partner proteins to initiate and maintain terminal differentiation programs, by coregulating a vast array of functionally distinct identity determinants of specific neuron types. Mouse orthologs of unc-86, the Brn3 genes, have been shown to fulfill a similar function in initiating and maintaining neuronal identity in specific parts of the mouse brain and similar functions appear to be carried out by the sole Drosophila ortholog, Acj6. The terminal selector function of UNC-86 in many different neuron types provides a paradigm for neuronal identity regulation across phylogeny. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Invertebrate Organogenesis > Worms Nervous System Development > Vertebrates: Regional Development.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , POU Domain Factors/genetics , Transcription Factor Brn-3C/genetics , Animals , Caenorhabditis elegans/classification , Caenorhabditis elegans/cytology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation , Cell Lineage/genetics , Conserved Sequence , Drosophila Proteins/metabolism , Drosophila melanogaster/classification , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nervous System/cytology , Nervous System/growth & development , Nervous System/metabolism , Neurons/cytology , Neurons/metabolism , POU Domain Factors/metabolism , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Transcription Factor Brn-3C/metabolism
17.
Nature ; 577(7790): 392-398, 2020 01.
Article in English | MEDLINE | ID: mdl-31915380

ABSTRACT

More than twelve morphologically and physiologically distinct subtypes of primary somatosensory neuron report salient features of our internal and external environments1-4. It is unclear how specialized gene expression programs emerge during development to endow these subtypes with their unique properties. To assess the developmental progression of transcriptional maturation of each subtype of principal somatosensory neuron, we generated a transcriptomic atlas of cells traversing the primary somatosensory neuron lineage in mice. Here we show that somatosensory neurogenesis gives rise to neurons in a transcriptionally unspecialized state, characterized by co-expression of transcription factors that become restricted to select subtypes as development proceeds. Single-cell transcriptomic analyses of sensory neurons from mutant mice lacking transcription factors suggest that these broad-to-restricted transcription factors coordinate subtype-specific gene expression programs in subtypes in which their expression is maintained. We also show that neuronal targets are involved in this process; disruption of the prototypic target-derived neurotrophic factor NGF leads to aberrant subtype-restricted patterns of transcription factor expression. Our findings support a model in which cues that emanate from intermediate and final target fields promote neuronal diversification in part by transitioning cells from a transcriptionally unspecialized state to transcriptionally distinct subtypes by modulating the selection of subtype-restricted transcription factors.


Subject(s)
Neurogenesis , Neurons/physiology , Animals , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nerve Growth Factor/metabolism , Neurons/cytology , RNA/analysis , RNA/genetics , Single-Cell Analysis , Transcription Factor Brn-3B/genetics , Transcription Factor Brn-3B/metabolism , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism
18.
Arch Biochem Biophys ; 680: 108224, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31830441

ABSTRACT

Pou4f3 plays an important role in the development of hair cells in the inner ear sensory epithelia. Autophagy is related to the auditory damage. However, the role and mechanism of Pou4f3 on drug-induced ototoxicity are incompletely understood. Hence, this study aimed to explore the effects of Pou4f3 on the apoptosis of cochlear hair cells (CHCs) and to explore whether autophagy was involved in this process. The cisplatin was used to produce a loss of CHCs to create a murine model of deafness. The AAV vectors were delivered into the scala media through the lateral wall. Compared with the control mice, the cisplatin-treated mice exhibited significantly enhanced apoptosis and autophagy in the cochleae, accompanied by a notably decreased Pou4f3 levels. Both mutation and knockdown of Pou4f3 promoted the apoptosis- and autophagy-related protein levels, and enhanced the cisplatin-induced levels of apoptosis- and autophagy-related proteins. Furthermore, the autophagy activator rapamycin promoted the apoptosis and autophagy in the cochlea. In addition, the autophagy inhibitor 3-MA overturned the promoting effect of Pou4f3 knockdown on the apoptosis and autophagy. Collectively, in cisplatin-induced deafness mice, the Pou4f3 gene mutation facilitated apoptosis of cochlear hair cells, at least partially, through inducing autophagy.


Subject(s)
Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Autophagy/drug effects , Cisplatin/adverse effects , Deafness/chemically induced , Homeodomain Proteins/genetics , Transcription Factor Brn-3C/genetics , Animals , Deafness/genetics , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation
19.
Hear Res ; 376: 58-68, 2019 05.
Article in English | MEDLINE | ID: mdl-30665849

ABSTRACT

Hereditary hearing loss is both clinically and genetically very heterogeneous. Despite the large number of genes that have been associated with the condition, many cases remain unexplained. Novel gene associations with hearing loss are to be expected but also are defects of regulatory regions of the genome which are currently not routinely addressed in molecular genetic testing and research. Inheritance patterns other than monogenic might be more common than assumed in isolated cases and diagnoses might have been missed because of misinterpretation of identified DNA variants. This review summarizes current insights in the genetics of hearing loss, the next steps that are being taken in research, and their challenges. Furthermore, genotype-phenotype correlations and modifying factors are discussed as these are instrumental in counselling hearing impaired individuals and/or their family members.


Subject(s)
Hearing Loss/genetics , Animals , Connexin 26 , Connexins/genetics , DNA Mutational Analysis , Extracellular Matrix Proteins/genetics , GPI-Linked Proteins/genetics , Genetic Association Studies , Genetic Variation , Homeodomain Proteins/genetics , Humans , Membrane Proteins/genetics , Mice , Molecular Biology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Transcription Factor Brn-3C/genetics , Exome Sequencing
20.
Dev Biol ; 446(2): 133-141, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30605626

ABSTRACT

Damage or loss of auditory hair cells leads to irreversible sensorineural hearing loss in human, thus regeneration of these cells to reconstruct auditory sensory epithelium holds the promise for the treatment of deafness. Regulatory factors involved in the development of auditory sensory epithelium play crucial roles in hair cell regeneration and hearing restoration. Here, we first focus on the transcription factor Atoh1 which is critical for hair cell development and regeneration, and comprehensively summarize the current understanding of the protein structure, target binding motif, developmental expression pattern, functional role, and upstream and downstream regulatory mechanism of Atoh1 in the context of controlling the cell fate commitment to hair cells or transdifferentiation from supporting cells. We also discuss cellular context dependency of Atoh1 in hair cell induction which should be taken into consideration when using Atoh1 gene therapy for hair cell regeneration. Next, we review the roles of Gfi1, Pou4f3, and Barhl1 in hair cell maturation and maintenance, and suggest that manipulation of these genes and their downstream targets will be helpful for the generation of functional hair cells with long-term viability. Finally, we provide an overview of the interplay between Notch, Wnt, Shh, and FGF signaling pathways during auditory sensory epithelium development. By analyzing crosstalk between these pathways, we suggest that combination of Wnt signaling activation with Hey1 and Hey2 inhibition will be crucial for hair cell regeneration and hearing restoration. Furthermore, this review highlights the importance of deeper understanding of the cellular context for hair cell development and the interconnection between these key regulators in developing new strategies to treat sensorineural hearing loss.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Ear, Inner/metabolism , Gene Expression Regulation, Developmental , Organ of Corti/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ear, Inner/embryology , Ear, Inner/growth & development , Hair Cells, Auditory/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ of Corti/embryology , Organ of Corti/growth & development , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...